General Information of Drug Off-Target (DOT) (ID: OTCYBO6X)

DOT Name Transcription factor Jun (JUN)
Synonyms Activator protein 1; AP1; Proto-oncogene c-Jun; Transcription factor AP-1 subunit Jun; V-jun avian sarcoma virus 17 oncogene homolog; p39
Gene Name JUN
UniProt ID
JUN_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
1A02; 1FOS; 1JNM; 1JUN; 1S9K; 1T2K; 5FV8; 5T01; 6Y3V
Pfam ID
PF00170 ; PF03957
Sequence
MTAKMETTFYDDALNASFLPSESGPYGYSNPKILKQSMTLNLADPVGSLKPHLRAKNSDL
LTSPDVGLLKLASPELERLIIQSSNGHITTTPTPTQFLCPKNVTDEQEGFAEGFVRALAE
LHSQNTLPSVTSAAQPVNGAGMVAPAVASVAGGSGSGGFSASLHSEPPVYANLSNFNPGA
LSSGGGAPSYGAAGLAFPAQPQQQQQPPHHLPQQMPVQHPRLQALKEEPQTVPEMPGETP
PLSPIDMESQERIKAERKRMRNRIAASKCRKRKLERIARLEEKVKTLKAQNSELASTANM
LREQVAQLKQKVMNHVNSGCQLMLTQQLQTF
Function
Transcription factor that recognizes and binds to the AP-1 consensus motif 5'-TGA[GC]TCA-3'. Heterodimerizes with proteins of the FOS family to form an AP-1 transcription complex, thereby enhancing its DNA binding activity to the AP-1 consensus sequence 5'-TGA[GC]TCA-3' and enhancing its transcriptional activity. Together with FOSB, plays a role in activation-induced cell death of T cells by binding to the AP-1 promoter site of FASLG/CD95L, and inducing its transcription in response to activation of the TCR/CD3 signaling pathway. Promotes activity of NR5A1 when phosphorylated by HIPK3 leading to increased steroidogenic gene expression upon cAMP signaling pathway stimulation. Involved in activated KRAS-mediated transcriptional activation of USP28 in colorectal cancer (CRC) cells. Binds to the USP28 promoter in colorectal cancer (CRC) cells ; (Microbial infection) Upon Epstein-Barr virus (EBV) infection, binds to viral BZLF1 Z promoter and activates viral BZLF1 expression.
Tissue Specificity Expressed in the developing and adult prostate and prostate cancer cells.
KEGG Pathway
Endocrine resistance (hsa01522 )
MAPK sig.ling pathway (hsa04010 )
ErbB sig.ling pathway (hsa04012 )
cAMP sig.ling pathway (hsa04024 )
Mitophagy - animal (hsa04137 )
Apoptosis (hsa04210 )
Wnt sig.ling pathway (hsa04310 )
Osteoclast differentiation (hsa04380 )
Focal adhesion (hsa04510 )
Tight junction (hsa04530 )
Toll-like receptor sig.ling pathway (hsa04620 )
NOD-like receptor sig.ling pathway (hsa04621 )
C-type lectin receptor sig.ling pathway (hsa04625 )
IL-17 sig.ling pathway (hsa04657 )
Th1 and Th2 cell differentiation (hsa04658 )
Th17 cell differentiation (hsa04659 )
T cell receptor sig.ling pathway (hsa04660 )
B cell receptor sig.ling pathway (hsa04662 )
TNF sig.ling pathway (hsa04668 )
Neurotrophin sig.ling pathway (hsa04722 )
GnRH sig.ling pathway (hsa04912 )
Estrogen sig.ling pathway (hsa04915 )
Oxytocin sig.ling pathway (hsa04921 )
Relaxin sig.ling pathway (hsa04926 )
Non-alcoholic fatty liver disease (hsa04932 )
AGE-RAGE sig.ling pathway in diabetic complications (hsa04933 )
Cocaine addiction (hsa05030 )
Amphetamine addiction (hsa05031 )
Epithelial cell sig.ling in Helicobacter pylori infection (hsa05120 )
Pathogenic Escherichia coli infection (hsa05130 )
Shigellosis (hsa05131 )
Salmonella infection (hsa05132 )
Pertussis (hsa05133 )
Yersinia infection (hsa05135 )
Leishmaniasis (hsa05140 )
Chagas disease (hsa05142 )
Hepatitis B (hsa05161 )
Measles (hsa05162 )
Human T-cell leukemia virus 1 infection (hsa05166 )
Kaposi sarcoma-associated herpesvirus infection (hsa05167 )
Epstein-Barr virus infection (hsa05169 )
Human immunodeficiency virus 1 infection (hsa05170 )
Coro.virus disease - COVID-19 (hsa05171 )
Pathways in cancer (hsa05200 )
Viral carcinogenesis (hsa05203 )
Chemical carcinogenesis - receptor activation (hsa05207 )
Chemical carcinogenesis - reactive oxygen species (hsa05208 )
Colorectal cancer (hsa05210 )
Re.l cell carcinoma (hsa05211 )
Breast cancer (hsa05224 )
Choline metabolism in cancer (hsa05231 )
PD-L1 expression and PD-1 checkpoint pathway in cancer (hsa05235 )
Inflammatory bowel disease (hsa05321 )
Rheumatoid arthritis (hsa05323 )
Lipid and atherosclerosis (hsa05417 )
Fluid shear stress and atherosclerosis (hsa05418 )
Reactome Pathway
