Oncogene Induced Senescence (Homo sapiens)

From WikiPathways

Jump to: navigation, search
11, 12, 14, 22, 23, 28...5186616, 92618610315, 24, 40, 90, 93...12, 45, 57, 94, 951114928611, 1411, 1459, 10323, 44, 69, 75, 1115986nucleoplasmcytosolCDK4 TNRC6C EIF2C3 TFDP2 miR-24NonendonucleolyticRISCTNRC6B ERF:ETS2 Genep-T38-ETS1 ID1 CDKN2A gene UBB(153-228) UBC(533-608) CDKN2A gene p-T38-ETS1 UBC(229-304) p-T72-ETS2 miR-24-2 ADPCDK4,CDK6ETS2p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4E2F1,E2F2,E2F3:TFDP1,TFDP2E2F3 EIF2C1 ETS1 p-S166,S188-MDM2dimer,p-S166,S188-MDM2:MDM4TP53 CDK4 p16INK4A/p14ARFmRNA: miR-24NonendonucleolyticRISCETS1/ETS2TP53 p16INK4A p-T202,Y204-MAPK3 p-S166,S188-MDM2 p-T526-ERFTNRC6C Mitotic G1 phase andG1/S transitionmiR-24-2 CDKN2C TNRC6A INK4p16INK4ASP1 p16INK4A/p14ARF mRNAUbp14ARF mRNA UBC(305-380) p16INK4A E2F1 SP1TNRC6A p16INK4A mRNAp-T72-ETS2:CDKN2Agenep14ARF ETS2 GeneMDM4 ID1TFDP1 PolyUb-TP53 RB1ERF p-T72-ETS2Oxidative StressInduced SenescencemiR-24-1 Intrinsic Pathwayfor ApoptosisCDK4,CDK6:INK4DNA Damage/TelomereStress InducedSenescenceUBA52(1-76) p14ARFTP53 TetramerOncogenic MAPKsignalingp-T38-ETS1p-T38-ETS1:CDKN2AgeneEIF2C1 Cell CycleCheckpointsp14ARF CDKN2B ERFEIF2C4 MDM4 DP1/2: E2F1/2/3:SP1: CDKN2A GeneTFDP2 p16INK4A mRNA CDK6 p-S166,S188-MDM2dimer,p-S166,S188-MDM2,MDM4:TP53p-T38-ETS1/p-T72-ETS2:CDKN2AGenep-S166,S188-MDM2 CDKN2A gene TFDP1 CDKN2C EIF2C4 E2F1 MDM4 p14ARF mRNA TP53 EIF2C3 ATPp-S166,S188-MDM2 UBB(1-76) p14ARFE2F3 CDKN2D p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4:TP53p-T72-ETS2 UBC(1-76) p-T,Y MAPK dimersp-T38-ETS1 p-T526-ERFPolyUb-TP53 Tetramerp-T38-ETS1/p-T72-ETS2ATPp-T72-ETS2 CDKN2A gene ADPUBC(609-684) p-T72-ETS2:ID1MOV10 p-T185,Y187-MAPK1 p14ARF mRNACDKN2B E2F2 CDK6 miR-24-1 TNRC6B p-T72-ETS2 MDM4 UBC(381-456) MOV10 CDKN2A genep16INK4A mRNA p-S166,S188-MDM2 CDKN2D UBB(77-152) ETS2 Gene UBC(153-228) E2F2 UBC(77-152) ETS2 RPS27A(1-76) UBC(457-532) 1, 4, 8, 10, 17...36, 65, 7259, 1038613, 19, 33, 34, 42...86149215, 38, 47867, 25, 31, 1145, 9, 16, 35, 68...2, 3, 23, 32, 44...


Description

Oncogene-induced senescence (OIS) 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: p16INK4A and p14ARF. p16INK4A and p14ARF 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. p16INK4A 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 p16INK4A 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, p15INK4B, is located in the vicinity of CDKN2A locus, at the chromosome band 9p21. p15INK4B, together with p16INK4A, 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 p15INK4B gene expression (Vijayachandra et al. 2003). TGF-beta-induced p15INK4B 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 p16INK4A 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 p16INK4A transcription (Ohtani et al. 2004).

Transcription of p14ARF is stimulated by binding of E2F transcription factors (E2F1, E2F2 or E2F3) in complex with SP1 to p14ARF 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.
While OIS has been studied to considerable detail in cultured cells, establishment of in vivo role of OIS has been difficult due to lack of specific biomarkers and its interconnectedness with other senescence pathways (Baek and Ryeom 2017, reviewed in Sharpless and Sherr 2015). View original pathway at Reactome.</div>

