General Information of Drug Off-Target (DOT) (ID: OTOB6J6R)

DOT Name Ribonucleoside-diphosphate reductase subunit M2 (RRM2)
Synonyms EC 1.17.4.1; Ribonucleotide reductase small chain; Ribonucleotide reductase small subunit
Gene Name RRM2
UniProt ID
RIR2_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
2UW2; 3OLJ; 3VPM; 3VPN; 3VPO
EC Number
1.17.4.1
Pfam ID
PF00268
Sequence
MLSLRVPLAPITDPQQLQLSPLKGLSLVDKENTPPALSGTRVLASKTARRIFQEPTEPKT
KAAAPGVEDEPLLRENPRRFVIFPIEYHDIWQMYKKAEASFWTAEEVDLSKDIQHWESLK
PEERYFISHVLAFFAASDGIVNENLVERFSQEVQITEARCFYGFQIAMENIHSEMYSLLI
DTYIKDPKEREFLFNAIETMPCVKKKADWALRWIGDKEATYGERVVAFAAVEGIFFSGSF
ASIFWLKKRGLMPGLTFSNELISRDEGLHCDFACLMFKHLVHKPSEERVREIIINAVRIE
QEFLTEALPVKLIGMNCTLMKQYIEFVADRLMLELGFSKVFRVENPFDFMENISLEGKTN
FFEKRVGEYQRMGVMSSPTENSFTLDADF
Function Provides the precursors necessary for DNA synthesis. Catalyzes the biosynthesis of deoxyribonucleotides from the corresponding ribonucleotides. Inhibits Wnt signaling.
KEGG Pathway
Purine metabolism (hsa00230 )
Pyrimidine metabolism (hsa00240 )
Glutathione metabolism (hsa00480 )
Drug metabolism - other enzymes (hsa00983 )
Metabolic pathways (hsa01100 )
Nucleotide metabolism (hsa01232 )
p53 sig.ling pathway (hsa04115 )
Reactome Pathway
G1/S-Specific Transcription (R-HSA-69205 )
Transcriptional Regulation by E2F6 (R-HSA-8953750 )
Interconversion of nucleotide di- and triphosphates (R-HSA-499943 )
BioCyc Pathway
MetaCyc:HS10398-MONOMER

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Drug Response of 2 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Gemcitabine DMSE3I7 Approved Ribonucleoside-diphosphate reductase subunit M2 (RRM2) affects the response to substance of Gemcitabine. [47]
Vinblastine DM5TVS3 Approved Ribonucleoside-diphosphate reductase subunit M2 (RRM2) affects the response to substance of Vinblastine. [48]
------------------------------------------------------------------------------------
2 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 Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [1]
Coumarin DM0N8ZM Investigative Coumarin affects the phosphorylation of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [44]
------------------------------------------------------------------------------------
47 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Ciclosporin DMAZJFX Approved Ciclosporin decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [2]
Tretinoin DM49DUI Approved Tretinoin decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [3]
Acetaminophen DMUIE76 Approved Acetaminophen decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [4]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [5]
Cisplatin DMRHGI9 Approved Cisplatin increases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [6]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [7]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [8]
Quercetin DM3NC4M Approved Quercetin decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [9]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [10]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [11]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [11]
Triclosan DMZUR4N Approved Triclosan decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [12]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [13]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [14]
Marinol DM70IK5 Approved Marinol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [15]
Zoledronate DMIXC7G Approved Zoledronate decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [16]
Phenobarbital DMXZOCG Approved Phenobarbital affects the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [17]
Progesterone DMUY35B Approved Progesterone decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [18]
Fluorouracil DMUM7HZ Approved Fluorouracil decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [19]
Demecolcine DMCZQGK Approved Demecolcine decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [20]
Niclosamide DMJAGXQ Approved Niclosamide decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [21]
Cannabidiol DM0659E Approved Cannabidiol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [22]
Troglitazone DM3VFPD Approved Troglitazone decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [23]
Diethylstilbestrol DMN3UXQ Approved Diethylstilbestrol increases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [24]
Azathioprine DMMZSXQ Approved Azathioprine decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [25]
Ethanol DMDRQZU Approved Ethanol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [26]
Cytarabine DMZD5QR Approved Cytarabine increases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [27]
Piroxicam DMTK234 Approved Piroxicam decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [28]
Dasatinib DMJV2EK Approved Dasatinib decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [29]
Lucanthone DMZLBUO Approved Lucanthone decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [30]
Methamphetamine DMPM4SK Approved Methamphetamine decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [31]
Palbociclib DMD7L94 Approved Palbociclib decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [32]
Bicalutamide DMZMSPF Approved Bicalutamide decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [33]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [34]
Tamibarotene DM3G74J Phase 3 Tamibarotene affects the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [3]
Genistein DM0JETC Phase 2/3 Genistein decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [35]
GSK2110183 DMZHB37 Phase 2 GSK2110183 decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [36]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [37]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [38]
TAK-114 DMTXE19 Phase 1 TAK-114 decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [39]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [40]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [41]
Celastrol DMWQIJX Preclinical Celastrol decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [42]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [43]
Coumestrol DM40TBU Investigative Coumestrol increases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [45]
Dibutyl phthalate DMEDGKO Investigative Dibutyl phthalate increases the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [39]
Aminohippuric acid DMUN54G Investigative Aminohippuric acid affects the expression of Ribonucleoside-diphosphate reductase subunit M2 (RRM2). [46]
------------------------------------------------------------------------------------
⏷ Show the Full List of 47 Drug(s)

References

1 Integrative omics data analyses of repeated dose toxicity of valproic acid in vitro reveal new mechanisms of steatosis induction. Toxicology. 2018 Jan 15;393:160-170.
