General Information of Drug Off-Target (DOT) (ID: OTY183WJ)

DOT Name Suppressor of cytokine signaling 3 (SOCS3)
Synonyms SOCS-3; Cytokine-inducible SH2 protein 3; CIS-3; STAT-induced STAT inhibitor 3; SSI-3
Gene Name SOCS3
UniProt ID
SOCS3_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
Pfam ID
PF00017
Sequence
MVTHSKFPAAGMSRPLDTSLRLKTFSSKSEYQLVVNAVRKLQESGFYWSAVTGGEANLLL
SAEPAGTFLIRDSSDQRHFFTLSVKTQSGTKNLRIQCEGGSFSLQSDPRSTQPVPRFDCV
LKLVHHYMPPPGAPSFPSPPTEPSSEVPEQPSAQPLPGSPPRRAYYIYSGGEKIPLVLSR
PLSSNVATLQHLCRKTVNGHLDSYEKVTQLPGPIREFLDQYDAPL
Function
SOCS family proteins form part of a classical negative feedback system that regulates cytokine signal transduction. SOCS3 is involved in negative regulation of cytokines that signal through the JAK/STAT pathway. Inhibits cytokine signal transduction by binding to tyrosine kinase receptors including IL6ST/gp130, LIF, erythropoietin, insulin, IL12, GCSF and leptin receptors. Binding to JAK2 inhibits its kinase activity and regulates IL6 signaling. Suppresses fetal liver erythropoiesis. Regulates onset and maintenance of allergic responses mediated by T-helper type 2 cells. Probable substrate recognition component of a SCF-like ECS (Elongin BC-CUL2/5-SOCS-box protein) E3 ubiquitin-protein ligase complex which mediates the ubiquitination and subsequent proteasomal degradation of target proteins.
Tissue Specificity Widely expressed with high expression in heart, placenta, skeletal muscle, peripheral blood leukocytes, fetal and adult lung, and fetal liver and kidney. Lower levels in thymus.
KEGG Pathway
Ubiquitin mediated proteolysis (hsa04120 )
Osteoclast differentiation (hsa04380 )
JAK-STAT sig.ling pathway (hsa04630 )
TNF sig.ling pathway (hsa04668 )
Insulin sig.ling pathway (hsa04910 )
Prolactin sig.ling pathway (hsa04917 )
Adipocytokine sig.ling pathway (hsa04920 )
Type II diabetes mellitus (hsa04930 )
Insulin resistance (hsa04931 )
Non-alcoholic fatty liver disease (hsa04932 )
Growth hormone synthesis, secretion and action (hsa04935 )
Hepatitis C (hsa05160 )
Influenza A (hsa05164 )
Herpes simplex virus 1 infection (hsa05168 )
Reactome Pathway
Signaling by Leptin (R-HSA-2586552 )
Interleukin-4 and Interleukin-13 signaling (R-HSA-6785807 )
Interferon gamma signaling (R-HSA-877300 )
Regulation of IFNG signaling (R-HSA-877312 )
PTK6 Activates STAT3 (R-HSA-8849474 )
RUNX1 regulates transcription of genes involved in differentiation of keratinocytes (R-HSA-8939242 )
Neddylation (R-HSA-8951664 )
Interferon alpha/beta signaling (R-HSA-909733 )
Regulation of IFNA/IFNB signaling (R-HSA-912694 )
Signaling by CSF3 (G-CSF) (R-HSA-9674555 )
Inactivation of CSF3 (G-CSF) signaling (R-HSA-9705462 )
Growth hormone receptor signaling (R-HSA-982772 )
Antigen processing (R-HSA-983168 )
Interleukin-6 signaling (R-HSA-1059683 )

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
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 Suppressor of cytokine signaling 3 (SOCS3). [1]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene increases the methylation of Suppressor of cytokine signaling 3 (SOCS3). [26]
------------------------------------------------------------------------------------
45 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 Suppressor of cytokine signaling 3 (SOCS3). [2]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [3]
Cisplatin DMRHGI9 Approved Cisplatin increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [4]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [5]
Arsenic DMTL2Y1 Approved Arsenic increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [6]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [7]
Vorinostat DMWMPD4 Approved Vorinostat decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [8]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [9]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [10]
Progesterone DMUY35B Approved Progesterone increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [11]
Panobinostat DM58WKG Approved Panobinostat decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [8]
Fulvestrant DM0YZC6 Approved Fulvestrant decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [12]
Demecolcine DMCZQGK Approved Demecolcine increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [13]
Niclosamide DMJAGXQ Approved Niclosamide increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [14]
Bortezomib DMNO38U Approved Bortezomib increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [15]
Malathion DMXZ84M Approved Malathion decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [16]
Azacitidine DMTA5OE Approved Azacitidine affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [17]
Simvastatin DM30SGU Approved Simvastatin increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [18]
Ibuprofen DM8VCBE Approved Ibuprofen affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [19]