Oxidative Stress Induced Senescence (R-HSA-2559580 )
Senescence-Associated Secretory Phenotype (SASP) (R-HSA-2559582 )
FCERI mediated MAPK activation (R-HSA-2871796 )
Activation of the AP-1 family of transcription factors (R-HSA-450341 )
Activation of anterior HOX genes in hindbrain development during early embryogenesis (R-HSA-5617472 )
MAPK6/MAPK4 signaling (R-HSA-5687128 )
TP53 Regulates Transcription of DNA Repair Genes (R-HSA-6796648 )
Deregulated CDK5 triggers multiple neurodegenerative pathways in Alzheimer's disease models (R-HSA-8862803 )
Regulation of PTEN gene transcription (R-HSA-8943724 )
Estrogen-dependent gene expression (R-HSA-9018519 )
WNT5 (R-HSA-9673324 )
Pre-NOTCH Transcription and Translation (R-HSA-1912408 )

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 4 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Morphine DMRMS0L Approved Transcription factor Jun (JUN) increases the Hyperpathia ADR of Morphine. [94]
Fructose DM43AN2 Approved Transcription factor Jun (JUN) increases the Metabolism and nutrition disorders ADR of Fructose. [94]
Chloroxine DMFZBMQ Approved Transcription factor Jun (JUN) increases the Apoptosis ADR of Chloroxine. [94]
TAM-67 DM26SE7 Investigative Transcription factor Jun (JUN) increases the Gene mutation ADR of TAM-67. [94]
------------------------------------------------------------------------------------
78 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate increases the expression of Transcription factor Jun (JUN). [1]
Ciclosporin DMAZJFX Approved Ciclosporin decreases the expression of Transcription factor Jun (JUN). [2]
Tretinoin DM49DUI Approved Tretinoin increases the expression of Transcription factor Jun (JUN). [3]
Acetaminophen DMUIE76 Approved Acetaminophen decreases the expression of Transcription factor Jun (JUN). [4]
Doxorubicin DMVP5YE Approved Doxorubicin increases the expression of Transcription factor Jun (JUN). [5]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate increases the expression of Transcription factor Jun (JUN). [6]
Cisplatin DMRHGI9 Approved Cisplatin decreases the expression of Transcription factor Jun (JUN). [7]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of Transcription factor Jun (JUN). [8]
Arsenic DMTL2Y1 Approved Arsenic increases the expression of Transcription factor Jun (JUN). [9]
Quercetin DM3NC4M Approved Quercetin increases the expression of Transcription factor Jun (JUN). [10]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Transcription factor Jun (JUN). [11]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide decreases the expression of Transcription factor Jun (JUN). [12]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Transcription factor Jun (JUN). [13]
Vorinostat DMWMPD4 Approved Vorinostat decreases the expression of Transcription factor Jun (JUN). [14]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Transcription factor Jun (JUN). [15]
Decitabine DMQL8XJ Approved Decitabine affects the expression of Transcription factor Jun (JUN). [17]
Marinol DM70IK5 Approved Marinol decreases the expression of Transcription factor Jun (JUN). [18]
Phenobarbital DMXZOCG Approved Phenobarbital affects the expression of Transcription factor Jun (JUN). [19]
Menadione DMSJDTY Approved Menadione increases the expression of Transcription factor Jun (JUN). [20]
Fluorouracil DMUM7HZ Approved Fluorouracil increases the expression of Transcription factor Jun (JUN). [21]
Panobinostat DM58WKG Approved Panobinostat increases the expression of Transcription factor Jun (JUN). [22]
Fulvestrant DM0YZC6 Approved Fulvestrant increases the expression of Transcription factor Jun (JUN). [23]
Demecolcine DMCZQGK Approved Demecolcine increases the expression of Transcription factor Jun (JUN). [24]
Bortezomib DMNO38U Approved Bortezomib decreases the expression of Transcription factor Jun (JUN). [11]
Troglitazone DM3VFPD Approved Troglitazone increases the expression of Transcription factor Jun (JUN). [27]
Azathioprine DMMZSXQ Approved Azathioprine increases the expression of Transcription factor Jun (JUN). [29]
