General Information of Drug Off-Target (DOT) (ID: OTITK1XD)

DOT Name Runt-related transcription factor 3 (RUNX3)
Synonyms
Acute myeloid leukemia 2 protein; Core-binding factor subunit alpha-3; CBF-alpha-3; Oncogene AML-2; Polyomavirus enhancer-binding protein 2 alpha C subunit; PEA2-alpha C; PEBP2-alpha C; SL3-3 enhancer factor 1 alpha C subunit; SL3/AKV core-binding factor alpha C subunit
Gene Name RUNX3
UniProt ID
RUNX3_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
5W69
Pfam ID
PF00853 ; PF08504
Sequence
MRIPVDPSTSRRFTPPSPAFPCGGGGGKMGENSGALSAQAAVGPGGRARPEVRSMVDVLA
DHAGELVRTDSPNFLCSVLPSHWRCNKTLPVAFKVVALGDVPDGTVVTVMAGNDENYSAE
LRNASAVMKNQVARFNDLRFVGRSGRGKSFTLTITVFTNPTQVATYHRAIKVTVDGPREP
RRHRQKLEDQTKPFPDRFGDLERLRMRVTPSTPSPRGSLSTTSHFSSQPQTPIQGTSELN
PFSDPRQFDRSFPTLPTLTESRFPDPRMHYPGAMSAAFPYSATPSGTSISSLSVAGMPAT
SRFHHTYLPPPYPGAPQNQSGPFQANPSPYHLYYGTSSGSYQFSMVAGSSSGGDRSPTRM
LASCTSSAASVAAGNLMNPSLGGQSDGVEADGSHSNSPTALSTPGRMDEAVWRPY
Function
Forms the heterodimeric complex core-binding factor (CBF) with CBFB. RUNX members modulate the transcription of their target genes through recognizing the core consensus binding sequence 5'-TGTGGT-3', or very rarely, 5'-TGCGGT-3', within their regulatory regions via their runt domain, while CBFB is a non-DNA-binding regulatory subunit that allosterically enhances the sequence-specific DNA-binding capacity of RUNX. The heterodimers bind to the core site of a number of enhancers and promoters, including murine leukemia virus, polyomavirus enhancer, T-cell receptor enhancers, LCK, IL3 and GM-CSF promoters. May be involved in the control of cellular proliferation and/or differentiation. In association with ZFHX3, up-regulates CDKN1A promoter activity following TGF-beta stimulation. CBF complexes repress ZBTB7B transcription factor during cytotoxic (CD8+) T cell development. They bind to RUNX-binding sequence within the ZBTB7B locus acting as transcriptional silencer and allowing for cytotoxic T cell differentiation. CBF complexes binding to the transcriptional silencer is essential for recruitment of nuclear protein complexes that catalyze epigenetic modifications to establish epigenetic ZBTB7B silencing.
Tissue Specificity Expressed in gastric cancer tissues (at protein level).
KEGG Pathway
Th1 and Th2 cell differentiation (hsa04658 )
Epstein-Barr virus infection (hsa05169 )
Reactome Pathway
RUNX3 regulates CDKN1A transcription (R-HSA-8941855 )
RUNX3 regulates NOTCH signaling (R-HSA-8941856 )
Regulation of RUNX3 expression and activity (R-HSA-8941858 )
RUNX3 Regulates Immune Response and Cell Migration (R-HSA-8949275 )
RUNX3 regulates WNT signaling (R-HSA-8951430 )
RUNX3 regulates YAP1-mediated transcription (R-HSA-8951671 )
RUNX3 regulates RUNX1-mediated transcription (R-HSA-8951911 )
RUNX3 regulates p14-ARF (R-HSA-8951936 )
RUNX3 regulates BCL2L11 (BIM) transcription (R-HSA-8952158 )
Binding of TCF/LEF (R-HSA-4411364 )

