Transcriptional regulation by RUNX1 (Homo sapiens)

From WikiPathways

Jump to: navigation, search
23, 29, 32, 46, 51...13211, 18243, 12910182642, 3, 43, 53, 129...125116167192, 19816174, 1231, 11, 1821619318411427, 131991, 11106, 132118435101611411416170171101741711987435101717074, 1231671711711141141849364, 741127, 1311043, 1291847011427, 131123plasma membranenucleoplasmcytoplasmcell junctionlysosomal lumenmitochondrial outer membraneTCF12 PSMD10 GATA3 HIST1H2AB H2AFJ FOXP3 HIST2H2AC PHC1 RUNX1:CBFB:KMT2APSMB2 ESR1 H2AFZ PSMA5 HIST1H2AC LMO2 RUNX2-P2 HIST1H2AJ RUNX1 PSMB6 PSMC5 Me3K5-H3F3A Regulation of RUNX1Expression andActivityESR1 RUNX1 ELF1 PSME1 SMARCC1 H2BFS p-S,T-EP300 SMARCD1 RUNX1:CBFB:SWI/SNFPSMD5 UBC(457-532) ESTG PSMD13 CBFB CBFB CBX8 RUNX1 UBC(153-228) HIST2H2AA3 CBFB CBFB RSPO3 gene CBX2 HIST1H2BJ CTSL1(114-288) GATA3 BMI1 ARID1B PSMC6 RUNX1:CBFB:SERPINB13genePSMB3 SMARCE1 SOCS3 GeneDNA Double StrandBreak ResponseAXIN1PolyUb-TP73 tetramerRUNX1 RUNX1 PSMD6 ACTL6A RING1 HIST1H4 p-Y407-YAP1:TP73tetramerp-S249,S273,T276-RUNX1:CBFBHIST1H2BC HIST1H2BL HIST2H3A ELF2 GATA3 UBC(609-684) OCLN geneHIST2H2AA3 RUNX1 ACTL6A GATA1 HIST1H2AJ AdoMetCBFB RUNX1:CBFB:(PRC1.4,PRC1.5)RYBP TAL1 core complexTP73 KCTD6 gene ITCHHIST1H2BD PSMA4 OCLNLDB1 CSNK2A1 UBC(305-380) LMO1 RUNX1 CBFB TCF12 HIST1H2AB PSMB9 SMARCA2 HIST1H2AC CBFB H2BFS PAX5 RUNX1 SERPINB13 RUNX1 YAF2 SERPINB13:cathepsinLIL3 gene p-S456-ABL1RUNX1:CBFB:LIFR geneCBFB SOCS4 gene ARID2 H2BFS HIST3H2BB CSNK2A2 SERPINB13 RUNX1:CBFB:LGALS3geneRUNX1:CBFB:ELF2SPI1 Gene PSMA2 HIST2H3A RUNX1 HIST2H2AC ELF1 HIST1H2BM SERPINB13 genePolyUb-TP73 LMO1 HIST1H2BN PSMD8 ACTL6B CBFB RUNX1:CBFB:EP300RUNX1 H2AFX HIST1H2AD YAP1 PSME4 CBFB PSMF1 RSPO3 geneGATA1 RUNX1 PAX5 H2AFX ESR1 AXIN1 gene PHC2 CBFB LIFRPSMB8 CBFB RUNX1 HIST2H2AA3 H2AFJ AUTS2 HIST1H3A PSMA1 HIST1H2BD SPI1 Gene (PRC1.4,PRC1.5)HIST1H2BH RUNX1:CBFB:PAX5HIST1H2BL ELF2PSMD11 KCTD6 genePSMA6 H2AFZ UBB(1-76) KMT2A ADPARID2 PSMD7 TP73 GATA2 GPAM geneELF1CSF2ESR1:estrogen:AXIN1genePSMB1 BLK geneCREBBP YAP1SHFM1 PSMD2 CBFB CBFB CBFB PSMB5 RUNX1:CBFB:KMT2A:SPI1 gene:H3K4me3-NucleosomeUBC(229-304) CBFB LGALS3 gene CTSK:SERPINB13SMARCD3 ESTG LGALS3GATA2 SERPINB13RUNX1 KMT2A HIST1H3A RUNX1 regulatesgenes involved inmegakaryocytedifferentiation andplatelet functionPSMC1 TJP1 geneCBFB CBX4 YAF2 RUNX1:CBFB:ELF,ELF2,PAX5:BLK genePSMD1 CSNK2B CTSKPHC1 CSNK2A2 HIST1H2BA RUNX1 RNF2 TCF12 H3F3A CTSK EP300 CBFB PSMC2 CLDN5 gene PHC3 LMO2 SMARCD1 CBX6 PSMB10 SMARCB1 CBFB HIST2H2AC CBFB HIST1H2BM SMARCA2 CBFB UBC(1-76) UbESTG RUNX2-P1 p-S249,S273,T276-RUNX1:CBFB:p-S,T-EP300MYB gene ESTG CSF2 geneCathepsin LLDB1 LGALS3 genePSMD9 PAX5Me3K5-HIST2H3A PHC3 ATPRING1 CBFB ITCH geneAXIN1 gene HIST1H2BN CBFB ESR1 RUNX1 ELF1 PSMA8 H2AFB1 SPI1 Gene Transcriptionalregulation by RUNX2PHC2 PSMA7 LMO2 PCGF5 SMARCD3 ATPCBX6 p-S249,S273,T276-RUNX1 CBFB TCF3 CBFB AUTS2 H3F3A HIST1H2BO RUNX1 HIST1H2BO PRKCB geneCBFB CDK7 RUNX1 CBFB KCTD6HIST1H2BK CREBBP HIPK2PSMD4 RUNX1:CBFB:ELF1,RUNX1:CBFB:ELF2,RUNX1:CBFB:PAX5HIST1H2BL RUNX1:CBFB:ESR1:estrogen:KCTD6 geneRUNX1:CBFB:FOXP3:RSPO3 genePSMD12 SMARCC1 PSMB7 PSMD14 ARID1A AXIN1 geneCSNK2B ESR1 RUNX1:CBFB:SOCS4geneELF2 UBB(77-152) TAL1 CBFB HIST1H2AC HIST1H4 TJP1ESR1 RUNX1:CBFB:FOXP3CREBBPLMO1 PAX5 RUNX1:CBFB:OCLN geneRUNX1 SWI/SNF chromatinremodelling complexUBC(533-608) RUNX1:CBFB:YAP1GATA3 RUNX1 TAL1 HIST1H2BB TP73 H2AFV ADPPSMC4 SOCS4H2AFX MYB geneSERPINB13 gene p-Y407-YAP1 CLDN5RUNX1 H2AFB1 Degradation ofbeta-catenin by thedestruction complexUBC(381-456) RNF2 IL3 geneELF2 EP300SPI1 gene:NucleosomeSMARCA4 RUNX1:CBFB:ESR1:estrogenLGALS3 gene ESTG RUNX1 BLK gene RYBP SCMH1-2 CBFB H2AFB1 TP73 TetramerSMARCC2 SMARCB1 RUNX1 CBFB CBFB ESTG TCF3 SMARCE1 H2AFJ HIST1H2AB HIST1H2AD CTSL2 RUNX1:CBFB:KMT2A:SPI1 gene:NucleosomeHIST1H4 RUNX1 RUNX1:CBFB:CLDN5geneHIST1H2BH ELF1 CBFB IL3YAP1 TJP1 gene CBX8 CBX2 SMARCC2 PSME3 HIST2H2BE H2AFV PSMA3 HIST1H2BA SMARCD2 KMT2ACSNK2A1 RUNX1:CBFB:ESR1:estrogen:GPAM geneSPI1SMARCA4 p-S249,S273,T276-RUNX1 ITCH gene LIFR geneHIST1H2AJ PCGF5 HIST1H2BK ARID1A TAL1 26S proteasomeRUNX1 RPS27A(1-76) RUNX1:CBFB:ELF1CTSL1(114-288) RUNX1 RUNX1 UBA52(1-76) RUNX1:CBFB:TAL1 corecomplexPSMB4 GPAM(1-828)HIST1H2BA HIST1H2BH HIST3H2BB CBFB PRKCB gene RUNX1 RUNX1 and FOXP3control thedevelopment ofregulatory Tlymphocytes (Tregs)CSF2 gene MYBPSMB11 RUNX2:CBFB:LGALS3geneTCF3 LMO2 HIST1H2AD HIST1H2BC UBC(77-152) OCLN gene MNAT1 HIST1H2BB LIFR gene RUNX1 RUNX1 SOCS3SOCS4 geneRUNX1:CBFB:SOCS3geneHIST1H2BN Signaling by NOTCH1KMT2A AdoHcyBLKHIST1H2BO HIST3H2BB Me3K5-HIST1H3A CTSL2 CBFB SMARCD2 p-Y407-YAP1SCMH1-2 CAKCBFB MYB PRKCBUBB(153-228) ACTL6B HIST1H2BK CBFB BMI1 ARID1B RUNX1:CBFBRUNX1:CBFB:CREBBP:CSF2 geneMYB RUNX1:CBFB:CREBBPPSME2 HIST1H2BJ RUNX1 HIST1H2BM HIST2H2BE HIST1H2BC GPAM gene TCF3 PBRM1 RUNX1:CBFB:GATA3-TAL1 core complex:MYB genePBRM1 RUNX1:CBFB:TJP1 genePSMC3 H2AFV RUNX1 RUNX1:CBFB:GATA3-TAL1 core complex,MYB:RUNX1:CBFB:GATA3-TAL1 core complexCCNH LDB1 TAL1 HIST1H2BJ H2AFZ LDB1 LMO1 SOCS3 Gene CBX4 RUNX1:CBFB:ESR1:estrogen:AXIN1 geneRUNX1:CBFB:ELF1:IL3geneRUNX1 RUNX1 RUNX1 CBFB RSPO3CBFB TCF12 TP73 TetramerRUNX1:CBFB:YAP1:ITCHgeneHIST1H2BB FOXP3 RUNX1:CBFB:PRKCBgeneHIST1H2BD ESR1:ESTGCLDN5 geneHIST2H2BE PSMD3 341, 11, 182117161321144, 15, 21, 36, 42...6, 8, 17, 22, 24...10117143, 1293574171101711671, 117, 9, 12-14, 20...12027, 13119813217711427, 1311841985, 18, 19, 28, 52...701, 11, 182137177114182161701043, 129119374, 123184459934, 14440, 63, 75, 113, 137...16111074, 12327, 1311502, 173


Description

The RUNX1 (AML1) transcription factor is a master regulator of hematopoiesis (Ichikawa et al. 2004) that is frequently translocated in acute myeloid leukemia (AML), resulting in formation of fusion proteins with altered transactivation profiles (Lam and Zhang 2012, Ichikawa et al. 2013). In addition to RUNX1, its heterodimerization partner CBFB is also frequently mutated in AML (Shigesada et al. 2004, Mangan and Speck 2011).
The core domain of CBFB binds to the Runt domain of RUNX1, resulting in formation of the RUNX1:CBFB heterodimer. CBFB does not interact with DNA directly. The Runt domain of RUNX1 mediated both DNA binding and heterodimerization with CBFB (Tahirov et al. 2001), while RUNX1 regions that flank the Runt domain are involved in transactivation (reviewed in Zhang et al. 2003) and negative regulation (autoinhibition). CBFB facilitates RUNX1 binding to DNA by stabilizing Runt domain regions that interact with the major and minor grooves of the DNA (Tahirov et al. 2001, Backstrom et al. 2002, Bartfeld et al. 2002). The transactivation domain of RUNX1 is located C-terminally to the Runt domain and is followed by the negative regulatory domain. Autoinhibiton of RUNX1 is relieved by interaction with CBFB (Kanno et al. 1998).
Transcriptional targets of the RUNX1:CBFB complex involve genes that regulate self-renewal of hematopoietic stem cells (HSCs) (Zhao et al. 2014), as well as commitment and differentiation of many hematopoietic progenitors, including myeloid (Friedman 2009) and megakaryocytic progenitors (Goldfarb 2009), regulatory T lymphocytes (Wong et al. 2011) and B lymphocytes (Boller and Grosschedl 2014).
RUNX1 binds to promoters of many genes involved in ribosomal biogenesis (Ribi) and is thought to stimulate their transcription. RUNX1 loss-of-function decreases ribosome biogenesis and translation in hematopoietic stem and progenitor cells (HSPCs). RUNX1 loss-of-function is therefore associated with a slow growth, but at the same time it results in reduced apoptosis and increases resistance of cells to genotoxic and endoplasmic reticulum stress, conferring an overall selective advantage to RUNX1 deficient HSPCs (Cai et al. 2015).
RUNX1 is implicated as a tumor suppressor in breast cancer. RUNX1 forms a complex with the activated estrogen receptor alpha (ESR1) and regulates expression of estrogen-responsive genes (Chimge and Frenkel 2013).
RUNX1 is overexpressed in epithelial ovarian carcinoma where it may contribute to cell proliferation, migration and invasion (Keita et al. 2013).
RUNX1 may cooperate with TP53 in transcriptional activation of TP53 target genes upon DNA damage (Wu et al. 2013).
RUNX1 is needed for the maintenance of skeletal musculature (Wang et al. 2005).
During mouse embryonic development, Runx1 is expressed in most nociceptive sensory neurons, which are involved in the perception of pain. In adult mice, Runx1 is expressed only in nociceptive sensory neurons that express the Ret receptor and is involved in regulation of expression of genes encoding ion channels (sodium-gated, ATP-gated and hydrogen ion-gated) and receptors (thermal receptors, opioid receptor MOR and the Mrgpr class of G protein coupled receptors). Mice lacking Runx1 show defective perception of thermal and neuropathic pain (Chen CL et al. 2006). Runx1 is thought to activate the neuronal differentiation of nociceptive dorsal root ganglion cells during embryonal development possibly through repression of Hes1 expression (Kobayashi et al. 2012). In chick and mouse embryos, Runx1 expression is restricted to the dorso-medial domain of the dorsal root ganglion, to TrkA-positive cutaneous sensory neurons. Runx3 expression in chick and mouse embryos is restricted to ventro-lateral domain of the dorsal root ganglion, to TrkC-positive proprioceptive neurons (Chen AI et al. 2006, Kramer et al. 2006). RUNX1 mediated regulation of neuronally expressed genes will be annotated when mechanistic details become available. View original pathway at Reactome.