Diphenylpyraline DMW4X37 Approved Diphenylpyraline decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [20]
Rofecoxib DM3P5DA Approved Rofecoxib increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [19]
Tofacitinib DMBS370 Approved Tofacitinib decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [21]
Pyrimethamine DM5X7VY Approved Pyrimethamine decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [22]
Fedratinib DM4ZBK6 Approved Fedratinib decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [22]
Isoflavone DM7U58J Phase 4 Isoflavone increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
SNDX-275 DMH7W9X Phase 3 SNDX-275 decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [8]
Resveratrol DM3RWXL Phase 3 Resveratrol increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [24]
Genistein DM0JETC Phase 2/3 Genistein increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [25]
Belinostat DM6OC53 Phase 2 Belinostat decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [8]
phorbol 12-myristate 13-acetate DMJWD62 Phase 2 phorbol 12-myristate 13-acetate increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [27]
PF-3758309 DM36PKZ Phase 1 PF-3758309 decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [28]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [29]
Trichostatin A DM9C8NX Investigative Trichostatin A decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [30]
Formaldehyde DM7Q6M0 Investigative Formaldehyde increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [31]
Milchsaure DM462BT Investigative Milchsaure increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [32]
Hexadecanoic acid DMWUXDZ Investigative Hexadecanoic acid affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [33]
Phencyclidine DMQBEYX Investigative Phencyclidine increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [34]
Forskolin DM6ITNG Investigative Forskolin increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
Apigenin DMI3491 Investigative Apigenin increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
1,6-hexamethylene diisocyanate DMLB3RT Investigative 1,6-hexamethylene diisocyanate affects the expression of Suppressor of cytokine signaling 3 (SOCS3). [35]
Flavone DMEQH6J Investigative Flavone increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
Fenthion DMKEG49 Investigative Fenthion increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [36]
ROLIPRAM DMJ03UM Investigative ROLIPRAM increases the expression of Suppressor of cytokine signaling 3 (SOCS3). [23]
4-[1-(4-hydroxyphenyl)-2-phenylbut-1-enyl]phenol DMTMLXU Investigative 4-[1-(4-hydroxyphenyl)-2-phenylbut-1-enyl]phenol decreases the expression of Suppressor of cytokine signaling 3 (SOCS3). [5]
------------------------------------------------------------------------------------
⏷ 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
MG-132 DMKA2YS Preclinical MG-132 decreases the degradation of Suppressor of cytokine signaling 3 (SOCS3). [23]
------------------------------------------------------------------------------------

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 Retinoic acid and its 4-oxo metabolites are functionally active in human skin cells in vitro. J Invest Dermatol. 2005 Jul;125(1):143-53.
3 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
4 Systematic transcriptome-based comparison of cellular adaptive stress response activation networks in hepatic stem cell-derived progeny and primary human hepatocytes. Toxicol In Vitro. 2021 Jun;73:105107. doi: 10.1016/j.tiv.2021.105107. Epub 2021 Feb 3.
5 Molecular mechanism of action of bisphenol and bisphenol A mediated by oestrogen receptor alpha in growth and apoptosis of breast cancer cells. Br J Pharmacol. 2013 May;169(1):167-78.
6 Activation of inflammation/NF-kappaB signaling in infants born to arsenic-exposed mothers. PLoS Genet. 2007 Nov;3(11):e207.
7 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.
8 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.
9 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.
10 Global molecular effects of tocilizumab therapy in rheumatoid arthritis synovium. Arthritis Rheumatol. 2014 Jan;66(1):15-23.
11 Progesterone promotes differentiation of human cord blood fetal T cells into T regulatory cells but suppresses their differentiation into Th17 cells. J Immunol. 2011 Aug 15;187(4):1778-87. doi: 10.4049/jimmunol.1003919. Epub 2011 Jul 18.
12 Arsenite and cadmium promote the development of mammary tumors. Carcinogenesis. 2020 Jul 14;41(7):1005-1014. doi: 10.1093/carcin/bgz176.
13 Characterization of formaldehyde's genotoxic mode of action by gene expression analysis in TK6 cells. Arch Toxicol. 2013 Nov;87(11):1999-2012.
14 Mitochondrial Uncoupling Induces Epigenome Remodeling and Promotes Differentiation in Neuroblastoma. Cancer Res. 2023 Jan 18;83(2):181-194. doi: 10.1158/0008-5472.CAN-22-1029.