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Regulation of Drug Effects of 1 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Doxorubicin DMVP5YE Approved Runt-related transcription factor 3 (RUNX3) decreases the export of Doxorubicin. [11]
------------------------------------------------------------------------------------
This DOT Affected the Drug Response of 3 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Etoposide DMNH3PG Approved Runt-related transcription factor 3 (RUNX3) increases the response to substance of Etoposide. [11]
Mitomycin DMH0ZJE Approved Runt-related transcription factor 3 (RUNX3) increases the response to substance of Mitomycin. [11]
Mitoxantrone DMM39BF Approved Runt-related transcription factor 3 (RUNX3) affects the response to substance of Mitoxantrone. [12]
------------------------------------------------------------------------------------
4 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate increases the methylation of Runt-related transcription factor 3 (RUNX3). [1]
Arsenic DMTL2Y1 Approved Arsenic affects the methylation of Runt-related transcription factor 3 (RUNX3). [4]
Azacitidine DMTA5OE Approved Azacitidine affects the methylation of Runt-related transcription factor 3 (RUNX3). [7]
TAK-243 DM4GKV2 Phase 1 TAK-243 increases the sumoylation of Runt-related transcription factor 3 (RUNX3). [10]
------------------------------------------------------------------------------------
10 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Tretinoin DM49DUI Approved Tretinoin increases the expression of Runt-related transcription factor 3 (RUNX3). [2]
Cisplatin DMRHGI9 Approved Cisplatin decreases the expression of Runt-related transcription factor 3 (RUNX3). [3]
Decitabine DMQL8XJ Approved Decitabine increases the expression of Runt-related transcription factor 3 (RUNX3). [5]
Panobinostat DM58WKG Approved Panobinostat increases the expression of Runt-related transcription factor 3 (RUNX3). [6]
Amphotericin B DMTAJQE Approved Amphotericin B decreases the expression of Runt-related transcription factor 3 (RUNX3). [3]
Cyclophosphamide DM4O2Z7 Approved Cyclophosphamide decreases the expression of Runt-related transcription factor 3 (RUNX3). [3]
Gentamicin DMKINJO Approved Gentamicin decreases the expression of Runt-related transcription factor 3 (RUNX3). [3]
SNDX-275 DMH7W9X Phase 3 SNDX-275 increases the expression of Runt-related transcription factor 3 (RUNX3). [6]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene increases the expression of Runt-related transcription factor 3 (RUNX3). [8]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Runt-related transcription factor 3 (RUNX3). [9]
------------------------------------------------------------------------------------
⏷ Show the Full List of 10 Drug(s)

References

1 Nuclear and Mitochondrial DNA Methylation Patterns Induced by Valproic Acid in Human Hepatocytes. Chem Res Toxicol. 2017 Oct 16;30(10):1847-1854. doi: 10.1021/acs.chemrestox.7b00171. Epub 2017 Sep 13.
2 Transcriptional and Metabolic Dissection of ATRA-Induced Granulocytic Differentiation in NB4 Acute Promyelocytic Leukemia Cells. Cells. 2020 Nov 5;9(11):2423. doi: 10.3390/cells9112423.
3 Effect of nephrotoxicants and hepatotoxicants on gene expression profile in human peripheral blood mononuclear cells. Biochem Biophys Res Commun. 2010 Oct 15;401(2):245-50. doi: 10.1016/j.bbrc.2010.09.039. Epub 2010 Sep 16.
4 Prenatal arsenic exposure and the epigenome: identifying sites of 5-methylcytosine alterations that predict functional changes in gene expression in newborn cord blood and subsequent birth outcomes. Toxicol Sci. 2015 Jan;143(1):97-106. doi: 10.1093/toxsci/kfu210. Epub 2014 Oct 10.
5 Chemical genomic screening for methylation-silenced genes in gastric cancer cell lines using 5-aza-2'-deoxycytidine treatment and oligonucleotide microarray. Cancer Sci. 2006 Jan;97(1):64-71.
6 A transcriptome-based classifier to identify developmental toxicants by stem cell testing: design, validation and optimization for histone deacetylase inhibitors. Arch Toxicol. 2015 Sep;89(9):1599-618.
7 Promoter hypermethylation of the RUNX3 gene in esophageal squamous cell carcinoma. Cancer Invest. 2007 Dec;25(8):685-90. doi: 10.1080/07357900701561131.
8 Transcriptional signature of human macrophages exposed to the environmental contaminant benzo(a)pyrene. Toxicol Sci. 2010 Apr;114(2):247-59.
9 Bromodomain-containing protein 4 (BRD4) regulates RNA polymerase II serine 2 phosphorylation in human CD4+ T cells. J Biol Chem. 2012 Dec 14;287(51):43137-55.
10 Inhibiting ubiquitination causes an accumulation of SUMOylated newly synthesized nuclear proteins at PML bodies. J Biol Chem. 2019 Oct 18;294(42):15218-15234. doi: 10.1074/jbc.RA119.009147. Epub 2019 Jul 8.
11 Tumor suppressor gene Runx3 sensitizes gastric cancer cells to chemotherapeutic drugs by downregulating Bcl-2, MDR-1 and MRP-1. Int J Cancer. 2005 Aug 10;116(1):155-60. doi: 10.1002/ijc.20919.
12 Gene expression profiling of 30 cancer cell lines predicts resistance towards 11 anticancer drugs at clinically achieved concentrations. Int J Cancer. 2006 Apr 1;118(7):1699-712. doi: 10.1002/ijc.21570.