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 8878171
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. Mao S, Frank RC, Zhang J, Miyazaki Y, Nimer SD.; ''Functional and physical interactions between AML1 proteins and an ETS protein, MEF: implications for the pathogenesis of t(8;21)-positive leukemias.''; PubMed Europe PMC Scholia
  2. Hoang T, Lambert JA, Martin R.; ''SCL/TAL1 in Hematopoiesis and Cellular Reprogramming.''; PubMed Europe PMC Scholia
  3. Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB, Dastur A, Amzallag A, Ramaswamy S, Tesar B, Jenkins CE, Hannett NM, McMillin D, Sanda T, Sim T, Kim ND, Look T, Mitsiades CS, Weng AP, Brown JR, Benes CH, Marto JA, Young RA, Gray NS.; ''Targeting transcription regulation in cancer with a covalent CDK7 inhibitor.''; PubMed Europe PMC Scholia
  4. Challen GA, Goodell MA.; ''Runx1 isoforms show differential expression patterns during hematopoietic development but have similar functional effects in adult hematopoietic stem cells.''; PubMed Europe PMC Scholia
  5. Block KL, Poncz M.; ''Platelet glycoprotein IIb gene expression as a model of megakaryocyte-specific expression.''; PubMed Europe PMC Scholia
  6. Kundu M, Javed A, Jeon JP, Horner A, Shum L, Eckhaus M, Muenke M, Lian JB, Yang Y, Nuckolls GH, Stein GS, Liu PP.; ''Cbfbeta interacts with Runx2 and has a critical role in bone development.''; PubMed Europe PMC Scholia
  7. Chastagner P, Israël A, Brou C.; ''AIP4/Itch regulates Notch receptor degradation in the absence of ligand.''; PubMed Europe PMC Scholia
  8. Zhang YY, Li X, Qian SW, Guo L, Huang HY, He Q, Liu Y, Ma CG, Tang QQ.; ''Down-regulation of type I Runx2 mediated by dexamethasone is required for 3T3-L1 adipogenesis.''; PubMed Europe PMC Scholia
  9. Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M.; ''The Notch target genes Hey1 and Hey2 are required for embryonic vascular development.''; PubMed Europe PMC Scholia
  10. Huang G, Zhao X, Wang L, Elf S, Xu H, Zhao X, Sashida G, Zhang Y, Liu Y, Lee J, Menendez S, Yang Y, Yan X, Zhang P, Tenen DG, Osato M, Hsieh JJ, Nimer SD.; ''The ability of MLL to bind RUNX1 and methylate H3K4 at PU.1 regulatory regions is impaired by MDS/AML-associated RUNX1/AML1 mutations.''; PubMed Europe PMC Scholia
  11. Cho JY, Akbarali Y, Zerbini LF, Gu X, Boltax J, Wang Y, Oettgen P, Zhang DE, Libermann TA.; ''Isoforms of the Ets transcription factor NERF/ELF-2 physically interact with AML1 and mediate opposing effects on AML1-mediated transcription of the B cell-specific blk gene.''; PubMed Europe PMC Scholia
  12. Lai EC, Deblandre GA, Kintner C, Rubin GM.; ''Drosophila neuralized is a ubiquitin ligase that promotes the internalization and degradation of delta.''; PubMed Europe PMC Scholia
  13. Pan D, Rubin GM.; ''Kuzbanian controls proteolytic processing of Notch and mediates lateral inhibition during Drosophila and vertebrate neurogenesis.''; PubMed Europe PMC Scholia
  14. Bray SJ, Takada S, Harrison E, Shen SC, Ferguson-Smith AC.; ''The atypical mammalian ligand Delta-like homologue 1 (Dlk1) can regulate Notch signalling in Drosophila.''; PubMed Europe PMC Scholia
  15. Browne G, Dragon JA, Hong D, Messier TL, Gordon JA, Farina NH, Boyd JR, VanOudenhove JJ, Perez AW, Zaidi SK, Stein JL, Stein GS, Lian JB.; ''MicroRNA-378-mediated suppression of Runx1 alleviates the aggressive phenotype of triple-negative MDA-MB-231 human breast cancer cells.''; PubMed Europe PMC Scholia
  16. Qadi AA, Taberlay PC, Phillips JL, Young A, West AC, Brettingham-Moore KH, Dickinson JL, Holloway AF.; ''The Leukemia Inhibitory Factor Receptor Gene Is a Direct Target of RUNX1.''; PubMed Europe PMC Scholia
  17. Zhou G, Zheng Q, Engin F, Munivez E, Chen Y, Sebald E, Krakow D, Lee B.; ''Dominance of SOX9 function over RUNX2 during skeletogenesis.''; PubMed Europe PMC Scholia
  18. Jalagadugula G, Mao G, Kaur G, Dhanasekaran DN, Rao AK.; ''Platelet protein kinase C-theta deficiency with human RUNX1 mutation: PRKCQ is a transcriptional target of RUNX1.''; PubMed Europe PMC Scholia
  19. Herglotz J, Kuvardina ON, Kolodziej S, Kumar A, Hussong H, Grez M, Lausen J.; ''Histone arginine methylation keeps RUNX1 target genes in an intermediate state.''; PubMed Europe PMC Scholia
  20. Welcker M, Clurman BE.; ''FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation.''; PubMed Europe PMC Scholia
  21. Sroczynska P, Lancrin C, Kouskoff V, Lacaud G.; ''The differential activities of Runx1 promoters define milestones during embryonic hematopoiesis.''; PubMed Europe PMC Scholia
  22. Le Marer N.; ''GALECTIN-3 expression in differentiating human myeloid cells.''; PubMed Europe PMC Scholia
  23. Chimge NO, Frenkel B.; ''The RUNX family in breast cancer: relationships with estrogen signaling.''; PubMed Europe PMC Scholia
  24. Roca H, Phimphilai M, Gopalakrishnan R, Xiao G, Franceschi RT.; ''Cooperative interactions between RUNX2 and homeodomain protein-binding sites are critical for the osteoblast-specific expression of the bone sialoprotein gene.''; PubMed Europe PMC Scholia
  25. Sato M, Morii E, Komori T, Kawahata H, Sugimoto M, Terai K, Shimizu H, Yasui T, Ogihara H, Yasui N, Ochi T, Kitamura Y, Ito Y, Nomura S.; ''Transcriptional regulation of osteopontin gene in vivo by PEBP2alphaA/CBFA1 and ETS1 in the skeletal tissues.''; PubMed Europe PMC Scholia
  26. Itoh M, Kim CH, Palardy G, Oda T, Jiang YJ, Maust D, Yeo SY, Lorick K, Wright GJ, Ariza-McNaughton L, Weissman AM, Lewis J, Chandrasekharappa SC, Chitnis AB.; ''Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by Delta.''; PubMed Europe PMC Scholia
  27. Aikawa Y, Nguyen LA, Isono K, Takakura N, Tagata Y, Schmitz ML, Koseki H, Kitabayashi I.; ''Roles of HIPK1 and HIPK2 in AML1- and p300-dependent transcription, hematopoiesis and blood vessel formation.''; PubMed Europe PMC Scholia
  28. Jalagadugula G, Mao G, Kaur G, Goldfinger LE, Dhanasekaran DN, Rao AK.; ''Regulation of platelet myosin light chain (MYL9) by RUNX1: implications for thrombocytopenia and platelet dysfunction in RUNX1 haplodeficiency.''; PubMed Europe PMC Scholia
  29. Chen CL, Broom DC, Liu Y, de Nooij JC, Li Z, Cen C, Samad OA, Jessell TM, Woolf CJ, Ma Q.; ''Runx1 determines nociceptive sensory neuron phenotype and is required for thermal and neuropathic pain.''; PubMed Europe PMC Scholia
  30. Robledo RF, Rajan L, Li X, Lufkin T.; ''The Dlx5 and Dlx6 homeobox genes are essential for craniofacial, axial, and appendicular skeletal development.''; PubMed Europe PMC Scholia
  31. Wallberg AE, Pedersen K, Lendahl U, Roeder RG.; ''p300 and PCAF act cooperatively to mediate transcriptional activation from chromatin templates by notch intracellular domains in vitro.''; PubMed Europe PMC Scholia
  32. Zhao X, Chen A, Yan X, Zhang Y, He F, Hayashi Y, Dong Y, Rao Y, Li B, Conway RM, Maiques-Diaz A, Elf SE, Huang N, Zuber J, Xiao Z, Tse W, Tenen DG, Wang Q, Chen W, Mulloy JC, Nimer SD, Huang G.; ''Downregulation of RUNX1/CBFβ by MLL fusion proteins enhances hematopoietic stem cell self-renewal.''; PubMed Europe PMC Scholia
  33. Takeda S, Bonnamy JP, Owen MJ, Ducy P, Karsenty G.; ''Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice.''; PubMed Europe PMC Scholia
  34. Wei SJ, Williams JG, Dang H, Darden TA, Betz BL, Humble MM, Chang FM, Trempus CS, Johnson K, Cannon RE, Tennant RW.; ''Identification of a specific motif of the DSS1 protein required for proteasome interaction and p53 protein degradation.''; PubMed Europe PMC Scholia
  35. Recouvreux MS, Grasso EN, Echeverria PC, Rocha-Viegas L, Castilla LH, Schere-Levy C, Tocci JM, Kordon EC, Rubinstein N.; ''RUNX1 and FOXP3 interplay regulates expression of breast cancer related genes.''; PubMed Europe PMC Scholia
  36. Huang H, Woo AJ, Waldon Z, Schindler Y, Moran TB, Zhu HH, Feng GS, Steen H, Cantor AB.; ''A Src family kinase-Shp2 axis controls RUNX1 activity in megakaryocyte and T-lymphocyte differentiation.''; PubMed Europe PMC Scholia
  37. Jaruga A, Hordyjewska E, Kandzierski G, Tylzanowski P.; ''Cleidocranial dysplasia and RUNX2-clinical phenotype-genotype correlation.''; PubMed Europe PMC Scholia
  38. Song R, Koo BK, Yoon KJ, Yoon MJ, Yoo KW, Kim HT, Oh HJ, Kim YY, Han JK, Kim CH, Kong YY.; ''Neuralized-2 regulates a Notch ligand in cooperation with Mind bomb-1.''; PubMed Europe PMC Scholia
  39. Li XQ, Du X, Li DM, Kong PZ, Sun Y, Liu PF, Wang QS, Feng YM.; ''ITGBL1 Is a Runx2 Transcriptional Target and Promotes Breast Cancer Bone Metastasis by Activating the TGFβ Signaling Pathway.''; PubMed Europe PMC Scholia
  40. Kitoh A, Ono M, Naoe Y, Ohkura N, Yamaguchi T, Yaguchi H, Kitabayashi I, Tsukada T, Nomura T, Miyachi Y, Taniuchi I, Sakaguchi S.; ''Indispensable role of the Runx1-Cbfbeta transcription complex for in vivo-suppressive function of FoxP3+ regulatory T cells.''; PubMed Europe PMC Scholia
  41. McGill MA, Dho SE, Weinmaster G, McGlade CJ.; ''Numb regulates post-endocytic trafficking and degradation of Notch1.''; PubMed Europe PMC Scholia
  42. Telfer JC, Rothenberg EV.; ''Expression and function of a stem cell promoter for the murine CBFalpha2 gene: distinct roles and regulation in natural killer and T cell development.''; PubMed Europe PMC Scholia
  43. Sanda T, Lawton LN, Barrasa MI, Fan ZP, Kohlhammer H, Gutierrez A, Ma W, Tatarek J, Ahn Y, Kelliher MA, Jamieson CH, Staudt LM, Young RA, Look AT.; ''Core transcriptional regulatory circuit controlled by the TAL1 complex in human T cell acute lymphoblastic leukemia.''; PubMed Europe PMC Scholia
  44. Eiraku M, Tohgo A, Ono K, Kaneko M, Fujishima K, Hirano T, Kengaku M.; ''DNER acts as a neuron-specific Notch ligand during Bergmann glial development.''; PubMed Europe PMC Scholia
  45. Lange S, Perera S, Teh P, Chen J.; ''Obscurin and KCTD6 regulate cullin-dependent small ankyrin-1 (sAnk1.5) protein turnover.''; PubMed Europe PMC Scholia
  46. Bäckström S, Wolf-Watz M, Grundström C, Härd T, Grundström T, Sauer UH.; ''The RUNX1 Runt domain at 1.25A resolution: a structural switch and specifically bound chloride ions modulate DNA binding.''; PubMed Europe PMC Scholia
  47. Jarriault S, Brou C, Logeat F, Schroeter EH, Kopan R, Israel A.; ''Signalling downstream of activated mammalian Notch.''; PubMed Europe PMC Scholia
  48. Palomero T, Lim WK, Odom DT, Sulis ML, Real PJ, Margolin A, Barnes KC, O'Neil J, Neuberg D, Weng AP, Aster JC, Sigaux F, Soulier J, Look AT, Young RA, Califano A, Ferrando AA.; ''NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth.''; PubMed Europe PMC Scholia
  49. Medina MA, Ugarte GD, Vargas MF, Avila ME, Necuñir D, Elorza AA, Gutiérrez SE, De Ferrari GV.; ''Alternative RUNX1 Promoter Regulation by Wnt/β-Catenin Signaling in Leukemia Cells and Human Hematopoietic Progenitors.''; PubMed Europe PMC Scholia
  50. Long F.; ''Building strong bones: molecular regulation of the osteoblast lineage.''; PubMed Europe PMC Scholia
  51. Mangan JK, Speck NA.; ''RUNX1 mutations in clonal myeloid disorders: from conventional cytogenetics to next generation sequencing, a story 40 years in the making.''; PubMed Europe PMC Scholia
  52. Debili N, Kieffer N, Nakazawa M, Guichard J, Titeux M, Cramer E, Breton-Gorius J, Vainchenker W.; ''Expression of platelet glycoprotein Ib by cultured human megakaryocytes: ultrastructural localization and biosynthesis.''; PubMed Europe PMC Scholia
  53. Wilson NK, Foster SD, Wang X, Knezevic K, Schütte J, Kaimakis P, Chilarska PM, Kinston S, Ouwehand WH, Dzierzak E, Pimanda JE, de Bruijn MF, Göttgens B.; ''Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators.''; PubMed Europe PMC Scholia
  54. Yoshida CA, Furuichi T, Fujita T, Fukuyama R, Kanatani N, Kobayashi S, Satake M, Takada K, Komori T.; ''Core-binding factor beta interacts with Runx2 and is required for skeletal development.''; PubMed Europe PMC Scholia
  55. Wysokinski D, Blasiak J, Pawlowska E.; ''Role of RUNX2 in Breast Carcinogenesis.''; PubMed Europe PMC Scholia
  56. Wu D, Ozaki T, Yoshihara Y, Kubo N, Nakagawara A.; ''Runt-related transcription factor 1 (RUNX1) stimulates tumor suppressor p53 protein in response to DNA damage through complex formation and acetylation.''; PubMed Europe PMC Scholia
  57. Leimeister C, Schumacher N, Steidl C, Gessler M.; ''Analysis of HeyL expression in wild-type and Notch pathway mutant mouse embryos.''; PubMed Europe PMC Scholia
  58. Xu G, Kanezaki R, Toki T, Watanabe S, Takahashi Y, Terui K, Kitabayashi I, Ito E.; ''Physical association of the patient-specific GATA1 mutants with RUNX1 in acute megakaryoblastic leukemia accompanying Down syndrome.''; PubMed Europe PMC Scholia
  59. Zhou S, Fujimuro M, Hsieh JJ, Chen L, Miyamoto A, Weinmaster G, Hayward SD.; ''SKIP, a CBF1-associated protein, interacts with the ankyrin repeat domain of NotchIC To facilitate NotchIC function.''; PubMed Europe PMC Scholia
  60. Otto F, Kanegane H, Mundlos S.; ''Mutations in the RUNX2 gene in patients with cleidocranial dysplasia.''; PubMed Europe PMC Scholia
  61. Tandon M, Chen Z, Pratap J.; ''Runx2 activates PI3K/Akt signaling via mTORC2 regulation in invasive breast cancer cells.''; PubMed Europe PMC Scholia
  62. Maier MM, Gessler M.; ''Comparative analysis of the human and mouse Hey1 promoter: Hey genes are new Notch target genes.''; PubMed Europe PMC Scholia
  63. Sakaguchi S.; ''Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses.''; PubMed Europe PMC Scholia
  64. Rossi M, De Laurenzi V, Munarriz E, Green DR, Liu YC, Vousden KH, Cesareni G, Melino G.; ''The ubiquitin-protein ligase Itch regulates p73 stability.''; PubMed Europe PMC Scholia
  65. Komeno Y, Yan M, Matsuura S, Lam K, Lo MC, Huang YJ, Tenen DG, Downing JR, Zhang DE.; ''Runx1 exon 6-related alternative splicing isoforms differentially regulate hematopoiesis in mice.''; PubMed Europe PMC Scholia
  66. Luo B, Aster JC, Hasserjian RP, Kuo F, Sklar J.; ''Isolation and functional analysis of a cDNA for human Jagged2, a gene encoding a ligand for the Notch1 receptor.''; PubMed Europe PMC Scholia
  67. Grbavec D, Stifani S.; ''Molecular interaction between TLE1 and the carboxyl-terminal domain of HES-1 containing the WRPW motif.''; PubMed Europe PMC Scholia
  68. Goldfarb AN.; ''Megakaryocytic programming by a transcriptional regulatory loop: A circle connecting RUNX1, GATA-1, and P-TEFb.''; PubMed Europe PMC Scholia
  69. Ben-Ami O, Pencovich N, Lotem J, Levanon D, Groner Y.; ''A regulatory interplay between miR-27a and Runx1 during megakaryopoiesis.''; PubMed Europe PMC Scholia
  70. Chimge NO, Little GH, Baniwal SK, Adisetiyo H, Xie Y, Zhang T, O'Laughlin A, Liu ZY, Ulrich P, Martin A, Mhawech-Fauceglia P, Ellis MJ, Tripathy D, Groshen S, Liang C, Li Z, Schones DE, Frenkel B.; ''RUNX1 prevents oestrogen-mediated AXIN1 suppression and β-catenin activation in ER-positive breast cancer.''; PubMed Europe PMC Scholia