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 2559585
Reactome-version 
Reactome version: 75
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. 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
  2. Depoortere F, Van Keymeulen A, Lukas J, Costagliola S, Bartkova J, Dumont JE, Bartek J, Roger PP, Dremier S.; ''A requirement for cyclin D3-cyclin-dependent kinase (cdk)-4 assembly in the cyclic adenosine monophosphate-dependent proliferation of thyrocytes.''; PubMed Europe PMC Scholia
  3. Chittenden T, Livingston DM, Kaelin WG.; ''The T/E1A-binding domain of the retinoblastoma product can interact selectively with a sequence-specific DNA-binding protein.''; PubMed Europe PMC Scholia
  4. 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
  5. Maertens O, Cichowski K.; ''An expanding role for RAS GTPase activating proteins (RAS GAPs) in cancer.''; PubMed Europe PMC Scholia
  6. Komori H, Enomoto M, Nakamura M, Iwanaga R, Ohtani K.; ''Distinct E2F-mediated transcriptional program regulates p14ARF gene expression.''; PubMed Europe PMC Scholia
  7. Chen J, Marechal V, Levine AJ.; ''Mapping of the p53 and mdm-2 interaction domains.''; PubMed Europe PMC Scholia
  8. 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
  9. Lavoie H, Therrien M.; ''Regulation of RAF protein kinases in ERK signalling.''; PubMed Europe PMC Scholia
  10. 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
  11. 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
  12. Seidel JJ, Graves BJ.; ''An ERK2 docking site in the Pointed domain distinguishes a subset of ETS transcription factors.''; PubMed Europe PMC Scholia
  13. Harley CB, Futcher AB, Greider CW.; ''Telomeres shorten during ageing of human fibroblasts.''; PubMed Europe PMC Scholia
  14. 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
  15. Cheng Q, Cross B, Li B, Chen L, Li Z, Chen J.; ''Regulation of MDM2 E3 ligase activity by phosphorylation after DNA damage.''; PubMed Europe PMC Scholia
  16. Stephen AG, Esposito D, Bagni RK, McCormick F.; ''Dragging ras back in the ring.''; PubMed Europe PMC Scholia
  17. 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
  18. 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
  19. 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
  20. 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
  21. 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
  22. Sharpless NE, Sherr CJ.; ''Forging a signature of in vivo senescence.''; PubMed Europe PMC Scholia
  23. 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
  24. Li M, Luo J, Brooks CL, Gu W.; ''Acetylation of p53 inhibits its ubiquitination by Mdm2.''; PubMed Europe PMC Scholia
  25. Oliner JD, Kinzler KW, Meltzer PS, George DL, Vogelstein B.; ''Amplification of a gene encoding a p53-associated protein in human sarcomas.''; PubMed Europe PMC Scholia
  26. 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
  27. White A, Pargellis CA, Studts JM, Werneburg BG, Farmer BT.; ''Molecular basis of MAPK-activated protein kinase 2:p38 assembly.''; PubMed Europe PMC Scholia
  28. 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
  29. 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
  30. Weinmann AS, Bartley SM, Zhang T, Zhang MQ, Farnham PJ.; ''Use of chromatin immunoprecipitation to clone novel E2F target promoters.''; PubMed Europe PMC Scholia
  31. Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW, Vogelstein B.; ''Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53.''; PubMed Europe PMC Scholia
  32. Lees JA, Saito M, Vidal M, Valentine M, Look T, Harlow E, Dyson N, Helin K.; ''The retinoblastoma protein binds to a family of E2F transcription factors.''; PubMed Europe PMC Scholia
  33. Lee JH, Paull TT.; ''ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex.''; PubMed Europe PMC Scholia
  34. 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
  35. Prior IA, Lewis PD, Mattos C.; ''A comprehensive survey of Ras mutations in cancer.''; PubMed Europe PMC Scholia
  36. Saelens X, Festjens N, Vande Walle L, van Gurp M, van Loo G, Vandenabeele P.; ''Toxic proteins released from mitochondria in cell death.''; PubMed Europe PMC Scholia
  37. 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
  38. Pant V, Xiong S, Iwakuma T, Quintás-Cardama A, Lozano G.; ''Heterodimerization of Mdm2 and Mdm4 is critical for regulating p53 activity during embryogenesis but dispensable for p53 and Mdm2 stability.''; PubMed Europe PMC Scholia
  39. 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
  40. Wu X, Bayle JH, Olson D, Levine AJ.; ''The p53-mdm-2 autoregulatory feedback loop.''; PubMed Europe PMC Scholia
  41. 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
  42. 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
  43. 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
  44. 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
  45. 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
  46. 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
  47. Huang L, Yan Z, Liao X, Li Y, Yang J, Wang ZG, Zuo Y, Kawai H, Shadfan M, Ganapathy S, Yuan ZM.; ''The p53 inhibitors MDM2/MDMX complex is required for control of p53 activity in vivo.''; PubMed Europe PMC Scholia
  48. 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
  49. 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
  50. 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
  51. Lindström MS, Klangby U, Inoue R, Pisa P, Wiman KG, Asker CE.; ''Immunolocalization of human p14(ARF) to the granular component of the interphase nucleolus.''; PubMed Europe PMC Scholia
  52. Sadasivam S, DeCaprio JA.; ''The DREAM complex: master coordinator of cell cycle-dependent gene expression.''; PubMed Europe PMC Scholia
  53. de Lange T.; ''Shelterin: the protein complex that shapes and safeguards human telomeres.''; PubMed Europe PMC Scholia
  54. 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
  55. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR.; ''The E2F transcription factor is a cellular target for the RB protein.''; PubMed Europe PMC Scholia
  56. 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
  57. Jayaraman G, Srinivas R, Duggan C, Ferreira E, Swaminathan S, Somasundaram K, Williams J, Hauser C, Kurkinen M, Dhar R, Weitzman S, Buttice G, Thimmapaya B.; ''p300/cAMP-responsive element-binding protein interactions with ets-1 and ets-2 in the transcriptional activation of the human stromelysin promoter.''; PubMed Europe PMC Scholia
  58. 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
  59. 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
  60. Baek KH, Ryeom S.; ''Detection of Oncogene-Induced Senescence In Vivo.''; PubMed Europe PMC Scholia
  61. 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
  62. 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
  63. 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
  64. Zhang H, Cohen SN.; ''Smurf2 up-regulation activates telomere-dependent senescence.''; PubMed Europe PMC Scholia