Cytarabine DMZD5QR Approved Cytarabine increases the expression of Transcription factor Jun (JUN). [30]
Aspirin DM672AH Approved Aspirin increases the expression of Transcription factor Jun (JUN). [31]
Irinotecan DMP6SC2 Approved Irinotecan increases the expression of Transcription factor Jun (JUN). [32]
Diclofenac DMPIHLS Approved Diclofenac affects the expression of Transcription factor Jun (JUN). [15]
Nicotine DMWX5CO Approved Nicotine increases the expression of Transcription factor Jun (JUN). [33]
Indomethacin DMSC4A7 Approved Indomethacin increases the expression of Transcription factor Jun (JUN). [34]
Menthol DMG2KW7 Approved Menthol increases the expression of Transcription factor Jun (JUN). [35]
Ethinyl estradiol DMODJ40 Approved Ethinyl estradiol affects the expression of Transcription factor Jun (JUN). [36]
Cocaine DMSOX7I Approved Cocaine increases the expression of Transcription factor Jun (JUN). [37]
Obeticholic acid DM3Q1SM Approved Obeticholic acid increases the expression of Transcription factor Jun (JUN). [40]
Mitoxantrone DMM39BF Approved Mitoxantrone increases the expression of Transcription factor Jun (JUN). [41]
Capsaicin DMGMF6V Approved Capsaicin decreases the expression of Transcription factor Jun (JUN). [43]
Methamphetamine DMPM4SK Approved Methamphetamine increases the activity of Transcription factor Jun (JUN). [44]
Acocantherin DM7JT24 Approved Acocantherin decreases the expression of Transcription factor Jun (JUN). [45]
Thalidomide DM70BU5 Approved Thalidomide increases the expression of Transcription factor Jun (JUN). [46]
Sorafenib DMS8IFC Approved Sorafenib increases the expression of Transcription factor Jun (JUN). [48]
Fluoxetine DM3PD2C Approved Fluoxetine decreases the expression of Transcription factor Jun (JUN). [49]
Sertraline DM0FB1J Approved Sertraline increases the expression of Transcription factor Jun (JUN). [50]
Ritonavir DMU764S Approved Ritonavir increases the expression of Transcription factor Jun (JUN). [51]
Nefazodone DM4ZS8M Approved Nefazodone increases the expression of Transcription factor Jun (JUN). [52]
Cholecalciferol DMGU74E Approved Cholecalciferol increases the expression of Transcription factor Jun (JUN). [54]
Nilotinib DM7HXWT Approved Nilotinib increases the expression of Transcription factor Jun (JUN). [57]
Prasterone DM67VKL Approved Prasterone increases the expression of Transcription factor Jun (JUN). [58]
Masoprocol DMMVNZ0 Approved Masoprocol decreases the expression of Transcription factor Jun (JUN). [59]
Amphetamine DMSZQAK Approved Amphetamine increases the expression of Transcription factor Jun (JUN). [37]
Mechlorethamine DM0CVXA Approved Mechlorethamine increases the expression of Transcription factor Jun (JUN). [62]
Fotemustine DMV62ED Approved Fotemustine increases the expression of Transcription factor Jun (JUN). [39]
Selegiline DM6034S Approved Selegiline increases the expression of Transcription factor Jun (JUN). [64]
Urethane DM7NSI0 Phase 4 Urethane increases the expression of Transcription factor Jun (JUN). [65]
Isoflavone DM7U58J Phase 4 Isoflavone increases the expression of Transcription factor Jun (JUN). [66]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of Transcription factor Jun (JUN). [67]
Epigallocatechin gallate DMCGWBJ Phase 3 Epigallocatechin gallate increases the expression of Transcription factor Jun (JUN). [68]
Curcumin DMQPH29 Phase 3 Curcumin increases the expression of Transcription factor Jun (JUN). [69]
Fenretinide DMRD5SP Phase 3 Fenretinide increases the expression of Transcription factor Jun (JUN). [70]
Camptothecin DM6CHNJ Phase 3 Camptothecin increases the expression of Transcription factor Jun (JUN). [5]
Rigosertib DMOSTXF Phase 3 Rigosertib increases the expression of Transcription factor Jun (JUN). [71]
I3C DMIGFOR Phase 3 I3C increases the expression of Transcription factor Jun (JUN). [72]
Genistein DM0JETC Phase 2/3 Genistein increases the expression of Transcription factor Jun (JUN). [73]
Tocopherol DMBIJZ6 Phase 2 Tocopherol decreases the expression of Transcription factor Jun (JUN). [74]