  71. Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, Shiina M, Sato K, Kumasaka T, Yamamoto M, Ishii S, Ogata K.; ''Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta.''; PubMed Europe PMC Scholia
  72. Nguyen LA, Pandolfi PP, Aikawa Y, Tagata Y, Ohki M, Kitabayashi I.; ''Physical and functional link of the leukemia-associated factors AML1 and PML.''; PubMed Europe PMC Scholia
  73. Perissi V, Aggarwal A, Glass CK, Rose DW, Rosenfeld MG.; ''A corepressor/coactivator exchange complex required for transcriptional activation by nuclear receptors and other regulated transcription factors.''; PubMed Europe PMC Scholia
  74. Levy D, Reuven N, Shaul Y.; ''A regulatory circuit controlling Itch-mediated p73 degradation by Runx.''; PubMed Europe PMC Scholia
  75. Shevach EM.; ''Regulatory T cells in autoimmmunity*.''; PubMed Europe PMC Scholia
  76. Schroeter EH, Kisslinger JA, Kopan R.; ''Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain.''; PubMed Europe PMC Scholia
  77. Boller S, Grosschedl R.; ''The regulatory network of B-cell differentiation: a focused view of early B-cell factor 1 function.''; PubMed Europe PMC Scholia
  78. Wong WF, Kohu K, Chiba T, Sato T, Satake M.; ''Interplay of transcription factors in T-cell differentiation and function: the role of Runx.''; PubMed Europe PMC Scholia
  79. Hartmann D, de Strooper B, Serneels L, Craessaerts K, Herreman A, Annaert W, Umans L, Lübke T, Lena Illert A, von Figura K, Saftig P.; ''The disintegrin/metalloprotease ADAM 10 is essential for Notch signalling but not for alpha-secretase activity in fibroblasts.''; PubMed Europe PMC Scholia
  80. Koutelou E, Sato S, Tomomori-Sato C, Florens L, Swanson SK, Washburn MP, Kokkinaki M, Conaway RC, Conaway JW, Moschonas NK.; ''Neuralized-like 1 (Neurl1) targeted to the plasma membrane by N-myristoylation regulates the Notch ligand Jagged1.''; PubMed Europe PMC Scholia
  81. Bee T, Swiers G, Muroi S, Pozner A, Nottingham W, Santos AC, Li PS, Taniuchi I, de Bruijn MF.; ''Nonredundant roles for Runx1 alternative promoters reflect their activity at discrete stages of developmental hematopoiesis.''; PubMed Europe PMC Scholia
  82. Cordle J, Redfieldz C, Stacey M, van der Merwe PA, Willis AC, Champion BR, Hambleton S, Handford PA.; ''Localization of the delta-like-1-binding site in human Notch-1 and its modulation by calcium affinity.''; PubMed Europe PMC Scholia
  83. Thomas DM, Carty SA, Piscopo DM, Lee JS, Wang WF, Forrester WC, Hinds PW.; ''The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation.''; PubMed Europe PMC Scholia
  84. De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS, Ray WJ, Goate A, Kopan R.; ''A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain.''; PubMed Europe PMC Scholia
  85. Kishi N, Tang Z, Maeda Y, Hirai A, Mo R, Ito M, Suzuki S, Nakao K, Kinoshita T, Kadesch T, Hui C, Artavanis-Tsakonas S, Okano H, Matsuno K.; ''Murine homologs of deltex define a novel gene family involved in vertebrate Notch signaling and neurogenesis.''; PubMed Europe PMC Scholia
  86. Underwood KF, D'Souza DR, Mochin-Peters M, Pierce AD, Kommineni S, Choe M, Bennett J, Gnatt A, Habtemariam B, MacKerell AD, Passaniti A.; ''Regulation of RUNX2 transcription factor-DNA interactions and cell proliferation by vitamin D3 (cholecalciferol) prohormone activity.''; PubMed Europe PMC Scholia
  87. Koo BK, Yoon MJ, Yoon KJ, Im SK, Kim YY, Kim CH, Suh PG, Jan YN, Kong YY.; ''An obligatory role of mind bomb-1 in notch signaling of mammalian development.''; PubMed Europe PMC Scholia
  88. Yang DC, Yang MH, Tsai CC, Huang TF, Chen YH, Hung SC.; ''Hypoxia inhibits osteogenesis in human mesenchymal stem cells through direct regulation of RUNX2 by TWIST.''; PubMed Europe PMC Scholia
  89. Keita M, Bachvarova M, Morin C, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Trinh XB, Bachvarov D.; ''The RUNX1 transcription factor is expressed in serous epithelial ovarian carcinoma and contributes to cell proliferation, migration and invasion.''; PubMed Europe PMC Scholia
  90. Rhyu MS, Jan LY, Jan YN.; ''Asymmetric distribution of numb protein during division of the sensory organ precursor cell confers distinct fates to daughter cells.''; PubMed Europe PMC Scholia
  91. Chen AI, de Nooij JC, Jessell TM.; ''Graded activity of transcription factor Runx3 specifies the laminar termination pattern of sensory axons in the developing spinal cord.''; PubMed Europe PMC Scholia
  92. Ichikawa M, Asai T, Chiba S, Kurokawa M, Ogawa S.; ''Runx1/AML-1 ranks as a master regulator of adult hematopoiesis.''; PubMed Europe PMC Scholia
  93. Hug BA, Ahmed N, Robbins JA, Lazar MA.; ''A chromatin immunoprecipitation screen reveals protein kinase Cbeta as a direct RUNX1 target gene.''; PubMed Europe PMC Scholia
  94. Karsenty G.; ''Transcriptional control of skeletogenesis.''; PubMed Europe PMC Scholia
  95. Bonnefoy A, Hoylaerts MF.; ''Thrombospondin-1 in von Willebrand factor function.''; PubMed Europe PMC Scholia
  96. Wu JQ, Seay M, Schulz VP, Hariharan M, Tuck D, Lian J, Du J, Shi M, Ye Z, Gerstein M, Snyder MP, Weissman S.; ''Tcf7 is an important regulator of the switch of self-renewal and differentiation in a multipotential hematopoietic cell line.''; PubMed Europe PMC Scholia
  97. Lee MH, Kim YJ, Yoon WJ, Kim JI, Kim BG, Hwang YS, Wozney JM, Chi XZ, Bae SC, Choi KY, Cho JY, Choi JY, Ryoo HM.; ''Dlx5 specifically regulates Runx2 type II expression by binding to homeodomain-response elements in the Runx2 distal promoter.''; PubMed Europe PMC Scholia
  98. Wang W, Schwemmers S, Hexner EO, Pahl HL.; ''AML1 is overexpressed in patients with myeloproliferative neoplasms and mediates JAK2V617F-independent overexpression of NF-E2.''; PubMed Europe PMC Scholia
  99. Bakshi R, Hassan MQ, Pratap J, Lian JB, Montecino MA, van Wijnen AJ, Stein JL, Imbalzano AN, Stein GS.; ''The human SWI/SNF complex associates with RUNX1 to control transcription of hematopoietic target genes.''; PubMed Europe PMC Scholia
  100. Ge T, Yin M, Yang M, Liu T, Lou G.; ''MicroRNA-302b suppresses human epithelial ovarian cancer cell growth by targeting RUNX1.''; PubMed Europe PMC Scholia
  101. Yu M, Mazor T, Huang H, Huang HT, Kathrein KL, Woo AJ, Chouinard CR, Labadorf A, Akie TE, Moran TB, Xie H, Zacharek S, Taniuchi I, Roeder RG, Kim CF, Zon LI, Fraenkel E, Cantor AB.; ''Direct recruitment of polycomb repressive complex 1 to chromatin by core binding transcription factors.''; PubMed Europe PMC Scholia
  102. Jilma-Stohlawetz P, Homoncik M, Jilma B, Knechtelsdorfer M, Unger P, Mannhalter C, Santoso S, Panzer S.; ''Glycoprotein Ib polymorphisms influence platelet plug formation under high shear rates.''; PubMed Europe PMC Scholia
  103. Shigesada K, van de Sluis B, Liu PP.; ''Mechanism of leukemogenesis by the inv(16) chimeric gene CBFB/PEBP2B-MHY11.''; PubMed Europe PMC Scholia
  104. Elagib KE, Racke FK, Mogass M, Khetawat R, Delehanty LL, Goldfarb AN.; ''RUNX1 and GATA-1 coexpression and cooperation in megakaryocytic differentiation.''; PubMed Europe PMC Scholia
  105. Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz DM, Olson EN, Justice MJ, Karsenty G.; ''A twist code determines the onset of osteoblast differentiation.''; PubMed Europe PMC Scholia
  106. Welss T, Sun J, Irving JA, Blum R, Smith AI, Whisstock JC, Pike RN, von Mikecz A, Ruzicka T, Bird PI, Abts HF.; ''Hurpin is a selective inhibitor of lysosomal cathepsin L and protects keratinocytes from ultraviolet-induced apoptosis.''; PubMed Europe PMC Scholia
  107. Aneja K, Jalagadugula G, Mao G, Singh A, Rao AK.; ''Mechanism of platelet factor 4 (PF4) deficiency with RUNX1 haplodeficiency: RUNX1 is a transcriptional regulator of PF4.''; PubMed Europe PMC Scholia
  108. Oshima M, Endoh M, Endo TA, Toyoda T, Nakajima-Takagi Y, Sugiyama F, Koseki H, Kyba M, Iwama A, Osawa M.; ''Genome-wide analysis of target genes regulated by HoxB4 in hematopoietic stem and progenitor cells developing from embryonic stem cells.''; PubMed Europe PMC Scholia
  109. Benedito R, Roca C, Sörensen I, Adams S, Gossler A, Fruttiger M, Adams RH.; ''The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis.''; PubMed Europe PMC Scholia
  110. Yoshida CA, Yamamoto H, Fujita T, Furuichi T, Ito K, Inoue K, Yamana K, Zanma A, Takada K, Ito Y, Komori T.; ''Runx2 and Runx3 are essential for chondrocyte maturation, and Runx2 regulates limb growth through induction of Indian hedgehog.''; PubMed Europe PMC Scholia
  111. Kobayashi A, Senzaki K, Ozaki S, Yoshikawa M, Shiga T.; ''Runx1 promotes neuronal differentiation in dorsal root ganglion.''; PubMed Europe PMC Scholia
  112. Pande S, Browne G, Padmanabhan S, Zaidi SK, Lian JB, van Wijnen AJ, Stein JL, Stein GS.; ''Oncogenic cooperation between PI3K/Akt signaling and transcription factor Runx2 promotes the invasive properties of metastatic breast cancer cells.''; PubMed Europe PMC Scholia
  113. Maloy KJ, Powrie F.; ''Regulatory T cells in the control of immune pathology.''; PubMed Europe PMC Scholia
  114. Miao YS, Zhao YY, Zhao LN, Wang P, Liu YH, Ma J, Xue YX.; ''MiR-18a increased the permeability of BTB via RUNX1 mediated down-regulation of ZO-1, occludin and claudin-5.''; PubMed Europe PMC Scholia
  115. Pierce AD, Anglin IE, Vitolo MI, Mochin MT, Underwood KF, Goldblum SE, Kommineni S, Passaniti A.; ''Glucose-activated RUNX2 phosphorylation promotes endothelial cell proliferation and an angiogenic phenotype.''; PubMed Europe PMC Scholia
  116. Koo BK, Yoon KJ, Yoo KW, Lim HS, Song R, So JH, Kim CH, Kong YY.; ''Mind bomb-2 is an E3 ligase for Notch ligand.''; PubMed Europe PMC Scholia
  117. Kimelman D, Xu W.; ''beta-catenin destruction complex: insights and questions from a structural perspective.''; PubMed Europe PMC Scholia
  118. Matsuno K, Eastman D, Mitsiades T, Quinn AM, Carcanciu ML, Ordentlich P, Kadesch T, Artavanis-Tsakonas S.; ''Human deltex is a conserved regulator of Notch signalling.''; PubMed Europe PMC Scholia
  119. Nam Y, Sliz P, Song L, Aster JC, Blacklow SC.; ''Structural basis for cooperativity in recruitment of MAML coactivators to Notch transcription complexes.''; PubMed Europe PMC Scholia
  120. Lukasik SM, Zhang L, Corpora T, Tomanicek S, Li Y, Kundu M, Hartman K, Liu PP, Laue TM, Biltonen RL, Speck NA, Bushweller JH.; ''Altered affinity of CBF beta-SMMHC for Runx1 explains its role in leukemogenesis.''; PubMed Europe PMC Scholia
  121. Sprinzak D, Lakhanpal A, Lebon L, Santat LA, Fontes ME, Anderson GA, Garcia-Ojalvo J, Elowitz MB.; ''Cis-interactions between Notch and Delta generate mutually exclusive signalling states.''; PubMed Europe PMC Scholia
  122. Mukherjee A, Veraksa A, Bauer A, Rosse C, Camonis J, Artavanis-Tsakonas S.; ''Regulation of Notch signalling by non-visual beta-arrestin.''; PubMed Europe PMC Scholia
  123. Levy D, Adamovich Y, Reuven N, Shaul Y.; ''Yap1 phosphorylation by c-Abl is a critical step in selective activation of proapoptotic genes in response to DNA damage.''; PubMed Europe PMC Scholia
  124. Hu QD, Ang BT, Karsak M, Hu WP, Cui XY, Duka T, Takeda Y, Chia W, Sankar N, Ng YK, Ling EA, Maciag T, Small D, Trifonova R, Kopan R, Okano H, Nakafuku M, Chiba S, Hirai H, Aster JC, Schachner M, Pallen CJ, Watanabe K, Xiao ZC.; ''F3/contactin acts as a functional ligand for Notch during oligodendrocyte maturation.''; PubMed Europe PMC Scholia
  125. Inoue T, Stuart J, Leno R, Maki CG.; ''Nuclear import and export signals in control of the p53-related protein p73.''; PubMed Europe PMC Scholia
  126. Naillat F, Yan W, Karjalainen R, Liakhovitskaia A, Samoylenko A, Xu Q, Sun Z, Shen B, Medvinsky A, Quaggin S, Vainio SJ.; ''Identification of the genes regulated by Wnt-4, a critical signal for commitment of the ovary.''; PubMed Europe PMC Scholia
  127. Tribioli C, Lufkin T.; ''The murine Bapx1 homeobox gene plays a critical role in embryonic development of the axial skeleton and spleen.''; PubMed Europe PMC Scholia
  128. Hoverter NP, Ting JH, Sundaresh S, Baldi P, Waterman ML.; ''A WNT/p21 circuit directed by the C-clamp, a sequence-specific DNA binding domain in TCFs.''; PubMed Europe PMC Scholia
  129. Mansour MR, Abraham BJ, Anders L, Berezovskaya A, Gutierrez A, Durbin AD, Etchin J, Lawton L, Sallan SE, Silverman LB, Loh ML, Hunger SP, Sanda T, Young RA, Look AT.; ''Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element.''; PubMed Europe PMC Scholia
  130. Kanno T, Kanno Y, Chen LF, Ogawa E, Kim WY, Ito Y.; ''Intrinsic transcriptional activation-inhibition domains of the polyomavirus enhancer binding protein 2/core binding factor alpha subunit revealed in the presence of the beta subunit.''; PubMed Europe PMC Scholia
  131. Wee HJ, Voon DC, Bae SC, Ito Y.; ''PEBP2-beta/CBF-beta-dependent phosphorylation of RUNX1 and p300 by HIPK2: implications for leukemogenesis.''; PubMed Europe PMC Scholia
  132. Jayakumar A, Kang Y, Frederick MJ, Pak SC, Henderson Y, Holton PR, Mitsudo K, Silverman GA, EL-Naggar AK, Brömme D, Clayman GL.; ''Inhibition of the cysteine proteinases cathepsins K and L by the serpin headpin (SERPINB13): a kinetic analysis.''; PubMed Europe PMC Scholia
  133. Li N, Zhang QY, Zou JL, Li ZW, Tian TT, Dong B, Liu XJ, Ge S, Zhu Y, Gao J, Shen L.; ''miR-215 promotes malignant progression of gastric cancer by targeting RUNX1.''; PubMed Europe PMC Scholia
  134. Chastagner P, Israël A, Brou C.; ''Itch/AIP4 mediates Deltex degradation through the formation of K29-linked polyubiquitin chains.''; PubMed Europe PMC Scholia
  135. Lam K, Zhang DE.; ''RUNX1 and RUNX1-ETO: roles in hematopoiesis and leukemogenesis.''; PubMed Europe PMC Scholia
  136. Zhuang M, Gao W, Xu J, Wang P, Shu Y.; ''The long non-coding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1.''; PubMed Europe PMC Scholia
  137. Ono M, Yaguchi H, Ohkura N, Kitabayashi I, Nagamura Y, Nomura T, Miyachi Y, Tsukada T, Sakaguchi S.; ''Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1.''; PubMed Europe PMC Scholia
  138. Drissi H, Luc Q, Shakoori R, Chuva De Sousa Lopes S, Choi JY, Terry A, Hu M, Jones S, Neil JC, Lian JB, Stein JL, Van Wijnen AJ, Stein GS.; ''Transcriptional autoregulation of the bone related CBFA1/RUNX2 gene.''; PubMed Europe PMC Scholia
  139. Jarriault S, Le Bail O, Hirsinger E, Pourquié O, Logeat F, Strong CF, Brou C, Seidah NG, Isra l A.; ''Delta-1 activation of notch-1 signaling results in HES-1 transactivation.''; PubMed Europe PMC Scholia
  140. Wang X, Blagden C, Fan J, Nowak SJ, Taniuchi I, Littman DR, Burden SJ.; ''Runx1 prevents wasting, myofibrillar disorganization, and autophagy of skeletal muscle.''; PubMed Europe PMC Scholia