  65. Salvesen GS, Duckett CS.; ''IAP proteins: blocking the road to death's door.''; PubMed Europe PMC Scholia
  66. Cheng M, Sexl V, Sherr CJ, Roussel MF.; ''Assembly of cyclin D-dependent kinase and titration of p27Kip1 regulated by mitogen-activated protein kinase kinase (MEK1).''; PubMed Europe PMC Scholia
  67. Glover JN, Harrison SC.; ''Crystal structure of the heterodimeric bZIP transcription factor c-Fos-c-Jun bound to DNA.''; PubMed Europe PMC Scholia
  68. Samatar AA, Poulikakos PI.; ''Targeting RAS-ERK signalling in cancer: promises and challenges.''; PubMed Europe PMC Scholia
  69. Hannon GJ, Beach D.; ''p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest.''; PubMed Europe PMC Scholia
  70. Lito P, Rosen N, Solit DB.; ''Tumor adaptation and resistance to RAF inhibitors.''; PubMed Europe PMC Scholia
  71. 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
  72. Wang X.; ''The expanding role of mitochondria in apoptosis.''; PubMed Europe PMC Scholia
  73. Moiseeva O, Bourdeau V, Roux A, Deschênes-Simard X, Ferbeyre G.; ''Mitochondrial dysfunction contributes to oncogene-induced senescence.''; PubMed Europe PMC Scholia
  74. Cobrinik D.; ''Pocket proteins and cell cycle control.''; PubMed Europe PMC Scholia
  75. 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
  76. 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
  77. Vijayachandra K, Lee J, Glick AB.; ''Smad3 regulates senescence and malignant conversion in a mouse multistage skin carcinogenesis model.''; PubMed Europe PMC Scholia
  78. Zhang H.; ''Life without kinase: cyclin E promotes DNA replication licensing and beyond.''; PubMed Europe PMC Scholia
  79. 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
  80. Wu L, Timmers C, Maiti B, Saavedra HI, Sang L, Chong GT, Nuckolls F, Giangrande P, Wright FA, Field SJ, Greenberg ME, Orkin S, Nevins JR, Robinson ML, Leone G.; ''The E2F1-3 transcription factors are essential for cellular proliferation.''; PubMed Europe PMC Scholia
  81. 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
  82. 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
  83. 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
  84. 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
  85. Bagchi S, Weinmann R, Raychaudhuri P.; ''The retinoblastoma protein copurifies with E2F-I, an E1A-regulated inhibitor of the transcription factor E2F.''; PubMed Europe PMC Scholia
  86. 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
  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. 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
  89. 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
  90. Maki CG.; ''Oligomerization is required for p53 to be efficiently ubiquitinated by MDM2.''; PubMed Europe PMC Scholia
  91. 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
  92. 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
  93. Fuchs SY, Adler V, Buschmann T, Wu X, Ronai Z.; ''Mdm2 association with p53 targets its ubiquitination.''; PubMed Europe PMC Scholia
  94. 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
  95. 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
  96. Connell-Crowley L, Harper JW, Goodrich DW.; ''Cyclin D1/Cdk4 regulates retinoblastoma protein-mediated cell cycle arrest by site-specific phosphorylation.''; PubMed Europe PMC Scholia
  97. Linares LK, Hengstermann A, Ciechanover A, Müller S, Scheffner M.; ''HdmX stimulates Hdm2-mediated ubiquitination and degradation of p53.''; PubMed Europe PMC Scholia
  98. 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
  99. 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
  100. Ferreira R, Magnaghi-Jaulin L, Robin P, Harel-Bellan A, Trouche D.; ''The three members of the pocket proteins family share the ability to repress E2F activity through recruitment of a histone deacetylase.''; PubMed Europe PMC Scholia
  101. Hiebert SW.; ''Regions of the retinoblastoma gene product required for its interaction with the E2F transcription factor are necessary for E2 promoter repression and pRb-mediated growth suppression.''; PubMed Europe PMC Scholia
  102. 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
  103. 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
  104. 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
  105. New L, Jiang Y, Han J.; ''Regulation of PRAK subcellular location by p38 MAP kinases.''; PubMed Europe PMC Scholia
  106. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D.; ''RAS oncogenes: weaving a tumorigenic web.''; PubMed Europe PMC Scholia
  107. 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
  108. Vidal A, Koff A.; ''Cell-cycle inhibitors: three families united by a common cause.''; PubMed Europe PMC Scholia
  109. 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
  110. Yu TW, Anderson D.; ''Reactive oxygen species-induced DNA damage and its modification: a chemical investigation.''; PubMed Europe PMC Scholia
  111. 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
  112. 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
  113. 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
  114. Momand J, Zambetti GP, Olson DC, George D, Levine AJ.; ''The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114856view16:36, 25 January 2021ReactomeTeamReactome version 75
113302view11:37, 2 November 2020ReactomeTeamReactome version 74
112514view15:47, 9 October 2020ReactomeTeamReactome version 73
101426view11:30, 1 November 2018ReactomeTeamreactome version 66
100964view21:07, 31 October 2018ReactomeTeamreactome version 65
100501view19:42, 31 October 2018ReactomeTeamreactome version 64
100047view16:25, 31 October 2018ReactomeTeamreactome version 63
99599view14:59, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
93939view13:46, 16 August 2017ReactomeTeamreactome version 61
93528view11:26, 9 August 2017ReactomeTeamreactome version 61
88060view14:01, 25 July 2016RyanmillerOntology Term : 'regulatory pathway' added !
88059view14:01, 25 July 2016RyanmillerOntology Term : 'cellular senescence pathway' added !
86627view09:22, 11 July 2016ReactomeTeamreactome version 56
83246view10:29, 18 November 2015ReactomeTeamVersion54
81351view12:52, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
ADPMetaboliteCHEBI:456216 (ChEBI)
ATPMetaboliteCHEBI:30616 (ChEBI)
CDK4 ProteinP11802 (Uniprot-TrEMBL)
CDK4,CDK6:INK4ComplexR-HSA-182579 (Reactome)
CDK4,CDK6ComplexR-HSA-69209 (Reactome)
CDK6 ProteinQ00534 (Uniprot-TrEMBL)
CDKN2A gene ProteinENSG00000147889 (Ensembl)
CDKN2A geneGeneProductENSG00000147889 (Ensembl)
CDKN2B ProteinP42772 (Uniprot-TrEMBL)
CDKN2C ProteinP42773 (Uniprot-TrEMBL)
CDKN2D ProteinP55273 (Uniprot-TrEMBL)
Cell Cycle CheckpointsPathwayR-HSA-69620 (Reactome) A hallmark of the human cell cycle in normal somatic cells is its precision. This remarkable fidelity is achieved by a number of signal transduction pathways, known as checkpoints, which monitor cell cycle progression ensuring an interdependency of S-phase and mitosis, the integrity of the genome and the fidelity of chromosome segregation.

