General Information of Drug Off-Target (DOT) (ID: OT0FSRW7)

DOT Name Regulator of G-protein signaling 2 (RGS2)
Synonyms RGS2; Cell growth-inhibiting gene 31 protein; G0/G1 switch regulatory protein 8
Gene Name RGS2
UniProt ID
RGS2_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
2AF0; 2V4Z; 4EKC; 4EKD
Pfam ID
PF00615
Sequence
MQSAMFLAVQHDCRPMDKSAGSGHKSEEKREKMKRTLLKDWKTRLSYFLQNSSTPGKPKT
GKKSKQQAFIKPSPEEAQLWSEAFDELLASKYGLAAFRAFLKSEFCEENIEFWLACEDFK
KTKSPQKLSSKARKIYTDFIEKEAPKEINIDFQTKTLIAQNIQEATSGCFTTAQKRVYSL
MENNSYPRFLESEFYQDLCKKPQITTEPHAT
Function
Regulates G protein-coupled receptor signaling cascades. Inhibits signal transduction by increasing the GTPase activity of G protein alpha subunits, thereby driving them into their inactive GDP-bound form. It is involved in the negative regulation of the angiotensin-activated signaling pathway. Plays a role in the regulation of blood pressure in response to signaling via G protein-coupled receptors and GNAQ. Plays a role in regulating the constriction and relaxation of vascular smooth muscle. Binds EIF2B5 and blocks its activity, thereby inhibiting the translation of mRNA into protein.
Tissue Specificity Expressed in acute myelogenous leukemia (AML) and in acute lymphoblastic leukemia (ALL).
KEGG Pathway
cGMP-PKG sig.ling pathway (hsa04022 )
Olfactory transduction (hsa04740 )
Oxytocin sig.ling pathway (hsa04921 )
Reactome Pathway
G alpha (q) signalling events (R-HSA-416476 )

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 1 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Mitoxantrone DMM39BF Approved Regulator of G-protein signaling 2 (RGS2) affects the response to substance of Mitoxantrone. [47]
------------------------------------------------------------------------------------
2 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate decreases the methylation of Regulator of G-protein signaling 2 (RGS2). [1]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene increases the methylation of Regulator of G-protein signaling 2 (RGS2). [34]
------------------------------------------------------------------------------------
45 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 Regulator of G-protein signaling 2 (RGS2). [2]
Tretinoin DM49DUI Approved Tretinoin increases the expression of Regulator of G-protein signaling 2 (RGS2). [3]
Acetaminophen DMUIE76 Approved Acetaminophen increases the expression of Regulator of G-protein signaling 2 (RGS2). [4]
Doxorubicin DMVP5YE Approved Doxorubicin increases the expression of Regulator of G-protein signaling 2 (RGS2). [5]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate increases the expression of Regulator of G-protein signaling 2 (RGS2). [6]
Cisplatin DMRHGI9 Approved Cisplatin increases the expression of Regulator of G-protein signaling 2 (RGS2). [7]
Estradiol DMUNTE3 Approved Estradiol increases the expression of Regulator of G-protein signaling 2 (RGS2). [8]
Quercetin DM3NC4M Approved Quercetin increases the expression of Regulator of G-protein signaling 2 (RGS2). [9]
Temozolomide DMKECZD Approved Temozolomide increases the expression of Regulator of G-protein signaling 2 (RGS2). [10]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Regulator of G-protein signaling 2 (RGS2). [11]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide affects the expression of Regulator of G-protein signaling 2 (RGS2). [12]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Regulator of G-protein signaling 2 (RGS2). [13]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Regulator of G-protein signaling 2 (RGS2). [14]
Decitabine DMQL8XJ Approved Decitabine increases the expression of Regulator of G-protein signaling 2 (RGS2). [15]
Marinol DM70IK5 Approved Marinol decreases the expression of Regulator of G-protein signaling 2 (RGS2). [16]
Progesterone DMUY35B Approved Progesterone increases the expression of Regulator of G-protein signaling 2 (RGS2). [17]
Fluorouracil DMUM7HZ Approved Fluorouracil decreases the expression of Regulator of G-protein signaling 2 (RGS2). [18]