2 Inter-laboratory comparison of human renal proximal tubule (HK-2) transcriptome alterations due to Cyclosporine A exposure and medium exhaustion. Toxicol In Vitro. 2009 Apr;23(3):486-99.
3 Differential modulation of PI3-kinase/Akt pathway during all-trans retinoic acid- and Am80-induced HL-60 cell differentiation revealed by DNA microarray analysis. Biochem Pharmacol. 2004 Dec 1;68(11):2177-86.
4 Multiple microRNAs function as self-protective modules in acetaminophen-induced hepatotoxicity in humans. Arch Toxicol. 2018 Feb;92(2):845-858.
5 Bringing in vitro analysis closer to in vivo: studying doxorubicin toxicity and associated mechanisms in 3D human microtissues with PBPK-based dose modelling. Toxicol Lett. 2018 Sep 15;294:184-192.
6 Low doses of cisplatin induce gene alterations, cell cycle arrest, and apoptosis in human promyelocytic leukemia cells. Biomark Insights. 2016 Aug 24;11:113-21.
7 Comparison of HepG2 and HepaRG by whole-genome gene expression analysis for the purpose of chemical hazard identification. Toxicol Sci. 2010 May;115(1):66-79.
8 Quantitative proteomics reveals a broad-spectrum antiviral property of ivermectin, benefiting for COVID-19 treatment. J Cell Physiol. 2021 Apr;236(4):2959-2975. doi: 10.1002/jcp.30055. Epub 2020 Sep 22.
9 Comparison of phenotypic and transcriptomic effects of false-positive genotoxins, true genotoxins and non-genotoxins using HepG2 cells. Mutagenesis. 2011 Sep;26(5):593-604.
10 Identification of transcriptome signatures and biomarkers specific for potential developmental toxicants inhibiting human neural crest cell migration. Arch Toxicol. 2016 Jan;90(1):159-80.
11 Effects of 1alpha,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011 May 18;10:58.
12 Transcriptome and DNA methylome dynamics during triclosan-induced cardiomyocyte differentiation toxicity. Stem Cells Int. 2018 Oct 29;2018:8608327.
13 Gene Expression Regulation and Pathway Analysis After Valproic Acid and Carbamazepine Exposure in a Human Embryonic Stem Cell-Based Neurodevelopmental Toxicity Assay. Toxicol Sci. 2015 Aug;146(2):311-20. doi: 10.1093/toxsci/kfv094. Epub 2015 May 15.
14 Methotrexate modulates folate phenotype and inflammatory profile in EA.hy 926 cells. Eur J Pharmacol. 2014 Jun 5;732:60-7.
15 JunD is involved in the antiproliferative effect of Delta9-tetrahydrocannabinol on human breast cancer cells. Oncogene. 2008 Aug 28;27(37):5033-44.
16 Interleukin-19 as a translational indicator of renal injury. Arch Toxicol. 2015 Jan;89(1):101-6.
17 Reproducible chemical-induced changes in gene expression profiles in human hepatoma HepaRG cells under various experimental conditions. Toxicol In Vitro. 2009 Apr;23(3):466-75. doi: 10.1016/j.tiv.2008.12.018. Epub 2008 Dec 30.
18 Effects of progesterone treatment on expression of genes involved in uterine quiescence. Reprod Sci. 2011 Aug;18(8):781-97.
19 Transcriptional profiling of MCF7 breast cancer cells in response to 5-Fluorouracil: relationship with cell cycle changes and apoptosis, and identification of novel targets of p53. Int J Cancer. 2006 Sep 1;119(5):1164-75.
20 Characterization of formaldehyde's genotoxic mode of action by gene expression analysis in TK6 cells. Arch Toxicol. 2013 Nov;87(11):1999-2012.
21 Computational discovery of niclosamide ethanolamine, a repurposed drug candidate that reduces growth of hepatocellular carcinoma cells initro and in mice by inhibiting cell division cycle 37 signaling. Gastroenterology. 2017 Jun;152(8):2022-2036.