Checkpoints are layers of control that act to delay CDK activation when defects in the division program occur. As the CDKs functioning at different points in the cell cycle are regulated by different means, the various checkpoints differ in the biochemical mechanisms by which they elicit their effect. However, all checkpoints share a common hierarchy of a sensor, signal transducers, and effectors that interact with the CDKs.

The stability of the genome in somatic cells contrasts to the almost universal genomic instability of tumor cells. There are a number of documented genetic lesions in checkpoint genes, or in cell cycle genes themselves, which result either directly in cancer or in a predisposition to certain cancer types. Indeed, restraint over cell cycle progression and failure to monitor genome integrity are likely prerequisites for the molecular evolution required for the development of a tumor. Perhaps most notable amongst these is the p53 tumor suppressor gene, which is mutated in >50% of human tumors. Thus, the importance of the checkpoint pathways to human biology is clear.

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.

DP1/2: E2F1/2/3: SP1: CDKN2A GeneComplexR-HSA-3209097 (Reactome)
E2F1 ProteinQ01094 (Uniprot-TrEMBL)
E2F1,E2F2,E2F3:TFDP1,TFDP2ComplexR-HSA-1227905 (Reactome)
E2F2 ProteinQ14209 (Uniprot-TrEMBL)
E2F3 ProteinO00716 (Uniprot-TrEMBL)
EIF2C1 ProteinQ9UL18 (Uniprot-TrEMBL)
EIF2C3 ProteinQ9H9G7 (Uniprot-TrEMBL)
EIF2C4 ProteinQ9HCK5 (Uniprot-TrEMBL)
ERF ProteinP50548 (Uniprot-TrEMBL)
ERF:ETS2 GeneComplexR-HSA-3209182 (Reactome)
ERFProteinP50548 (Uniprot-TrEMBL)
ETS1 ProteinP14921 (Uniprot-TrEMBL)
ETS1/ETS2ComplexR-HSA-3132719 (Reactome)
ETS2 Gene ProteinENSG00000157557 (Ensembl)
ETS2 GeneGeneProductENSG00000157557 (Ensembl)
ETS2 ProteinP15036 (Uniprot-TrEMBL)
ETS2ProteinP15036 (Uniprot-TrEMBL)
ID1 ProteinP41134 (Uniprot-TrEMBL)
ID1ProteinP41134 (Uniprot-TrEMBL)
INK4ComplexR-HSA-182588 (Reactome)
Intrinsic Pathway for ApoptosisPathwayR-HSA-109606 (Reactome) The intrinsic (Bcl-2 inhibitable or mitochondrial) pathway of apoptosis functions in response to various types of intracellular stress including growth factor withdrawal, DNA damage, unfolding stresses in the endoplasmic reticulum and death receptor stimulation. Following the reception of stress signals, proapoptotic BCL-2 family proteins are activated and subsequently interact with and inactivate antiapoptotic BCL-2 proteins. This interaction leads to the destabilization of the mitochondrial membrane and release of apoptotic factors. These factors induce the caspase proteolytic cascade, chromatin condensation, and DNA fragmentation, ultimately leading to cell death. The key players in the Intrinsic pathway are the Bcl-2 family of proteins that are critical death regulators residing immediately upstream of mitochondria. The Bcl-2 family consists of both anti- and proapoptotic members that possess conserved alpha-helices with sequence conservation clustered in BCL-2 Homology (BH) domains. Proapoptotic members are organized as follows:

1. "Multidomain" BAX family proteins such as BAX, BAK etc. that display sequence conservation in their BH1-3 regions. These proteins act downstream in mitochondrial disruption.

2. "BH3-only" proteins such as BID,BAD, NOXA, PUMA,BIM, and BMF have only the short BH3 motif. These act upstream in the pathway, detecting developmental death cues or intracellular damage. Anti-apoptotic members like Bcl-2, Bcl-XL and their relatives exhibit homology in all segments BH1-4. One of the critical functions of BCL-2/BCL-XL proteins is to maintain the integrity of the mitochondrial outer membrane.

MDM4 ProteinO15151 (Uniprot-TrEMBL)
MOV10 ProteinQ9HCE1 (Uniprot-TrEMBL)
Mitotic G1 phase and G1/S transitionPathwayR-HSA-453279 (Reactome) Mitotic G1-G1/S phase involves G1 phase of the mitotic interphase and G1/S transition, when a cell commits to DNA replication and divison genetic and cellular material to two daughter cells.

During early G1, cells can enter a quiescent G0 state. In quiescent cells, the evolutionarily conserved DREAM complex, consisting of the pocket protein family member p130 (RBL2), bound to E2F4 or E2F5, and the MuvB complex, represses transcription of cell cycle genes (reviewed by Sadasivam and DeCaprio 2013).

During early G1 phase in actively cycling cells, transcription of cell cycle genes is repressed by another pocket protein family member, p107 (RBL1), which forms a complex with E2F4 (Ferreira et al. 1998, Cobrinik 2005). RB1 tumor suppressor, the product of the retinoblastoma susceptibility gene, is the third member of the pocket protein family. RB1 binds to E2F transcription factors E2F1, E2F2 and E2F3 and inhibits their transcriptional activity, resulting in prevention of G1/S transition (Chellappan et al. 1991, Bagchi et al. 1991, Chittenden et al. 1991, Lees et al. 1993, Hiebert 1993, Wu et al. 2001). Once RB1 is phosphorylated on serine residue S795 by Cyclin D:CDK4/6 complexes, it can no longer associate with and inhibit E2F1-3. Thus, CDK4/6-mediated phosphorylation of RB1 leads to transcriptional activation of E2F1-3 target genes needed for the S phase of the cell cycle (Connell-Crowley et al. 1997). CDK2, in complex with cyclin E, contributes to RB1 inactivation and also activates proteins needed for the initiation of DNA replication (Zhang 2007). Expression of D type cyclins is regulated by extracellular mitogens (Cheng et al. 1998, Depoortere et al. 1998). Catalytic activities of CDK4/6 and CDK2 are controlled by CDK inhibitors of the INK4 family (Serrano et al. 1993, Hannon and Beach 1994, Guan et al. 1994, Guan et al. 1996, Parry et al. 1995) and the Cip/Kip family, respectively.

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 p16INK4A and p14ARF from the CDKN2A locus, where PCR1.4 mediated repression of p14ARF 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 p14ARF 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 p16INK4A (Agger et al. 2009, Barradas et al. 2009, Lin et al. 2012).


p16INK4A inhibits phosphorylation-mediated inactivation of RB family members by CDK4 and CDK6, leading to cell cycle arrest (Serrano et al. 1993). p14ARF 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).

PolyUb-TP53 ProteinP04637 (Uniprot-TrEMBL)
PolyUb-TP53 TetramerComplexR-HSA-3209186 (Reactome)
RB1ProteinP06400 (Uniprot-TrEMBL)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
SP1 ProteinP08047 (Uniprot-TrEMBL)
SP1ProteinP08047 (Uniprot-TrEMBL)
TFDP1 ProteinQ14186 (Uniprot-TrEMBL)
TFDP2 ProteinQ14188 (Uniprot-TrEMBL)
TNRC6A ProteinQ8NDV7 (Uniprot-TrEMBL)
TNRC6B ProteinQ9UPQ9 (Uniprot-TrEMBL)
TNRC6C ProteinQ9HCJ0 (Uniprot-TrEMBL)
TP53 ProteinP04637 (Uniprot-TrEMBL)
TP53 TetramerComplexR-HSA-3209194 (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)
UbComplexR-HSA-68524 (Reactome)
miR-24