Amphotericin B DMTAJQE Approved Amphotericin B decreases the expression of Regulator of G-protein signaling 2 (RGS2). [19]
Simvastatin DM30SGU Approved Simvastatin increases the expression of Regulator of G-protein signaling 2 (RGS2). [20]
Gemcitabine DMSE3I7 Approved Gemcitabine increases the expression of Regulator of G-protein signaling 2 (RGS2). [21]
Melphalan DMOLNHF Approved Melphalan increases the expression of Regulator of G-protein signaling 2 (RGS2). [22]
Acocantherin DM7JT24 Approved Acocantherin decreases the expression of Regulator of G-protein signaling 2 (RGS2). [23]
Lindane DMB8CNL Approved Lindane decreases the expression of Regulator of G-protein signaling 2 (RGS2). [24]
Enzalutamide DMGL19D Approved Enzalutamide decreases the expression of Regulator of G-protein signaling 2 (RGS2). [25]
Urethane DM7NSI0 Phase 4 Urethane increases the expression of Regulator of G-protein signaling 2 (RGS2). [26]
Dihydrotestosterone DM3S8XC Phase 4 Dihydrotestosterone increases the expression of Regulator of G-protein signaling 2 (RGS2). [25]
Seocalcitol DMKL9QO Phase 3 Seocalcitol increases the expression of Regulator of G-protein signaling 2 (RGS2). [27]
Rigosertib DMOSTXF Phase 3 Rigosertib increases the expression of Regulator of G-protein signaling 2 (RGS2). [28]
Coprexa DMA0WEK Phase 3 Coprexa increases the expression of Regulator of G-protein signaling 2 (RGS2). [29]
Tocopherol DMBIJZ6 Phase 2 Tocopherol increases the expression of Regulator of G-protein signaling 2 (RGS2). [30]
DNCB DMDTVYC Phase 2 DNCB increases the expression of Regulator of G-protein signaling 2 (RGS2). [31]
Lithium DMZ3OU6 Phase 2 Lithium increases the expression of Regulator of G-protein signaling 2 (RGS2). [33]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Regulator of G-protein signaling 2 (RGS2). [35]
Torcetrapib DMDHYM7 Discontinued in Phase 2 Torcetrapib increases the expression of Regulator of G-protein signaling 2 (RGS2). [36]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN increases the expression of Regulator of G-protein signaling 2 (RGS2). [37]
Scriptaid DM9JZ21 Preclinical Scriptaid affects the expression of Regulator of G-protein signaling 2 (RGS2). [38]
Bisphenol A DM2ZLD7 Investigative Bisphenol A increases the expression of Regulator of G-protein signaling 2 (RGS2). [39]
Trichostatin A DM9C8NX Investigative Trichostatin A increases the expression of Regulator of G-protein signaling 2 (RGS2). [40]
Formaldehyde DM7Q6M0 Investigative Formaldehyde increases the expression of Regulator of G-protein signaling 2 (RGS2). [41]
Sulforaphane DMQY3L0 Investigative Sulforaphane decreases the expression of Regulator of G-protein signaling 2 (RGS2). [42]
chloropicrin DMSGBQA Investigative chloropicrin increases the expression of Regulator of G-protein signaling 2 (RGS2). [43]
3R14S-OCHRATOXIN A DM2KEW6 Investigative 3R14S-OCHRATOXIN A decreases the expression of Regulator of G-protein signaling 2 (RGS2). [24]
methyl p-hydroxybenzoate DMO58UW Investigative methyl p-hydroxybenzoate increases the expression of Regulator of G-protein signaling 2 (RGS2). [44]
Paraquat DMR8O3X Investigative Paraquat increases the expression of Regulator of G-protein signaling 2 (RGS2). [45]
Manganese DMKT129 Investigative Manganese increases the expression of Regulator of G-protein signaling 2 (RGS2). [46]
------------------------------------------------------------------------------------
⏷ Show the Full List of 45 Drug(s)
1 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
ANW-32821 DMMJOZD Phase 2 ANW-32821 increases the degradation of Regulator of G-protein signaling 2 (RGS2). [32]
------------------------------------------------------------------------------------

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 Integrative "-Omics" analysis in primary human hepatocytes unravels persistent mechanisms of cyclosporine A-induced cholestasis. Chem Res Toxicol. 2016 Dec 19;29(12):2164-2174.