15 The proapoptotic effect of zoledronic acid is independent of either the bone microenvironment or the intrinsic resistance to bortezomib of myeloma cells and is enhanced by the combination with arsenic trioxide. Exp Hematol. 2011 Jan;39(1):55-65.
16 Malathion induced cancer-linked gene expression in human lymphocytes. Environ Res. 2020 Mar;182:109131. doi: 10.1016/j.envres.2020.109131. Epub 2020 Jan 10.
17 The effect of azacitidine on interleukin-6 signaling and nuclear factor-kappaB activation and its in vitro and in vivo activity against multiple myeloma. Haematologica. 2008 Jun;93(6):860-9. doi: 10.3324/haematol.12261. Epub 2008 Apr 28.
18 Simvastatin inhibits IL-17 secretion by targeting multiple IL-17-regulatory cytokines and by inhibiting the expression of IL-17 transcription factor RORC in CD4+ lymphocytes. J Immunol. 2008 May 15;180(10):6988-96. doi: 10.4049/jimmunol.180.10.6988.
19 Rofecoxib modulates multiple gene expression pathways in a clinical model of acute inflammatory pain. Pain. 2007 Mar;128(1-2):136-47.
20 Controlled diesel exhaust and allergen coexposure modulates microRNA and gene expression in humans: Effects on inflammatory lung markers. J Allergy Clin Immunol. 2016 Dec;138(6):1690-1700. doi: 10.1016/j.jaci.2016.02.038. Epub 2016 Apr 24.
21 White-to-brown metabolic conversion of human adipocytes by JAK inhibition. Nat Cell Biol. 2015 Jan;17(1):57-67. doi: 10.1038/ncb3075. Epub 2014 Dec 8.
22 The antimicrobial drug pyrimethamine inhibits STAT3 transcriptional activity by targeting the enzyme dihydrofolate reductase. J Biol Chem. 2022 Feb;298(2):101531. doi: 10.1016/j.jbc.2021.101531. Epub 2021 Dec 23.
23 Flavanoids induce expression of the suppressor of cytokine signalling 3 (SOCS3) gene and suppress IL-6-activated signal transducer and activator of transcription 3 (STAT3) activation in vascular endothelial cells. Biochem J. 2013 Sep 1;454(2):283-93. doi: 10.1042/BJ20130481.
24 Resveratrol induces nuclear factor-B activity in human cardiac cells. Int J Cardiol. 2013 Sep 10;167(6):2507-16. doi: 10.1016/j.ijcard.2012.06.006. Epub 2012 Jun 27.
25 Estrogen receptor-alpha regulates SOCS-3 expression in human breast cancer cells. Biochem Biophys Res Commun. 2005 Sep 16;335(1):168-74. doi: 10.1016/j.bbrc.2005.07.057.
26 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.
27 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.
28 Inhibition of neuroblastoma proliferation by PF-3758309, a small-molecule inhibitor that targets p21-activated kinase 4. Oncol Rep. 2017 Nov;38(5):2705-2716. doi: 10.3892/or.2017.5989. Epub 2017 Sep 22.
29 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.
30 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.
31 Gene expression changes in primary human nasal epithelial cells exposed to formaldehyde in vitro. Toxicol Lett. 2010 Oct 5;198(2):289-95.
32 Transcriptional profiling of lactic acid treated reconstructed human epidermis reveals pathways underlying stinging and itch. Toxicol In Vitro. 2019 Jun;57:164-173.
33 Combined effects of arsenic and palmitic acid on oxidative stress and lipid metabolism disorder in human hepatoma HepG2 cells. Sci Total Environ. 2021 May 15;769:144849. doi: 10.1016/j.scitotenv.2020.144849. Epub 2021 Jan 19.
34 Microarray Analysis of Gene Expression Alteration in Human Middle Ear Epithelial Cells Induced by Asian Sand Dust. Clin Exp Otorhinolaryngol. 2015 Dec;8(4):345-53. doi: 10.3342/ceo.2015.8.4.345. Epub 2015 Nov 10.
35 Gene profiles of a human alveolar epithelial cell line after in vitro exposure to respiratory (non-)sensitizing chemicals: identification of discriminating genetic markers and pathway analysis. Toxicol Lett. 2009 Feb 25;185(1):16-22. doi: 10.1016/j.toxlet.2008.11.017. Epub 2008 Dec 6.
36 Fenthion and terbufos induce DNA damage, the expression of tumor-related genes, and apoptosis in HEPG2 cells. Environ Mol Mutagen. 2011 Aug;52(7):529-37. doi: 10.1002/em.20652. Epub 2011 Apr 27.