  141. Brou C, Logeat F, Gupta N, Bessia C, LeBail O, Doedens JR, Cumano A, Roux P, Black RA, Israël A.; ''A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin-metalloprotease TACE.''; PubMed Europe PMC Scholia
  142. Wu G, Lyapina S, Das I, Li J, Gurney M, Pauley A, Chui I, Deshaies RJ, Kitajewski J.; ''SEL-10 is an inhibitor of notch signaling that targets notch for ubiquitin-mediated protein degradation.''; PubMed Europe PMC Scholia
  143. Baladrón V, Ruiz-Hidalgo MJ, Nueda ML, Díaz-Guerra MJ, García-Ramírez JJ, Bonvini E, Gubina E, Laborda J.; ''dlk acts as a negative regulator of Notch1 activation through interactions with specific EGF-like repeats.''; PubMed Europe PMC Scholia
  144. Voges D, Zwickl P, Baumeister W.; ''The 26S proteasome: a molecular machine designed for controlled proteolysis.''; PubMed Europe PMC Scholia
  145. Huppert SS, Le A, Schroeter EH, Mumm JS, Saxena MT, Milner LA, Kopan R.; ''Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1.''; PubMed Europe PMC Scholia
  146. Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V, Amling M, Karsenty G.; ''A Cbfa1-dependent genetic pathway controls bone formation beyond embryonic development.''; PubMed Europe PMC Scholia
  147. Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, Ruas JL, Poellinger L, Lendahl U, Bondesson M.; ''Hypoxia requires notch signaling to maintain the undifferentiated cell state.''; PubMed Europe PMC Scholia
  148. Oberg C, Li J, Pauley A, Wolf E, Gurney M, Lendahl U.; ''The Notch intracellular domain is ubiquitinated and negatively regulated by the mammalian Sel-10 homolog.''; PubMed Europe PMC Scholia
  149. Fisher AL, Ohsako S, Caudy M.; ''The WRPW motif of the hairy-related basic helix-loop-helix repressor proteins acts as a 4-amino-acid transcription repression and protein-protein interaction domain.''; PubMed Europe PMC Scholia
  150. Ciccia A, Elledge SJ.; ''The DNA damage response: making it safe to play with knives.''; PubMed Europe PMC Scholia
  151. Karsenty G, Olson EN.; ''Bone and Muscle Endocrine Functions: Unexpected Paradigms of Inter-organ Communication.''; PubMed Europe PMC Scholia
  152. van Tetering G, van Diest P, Verlaan I, van der Wall E, Kopan R, Vooijs M.; ''Metalloprotease ADAM10 is required for Notch1 site 2 cleavage.''; PubMed Europe PMC Scholia
  153. Tijssen MR, Cvejic A, Joshi A, Hannah RL, Ferreira R, Forrai A, Bellissimo DC, Oram SH, Smethurst PA, Wilson NK, Wang X, Ottersbach K, Stemple DL, Green AR, Ouwehand WH, Göttgens B.; ''Genome-wide analysis of simultaneous GATA1/2, RUNX1, FLI1, and SCL binding in megakaryocytes identifies hematopoietic regulators.''; PubMed Europe PMC Scholia
  154. Ichikawa M, Yoshimi A, Nakagawa M, Nishimoto N, Watanabe-Okochi N, Kurokawa M.; ''A role for RUNX1 in hematopoiesis and myeloid leukemia.''; PubMed Europe PMC Scholia
  155. Li XQ, Lu JT, Tan CC, Wang QS, Feng YM.; ''RUNX2 promotes breast cancer bone metastasis by increasing integrin α5-mediated colonization.''; PubMed Europe PMC Scholia
  156. Zhang L, Lukasik SM, Speck NA, Bushweller JH.; ''Structural and functional characterization of Runx1, CBF beta, and CBF beta-SMMHC.''; PubMed Europe PMC Scholia
  157. Kao HY, Ordentlich P, Koyano-Nakagawa N, Tang Z, Downes M, Kintner CR, Evans RM, Kadesch T.; ''A histone deacetylase corepressor complex regulates the Notch signal transduction pathway.''; PubMed Europe PMC Scholia
  158. Shimizu K, Chiba S, Saito T, Kumano K, Hirai H.; ''Physical interaction of Delta1, Jagged1, and Jagged2 with Notch1 and Notch3 receptors.''; PubMed Europe PMC Scholia
  159. Kramer I, Sigrist M, de Nooij JC, Taniuchi I, Jessell TM, Arber S.; ''A role for Runx transcription factor signaling in dorsal root ganglion sensory neuron diversification.''; PubMed Europe PMC Scholia
  160. Ghozi MC, Bernstein Y, Negreanu V, Levanon D, Groner Y.; ''Expression of the human acute myeloid leukemia gene AML1 is regulated by two promoter regions.''; PubMed Europe PMC Scholia
  161. Oakford PC, James SR, Qadi A, West AC, Ray SN, Bert AG, Cockerill PN, Holloway AF.; ''Transcriptional and epigenetic regulation of the GM-CSF promoter by RUNX1.''; PubMed Europe PMC Scholia
  162. Lengner CJ, Hassan MQ, Serra RW, Lepper C, van Wijnen AJ, Stein JL, Lian JB, Stein GS.; ''Nkx3.2-mediated repression of Runx2 promotes chondrogenic differentiation.''; PubMed Europe PMC Scholia
  163. Cauwenberghs N, Vanhoorelbeke K, Vauterin S, Deckmyn H.; ''Structural determinants within platelet glycoprotein Ibalpha involved in its binding to von Willebrand factor.''; PubMed Europe PMC Scholia
  164. Mortus JR, Zhang Y, Hughes DP.; ''Developmental pathways hijacked by osteosarcoma.''; PubMed Europe PMC Scholia
  165. Arnett KL, Hass M, McArthur DG, Ilagan MX, Aster JC, Kopan R, Blacklow SC.; ''Structural and mechanistic insights into cooperative assembly of dimeric Notch transcription complexes.''; PubMed Europe PMC Scholia
  166. Bluteau D, Gilles L, Hilpert M, Antony-Debré I, James C, Debili N, Camara-Clayette V, Wagner-Ballon O, Cordette-Lagarde V, Robert T, Ripoche H, Gonin P, Swierczek S, Prchal J, Vainchenker W, Favier R, Raslova H.; ''Down-regulation of the RUNX1-target gene NR4A3 contributes to hematopoiesis deregulation in familial platelet disorder/acute myelogenous leukemia.''; PubMed Europe PMC Scholia
  167. Boyapati A, Ren B, Zhang DE.; ''SERPINB13 is a novel RUNX1 target gene.''; PubMed Europe PMC Scholia
  168. Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, Bates DL, Guo L, Han A, Ziegler SF, Mathis D, Benoist C, Chen L, Rao A.; ''FOXP3 controls regulatory T cell function through cooperation with NFAT.''; PubMed Europe PMC Scholia
  169. Yang LT, Nichols JT, Yao C, Manilay JO, Robey EA, Weinmaster G.; ''Fringe glycosyltransferases differentially modulate Notch1 proteolysis induced by Delta1 and Jagged1.''; PubMed Europe PMC Scholia
  170. Török K, Dezső B, Bencsik A, Uzonyi B, Erdei A.; ''Complement receptor type 1 (CR1/CD35) expressed on activated human CD4+ T cells contributes to generation of regulatory T cells.''; PubMed Europe PMC Scholia
  171. Stender JD, Kim K, Charn TH, Komm B, Chang KC, Kraus WL, Benner C, Glass CK, Katzenellenbogen BS.; ''Genome-wide analysis of estrogen receptor alpha DNA binding and tethering mechanisms identifies Runx1 as a novel tethering factor in receptor-mediated transcriptional activation.''; PubMed Europe PMC Scholia
  172. Fontana L, Pelosi E, Greco P, Racanicchi S, Testa U, Liuzzi F, Croce CM, Brunetti E, Grignani F, Peschle C.; ''MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation.''; PubMed Europe PMC Scholia
  173. Lahlil R, Lécuyer E, Herblot S, Hoang T.; ''SCL assembles a multifactorial complex that determines glycophorin A expression.''; PubMed Europe PMC Scholia
  174. Friedman AD.; ''Cell cycle and developmental control of hematopoiesis by Runx1.''; PubMed Europe PMC Scholia
  175. Zhao X, Jankovic V, Gural A, Huang G, Pardanani A, Menendez S, Zhang J, Dunne R, Xiao A, Erdjument-Bromage H, Allis CD, Tempst P, Nimer SD.; ''Methylation of RUNX1 by PRMT1 abrogates SIN3A binding and potentiates its transcriptional activity.''; PubMed Europe PMC Scholia
  176. Peterson LF, Boyapati A, Ranganathan V, Iwama A, Tenen DG, Tsai S, Zhang DE.; ''The hematopoietic transcription factor AML1 (RUNX1) is negatively regulated by the cell cycle protein cyclin D3.''; PubMed Europe PMC Scholia
  177. Joerger AC, Rajagopalan S, Natan E, Veprintsev DB, Robinson CV, Fersht AR.; ''Structural evolution of p53, p63, and p73: implication for heterotetramer formation.''; PubMed Europe PMC Scholia
  178. Paroush Z, Finley RL, Kidd T, Wainwright SM, Ingham PW, Brent R, Ish-Horowicz D.; ''Groucho is required for Drosophila neurogenesis, segmentation, and sex determination and interacts directly with hairy-related bHLH proteins.''; PubMed Europe PMC Scholia
  179. Ducy P, Karsenty G.; ''Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene.''; PubMed Europe PMC Scholia
  180. Wang Q, Stacy T, Miller JD, Lewis AF, Gu TL, Huang X, Bushweller JH, Bories JC, Alt FW, Ryan G, Liu PP, Wynshaw-Boris A, Binder M, Marín-Padilla M, Sharpe AH, Speck NA.; ''The CBFbeta subunit is essential for CBFalpha2 (AML1) function in vivo.''; PubMed Europe PMC Scholia
  181. Teplyuk NM, Galindo M, Teplyuk VI, Pratap J, Young DW, Lapointe D, Javed A, Stein JL, Lian JB, Stein GS, van Wijnen AJ.; ''Runx2 regulates G protein-coupled signaling pathways to control growth of osteoblast progenitors.''; PubMed Europe PMC Scholia
  182. Libermann TA, Pan Z, Akbarali Y, Hetherington CJ, Boltax J, Yergeau DA, Zhang DE.; ''AML1 (CBFalpha2) cooperates with B cell-specific activating protein (BSAP/PAX5) in activation of the B cell-specific BLK gene promoter.''; PubMed Europe PMC Scholia
  183. Kammerer M, Gutzwiller S, Stauffer D, Delhon I, Seltenmeyer Y, Fournier B.; ''Estrogen Receptor α (ERα) and Estrogen Related Receptor α (ERRα) are both transcriptional regulators of the Runx2-I isoform.''; PubMed Europe PMC Scholia
  184. Scheitz CJ, Lee TS, McDermitt DJ, Tumbar T.; ''Defining a tissue stem cell-driven Runx1/Stat3 signalling axis in epithelial cancer.''; PubMed Europe PMC Scholia
  185. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G.; ''Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation.''; PubMed Europe PMC Scholia
  186. Cai X, Gao L, Teng L, Ge J, Oo ZM, Kumar AR, Gilliland DG, Mason PJ, Tan K, Speck NA.; ''Runx1 Deficiency Decreases Ribosome Biogenesis and Confers Stress Resistance to Hematopoietic Stem and Progenitor Cells.''; PubMed Europe PMC Scholia
  187. Kuo YH, Zaidi SK, Gornostaeva S, Komori T, Stein GS, Castilla LH.; ''Runx2 induces acute myeloid leukemia in cooperation with Cbfbeta-SMMHC in mice.''; PubMed Europe PMC Scholia
  188. Burns CE, Traver D, Mayhall E, Shepard JL, Zon LI.; ''Hematopoietic stem cell fate is established by the Notch-Runx pathway.''; PubMed Europe PMC Scholia
  189. Perissi V, Scafoglio C, Zhang J, Ohgi KA, Rose DW, Glass CK, Rosenfeld MG.; ''TBL1 and TBLR1 phosphorylation on regulated gene promoters overcomes dual CtBP and NCoR/SMRT transcriptional repression checkpoints.''; PubMed Europe PMC Scholia
  190. Rebolledo-Jaramillo B, Alarcon RA, Fernandez VI, Gutierrez SE.; ''Cis-regulatory elements are harbored in Intron5 of the RUNX1 gene.''; PubMed Europe PMC Scholia
  191. Fryer CJ, White JB, Jones KA.; ''Mastermind recruits CycC:CDK8 to phosphorylate the Notch ICD and coordinate activation with turnover.''; PubMed Europe PMC Scholia
  192. Vladimirova V, Waha A, Lückerath K, Pesheva P, Probstmeier R.; ''Runx2 is expressed in human glioma cells and mediates the expression of galectin-3.''; PubMed Europe PMC Scholia
  193. Ito Y, Bae SC, Chuang LS.; ''The RUNX family: developmental regulators in cancer.''; PubMed Europe PMC Scholia
  194. Fryer CJ, Lamar E, Turbachova I, Kintner C, Jones KA.; ''Mastermind mediates chromatin-specific transcription and turnover of the Notch enhancer complex.''; PubMed Europe PMC Scholia
  195. Li L, Milner LA, Deng Y, Iwata M, Banta A, Graf L, Marcovina S, Friedman C, Trask BJ, Hood L, Torok-Storb B.; ''The human homolog of rat Jagged1 expressed by marrow stroma inhibits differentiation of 32D cells through interaction with Notch1.''; PubMed Europe PMC Scholia
  196. Rho JK, Kim JH, Yu J, Choe SY.; ''Correlation between cellular localization of TEL/AML1 fusion protein and repression of AML1-mediated transactivation of CR1 gene.''; PubMed Europe PMC Scholia
  197. Kim JH, Lee S, Rho JK, Choe SY.; ''AML1, the target of chromosomal rearrangements in human leukemia, regulates the expression of human complement receptor type 1 (CR1) gene.''; PubMed Europe PMC Scholia
  198. Zhang HY, Jin L, Stilling GA, Ruebel KH, Coonse K, Tanizaki Y, Raz A, Lloyd RV.; ''RUNX1 and RUNX2 upregulate Galectin-3 expression in human pituitary tumors.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114652view16:12, 25 January 2021ReactomeTeamReactome version 75
113100view11:16, 2 November 2020ReactomeTeamReactome version 74
112334view15:25, 9 October 2020ReactomeTeamReactome version 73
101233view11:13, 1 November 2018ReactomeTeamreactome version 66
100771view20:40, 31 October 2018ReactomeTeamreactome version 65
100315view19:17, 31 October 2018ReactomeTeamreactome version 64
99860view16:00, 31 October 2018ReactomeTeamreactome version 63
99417view14:35, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99101view12:39, 31 October 2018ReactomeTeamreactome version 62
93561view11:27, 9 August 2017ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
(PRC1.4,PRC1.5)ComplexR-HSA-8937992 (Reactome)
26S proteasomeComplexR-HSA-68819 (Reactome)
ACTL6A ProteinO96019 (Uniprot-TrEMBL)
ACTL6B ProteinO94805 (Uniprot-TrEMBL)
ADPMetaboliteCHEBI:456216 (ChEBI)
ARID1A ProteinO14497 (Uniprot-TrEMBL)
ARID1B ProteinQ8NFD5 (Uniprot-TrEMBL)
ARID2 ProteinQ68CP9 (Uniprot-TrEMBL)
ATPMetaboliteCHEBI:30616 (ChEBI)
AUTS2 ProteinQ8WXX7 (Uniprot-TrEMBL)
AXIN1 gene ProteinENSG00000103126 (Ensembl)
AXIN1 geneGeneProductENSG00000103126 (Ensembl)
AXIN1ProteinO15169 (Uniprot-TrEMBL)
AdoHcyMetaboliteCHEBI:16680 (ChEBI)
AdoMetMetaboliteCHEBI:15414 (ChEBI)
BLK gene ProteinENSG00000136573 (Ensembl)
BLK geneGeneProductENSG00000136573 (Ensembl)
BLKProteinP51451 (Uniprot-TrEMBL)
BMI1 ProteinP35226 (Uniprot-TrEMBL)
CAKComplexR-HSA-69221 (Reactome)
CBFB ProteinQ13951 (Uniprot-TrEMBL)
CBX2 ProteinQ14781 (Uniprot-TrEMBL)
CBX4 ProteinO00257 (Uniprot-TrEMBL)
CBX6 ProteinO95503 (Uniprot-TrEMBL)
CBX8 ProteinQ9HC52 (Uniprot-TrEMBL)
CCNH ProteinP51946 (Uniprot-TrEMBL)
CDK7 ProteinP50613 (Uniprot-TrEMBL)
CLDN5 gene ProteinENSG00000184113 (Ensembl)
CLDN5 geneGeneProductENSG00000184113 (Ensembl)
CLDN5ProteinO00501 (Uniprot-TrEMBL)
CREBBP ProteinQ92793 (Uniprot-TrEMBL)
CREBBPProteinQ92793 (Uniprot-TrEMBL)
CSF2 gene ProteinENSG00000164400 (Ensembl)
CSF2 geneGeneProductENSG00000164400 (Ensembl)
CSF2ProteinP04141 (Uniprot-TrEMBL)
CSNK2A1 ProteinP68400 (Uniprot-TrEMBL)
CSNK2A2 ProteinP19784 (Uniprot-TrEMBL)
CSNK2B ProteinP67870 (Uniprot-TrEMBL)
CTSK ProteinP43235 (Uniprot-TrEMBL)
CTSK:SERPINB13ComplexR-HSA-8938113 (Reactome)
CTSKProteinP43235 (Uniprot-TrEMBL)
CTSL1(114-288) ProteinP07711 (Uniprot-TrEMBL)
CTSL2 ProteinO60911 (Uniprot-TrEMBL)
Cathepsin LComplexR-HSA-2029110 (Reactome)
DNA Double Strand Break ResponsePathwayR-HSA-5693606 (Reactome) DNA double strand break (DSB) response involves sensing of DNA DSBs by the MRN complex which triggers ATM activation. ATM phosphorylates a number of proteins involved in DNA damage checkpoint signaling, as well as proteins directly involved in the repair of DNA DSBs. For a recent review, please refer to Ciccia and Elledge, 2010.
Degradation of