3 Phenotypic characterization of retinoic acid differentiated SH-SY5Y cells by transcriptional profiling. PLoS One. 2013 May 28;8(5):e63862.
4 Gene expression analysis of precision-cut human liver slices indicates stable expression of ADME-Tox related genes. Toxicol Appl Pharmacol. 2011 May 15;253(1):57-69.
5 RNA sequence analysis of inducible pluripotent stem cell-derived cardiomyocytes reveals altered expression of DNA damage and cell cycle genes in response to doxorubicin. Toxicol Appl Pharmacol. 2018 Oct 1;356:44-53.
6 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
7 Real-time monitoring of cisplatin-induced cell death. PLoS One. 2011;6(5):e19714. doi: 10.1371/journal.pone.0019714. Epub 2011 May 16.
8 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.
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 Temozolomide induces activation of Wnt/-catenin signaling in glioma cells via PI3K/Akt pathway: implications in glioma therapy. Cell Biol Toxicol. 2020 Jun;36(3):273-278. doi: 10.1007/s10565-019-09502-7. Epub 2019 Nov 22.
11 Gene expression profile changes in NB4 cells induced by arsenic trioxide. Acta Pharmacol Sin. 2003 Jul;24(7):646-50.
12 Global gene expression analysis reveals differences in cellular responses to hydroxyl- and superoxide anion radical-induced oxidative stress in caco-2 cells. Toxicol Sci. 2010 Apr;114(2):193-203. doi: 10.1093/toxsci/kfp309. Epub 2009 Dec 31.
13 The exosome-like vesicles derived from androgen exposed-prostate stromal cells promote epithelial cells proliferation and epithelial-mesenchymal transition. Toxicol Appl Pharmacol. 2021 Jan 15;411:115384. doi: 10.1016/j.taap.2020.115384. Epub 2020 Dec 25.
14 The contribution of methotrexate exposure and host factors on transcriptional variance in human liver. Toxicol Sci. 2007 Jun;97(2):582-94.
15 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.
16 THC exposure of human iPSC neurons impacts genes associated with neuropsychiatric disorders. Transl Psychiatry. 2018 Apr 25;8(1):89. doi: 10.1038/s41398-018-0137-3.
17 Progesterone regulation of implantation-related genes: new insights into the role of oestrogen. Cell Mol Life Sci. 2007 Apr;64(7-8):1009-32.
18 Pharmacogenomic identification of novel determinants of response to chemotherapy in colon cancer. Cancer Res. 2006 Mar 1;66(5):2765-77.
19 Differential expression of microRNAs and their predicted targets in renal cells exposed to amphotericin B and its complex with copper (II) ions. Toxicol Mech Methods. 2017 Sep;27(7):537-543. doi: 10.1080/15376516.2017.1333554. Epub 2017 Jun 8.
20 Simvastatin inactivates beta1-integrin and extracellular signal-related kinase signaling and inhibits cell proliferation in head and neck squamous cell carcinoma cells. Cancer Sci. 2007 Jun;98(6):890-9.
21 Gene expression profiling of breast cancer cells in response to gemcitabine: NF-kappaB pathway activation as a potential mechanism of resistance. Breast Cancer Res Treat. 2007 Apr;102(2):157-72.
22 Bone marrow osteoblast damage by chemotherapeutic agents. PLoS One. 2012;7(2):e30758. doi: 10.1371/journal.pone.0030758. Epub 2012 Feb 17.
23 Ouabain impairs cell migration, and invasion and alters gene expression of human osteosarcoma U-2 OS cells. Environ Toxicol. 2017 Nov;32(11):2400-2413. doi: 10.1002/tox.22453. Epub 2017 Aug 10.
24 Transcriptome-based functional classifiers for direct immunotoxicity. Arch Toxicol. 2014 Mar;88(3):673-89.
25 LSD1 activates a lethal prostate cancer gene network independently of its demethylase function. Proc Natl Acad Sci U S A. 2018 May 1;115(18):E4179-E4188.
26 Ethyl carbamate induces cell death through its effects on multiple metabolic pathways. Chem Biol Interact. 2017 Nov 1;277:21-32.