Nonendonucleolytic

RISC
ComplexR-HSA-3209134 (Reactome) The RNA-induced silencing complex contains an Argonaute (AGO) protein, whose PAZ domain binds the 3' end of the miRNA. The PIWI domain of AGO is responsible for cleavage of target RNAs, that is, RNAs complementary to the miRNA. Only AGO2 (EIF2C2) is capable of cleavage, however. AGO1 (EIF2C1), AGO3 (EIF2C3), and AGO4 (EIF2C4) repress translation of target RNAs by binding without cleavage. In vivo, cleavage by AGO2 and repression of translation by all AGOs require interaction with a TNRC6 protein (GW182 protein) and MOV10. The interaction with TNRC6 proteins is also responsible for localizing the AGO complex to Processing Bodies (P-bodies). Tethering of the C-terminal domain of a TNRC6 protein to a mRNA is sufficient to cause repression of translation.
miR-24-1 ProteinMI0000080 (miRBase mature sequence)
miR-24-2 ProteinMI0000081 (miRBase mature sequence)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2,MDM4:TP53
ComplexR-HSA-6804885 (Reactome)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2:MDM4
ComplexR-HSA-6804745 (Reactome)
p-S166,S188-MDM2 ProteinQ00987 (Uniprot-TrEMBL)
p-T,Y MAPK dimersComplexR-HSA-198701 (Reactome)
p-T185,Y187-MAPK1 ProteinP28482 (Uniprot-TrEMBL)
p-T202,Y204-MAPK3 ProteinP27361 (Uniprot-TrEMBL)
p-T38-ETS1 ProteinP14921 (Uniprot-TrEMBL)
p-T38-ETS1/

p-T72-ETS2:CDKN2A

Gene
ComplexR-HSA-3200029 (Reactome)
p-T38-ETS1/ p-T72-ETS2ComplexR-HSA-3132724 (Reactome)
p-T38-ETS1:CDKN2A geneComplexR-HSA-8979083 (Reactome)
p-T38-ETS1ProteinP14921 (Uniprot-TrEMBL)
p-T526-ERFProteinP50548 (Uniprot-TrEMBL)
p-T72-ETS2 ProteinP15036 (Uniprot-TrEMBL)
p-T72-ETS2:CDKN2A geneComplexR-HSA-8979081 (Reactome)
p-T72-ETS2:ID1ComplexR-HSA-3209172 (Reactome)
p-T72-ETS2ProteinP15036 (Uniprot-TrEMBL)
p14ARF ProteinQ8N726 (Uniprot-TrEMBL)
p14ARF mRNA ProteinENST00000579755 (Ensembl)
p14ARF mRNARnaENST00000579755 (Ensembl)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4:TP53ComplexR-HSA-6804999 (Reactome)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4ComplexR-HSA-6804995 (Reactome)
p14ARFProteinQ8N726 (Uniprot-TrEMBL)
p16INK4A ProteinP42771 (Uniprot-TrEMBL)
p16INK4A mRNA ProteinENST00000304494 (Ensembl)
p16INK4A mRNARnaENST00000304494 (Ensembl)
p16INK4A/p14ARF