beta-catenin by the

destruction complex
PathwayR-HSA-195253 (Reactome) The beta-catenin destruction complex plays a key role in the canonical Wnt signaling pathway. In the absence of Wnt signaling, this complex controls the levels of cytoplamic beta-catenin. Beta-catenin associates with and is phosphorylated by the destruction complex. Phosphorylated beta-catenin is recognized and ubiquitinated by the SCF-beta TrCP ubiquitin ligase complex and is subsequently degraded by the proteasome (reviewed in Kimelman and Xu, 2006).
ELF1 ProteinP32519 (Uniprot-TrEMBL)
ELF1ProteinP32519 (Uniprot-TrEMBL)
ELF2 ProteinQ15723 (Uniprot-TrEMBL)
ELF2ProteinQ15723 (Uniprot-TrEMBL)
EP300 ProteinQ09472 (Uniprot-TrEMBL)
EP300ProteinQ09472 (Uniprot-TrEMBL)
ESR1 ProteinP03372 (Uniprot-TrEMBL)
ESR1:ESTGComplexR-HSA-1254381 (Reactome)
ESR1:estrogen:AXIN1 geneComplexR-HSA-8932051 (Reactome)
ESTG MetaboliteCHEBI:50114 (ChEBI)
FOXP3 ProteinQ9BZS1 (Uniprot-TrEMBL)
GATA1 ProteinP15976 (Uniprot-TrEMBL)
GATA2 ProteinP23769 (Uniprot-TrEMBL)
GATA3 ProteinP23771 (Uniprot-TrEMBL)
GPAM gene ProteinENSG00000119927 (Ensembl)
GPAM geneGeneProductENSG00000119927 (Ensembl)
GPAM(1-828)ProteinQ9HCL2 (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)
HIPK2ProteinQ9H2X6 (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)
IL3 gene ProteinENSG00000164399 (Ensembl)
IL3 geneGeneProductENSG00000164399 (Ensembl)
IL3ProteinP08700 (Uniprot-TrEMBL)
ITCH gene ProteinENSG00000078747 (Ensembl)
ITCH geneGeneProductENSG00000078747 (Ensembl)
ITCHProteinQ96J02 (Uniprot-TrEMBL)
KCTD6 gene ProteinENSG00000168301 (Ensembl)
KCTD6 geneGeneProductENSG00000168301 (Ensembl)
KCTD6ProteinQ8NC69 (Uniprot-TrEMBL)
KMT2A ProteinQ03164 (Uniprot-TrEMBL)
KMT2AProteinQ03164 (Uniprot-TrEMBL)
LDB1 ProteinQ86U70 (Uniprot-TrEMBL)
LGALS3 gene ProteinENSG00000131981 (Ensembl)
LGALS3 geneGeneProductENSG00000131981 (Ensembl)
LGALS3ProteinP17931 (Uniprot-TrEMBL)
LIFR gene ProteinENSG00000113594 (Ensembl)
LIFR geneGeneProductENSG00000113594 (Ensembl)
LIFRProteinP42702 (Uniprot-TrEMBL)
LMO1 ProteinP25800 (Uniprot-TrEMBL)
LMO2 ProteinP25791 (Uniprot-TrEMBL)
MNAT1 ProteinP51948 (Uniprot-TrEMBL)
MYB ProteinP10242 (Uniprot-TrEMBL)
MYB gene ProteinENSG00000118513 (Ensembl)
MYB geneGeneProductENSG00000118513 (Ensembl)
MYBProteinP10242 (Uniprot-TrEMBL)
Me3K5-H3F3A ProteinP84243 (Uniprot-TrEMBL)
Me3K5-HIST1H3A ProteinP68431 (Uniprot-TrEMBL)
Me3K5-HIST2H3A ProteinQ71DI3 (Uniprot-TrEMBL)
OCLN gene ProteinENSG00000197822 (Ensembl)
OCLN geneGeneProductENSG00000197822 (Ensembl)
OCLNProteinQ16625 (Uniprot-TrEMBL)
PAX5 ProteinQ02548 (Uniprot-TrEMBL)
PAX5ProteinQ02548 (Uniprot-TrEMBL)
PBRM1 ProteinQ86U86 (Uniprot-TrEMBL)
PCGF5 ProteinQ86SE9 (Uniprot-TrEMBL)
PHC1 ProteinP78364 (Uniprot-TrEMBL)
PHC2 ProteinQ8IXK0 (Uniprot-TrEMBL)
PHC3 ProteinQ8NDX5 (Uniprot-TrEMBL)
PRKCB gene ProteinENSG00000166501 (Ensembl)
PRKCB geneGeneProductENSG00000166501 (Ensembl)
PRKCBProteinP05771 (Uniprot-TrEMBL)
PSMA1 ProteinP25786 (Uniprot-TrEMBL)
PSMA2 ProteinP25787 (Uniprot-TrEMBL)
PSMA3 ProteinP25788 (Uniprot-TrEMBL)
PSMA4 ProteinP25789 (Uniprot-TrEMBL)
PSMA5 ProteinP28066 (Uniprot-TrEMBL)
PSMA6 ProteinP60900 (Uniprot-TrEMBL)
PSMA7 ProteinO14818 (Uniprot-TrEMBL)
PSMA8 ProteinQ8TAA3 (Uniprot-TrEMBL)
PSMB1 ProteinP20618 (Uniprot-TrEMBL)
PSMB10 ProteinP40306 (Uniprot-TrEMBL)
PSMB11 ProteinA5LHX3 (Uniprot-TrEMBL)
PSMB2 ProteinP49721 (Uniprot-TrEMBL)
PSMB3 ProteinP49720 (Uniprot-TrEMBL)
PSMB4 ProteinP28070 (Uniprot-TrEMBL)
PSMB5 ProteinP28074 (Uniprot-TrEMBL)
PSMB6 ProteinP28072 (Uniprot-TrEMBL)
PSMB7 ProteinQ99436 (Uniprot-TrEMBL)
PSMB8 ProteinP28062 (Uniprot-TrEMBL)
PSMB9 ProteinP28065 (Uniprot-TrEMBL)
PSMC1 ProteinP62191 (Uniprot-TrEMBL)
PSMC2 ProteinP35998 (Uniprot-TrEMBL)
PSMC3 ProteinP17980 (Uniprot-TrEMBL)
PSMC4 ProteinP43686 (Uniprot-TrEMBL)
PSMC5 ProteinP62195 (Uniprot-TrEMBL)
PSMC6 ProteinP62333 (Uniprot-TrEMBL)
PSMD1 ProteinQ99460 (Uniprot-TrEMBL)
PSMD10 ProteinO75832 (Uniprot-TrEMBL)
PSMD11 ProteinO00231 (Uniprot-TrEMBL)
PSMD12 ProteinO00232 (Uniprot-TrEMBL)
PSMD13 ProteinQ9UNM6 (Uniprot-TrEMBL)
PSMD14 ProteinO00487 (Uniprot-TrEMBL)
PSMD2 ProteinQ13200 (Uniprot-TrEMBL)
PSMD3 ProteinO43242 (Uniprot-TrEMBL)
PSMD4 ProteinP55036 (Uniprot-TrEMBL)
PSMD5 ProteinQ16401 (Uniprot-TrEMBL)
PSMD6 ProteinQ15008 (Uniprot-TrEMBL)
PSMD7 ProteinP51665 (Uniprot-TrEMBL)
PSMD8 ProteinP48556 (Uniprot-TrEMBL)
PSMD9 ProteinO00233 (Uniprot-TrEMBL)
PSME1 ProteinQ06323 (Uniprot-TrEMBL)
PSME2 ProteinQ9UL46 (Uniprot-TrEMBL)
PSME3 ProteinP61289 (Uniprot-TrEMBL)
PSME4 ProteinQ14997 (Uniprot-TrEMBL)
PSMF1 ProteinQ92530 (Uniprot-TrEMBL)
PolyUb-TP73 ProteinO15350 (Uniprot-TrEMBL)
PolyUb-TP73 tetramerComplexR-HSA-8957254 (Reactome)
RING1 ProteinQ06587 (Uniprot-TrEMBL)
RNF2 ProteinQ99496 (Uniprot-TrEMBL)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
RSPO3 gene ProteinENSG00000146374 (Ensembl)
RSPO3 geneGeneProductENSG00000146374 (Ensembl)
RSPO3ProteinQ9BXY4 (Uniprot-TrEMBL)
RUNX1 ProteinQ01196 (Uniprot-TrEMBL)
RUNX1 and FOXP3

control the development of regulatory T

lymphocytes (Tregs)
PathwayR-HSA-8877330 (Reactome) The complex of CBFB and RUNX1 (AML1) controls transcription of the FOXP3 gene. FOXP3 is a transcription factor that acts as a key regulator of development and function of regulatory T lymphocytes (Tregs). Tregs are CD25+CD4+ T lymphocytes involved in suppression of aberrant immune responses seen in autoimmune diseases and allergies. FOXP3 can bind to RUNX1 and control transcriptional activity of the RUNX1:CBFB complex. RUNX1 stimulates transcription of IL2 and IFNG1 (IFN-gamma), and the expression of these two genes is repressed upon binding of FOXP3 to RUNX1. The complex of FOXP3 and RUNX1, on the other hand, stimulates transcription of cell surface markers of Tregs, such as CD25, CTLA-4 and GITR. In the absence of FOXP3, RUNX1 represses transcription of these genes (Shevach 2000, Maloy and Powrie 2001, Sakaguchi 2004, Ono et al. 2007, Kitoh et al. 2009).
The RUNX1:CBFB complex directly stimulates transcription of the CR1 gene, encoding Complement receptor type 1 (CD35) (Kim et al. 1999, Rho et al. 2002). Expression of CR1 on the surface of activated T cells contributes to generation of Tregs (Torok et al. 2015).
RUNX1 regulates

genes involved in megakaryocyte differentiation and

platelet function
PathwayR-HSA-8936459 (Reactome) In human hematopoietic progenitors, RUNX1 and its partner CBFB are up-regulated at the onset of megakaryocytic differentiation and down-regulated at the onset of erythroid differentiation. The complex of RUNX1 and CBFB cooperates with the transcription factor GATA1 in the transactivation of megakaryocyte-specific genes. In addition, RUNX1 and GATA1 physically interact (Elagib et al. 2003), and this interaction involves the zinc finger domain of GATA1 (Xu et al. 2006). Other components of the RUNX1:CBFB activating complex at megakaryocytic promoters are GATA1 heterodimerization partner, ZFPM1 (FOG1), histone acetyltransferases EP300 (p300) and KAT2B (PCAF), the WDR5-containing histone methyltransferase MLL complex and the arginine methyltransferase PRMT1 (Herglotz et al. 2013). In the absence of PRMT1, the transcriptional repressor complex can form at megakaryocytic promoters, as RUNX1 that is not arginine methylated can bind to SIN3A/SIN3B co-repressors (Zhao et al. 2008). Besides SIN3A/SIN3B, the RUNX1:CBFB repressor complex at megakaryocytic promoters also includes histone deacetylase HDAC1 and histone arginine methyltransferase PRMT6 (Herglotz et al. 2013).
Megakaryocytic promoters regulated by the described RUNX1:CBFB activating and repressing complexes include ITGA2B, GP1BA, THBS1 and MIR27A (Herglotz et al. 2013). ITGA2B is only expressed in maturing megakaryocytes and platelets and is involved in platelet aggregation (Block and Poncz 1995). GP1BA is expressed at the cell surface membrane of maturing megakaryocytes and platelets and participates in formation of platelet plugs (Cauwenberghs et al. 2000, Jilma-Stohlawetz et al. 2003, Debili et al. 1990). THBS1 homotrimers contribute to stabilization of the platelet aggregate (Bonnefoy and Hoylaerts 2008). MIR27A is a negative regulator of RUNX1 mRNA translation and may be involved in erythroid/megakaryocytic lineage determination (Ben-Ami et al. 2009).
The RUNX1:CBFB complex stimulates transcription of the PF4 gene, encoding a component of platelet alpha granules (Aneja et al. 2011), the NR4A3 gene, associated with the familial platelet disorder (FPD) (Bluteau et al. 2011), the PRKCQ gene, associated with inherited thrombocytopenia (Jalagadugula et al. 2011), the MYL9 gene, involved in thrombopoiesis (Jalagadugula et al. 2010), and the NFE2 gene, a regulator of erythroid and megakaryocytic maturation and differentiation (Wang et al. 2010).
RUNX1:CBFB:(PRC1.4,PRC1.5)ComplexR-HSA-8937995 (Reactome)
RUNX1:CBFB:CLDN5 geneComplexR-HSA-8935998 (Reactome)
RUNX1:CBFB:CREBBP:CSF2 geneComplexR-HSA-8938236 (Reactome)
RUNX1:CBFB:CREBBPComplexR-HSA-8938230 (Reactome)
RUNX1:CBFB:ELF,ELF2,PAX5:BLK geneComplexR-HSA-8938969 (Reactome)
RUNX1:CBFB:ELF1,RUNX1:CBFB:ELF2,RUNX1:CBFB:PAX5ComplexR-HSA-8938964 (Reactome)
RUNX1:CBFB:ELF1:IL3 geneComplexR-HSA-8938942 (Reactome)
RUNX1:CBFB:ELF1ComplexR-HSA-8938916 (Reactome)
RUNX1:CBFB:ELF2ComplexR-HSA-8938928 (Reactome)
RUNX1:CBFB:EP300ComplexR-HSA-8878064 (Reactome)
RUNX1:CBFB:ESR1:estrogen:AXIN1 geneComplexR-HSA-8932085 (Reactome)
RUNX1:CBFB:ESR1:estrogen:GPAM geneComplexR-HSA-8932004 (Reactome)
RUNX1:CBFB:ESR1:estrogen:KCTD6 geneComplexR-HSA-8932035 (Reactome)
RUNX1:CBFB:ESR1:estrogenComplexR-HSA-8931986 (Reactome)
RUNX1:CBFB:FOXP3:RSPO3 geneComplexR-HSA-8877881 (Reactome)
RUNX1:CBFB:FOXP3ComplexR-HSA-8877193 (Reactome)
RUNX1:CBFB:GATA3-TAL1 core complex,MYB:RUNX1:CBFB:GATA3-TAL1 core complexComplexR-HSA-8956602 (Reactome)
RUNX1:CBFB:GATA3-TAL1 core complex:MYB geneComplexR-HSA-8956585 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:H3K4me3-NucleosomeComplexR-HSA-8865496 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:NucleosomeComplexR-HSA-8865490 (Reactome)
RUNX1:CBFB:KMT2AComplexR-HSA-8865480 (Reactome)
RUNX1:CBFB:LGALS3 geneComplexR-HSA-8938391 (Reactome)
RUNX1:CBFB:LIFR geneComplexR-HSA-8934703 (Reactome)
RUNX1:CBFB:OCLN geneComplexR-HSA-8935979 (Reactome)
RUNX1:CBFB:PAX5ComplexR-HSA-8938948 (Reactome)
RUNX1:CBFB:PRKCB geneComplexR-HSA-8939061 (Reactome)
RUNX1:CBFB:SERPINB13 geneComplexR-HSA-8938054 (Reactome)
RUNX1:CBFB:SOCS3 geneComplexR-HSA-8955750 (Reactome)
RUNX1:CBFB:SOCS4 geneComplexR-HSA-8955821 (Reactome)
RUNX1:CBFB:SWI/SNFComplexR-HSA-8938220 (Reactome)
RUNX1:CBFB:TAL1 core complexComplexR-HSA-8956549 (Reactome)
RUNX1:CBFB:TJP1 geneComplexR-HSA-8935959 (Reactome)
RUNX1:CBFB:YAP1:ITCH geneComplexR-HSA-8956650 (Reactome)
RUNX1:CBFB:YAP1ComplexR-HSA-8956638 (Reactome)
RUNX1:CBFBComplexR-HSA-8865330 (Reactome)
RUNX2-P1 ProteinQ13950-1 (Uniprot-TrEMBL)
RUNX2-P2 ProteinQ13950-2 (Uniprot-TrEMBL)
RUNX2:CBFB:LGALS3 geneComplexR-HSA-8938367 (Reactome)
RYBP ProteinQ8N488 (Uniprot-TrEMBL)
Regulation of RUNX1