27 Expression profiling in squamous carcinoma cells reveals pleiotropic effects of vitamin D3 analog EB1089 signaling on cell proliferation, differentiation, and immune system regulation. Mol Endocrinol. 2002 Jun;16(6):1243-56.
28 ON 01910.Na is selectively cytotoxic for chronic lymphocytic leukemia cells through a dual mechanism of action involving PI3K/AKT inhibition and induction of oxidative stress. Clin Cancer Res. 2012 Apr 1;18(7):1979-91. doi: 10.1158/1078-0432.CCR-11-2113. Epub 2012 Feb 20.
29 Copper deprivation enhances the chemosensitivity of pancreatic cancer to rapamycin by mTORC1/2 inhibition. Chem Biol Interact. 2023 Sep 1;382:110546. doi: 10.1016/j.cbi.2023.110546. Epub 2023 Jun 7.
30 Selenium and vitamin E: cell type- and intervention-specific tissue effects in prostate cancer. J Natl Cancer Inst. 2009 Mar 4;101(5):306-20.
31 Upregulation of genes orchestrating keratinocyte differentiation, including the novel marker gene ID2, by contact sensitizers in human bulge-derived keratinocytes. J Biochem Mol Toxicol. 2010 Jan-Feb;24(1):10-20.
32 Cholesterol increases protein levels of the E3 ligase MARCH6 and thereby stimulates protein degradation. J Biol Chem. 2019 Feb 15;294(7):2436-2448. doi: 10.1074/jbc.RA118.005069. Epub 2018 Dec 13.
33 A genetic network model of cellular responses to lithium treatment and cocaine abuse in bipolar disorder. BMC Syst Biol. 2010 Nov 19;4:158. doi: 10.1186/1752-0509-4-158.
34 Air pollution and DNA methylation alterations in lung cancer: A systematic and comparative study. Oncotarget. 2017 Jan 3;8(1):1369-1391. doi: 10.18632/oncotarget.13622.
35 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.
36 Clarifying off-target effects for torcetrapib using network pharmacology and reverse docking approach. BMC Syst Biol. 2012 Dec 10;6:152.
37 Chemical stresses fail to mimic the unfolded protein response resulting from luminal load with unfolded polypeptides. J Biol Chem. 2018 Apr 13;293(15):5600-5612.
38 Histone deacetylase inhibitor scriptaid induces cell cycle arrest and epigenetic change in colon cancer cells. Int J Oncol. 2008 Oct;33(4):767-76.
39 Bisphenol A and bisphenol S induce distinct transcriptional profiles in differentiating human primary preadipocytes. PLoS One. 2016 Sep 29;11(9):e0163318.
40 From transient transcriptome responses to disturbed neurodevelopment: role of histone acetylation and methylation as epigenetic switch between reversible and irreversible drug effects. Arch Toxicol. 2014 Jul;88(7):1451-68.
41 Gene expression changes in primary human nasal epithelial cells exposed to formaldehyde in vitro. Toxicol Lett. 2010 Oct 5;198(2):289-95.
42 Transcriptome and DNA methylation changes modulated by sulforaphane induce cell cycle arrest, apoptosis, DNA damage, and suppression of proliferation in human liver cancer cells. Food Chem Toxicol. 2020 Feb;136:111047. doi: 10.1016/j.fct.2019.111047. Epub 2019 Dec 12.
43 Transcriptomic analysis of human primary bronchial epithelial cells after chloropicrin treatment. Chem Res Toxicol. 2015 Oct 19;28(10):1926-35.
44 Transcriptome dynamics of alternative splicing events revealed early phase of apoptosis induced by methylparaben in H1299 human lung carcinoma cells. Arch Toxicol. 2020 Jan;94(1):127-140. doi: 10.1007/s00204-019-02629-w. Epub 2019 Nov 20.
45 CD34+ derived macrophage and dendritic cells display differential responses to paraquat. Toxicol In Vitro. 2021 Sep;75:105198. doi: 10.1016/j.tiv.2021.105198. Epub 2021 Jun 9.
46 Gene expression profiling of human primary astrocytes exposed to manganese chloride indicates selective effects on several functions of the cells. Neurotoxicology. 2007 May;28(3):478-89.
47 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.