mRNA: miR-24 Nonendonucleolytic

RISC
ComplexR-HSA-3209131 (Reactome)
p16INK4A/p14ARF mRNAComplexR-HSA-3209130 (Reactome)
p16INK4AProteinP42771 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
ADPArrowR-HSA-3132737 (Reactome)
ADPArrowR-HSA-3209160 (Reactome)
ATPR-HSA-3132737 (Reactome)
ATPR-HSA-3209160 (Reactome)
CDK4,CDK6:INK4ArrowR-HSA-182594 (Reactome)
CDK4,CDK6R-HSA-182594 (Reactome)
CDKN2A geneR-HSA-3200023 (Reactome)
CDKN2A geneR-HSA-3209096 (Reactome)
CDKN2A geneR-HSA-3209098 (Reactome)
CDKN2A geneR-HSA-3209109 (Reactome)
CDKN2A geneR-HSA-8979082 (Reactome)
DP1/2: E2F1/2/3: SP1: CDKN2A GeneArrowR-HSA-3209096 (Reactome)
DP1/2: E2F1/2/3: SP1: CDKN2A GeneArrowR-HSA-3209109 (Reactome)
E2F1,E2F2,E2F3:TFDP1,TFDP2R-HSA-3209096 (Reactome)
ERF:ETS2 GeneArrowR-HSA-3209177 (Reactome)
ERF:ETS2 GeneTBarR-HSA-3209179 (Reactome)
ERFR-HSA-3209160 (Reactome)
ERFR-HSA-3209177 (Reactome)
ETS1/ETS2R-HSA-3132737 (Reactome)
ETS2 GeneR-HSA-3209177 (Reactome)
ETS2 GeneR-HSA-3209179 (Reactome)
ETS2ArrowR-HSA-3209179 (Reactome)
ID1R-HSA-3209165 (Reactome)
INK4R-HSA-182594 (Reactome)
PolyUb-TP53 TetramerArrowR-HSA-6804879 (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 and their inhibitory phosphorylation of the RB family (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 p16INK4A (Parry et al. 1995).
R-HSA-3132737 (Reactome) Both ETS1 and ETS2 contain a consensus site (PLLTP) for MAPK3 and MAPK1 (ERK1 and ERK2, respectively) in the vicinity of the pointed domain, while the pointed domain contains a docking site needed for ERK1/2 binding to ETS1/2. ETS1 and ETS2 are able to collaborate with RAS in superactivating the promoters that contain RREs (RAS response elements) that include ETS-binding sites. The cooperation of ETS1 and ETS2 with RAS activation is dependent on the phosphorylation of PLLTP threonine residue (T38 in ETS1; T72 in ETS2) (Yang et al. 1996, Seidel et al. 2002). Phosphorylation of ETS1 and ETS2 by ERK1/2 induces a conformational change that increases their affinity for the TAZ domain of the transcriptional coactivator CREBBP (CBP) and the transcriptional activation of RREs (Foulds et al. 2004, Nelson et al. 2010), although ETS1/ETS2 may interact with CREBBP in the absence of phosphorylation (Jayaraman et al. 1999). Phosphorylation of serine residue S41 of ETS1 (corresponds to serine residue S75 of ETS2) may be necessary for full activation of ETS1/2 (Nelson et al. 2010).
R-HSA-3200023 (Reactome) ETS1 and ETS2, activated by RAS/RAF/MAP kinase cascade, bind the promoter of p16INK4A in the CDKN2A locus (Ohtani et al. 2001). CDKN2A locus also encodes p14ARF (p19ARF in mouse), but from a different promoter and in a different reading frame. While p16INK4A and p14ARF use different exon 1, (exon 1-alpha and exon 1-beta, respectively), they share exons 2 and 3. However, because the reading frames are different, there is no amino acid sequence similarity between the two proteins (Quelle et al. 1995).
R-HSA-3209096 (Reactome) E2F1, E2F2 or E2F3 forms a complex with the transcription factor SP1 on p14-ARF promoter (Parisi et al. 2002).
R-HSA-3209098 (Reactome) Phosphorylated ETS1 and ETS2 stimulate p16INK4A transcription, resulting in cell cycle arrest with arrested cells exhibiting high p16INK4A level and senescence-associated beta-galactosidase activity. It is possible that ETS2 is the main transmitter of RAS signaling to p16INK4A at the initiation of the senescence program, and that ETS1 maintains high p16INK4A level once the senescence is already established (Ohtani et al. 2001).
R-HSA-3209109 (Reactome) E2F1, E2F2 or E2F3 in complex with SP1 stimulates p14ARF transcription (Parisi et al. 2002). Therefore, increased activity of E2F1, E2F2 or E2F3, which may result from the loss of function of the tumor suppressor protein RB1, an inhibitor of E2F1/2/3, leads to an increased level of p14ARF (Komori et al. 2005).
R-HSA-3209111 (Reactome) MicroRNA miR-24 inhibits translation of p14ARF mRNA without causing mRNA degradation. This results in high p14ARF transcript level accompanied by low p14ARF protein level (To et al. 2012).
R-HSA-3209114 (Reactome) MicroRNA miR-24 inhibits translation of p16INK4A mRNA without causing mRNA degradation. This results in high p16INK4A transcript level accompanied by low p16INK4A protein level (Lal et al. 2008). p16INK4A 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).
R-HSA-3209151 (Reactome) MicroRNA miR-24 is able to bind both p16INK4A mRNA (Lal et al. 2008) and p14ARF mRNA (To et al. 2012) through their shared 3'UTR. miR-24 inhibits translation of p16INK4A and p14ARF mRNAs, but does not induce mRNA degradation, resulting in expression of high levels of p16INK4A and p14ARF transcripts, while protein levels of p16INK4A and p14ARF are low (Lal et al. 2008, To et al. 2012).
R-HSA-3209159 (Reactome) Phosphorylation of ERF at threonine T526 by activated ERKs triggers ERF export from the nucleus to the cytosol (Le Gallic et al. 2004), which is expected to relieve ERF-mediated inhibition of ETS2 transcription.
R-HSA-3209160 (Reactome) Activated ERKs (ERK1 i.e. MAPK3 and ERK2 i.e. MAPK1) phosphorylate ERF on threonine residue T526 and possibly other sites. The threonine T526 seems to be the dominant phosphorylation site and its functional relevance has been established (Sgouras et al. 1995, Le Gallic et al. 2004).
R-HSA-3209165 (Reactome) Binding of ID1 to ETS2 inhibits ETS2-mediated activation of p16INK4A transcription (Ohtani et al. 2001).
R-HSA-3209177 (Reactome) ERF binds to an ETS-binding site in the ETS2 promoter (Sgouras et al. 1995). Phosphorylation of ERF by activated MAPK1 (ERK2) or MAPK3 (ERK1) interferes with ERF-mediated upregulation of ETS2 (Le Gallic et al. 2004).
R-HSA-3209179 (Reactome) Binding of ERF to ETS2 promoter strongly represses ETS2 transcription (Sgouras et al. 1995).
R-HSA-6804879 (Reactome) MDM2 is an ubiquitin ligase whose expression is positively regulated by TP53 (p53) (Wu et al. 1993). MDM2 binds TP53 tetramer (Maki 1999) and promotes its ubiquitination and subsequent degradation (Fuchs et al. 1998). Formation of MDM2 homodimers (Cheng et al. 2011) or heterodimers with MDM4 (MDMX) is needed for efficient ubiquitination of TP53 (Linares et al. 2003). While MDM2-TP53 binding occurs at the amino-terminus of TP53, MDM2 ubiquitinates TP53 lysine residues at the carboxy-terminus. Acetylation of those lysines can inhibit MDM2-dependent ubiquitination (Li et al. 2002).
R-HSA-6804996 (Reactome) Binding of p14ARF to MDM2 decreases the half-life of MDM2, likely through promoting MDM2 degradation. Thus, p14ARF inhibits MDM2-mediated ubiquitination and degradation of TP53 (Zhang et al. 1998).
R-HSA-6804998 (Reactome) p14ARF forms a complex with TP53-bound MDM2 by interacting with the C-terminus of MDM2, while the N-terminus of MDM2 is involved in TP53 (p53) binding. p14ARF cannot associate with TP53 in the absence of MDM2 (Zhang et al. 1998).
R-HSA-8979082 (Reactome) ETS2, activated by RAS/RAF/MAP kinase cascade, binds the promoter of p16INK4A in the CDKN2A locus (Ohtani et al. 2001). CDKN2A locus also encodes p14ARF (p19ARF in mouse), but from a different promoter and in a different reading frame. While p16INK4A and p14ARF use different exon 1, (exon 1-alpha and exon 1-beta, respectively), they share exons 2 and 3. However, because the reading frames are different, there is no amino acid sequence similarity between the two proteins (Quelle et al. 1995).
R-HSA-9645672 (Reactome) p14ARF is mainly localized inside the nucleus, specifically the nucleolus (Zhang and Xiong 1999, Lindstrom et al. 2000), similar to its mouse orthologue p19ARF (Tao and Levine 1999).
RB1TBarR-HSA-3209109 (Reactome)
SP1R-HSA-3209096 (Reactome)
TP53 TetramerArrowR-HSA-6804996 (Reactome)
UbR-HSA-6804879 (Reactome)
miR-24