Expression and

Activity
PathwayR-HSA-8934593 (Reactome) At the level of transcription, expression of the RUNX1 transcription factor is regulated by two alternative promoters: a distal promoter, P1, and a proximal promoter, P2. P1 is more than 7 kb upstream of P2 (Ghozi et al. 1996). In mice, the Runx1 gene is preferentially transcribed from the proximal P2 promoter during generation of hematopoietic cells from hemogenic endothelium. In fully committed hematopoietic progenitors, the Runx1 gene is preferentially transcribed from the distal P1 promoter (Sroczynska et al. 2009, Bee et al. 2010). In human T cells, RUNX1 is preferentially transcribed from P1 throughout development, while developing natural killer cells transcribe RUNX1 predominantly from P2. Developing B cells transcribe low levels of RUNX1 from both promoters (Telfer and Rothenberg 2001).
RUNX1 mRNAs transcribed from alternative promoters differ in their 5'UTRs and splicing isoforms of RUNX1 have also been described. The function of alternative splice isoforms and alternative 5'UTRs has not been fully elucidated (Challen and Goodell 2010, Komeno et al. 2014).
During zebrafish hematopoiesis, RUNX1 expression increases in response to NOTCH signaling, but direct transcriptional regulation of RUNX1 by NOTCH has not been demonstrated (Burns et al. 2005). RUNX1 transcription also increases in response to WNT signaling. BothTCF7 and TCF4 bind the RUNX1 promoter (Wu et al. 2012, Hoverter et al. 2012), and RUNX1 transcription driven by the TCF binding element (TBE) in response to WNT3A treatment is inhibited by the dominant-negative mutant of TCF4 (Medina et al. 2016). In developing mouse ovary, Runx1 expression is positively regulated by Wnt4 signaling (Naillat et al. 2015).
Studies in mouse hematopoietic stem and progenitor cells imply that RUNX1 may be a direct transcriptional target of HOXB4 (Oshima et al. 2011).
Conserved cis-regulatory elements were recently identified in intron 5 of RUNX1. The RUNX1 breakpoints observed in acute myeloid leukemia (AML) with translocation (8;21), which result in expression of a fusion RUNX1-ETO protein, cluster in intron 5, in proximity to these not yet fully characterized cis regulatory elements (Rebolledo-Jaramillo et al. 2014).
At the level of translation, RUNX1 expression is regulated by various microRNAs which bind to the 3'UTR of RUNX1 mRNA and inhibit its translation through endonucleolytic and/or nonendonucleolytic mechanisms. MicroRNAs that target RUNX1 include miR-378 (Browne et al. 2016), miR-302b (Ge et al. 2014), miR-18a (Miao et al. 2015), miR-675 (Zhuang et al. 2014), miR-27a (Ben-Ami et al. 2009), miR-17, miR-20a, miR106 (Fontana et al. 2007) and miR-215 (Li et al. 2016).
At the posttranslational level, RUNX1 activity is regulated by postranslational modifications and binding to co-factors. SRC family kinases phosphorylate RUNX1 on multiple tyrosine residues in the negative regulatory domain, involved in autoinhibition of RUNX1. RUNX1 tyrosine phosphorylation correlates with reduced binding of RUNX1 to GATA1 and increased binding of RUNX1 to the SWI/SNF complex, leading to inhibition of RUNX1-mediated differentiation of T-cells and megakaryocytes. SHP2 (PTPN11) tyrosine phosphatase binds to RUNX1 and dephosphorylates it (Huang et al. 2012).
Formation of the complex with CBFB is necessary for the transcriptional activity of RUNX1 (Wang et al. 1996). Binding of CCND3 and probably other two cyclin D family members, CCND1 and CCND2, to RUNX1 inhibits its association with CBFB (Peterson et al. 2005), while binding to CDK6 interferes with binding of RUNX1 to DNA without affecting formation of the RUNX1:CBFB complex. Binding of RUNX1 to PML plays a role in subnuclear targeting of RUNX1 (Nguyen et al. 2005).
RUNX1 activity and protein levels vary during the cell cycle. RUNX1 protein levels increase from G1 to S and from S to G2 phases, with no increase in RUNX1 mRNA levels. CDK1-mediated phosphorylation of RUNX1 at the G2/M transition is implicated in reduction of RUNX1 transactivation potency and may promote RUNX1 protein degradation by the anaphase promoting complex (reviewed by Friedman 2009).
SCMH1-2 ProteinQ96GD3-2 (Uniprot-TrEMBL)
SERPINB13 ProteinQ9UIV8 (Uniprot-TrEMBL)
SERPINB13 gene ProteinENSG00000197641 (Ensembl)
SERPINB13 geneGeneProductENSG00000197641 (Ensembl)
SERPINB13:cathepsin LComplexR-HSA-8938111 (Reactome)
SERPINB13ProteinQ9UIV8 (Uniprot-TrEMBL)
SHFM1 ProteinP60896 (Uniprot-TrEMBL)
SMARCA2 ProteinP51531 (Uniprot-TrEMBL)
SMARCA4 ProteinP51532 (Uniprot-TrEMBL)
SMARCB1 ProteinQ12824 (Uniprot-TrEMBL)
SMARCC1 ProteinQ92922 (Uniprot-TrEMBL)
SMARCC2 ProteinQ8TAQ2 (Uniprot-TrEMBL)
SMARCD1 ProteinQ96GM5 (Uniprot-TrEMBL)
SMARCD2 ProteinQ92925 (Uniprot-TrEMBL)
SMARCD3 ProteinQ6STE5 (Uniprot-TrEMBL)
SMARCE1 ProteinQ969G3 (Uniprot-TrEMBL)
SOCS3 Gene ProteinENSG00000184557 (Ensembl)
SOCS3 GeneGeneProductENSG00000184557 (Ensembl)
SOCS3ProteinO14543 (Uniprot-TrEMBL)
SOCS4 gene ProteinENSG00000180008 (Ensembl)
SOCS4 geneGeneProductENSG00000180008 (Ensembl)
SOCS4ProteinQ8WXH5 (Uniprot-TrEMBL)
SPI1 Gene ProteinENSG00000066336 (Ensembl)
SPI1 gene:NucleosomeComplexR-HSA-8865492 (Reactome)
SPI1ProteinP17947 (Uniprot-TrEMBL)
SWI/SNF chromatin remodelling complexComplexR-HSA-5225604 (Reactome)
Signaling by NOTCH1PathwayR-HSA-1980143 (Reactome) NOTCH1 functions as both a transmembrane receptor presented on the cell surface and as a transcriptional regulator in the nucleus.

NOTCH1 receptor presented on the plasma membrane is activated by a membrane bound ligand expressed in trans on the surface of a neighboring cell. In trans, ligand binding triggers proteolytic cleavage of NOTCH1 and results in release of the NOTCH1 intracellular domain, NICD1, into the cytosol.

NICD1 translocates to the nucleus where it associates with RBPJ (also known as CSL or CBF) and mastermind-like (MAML) proteins (MAML1, MAML2 or MAML3; possibly also MAMLD1) to form NOTCH1 coactivator complex. NOTCH1 coactivator complex activates transcription of genes that possess RBPJ binding sites in their promoters.

TAL1 ProteinP17542 (Uniprot-TrEMBL)
TAL1 core complexComplexR-HSA-8956524 (Reactome)
TCF12 ProteinQ99081 (Uniprot-TrEMBL)
TCF3 ProteinP15923 (Uniprot-TrEMBL)
TJP1 gene ProteinENSG00000104067 (Ensembl)
TJP1 geneGeneProductENSG00000104067 (Ensembl)
TJP1ProteinQ07157 (Uniprot-TrEMBL)
TP73 ProteinO15350 (Uniprot-TrEMBL)
TP73 TetramerComplexR-HSA-6798077 (Reactome)
TP73 TetramerComplexR-HSA-8957246 (Reactome)
Transcriptional regulation by RUNX2PathwayR-HSA-8878166 (Reactome) RUNX2 (CBFA1 or AML3) transcription factor, similar to other RUNX family members, RUNX1 and RUNX3, can function in complex with CBFB (CBF-beta) (Kundu et al. 2002, Yoshida et al. 2002, Otto et al. 2002). RUNX2 mainly regulates transcription of genes involved in skeletal development (reviewed in Karsenty 2008). RUNX2 is involved in development of both intramembraneous and endochondral bones through regulation of osteoblast differentiation and chondrocyte maturation, respectively. RUNX2 stimulates transcription of the BGLAP gene (Ducy and Karsenty 1995, Ducy et al. 1997), which encodes Osteocalcin, a bone-derived hormone which is one of the most abundant non-collagenous proteins of the bone extracellular matrix (reviewed in Karsenty and Olson 2016). RUNX2 directly controls the expression of most genes associated with osteoblast differentiation and function (Sato et al. 1998, Ducy et al. 1999, Roce et al. 2005). RUNX2-mediated transcriptional regulation of several genes involved in GPCR (G protein coupled receptor) signaling is implicated in the control of growth of osteoblast progenitors (Teplyuk et al. 2009). RUNX2 promotes chondrocyte maturation by stimulating transcription of the IHH gene, encoding Indian hedgehog (Takeda et al. 2001, Yoshida et al. 2004). Germline loss-of-function mutations of the RUNX2 gene are associated with cleidocranial dysplasia syndrome (CCD), an autosomal skeletal disorder (reviewed in Jaruga et al. 2016). The function of RUNX2 is frequently disrupted in osteosarcoma (reviewed in Mortus et al. 2014). Vitamin D3 is implicated in regulation of transcriptional activity of the RUNX2:CBFB complex (Underwood et al. 2012).

RUNX2 expression is regulated by estrogen signaling, and RUNX2 is implicated in breast cancer development and metastasis (reviewed in Wysokinski et al. 2014). Besides estrogen receptor alpha (ESR1) and estrogen-related receptor alpha (ERRA) (Kammerer et al. 2013), RUNX2 transcription is also regulated by TWIST1 (Yang, Yang et al. 2011), glucocorticoid receptor (NR3C1) (Zhang et al. 2012), NKX3-2 (BAPX1) (Tribioli and Lufkin 1999, Lengner et al. 2005), DLX5 (Robledo et al. 2002, Lee et al. 2005) and MSX2 (Lee et al. 2005). RUNX2 can autoregulate, by directly inhibiting its own transcription (Drissi et al. 2000). Several E3 ubiquitin ligases target RUNX2 for proteasome-mediated degradation: FBXW7a (Kumar et al. 2015), STUB1 (CHIP) (Li et al. 2008), SMURF1 (Zhao et al. 2003, Yang et al. 2014), WWP1 (Jones et al. 2006), and SKP2 (Thacker et al. 2016). Besides formation of RUNX2:CBFB heterodimers, transcriptional activity of RUNX2 is regulated by binding to a number of other transcription factors, for example SOX9 (Zhou et al. 2006, TWIST1 (Bialek et al. 2004) and RB1 (Thomas et al. 2001).

RUNX2 regulates expression of several genes implicated in cell migration during normal development and bone metastasis of breast cancer cells. RUNX2 stimulates transcription of the ITGA5 gene, encoding Integrin alpha 5 (Li et al. 2016) and the ITGBL1 gene, encoding Integrin beta like protein 1 (Li et al. 2015). RUNX2 mediated transcription of the MMP13 gene, encoding Colagenase 3 (Matrix metalloproteinase 13), is stimulated by AKT mediated phosphorylation of RUNX2 (Pande et al. 2013). RUNX2 is implicated in positive regulation of AKT signaling by stimulating expression of AKT-activating TORC2 complex components MTOR and RICTOR, which may contribute to survival of breast cancer cells (Tandon et al. 2014).

RUNX2 inhibits CDKN1A transcription, thus preventing CDKN1A-induced cell cycle arrest. Phosphorylation of RUNX2 by CDK4 in response to high glucose enhances RUNX2-mediated repression of the CDKN1A gene in endothelial cells (Pierce et al. 2012). In mice, Runx2-mediated repression of Cdkn1a may contribute to the development of acute myeloid leukemia (AML) (Kuo et al. 2009). RUNX2 can stimulate transcription of the LGALS3 gene, encoding Galectin-3 (Vladimirova et al. 2008, Zhang et al. 2009). Galectin 3 is expressed in myeloid progenitors and its levels increase during the maturation process (Le Marer 2000).

For a review of RUNX2 function, please refer to Long 2012 and Ito et al. 2015.