22 Cannabidiol-induced transcriptomic changes and cellular senescence in human Sertoli cells. Toxicol Sci. 2023 Feb 17;191(2):227-238. doi: 10.1093/toxsci/kfac131.
23 Effects of ciglitazone and troglitazone on the proliferation of human stomach cancer cells. World J Gastroenterol. 2009 Jan 21;15(3):310-20.
24 Identification of biomarkers and outcomes of endocrine disruption in human ovarian cortex using In Vitro Models. Toxicology. 2023 Feb;485:153425. doi: 10.1016/j.tox.2023.153425. Epub 2023 Jan 5.
25 A transcriptomics-based in vitro assay for predicting chemical genotoxicity in vivo. Carcinogenesis. 2012 Jul;33(7):1421-9.
26 Chronic ethanol exposure increases goosecoid (GSC) expression in human embryonic carcinoma cell differentiation. J Appl Toxicol. 2014 Jan;34(1):66-75.
27 The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia. 2009 Jun;23(6):1019-28.
28 Apoptosis induced by piroxicam plus cisplatin combined treatment is triggered by p21 in mesothelioma. PLoS One. 2011;6(8):e23569.
29 Dasatinib reverses cancer-associated fibroblasts (CAFs) from primary lung carcinomas to a phenotype comparable to that of normal fibroblasts. Mol Cancer. 2010 Jun 27;9:168.
30 Lucanthone is a novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem. 2011 Feb 25;286(8):6602-13.
31 Methamphetamine alters the normal progression by inducing cell cycle arrest in astrocytes. PLoS One. 2014 Oct 7;9(10):e109603.
32 Cdk4/6 inhibition induces epithelial-mesenchymal transition and enhances invasiveness in pancreatic cancer cells. Mol Cancer Ther. 2012 Oct;11(10):2138-48. doi: 10.1158/1535-7163.MCT-12-0562. Epub 2012 Aug 6.
33 Microarray analysis of bicalutamide action on telomerase activity, p53 pathway and viability of prostate carcinoma cell lines. J Pharm Pharmacol. 2005 Jan;57(1):83-92.
34 Resveratrol-induced gene expression profiles in human prostate cancer cells. Cancer Epidemiol Biomarkers Prev. 2005 Mar;14(3):596-604. doi: 10.1158/1055-9965.EPI-04-0398.
35 A high concentration of genistein down-regulates activin A, Smad3 and other TGF-beta pathway genes in human uterine leiomyoma cells. Exp Mol Med. 2012 Apr 30;44(4):281-92.
36 Novel ATP-competitive Akt inhibitor afuresertib suppresses the proliferation of malignant pleural mesothelioma cells. Cancer Med. 2017 Nov;6(11):2646-2659. doi: 10.1002/cam4.1179. Epub 2017 Sep 27.
37 Transcriptional signature of human macrophages exposed to the environmental contaminant benzo(a)pyrene. Toxicol Sci. 2010 Apr;114(2):247-59.
38 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
39 In silico, in vitro and in vivo studies: Dibutyl phthalate promotes prostate cancer cell proliferation by activating Forkhead Box M1 and remission after Natura- pretreatment. Toxicology. 2023 Apr;488:153465. doi: 10.1016/j.tox.2023.153465. Epub 2023 Feb 23.
40 Cell-based two-dimensional morphological assessment system to predict cancer drug-induced cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol. 2019 Nov 15;383:114761. doi: 10.1016/j.taap.2019.114761. Epub 2019 Sep 15.
41 Endoplasmic reticulum stress impairs insulin signaling through mitochondrial damage in SH-SY5Y cells. Neurosignals. 2012;20(4):265-80.
42 Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell. 2006 Oct;10(4):321-30.
43 Bisphenol A induces DSB-ATM-p53 signaling leading to cell cycle arrest, senescence, autophagy, stress response, and estrogen release in human fetal lung fibroblasts. Arch Toxicol. 2018 Apr;92(4):1453-1469.
44 Quantitative phosphoproteomics reveal cellular responses from caffeine, coumarin and quercetin in treated HepG2 cells. Toxicol Appl Pharmacol. 2022 Aug 15;449:116110. doi: 10.1016/j.taap.2022.116110. Epub 2022 Jun 7.
45 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
46 Identification of molecular signatures predicting the carcinogenicity of polycyclic aromatic hydrocarbons (PAHs). Toxicol Lett. 2012 Jul 7;212(1):18-28. doi: 10.1016/j.toxlet.2012.04.013. Epub 2012 May 1.
47 14-3-3 regulation of and interaction with YAP1 in acquired gemcitabine resistance via promoting ribonucleotide reductase expression. Oncotarget. 2016 Apr 5;7(14):17726-36. doi: 10.18632/oncotarget.7394.
48 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.