Nonendonucleolytic

RISC
R-HSA-3209151 (Reactome)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2,MDM4:TP53
R-HSA-6804879 (Reactome)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2,MDM4:TP53
R-HSA-6804998 (Reactome)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2,MDM4:TP53
mim-catalysisR-HSA-6804879 (Reactome)
p-S166,S188-MDM2

dimer,

p-S166,S188-MDM2:MDM4
ArrowR-HSA-6804879 (Reactome)
p-T,Y MAPK dimersTBarR-HSA-3209177 (Reactome)
p-T,Y MAPK dimersmim-catalysisR-HSA-3132737 (Reactome)
p-T,Y MAPK dimersmim-catalysisR-HSA-3209160 (Reactome)
p-T38-ETS1/

p-T72-ETS2:CDKN2A

Gene
ArrowR-HSA-3209098 (Reactome)
p-T38-ETS1/ p-T72-ETS2ArrowR-HSA-3132737 (Reactome)
p-T38-ETS1:CDKN2A geneArrowR-HSA-3200023 (Reactome)
p-T38-ETS1R-HSA-3200023 (Reactome)
p-T526-ERFArrowR-HSA-3209159 (Reactome)
p-T526-ERFArrowR-HSA-3209160 (Reactome)
p-T526-ERFR-HSA-3209159 (Reactome)
p-T72-ETS2:CDKN2A geneArrowR-HSA-8979082 (Reactome)
p-T72-ETS2:ID1ArrowR-HSA-3209165 (Reactome)
p-T72-ETS2:ID1TBarR-HSA-8979082 (Reactome)
p-T72-ETS2R-HSA-3209165 (Reactome)
p-T72-ETS2R-HSA-8979082 (Reactome)
p14ARF mRNAArrowR-HSA-3209109 (Reactome)
p14ARF mRNAR-HSA-3209111 (Reactome)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4:TP53ArrowR-HSA-6804998 (Reactome)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4:TP53R-HSA-6804996 (Reactome)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4ArrowR-HSA-6804996 (Reactome)
p14ARF:p-S166,S188-MDM2 dimer,p-S166,S188-MDM2:MDM4TBarR-HSA-6804879 (Reactome)
p14ARFArrowR-HSA-3209111 (Reactome)
p14ARFArrowR-HSA-9645672 (Reactome)
p14ARFR-HSA-6804998 (Reactome)
p14ARFR-HSA-9645672 (Reactome)
p16INK4A mRNAArrowR-HSA-3209098 (Reactome)
p16INK4A mRNAR-HSA-3209114 (Reactome)
p16INK4A/p14ARF

mRNA: miR-24 Nonendonucleolytic

RISC
ArrowR-HSA-3209151 (Reactome)
p16INK4A/p14ARF

mRNA: miR-24 Nonendonucleolytic

RISC
TBarR-HSA-3209111 (Reactome)
p16INK4A/p14ARF

mRNA: miR-24 Nonendonucleolytic

RISC
TBarR-HSA-3209114 (Reactome)
p16INK4A/p14ARF mRNAR-HSA-3209151 (Reactome)
p16INK4AArrowR-HSA-3209114 (Reactome)

Personal tools