UBA52(1-76) ProteinP62987 (Uniprot-TrEMBL)
UBB(1-76) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(153-228) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(77-152) ProteinP0CG47 (Uniprot-TrEMBL)
UBC(1-76) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(153-228) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(229-304) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(305-380) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(381-456) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(457-532) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(533-608) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(609-684) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(77-152) ProteinP0CG48 (Uniprot-TrEMBL)
UbComplexR-HSA-113595 (Reactome)
YAF2 ProteinQ8IY57 (Uniprot-TrEMBL)
YAP1 ProteinP46937 (Uniprot-TrEMBL)
YAP1ProteinP46937 (Uniprot-TrEMBL)
p-S,T-EP300 ProteinQ09472 (Uniprot-TrEMBL)
p-S249,S273,T276-RUNX1 ProteinQ01196 (Uniprot-TrEMBL)
p-S249,S273,T276-RUNX1:CBFB:p-S,T-EP300ComplexR-HSA-8878073 (Reactome)
p-S249,S273,T276-RUNX1:CBFBComplexR-HSA-8878069 (Reactome)
p-S456-ABL1ProteinP00519 (Uniprot-TrEMBL)
p-Y407-YAP1 ProteinP46937 (Uniprot-TrEMBL)
p-Y407-YAP1:TP73 tetramerComplexR-HSA-8956675 (Reactome)
p-Y407-YAP1ProteinP46937 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
(PRC1.4,PRC1.5)R-HSA-8937989 (Reactome)
26S proteasomemim-catalysisR-HSA-8957265 (Reactome)
ADPArrowR-HSA-8878050 (Reactome)
ADPArrowR-HSA-8878054 (Reactome)
ADPArrowR-HSA-8956659 (Reactome)
ATPR-HSA-8878050 (Reactome)
ATPR-HSA-8878054 (Reactome)
ATPR-HSA-8956659 (Reactome)
AXIN1 geneR-HSA-8932070 (Reactome)
AXIN1 geneR-HSA-8932076 (Reactome)
AXIN1 geneR-HSA-8932084 (Reactome)
AXIN1ArrowR-HSA-8932076 (Reactome)
AdoHcyArrowR-HSA-8865498 (Reactome)
AdoMetR-HSA-8865498 (Reactome)
BLK geneR-HSA-8938965 (Reactome)
BLK geneR-HSA-8938987 (Reactome)
BLKArrowR-HSA-8938987 (Reactome)
CAKArrowR-HSA-8956568 (Reactome)
CLDN5 geneR-HSA-8936000 (Reactome)
CLDN5 geneR-HSA-8936007 (Reactome)
CLDN5ArrowR-HSA-8936007 (Reactome)
CREBBPR-HSA-8938231 (Reactome)
CSF2 geneR-HSA-8938228 (Reactome)
CSF2 geneR-HSA-8938251 (Reactome)
CSF2ArrowR-HSA-8938251 (Reactome)
CTSK:SERPINB13ArrowR-HSA-8938121 (Reactome)
CTSKR-HSA-8938121 (Reactome)
Cathepsin LR-HSA-8938108 (Reactome)
ELF1R-HSA-8938913 (Reactome)
ELF2R-HSA-8938930 (Reactome)
EP300R-HSA-8878056 (Reactome)
ESR1:ESTGR-HSA-8931981 (Reactome)
ESR1:ESTGR-HSA-8932070 (Reactome)
ESR1:estrogen:AXIN1 geneArrowR-HSA-8932070 (Reactome)
ESR1:estrogen:AXIN1 geneTBarR-HSA-8932076 (Reactome)
GPAM geneR-HSA-8932020 (Reactome)
GPAM geneR-HSA-8932021 (Reactome)
GPAM(1-828)ArrowR-HSA-8932020 (Reactome)
HIPK2mim-catalysisR-HSA-8878050 (Reactome)
HIPK2mim-catalysisR-HSA-8878054 (Reactome)
IL3 geneR-HSA-8938938 (Reactome)
IL3 geneR-HSA-8938981 (Reactome)
IL3ArrowR-HSA-8938981 (Reactome)
ITCH geneR-HSA-8956649 (Reactome)
ITCH geneR-HSA-8956652 (Reactome)
ITCHArrowR-HSA-8956652 (Reactome)
ITCHmim-catalysisR-HSA-8956684 (Reactome)
KCTD6 geneR-HSA-8932033 (Reactome)
KCTD6 geneR-HSA-8932037 (Reactome)
KCTD6ArrowR-HSA-8932033 (Reactome)
KMT2AR-HSA-8865482 (Reactome)
LGALS3 geneR-HSA-8938382 (Reactome)
LGALS3 geneR-HSA-8938389 (Reactome)
LGALS3ArrowR-HSA-8938382 (Reactome)
LIFR geneR-HSA-8934688 (Reactome)
LIFR geneR-HSA-8934690 (Reactome)
LIFRArrowR-HSA-8934690 (Reactome)
MYB geneR-HSA-8956586 (Reactome)
MYB geneR-HSA-8956608 (Reactome)
MYBArrowR-HSA-8956608 (Reactome)
OCLN geneR-HSA-8935980 (Reactome)
OCLN geneR-HSA-8935988 (Reactome)
OCLNArrowR-HSA-8935988 (Reactome)
PAX5R-HSA-8938951 (Reactome)
PRKCB geneR-HSA-8939054 (Reactome)
PRKCB geneR-HSA-8939066 (Reactome)
PRKCBArrowR-HSA-8939066 (Reactome)
PolyUb-TP73 tetramerArrowR-HSA-8956684 (Reactome)
PolyUb-TP73 tetramerR-HSA-8957265 (Reactome)
R-HSA-8865482 (Reactome) Histone methyltransferase KMT2A (MLL) binds to RUNX1 (AML1) both in the presence and absence of CBFB (Huang et al. 2011).
R-HSA-8865491 (Reactome) RUNX1 recruits histone methyltransferase KMT2A (MLL) to the SPI1 (PU.1) gene locus (Huang et al. 2011). This interaction was demonstrated by using endogenous mouse proteins and DNA, as well as by expressing recombinant human KMT2A and RUNX1 in mouse myeloid progenitor cell line 416B.
R-HSA-8865498 (Reactome) Once recruited to the SPI1 (PU.1) gene locus, KMT2A (MLL) histone methyltransferase trimethylates nucleosomes associated with the SPI1 promoter and the upstream regulatory element. KMT2A creates the H3K4Me3 mark on histone H3 at the SPI1 gene, a characteristic of transcriptionally active chromatin (Huang et al. 2011).
R-HSA-8865505 (Reactome) The SPI1 (PU.1) transcription factor represses self renewal and proliferation of HSCs (Fukuchi et al. 2008) and is needed for commitment of HSCs to specific hematopoietic lineages (Imperato et al. 2015), for example differentiation of lymphoid cells. SPI1 gene transcription is directly stimulated by the RUNX1:CBFB transcription factor complex, in the presence of the activating histone methyltransferase KMT2A (MLL) (Huang et al. 2011).
R-HSA-8877879 (Reactome) The complex of the RUNX1:CBFB heterodimer and FOXP3 binds the promoter of the RSPO3 gene, which encodes a WNT ligand (Recouvreux et al. 2016).
R-HSA-8877884 (Reactome) Binding of the RUNX1:CBFB complex and FOXP3 to the promoter of the RSPO3 gene stimulates RSPO3 transcription. RSPO3 functions as a WNT ligand and is a known breast cancer oncogene (Recouvreux et al. 2016).
R-HSA-8878050 (Reactome) HIPK2 can simultaneously phosphorylate RUNX1 and EP300 (p300) histone acetyltransferase bound to the RUNX1:CBFB complex. RUNX1 is phosphorylated by HIPK2 at serine residues S249 and S273 and threonine residue T276. EP300 is phosphorylated by HIPK2 at multiple serine and threonine residues that have not been thoroughly identified. HIPK2-mediated phosphorylation contributed to the activation of histone acetyltransferase activity of EP300 at RUNX1 target promoters (Aikawa et al. 2006, Wee et al. 2008).
R-HSA-8878054 (Reactome) Protein serine/threonine kinase HIPK2 phosphorylates RUNX1 upon formation of the RUNX1:CBFB complex. Serine residues S249 and S273 and threonine residue S276 of RUNX1 are phosphorylated by HIPK2. HIPK2-mediated phosphorylation of RUNX1 is implicated as an important regulatory step in hematopoiesis and is disrupted by leukemogenic mutations in CBFB (Aikawa et al. 2006, Wee et al. 2008).
R-HSA-8878056 (Reactome) The complex of CBFB and RUNX1 can bind histone acetyltransferase EP300 (p300). Formation of this complex does not depend on HIPK2-mediated phosphorylation of RUNX1. Histone acetyltransferase activity of EP300 probably contributes to transcriptional activation of RUNX1:CBFB target genes (Aikawa et al. 2006, Wee et al. 2008).
R-HSA-8931981 (Reactome) The RUNX1:CBFB complex binds the estrogen receptor alpha (ESR1). The interaction between RUNX1 and ESR1 is significantly enhanced upon ESR1 activation by estrogens (Stender et al. 2010).
R-HSA-8932020 (Reactome) GPAM gene expression is cooperatively stimulated by RUNX1 and ESR1, which form a complex and bind the GPAM gene enhancer (Stender et al. 2010). GPAM encodes a glycerol-3-phosphate acyltransferase whose high expression correlates with better overall survival in breast cancer (Brockmoller et al. 2012).
R-HSA-8932021 (Reactome) RUNX1 and ESR1 cooperatively bind to the enhancer of the GPAM gene, which contains both estrogen response elements and RUNX1 binding sites (Stender et al. 2010).
R-HSA-8932033 (Reactome) RUNX1 and ESR1, which form a complex that binds to the KCTD6 gene enhancer, cooperatively stimulate the expression of the KCTD6 gene (Stender et al. 2010).
R-HSA-8932037 (Reactome) RUNX1 and ESR1 cooperatively bind the KCTD6 gene enhancer, which contains both estrogen response elements and RUNX1 response elements (Stender et al. 2010).
R-HSA-8932070 (Reactome) Estrogen receptor alpha (ESR1) binds to estrogen response elements in the second intron of the AXIN1 gene (Chimge et al. 2016).
R-HSA-8932076 (Reactome) Transciption of the AXIN1 gene, which encodes a component of the beta-catenin (CTNNB1) destruction complex, is inhibited by binding of the activated estrogen receptor alpha (ESR1) to estrogen response elements in the second intron of AXIN1 (Chimge et al. 2016).
The AXIN1 gene expression is stimulated by cooperative binding of RUNX1 and estrogen receptor alpha (ESR1) to adjacent RUNX1 binding sites and estrogen response elements in the second intron of AXIN1 (Chimge et al. 2016).
R-HSA-8932084 (Reactome) RUNX1 and ESR1, which are known to form a complex (Stender et al. 2010), cooperatively bind to adjacent Runx binding sites and estrogen response elements, respectively, in the second intron of the AXIN1 gene (Chimge et al. 2016).
R-HSA-8934688 (Reactome) The RUNX1:CBFB complex binds both the general and the placental promoter of the LIFR gene (Qadi et al. 2016).
R-HSA-8934690 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the LIFR gene stimulates LIFR transcription. In contrast, the oncogenic fusion protein RUNX1-ETO, the product of t(8;21) translocation in acute myeloid leukemia (AML), represses transcription of the LIFR gene (Qadi et al. 2016). LIFR encodes the receptor for the leukemia inhibitory factor (LIF), a member of the interleukin-6 (IL6) cytokine family. Signaling by LIFR is implicated in hematopoiesis, embryo implantation, placental formation and nervous system development (Nicola and Babon 2015).
R-HSA-8935960 (Reactome) The RUNX1:CBFB complex binds the promoter of the TJP1 gene, which encodes the tight junction component ZO-1 (Miao et al. 2015).
R-HSA-8935972 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the TJP1 gene stimulates transcription of the tight junction component ZO-1 (Miao et al. 2015).
R-HSA-8935980 (Reactome) The RUNX1:CBFB complex binds to the promoter of the OCLN gene, encoding Occludin, a component of the tight junction (Miao et al. 2015).
R-HSA-8935988 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the OCLN gene, stimulates transcription of the tight junction component Occludin (Miao et al. 2015).
R-HSA-8936000 (Reactome) The RUNX1:CBFB complex binds the promoter of the CLDN5 gene, encoding the component of tight junctions Claudin-5 (Miao et al. 2015).
R-HSA-8936007 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the CLDN5 gene stimulates transcription of the tight junction component Claudin-5 (Miao et al. 2015).
R-HSA-8937989 (Reactome) The RUNX1:CBFB complex associates with the polycomb repressor complex 1 (PRC1), including components of the PRC1.4 complex (the defining subunit being BMI1) and the PRC1.5 complex (the defining subunit being PCGF5). It is possible that the RUNX1:CBFB can also associate with other PRC1 complexes. PRC1 complexes are recruited to many RUNX1:CBFB target promoters and they either positively or negatively affect the transcription of RUNX1 target genes. The definitive composition of RUNX1:CBFB:PRC1 complexes at different RUNX1 target promoters has not been determined (Yu et al. 2011).
R-HSA-8938053 (Reactome) The RUNX1:CBFB complex binds the RUNX1 element in the promoter of the SERPINB13 gene (Boyapati et al. 2011).
R-HSA-8938063 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the SERPINB13 gene inhibits SERPINB13 transcription and results in higher cathepsin K activity in cells, as SERPINB13 is an inhibitor of cathepsin K and L. Cathepsin K and L are associated with proliferation and invasiveness of cancer cells (Nomura and Katunuma 2005). SERPINB13 is frequently downregulated in head and neck cancers (Boyapati et al. 2011).
R-HSA-8938108 (Reactome) SERPINB13 binds the lysosomal cystein protease cathepsin L and inhibits its catalytic activity (Jayakumar et al. 2003, Welss et al. 2003). It is uncertain whether SERPINB13 functions in the lysosomes or exclusively in the cytosol, where it would inhibit the enzymatic activity of ectopic cathepsin L (Welss et al. 2003).
R-HSA-8938121 (Reactome) SERPINB13 binds the lysosomal cystein proteinase cathepsin K (CTSK) and ihbits its catalytic activity (Jayakumar et al. 2003).
R-HSA-8938217 (Reactome) The RUNX1:CBFB complex can bind the SWI/SNF chromatin remodeling complex by directly interacting with at least three subunits of the SWI/SNF: BRG1 (SMARCA4), BAF155 (SMARCC1) and INI1 (SMARCB1). RUNX1 recruits the SWI/SNF complex to many of its target promoters, which is associated with changes in histone modifications at these promoters, but precise changes and their effect on gene expression have not been fully elucidated (Bakshi et al. 2010).
R-HSA-8938228 (Reactome) The complex of the RUNX1:CBFB heterodimer and histone acetyltransferase CREBBP (CBP) binds the promoter of the CSF2 (GM-CSF) gene, encoding Granulocyte-macrophage colony-stimulating factor (Oakford et al. 2010). RUNX1 can also recruit the SWI/SNF complex to the CSF2 gene promoter (Bakshi et al. 2010).
R-HSA-8938231 (Reactome) The RUNX1:CBFB complex binds CREBBP (CBP) histone acetyltransferase. The interaction involves the C-terminal domain of RUNX1 (Oakford et al. 2010).
R-HSA-8938251 (Reactome) Binding of the RUNX1:CBFB complex associated with the histone acetyltransferase (HAT) CREBBP (CBP) to the promoter of the CSF2 gene, encoding Granulocyte-macrophage colony stimulating factor (GM-CSF), stimulates CSF2 transcription. HAT activity of CREBBP is required for RUNX1-mediated stimulation of CSF2 expression. GM-CSF is involved in myeloid cell differentiation (Oakford et al. 2010). It has not been fully elucidated how recruitment of the SWI/SNF complex to the CSF2 promoter by RUNX1 affects CSF2 transcription (Bakshi et al. 2010).
R-HSA-8938382 (Reactome) Binding of the RUNX1:CBFB or RUNX2:CBFB complex to RUNX response elements in the promoter of the LGALS3 gene stimulates LGLAS3 transcription. The LGALS3 gene encodes Galectin-3, which is highly expressed in pituitary tumors and glioma (Vladimirova et al. 2008, Zhang et al. 2009). Galectin-3 is expressed in myeloid progenitors and its levels increase during the maturation process (Le Marer 2000).
R-HSA-8938389 (Reactome) The RUNX1:CBFB complex binds to the RUNX1 response element in the promoter of the LGALS3 gene, encoding Galectin-3 (Zhang et al. 2009).
R-HSA-8938913 (Reactome) The RUNX1:CBFB complex binds to ELF1 (MEF), a member of the ETS family of transcription factors (Mao et al. 1999, Cho et al. 2004). The interaction involves the ETS-interacting subdomain (EID) in the C-terminal portion of the RUNX1 Runt domain and a region of ELF1 that is N-terminal to its ETS domain. ELF1 does not directly interact with CBFB (Mao et al. 1999).
R-HSA-8938930 (Reactome) The RUNX1:CBFB complex binds to ELF2 (NERF), a member of the ETS family of transcription factors. The interaction involves a basic amino acid region upstream of the ETS domain of ELF2 and the runt domain of RUNX1 (Cho et al. 2004).
R-HSA-8938938 (Reactome) RUNX1 (AML1) and ELF1 (MEF) bind to adjacent RUNX1- and ETS- response elements in the promoter of the IL3 gene, encoding interleukin-3. While RUNX1 and ELF1 can independently activate the IL3 gene transcription, formation of the complex between RUNX1 and ELF1 has a synergistic effect on IL3 expression (Mao et al. 1999).
R-HSA-8938951 (Reactome) The RUNX1:CBFB complex binds to PAX5, also known as BSAP (B-cell specific activating protein). The interaction involves the runt domain of RUNX1 and the paired DNA-binding domain of PAX5 (Libermann et al. 1999).
R-HSA-8938965 (Reactome) The RUNX1 response element in the promoter of the BLK gene, encoding B cell tyrosine kinase BLK, is adjacent to the ETS response element as well as the PAX5 (BSAP) response element. The RUNX1:CBFB complex can bind to the ETS family transcription factors ELF1 or ELF2, and this complex can bind to RUNX1 and ETS response elements in the BLK promoter (Cho et al. 2004). RUNX1 can also form a complex with PAX5 (BSAP), and this complex can bind to RUNX1 and PAX5 response elements in the BLK promoter (Libermann et al. 1999). It is not known whether a larger complex, consisting of ELF1 or ELF2, the RUNX1:CBFB complex and PAX5 is formed and if ELF1/ELF2, RUNX1 and PAX5 can simultaneously reside at the BLK promoter. PAX5 is known to contain and ETS binding region and it is possible that it can, through this region, interact with ELF1 and ELF2 (Cho et al. 2004).
R-HSA-8938981 (Reactome) Binding of the RUNX1:CBFB complex associated with ELF1, an ETS family member, to the promoter of the IL3 gene, encoding interleukin-3, stimulates IL3 transcription (Mao et al. 1999).
R-HSA-8938987 (Reactome) Binding of RUNX1 in complex with ELF1, ELF2 or PAX5 (BSAP) to the BLK gene promoter stimulates BLK transcription. The BLK gene encodes B-cell specific tyrosine kinase BLK (B lymphocyte kinase) involved in B cell receptor (BCR) signaling and B cell development and differentiation (Libermann et al. 1999, Cho et al. 2004).
R-HSA-8939054 (Reactome) The RUNX1:CBFB complex binds the promoter of the PRKCB gene, encoding protein kinase C beta (Hug et al. 2004).
R-HSA-8939066 (Reactome) Binding of the RUNX1:CBFB complex to the promoter of the PRKCB gene, encoding Protein kinase C-beta, stimulates PRKCB transcription. RUNX1-mediated upregulation of PRKCB expression may contribute to apoptosis of myeloid cells (Hug et al. 2004).
R-HSA-8955748 (Reactome) Based on studies in mouse keratinocytes, RUNX1, presumably in complex with CBFB, binds the SOCS3 gene (Scheitz et al. 2012). By sequence similarity, at least one Runx binding element is conserved between human and mouse SOCS3 gene loci.
R-HSA-8955822 (Reactome) Based on studies in mouse keratinocytes, RUNX1, presumably in complex with CBFB, binds the SOCS4 gene (Scheitz et al. 2012). Runx binding elements are found in the promoter region and enhancer elements downstream of the mouse Socs4 gen. In the human SOCS4 gene, Runx binding elements can be found in the first intron and downstream of the SOCS4 gene.
R-HSA-8955885 (Reactome) RUNX1, presumably in complex with CBFB, inhibits transcription of the SOCS3 gene. As SOCS3 is an inhibitor of STAT3, RUNX1-mediated repression of SOCS3 increases STAT3 activity, which is implicated in development of epithelial cancers (Scheitz et al. 2012).
R-HSA-8955893 (Reactome) RUNX1, presumably in complex with CBFB, inhibits transcription of the SOCS4 gene. As SOCS4 is an inhibitor of STAT3, RUNX1-mediated repression of SOCS4 increases STAT3 activity, which is implicated in development of epithelial cancers (Scheitz et al. 2012).
R-HSA-8956568 (Reactome) RUNX1, in complex with CBFB, binds to the core TAL1 complex consisting of TAL1 (SCL), TCF3 (E2A) or TCF12 (HEB), LMO1 or LMO2, LDB1 and GATA1, GATA2 or GATA3 (Wilson et al. 2010, Tijssen et al. 2011, Sanda et al. 2012, Mansour et al. 2014, Hoang et al. 2016). Assembly of the RUNX1- and GATA3-containing TAL1 complex is positively regulated by the CDK7-containing CAK complex (Kwiatkowski et al. 2014).
R-HSA-8956586 (Reactome) The TAL1 complex containing GATA3, RUNX1 and CBFB binds the MYB gene enhancer element (Sanda et al. 2012, Mansour et al. 2014). Binding of the RUNX1-containing TAL1 complex to the MYB gene enhancer can be facilitated by the presence of the MYB transcription factor (Mansour et al. 2014).
R-HSA-8956608 (Reactome) Expression of the MYB gene is stimulated by binding of the RUNX1-containing TAL1 complex to the MYB gene enhancer (Sanda et al. 2012, Mansour et al. 2014). MYB transcription factor encoded by the MYB gene can bind to the RUNX1-containing TAL1 complex at the MYB gene enhancer to further facilitate MYB gene expression, thus creating a positive feedback loop (Mansour et al. 2014). In addition, somatic mutations at the TAL1 gene enhancer enable binding of the TAL1 complex containing MYB, RUNX1 and MYB-associated histone acetyltransferase CBP (CREBBP) to the TAL1 gene enhancer, creating a super-enhancer, which further amplifies the positive feedback loop involved in the MYB gene regulation (Mansour et al. 2014).
R-HSA-8956639 (Reactome) RUNX1, presumably in complex with CBFB, binds to YAP1 (Levy, Adamovich et al. 2008; Levy, Reuven and Shaul 2008). Phosphorylation of YAP1 by ABL1 in response to DNA damage prevents binding of YAP1 to RUNX1 (Levy, Adamovich et al. 2008).
R-HSA-8956649 (Reactome) The complex of RUNX1, presumably associated with CBFB, and YAP1 binds the promoter of the ITCH gene, encoding the E3 ubiquitin-protein ligase ITCH (Levy, Reuven and Shaul 2008).
R-HSA-8956652 (Reactome) Expression of the ITCH gene, encoding the E3 ubiquitin-protein ligase ITCH, is directly stimulated by binding of the complex of RUNX1 and YAP1 to the ITCH promoter (Levy, Reuven and Shaul 2008).
ITCH is an important regulator of hematopoietic stem cell (HSC) function and homeostasis. ITCH reduces the proliferation rate of HSCs by downregulating NOTCH1 (Rathinam et al. 2011).
R-HSA-8956659 (Reactome) In response to DNA damage, ABL1 phosphorylates YAP1 on tyrosine residue Y407 (corresponds to Y357 in the YAP1 splicing isoform 3, known as YAP1-1beta, which was used in the study by Levy, Adamovich et al. 2008).
R-HSA-8956676 (Reactome) YAP1, phosphorylated on tyrosine residue Y407 (Y357 in the splicing isoform 3, known as YAP1-1beta) by the protein tyrosine kinase ABL1, activated in response to DNA damage, forms a complex with TP73. ABL1-phosphorylated YAP1 can no longer bind RUNX1 (Levy, Adamovich et al. 2008; Levy, Reuven and Shaul 2008). Binding of phosphorylated YAP1 to TP73 may target TP73 to promoters of pro-apoptotic target genes instead of cell cycle arrest genes (Levy, Adamovich et al. 2008).
R-HSA-8956684 (Reactome) The E3 ubiquitin-protein ligase ITCH polyubiquitinates TP73 (p73), targeting it for degradation. In response to DNA damage, ITCH levels are downregulated, allowing TP73 to accumulate (Rossi et al. 2005). Downregulation of ITCH in response to DNA damage is the consequence of DNA damage-induced phosphorylation of YAP1 by ABL1. YAP1 phosphorylated by ABL1 is no longer able to bind to RUNX1, and the complex of RUNX1 and YAP1 is needed for transcription of the ITCH gene (Levy, Reuven and Shaul 2008).
R-HSA-8957241 (Reactome) TP73 (p73) possesses both a nuclear localization signal (NLS) and a nuclear export signal (NES) and can shuttle between the nucleus and the cytosol (Inoue et al. 2002).
R-HSA-8957265 (Reactome) ITCH-mediated polyubiquitination of TP73 (p73) targets TP73 for proteasome-mediated degradation (Rossi et al. 2005).
RSPO3 geneR-HSA-8877879 (Reactome)
RSPO3 geneR-HSA-8877884 (Reactome)
RSPO3ArrowR-HSA-8877884 (Reactome)
RUNX1:CBFB:(PRC1.4,PRC1.5)ArrowR-HSA-8937989 (Reactome)
RUNX1:CBFB:CLDN5 geneArrowR-HSA-8936000 (Reactome)
RUNX1:CBFB:CLDN5 geneArrowR-HSA-8936007 (Reactome)
RUNX1:CBFB:CREBBP:CSF2 geneArrowR-HSA-8938228 (Reactome)
RUNX1:CBFB:CREBBP:CSF2 geneArrowR-HSA-8938251 (Reactome)
RUNX1:CBFB:CREBBPArrowR-HSA-8938231 (Reactome)
RUNX1:CBFB:CREBBPR-HSA-8938228 (Reactome)
RUNX1:CBFB:ELF,ELF2,PAX5:BLK geneArrowR-HSA-8938965 (Reactome)
RUNX1:CBFB:ELF,ELF2,PAX5:BLK geneArrowR-HSA-8938987 (Reactome)
RUNX1:CBFB:ELF1,RUNX1:CBFB:ELF2,RUNX1:CBFB:PAX5R-HSA-8938965 (Reactome)
RUNX1:CBFB:ELF1:IL3 geneArrowR-HSA-8938938 (Reactome)
RUNX1:CBFB:ELF1:IL3 geneArrowR-HSA-8938981 (Reactome)
RUNX1:CBFB:ELF1ArrowR-HSA-8938913 (Reactome)
RUNX1:CBFB:ELF1R-HSA-8938938 (Reactome)
RUNX1:CBFB:ELF2ArrowR-HSA-8938930 (Reactome)
RUNX1:CBFB:EP300ArrowR-HSA-8878056 (Reactome)
RUNX1:CBFB:EP300R-HSA-8878050 (Reactome)
RUNX1:CBFB:ESR1:estrogen:AXIN1 geneArrowR-HSA-8932076 (Reactome)
RUNX1:CBFB:ESR1:estrogen:AXIN1 geneArrowR-HSA-8932084 (Reactome)
RUNX1:CBFB:ESR1:estrogen:GPAM geneArrowR-HSA-8932020 (Reactome)
RUNX1:CBFB:ESR1:estrogen:GPAM geneArrowR-HSA-8932021 (Reactome)
RUNX1:CBFB:ESR1:estrogen:KCTD6 geneArrowR-HSA-8932033 (Reactome)
RUNX1:CBFB:ESR1:estrogen:KCTD6 geneArrowR-HSA-8932037 (Reactome)
RUNX1:CBFB:ESR1:estrogenArrowR-HSA-8931981 (Reactome)
RUNX1:CBFB:ESR1:estrogenR-HSA-8932021 (Reactome)
RUNX1:CBFB:ESR1:estrogenR-HSA-8932037 (Reactome)
RUNX1:CBFB:ESR1:estrogenR-HSA-8932084 (Reactome)
RUNX1:CBFB:FOXP3:RSPO3 geneArrowR-HSA-8877879 (Reactome)
RUNX1:CBFB:FOXP3:RSPO3 geneArrowR-HSA-8877884 (Reactome)
RUNX1:CBFB:FOXP3R-HSA-8877879 (Reactome)
RUNX1:CBFB:GATA3-TAL1 core complex,MYB:RUNX1:CBFB:GATA3-TAL1 core complexR-HSA-8956586 (Reactome)
RUNX1:CBFB:GATA3-TAL1 core complex:MYB geneArrowR-HSA-8956586 (Reactome)
RUNX1:CBFB:GATA3-TAL1 core complex:MYB geneArrowR-HSA-8956608 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:H3K4me3-NucleosomeArrowR-HSA-8865498 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:H3K4me3-NucleosomeArrowR-HSA-8865505 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:NucleosomeArrowR-HSA-8865491 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:NucleosomeR-HSA-8865498 (Reactome)
RUNX1:CBFB:KMT2A:SPI1 gene:Nucleosomemim-catalysisR-HSA-8865498 (Reactome)
RUNX1:CBFB:KMT2AArrowR-HSA-8865482 (Reactome)
RUNX1:CBFB:KMT2AR-HSA-8865491 (Reactome)
RUNX1:CBFB:LGALS3 geneArrowR-HSA-8938382 (Reactome)
RUNX1:CBFB:LGALS3 geneArrowR-HSA-8938389 (Reactome)
RUNX1:CBFB:LIFR geneArrowR-HSA-8934688 (Reactome)
RUNX1:CBFB:LIFR geneArrowR-HSA-8934690 (Reactome)
RUNX1:CBFB:OCLN geneArrowR-HSA-8935980 (Reactome)
RUNX1:CBFB:OCLN geneArrowR-HSA-8935988 (Reactome)
RUNX1:CBFB:PAX5ArrowR-HSA-8938951 (Reactome)
RUNX1:CBFB:PRKCB geneArrowR-HSA-8939054 (Reactome)
RUNX1:CBFB:PRKCB geneArrowR-HSA-8939066 (Reactome)
RUNX1:CBFB:SERPINB13 geneArrowR-HSA-8938053 (Reactome)
RUNX1:CBFB:SERPINB13 geneTBarR-HSA-8938063 (Reactome)
RUNX1:CBFB:SOCS3 geneArrowR-HSA-8955748 (Reactome)
RUNX1:CBFB:SOCS3 geneTBarR-HSA-8955885 (Reactome)
RUNX1:CBFB:SOCS4 geneArrowR-HSA-8955822 (Reactome)
RUNX1:CBFB:SOCS4 geneTBarR-HSA-8955893 (Reactome)
RUNX1:CBFB:SWI/SNFArrowR-HSA-8938217 (Reactome)
RUNX1:CBFB:TAL1 core complexArrowR-HSA-8956568 (Reactome)
RUNX1:CBFB:TJP1 geneArrowR-HSA-8935960 (Reactome)
RUNX1:CBFB:TJP1 geneArrowR-HSA-8935972 (Reactome)
RUNX1:CBFB:YAP1:ITCH geneArrowR-HSA-8956649 (Reactome)
RUNX1:CBFB:YAP1:ITCH geneArrowR-HSA-8956652 (Reactome)
RUNX1:CBFB:YAP1ArrowR-HSA-8956639 (Reactome)
RUNX1:CBFB:YAP1R-HSA-8956649 (Reactome)
RUNX1:CBFBR-HSA-8865482 (Reactome)
RUNX1:CBFBR-HSA-8878054 (Reactome)
RUNX1:CBFBR-HSA-8878056 (Reactome)
RUNX1:CBFBR-HSA-8931981 (Reactome)
RUNX1:CBFBR-HSA-8934688 (Reactome)
RUNX1:CBFBR-HSA-8935960 (Reactome)
RUNX1:CBFBR-HSA-8935980 (Reactome)
RUNX1:CBFBR-HSA-8936000 (Reactome)
RUNX1:CBFBR-HSA-8937989 (Reactome)
RUNX1:CBFBR-HSA-8938053 (Reactome)
RUNX1:CBFBR-HSA-8938217 (Reactome)
RUNX1:CBFBR-HSA-8938231 (Reactome)
RUNX1:CBFBR-HSA-8938389 (Reactome)
RUNX1:CBFBR-HSA-8938913 (Reactome)
RUNX1:CBFBR-HSA-8938930 (Reactome)
RUNX1:CBFBR-HSA-8938951 (Reactome)
RUNX1:CBFBR-HSA-8939054 (Reactome)
RUNX1:CBFBR-HSA-8955748 (Reactome)
RUNX1:CBFBR-HSA-8955822 (Reactome)
RUNX1:CBFBR-HSA-8956568 (Reactome)
RUNX1:CBFBR-HSA-8956639 (Reactome)
RUNX2:CBFB:LGALS3 geneArrowR-HSA-8938382 (Reactome)
SERPINB13 geneR-HSA-8938053 (Reactome)
SERPINB13 geneR-HSA-8938063 (Reactome)
SERPINB13:cathepsin LArrowR-HSA-8938108 (Reactome)
SERPINB13ArrowR-HSA-8938063 (Reactome)
SERPINB13R-HSA-8938108 (Reactome)
SERPINB13R-HSA-8938121 (Reactome)
SOCS3 GeneR-HSA-8955748 (Reactome)
SOCS3 GeneR-HSA-8955885 (Reactome)
SOCS3ArrowR-HSA-8955885 (Reactome)
SOCS4 geneR-HSA-8955822 (Reactome)
SOCS4 geneR-HSA-8955893 (Reactome)
SOCS4ArrowR-HSA-8955893 (Reactome)
SPI1 gene:NucleosomeR-HSA-8865491 (Reactome)
SPI1 gene:NucleosomeR-HSA-8865505 (Reactome)
SPI1ArrowR-HSA-8865505 (Reactome)
SWI/SNF chromatin remodelling complexR-HSA-8938217 (Reactome)
TAL1 core complexR-HSA-8956568 (Reactome)
TJP1 geneR-HSA-8935960 (Reactome)
TJP1 geneR-HSA-8935972 (Reactome)
TJP1ArrowR-HSA-8935972 (Reactome)
TP73 TetramerArrowR-HSA-8957241 (Reactome)
TP73 TetramerR-HSA-8956676 (Reactome)
TP73 TetramerR-HSA-8956684 (Reactome)
TP73 TetramerR-HSA-8957241 (Reactome)
UbArrowR-HSA-8957265 (Reactome)
UbR-HSA-8956684 (Reactome)
YAP1R-HSA-8956639 (Reactome)
YAP1R-HSA-8956659 (Reactome)
p-S249,S273,T276-RUNX1:CBFB:p-S,T-EP300ArrowR-HSA-8878050 (Reactome)
p-S249,S273,T276-RUNX1:CBFBArrowR-HSA-8878054 (Reactome)
p-S456-ABL1TBarR-HSA-8956639 (Reactome)
p-S456-ABL1mim-catalysisR-HSA-8956659 (Reactome)
p-Y407-YAP1:TP73 tetramerArrowR-HSA-8956676 (Reactome)
p-Y407-YAP1ArrowR-HSA-8956659 (Reactome)
p-Y407-YAP1R-HSA-8956676 (Reactome)
Personal tools