DNCB DMDTVYC Phase 2 DNCB increases the expression of Transcription factor Jun (JUN). [75]
Afimoxifene DMFORDT Phase 2 Afimoxifene increases the expression of Transcription factor Jun (JUN). [76]
phorbol 12-myristate 13-acetate DMJWD62 Phase 2 phorbol 12-myristate 13-acetate increases the expression of Transcription factor Jun (JUN). [77]
Delphinidin DMS2WIN Phase 2 Delphinidin increases the expression of Transcription factor Jun (JUN). [79]
Antroquinonol DMRGQVZ Phase 2 Antroquinonol decreases the expression of Transcription factor Jun (JUN). [81]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Transcription factor Jun (JUN). [83]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide increases the expression of Transcription factor Jun (JUN). [84]
Torcetrapib DMDHYM7 Discontinued in Phase 2 Torcetrapib increases the expression of Transcription factor Jun (JUN). [88]
AN-9 DMGL0Q2 Discontinued in Phase 2 AN-9 decreases the expression of Transcription factor Jun (JUN). [89]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN increases the expression of Transcription factor Jun (JUN). [90]
Taxifolin DMQJSF9 Preclinical Taxifolin decreases the expression of Transcription factor Jun (JUN). [91]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the expression of Transcription factor Jun (JUN). [92]
Trichostatin A DM9C8NX Investigative Trichostatin A increases the expression of Transcription factor Jun (JUN). [93]
------------------------------------------------------------------------------------
⏷ Show the Full List of 78 Drug(s)
20 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Methotrexate DM2TEOL Approved Methotrexate increases the phosphorylation of Transcription factor Jun (JUN). [16]
Niclosamide DMJAGXQ Approved Niclosamide increases the phosphorylation of Transcription factor Jun (JUN). [25]
Cannabidiol DM0659E Approved Cannabidiol increases the phosphorylation of Transcription factor Jun (JUN). [26]
Hydroquinone DM6AVR4 Approved Hydroquinone increases the phosphorylation of Transcription factor Jun (JUN). [28]
Paclitaxel DMLB81S Approved Paclitaxel increases the phosphorylation of Transcription factor Jun (JUN). [5]
Topotecan DMP6G8T Approved Topotecan increases the phosphorylation of Transcription factor Jun (JUN). [39]
Vinblastine DM5TVS3 Approved Vinblastine increases the phosphorylation of Transcription factor Jun (JUN). [42]
Lindane DMB8CNL Approved Lindane increases the phosphorylation of Transcription factor Jun (JUN). [47]
Colchicine DM2POTE Approved Colchicine increases the phosphorylation of Transcription factor Jun (JUN). [42]
Docetaxel DMDI269 Approved Docetaxel increases the phosphorylation of Transcription factor Jun (JUN). [53]
Dopamine DMPGUCF Approved Dopamine increases the phosphorylation of Transcription factor Jun (JUN). [55]
Cantharidin DMBP5N3 Approved Cantharidin increases the phosphorylation of Transcription factor Jun (JUN). [56]
Efavirenz DMC0GSJ Approved Efavirenz increases the phosphorylation of Transcription factor Jun (JUN). [60]
Hydroxychloroquine DMSIVND Approved Hydroxychloroquine increases the phosphorylation of Transcription factor Jun (JUN). [61]
Sorbitol DMAN0DE Approved Sorbitol increases the phosphorylation of Transcription factor Jun (JUN). [63]
NCX-4016 DMOX1CU Phase 2 NCX-4016 increases the phosphorylation of Transcription factor Jun (JUN). [80]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene increases the methylation of Transcription factor Jun (JUN). [82]
TAK-243 DM4GKV2 Phase 1 TAK-243 increases the sumoylation of Transcription factor Jun (JUN). [85]
Tetrandrine DMAOJBX Phase 1 Tetrandrine decreases the phosphorylation of Transcription factor Jun (JUN). [86]
Adaphostin DM16QSG Phase 1 Adaphostin increases the phosphorylation of Transcription factor Jun (JUN). [87]
------------------------------------------------------------------------------------
⏷ Show the Full List of 20 Drug(s)
2 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
Simvastatin DM30SGU Approved Simvastatin decreases the localization of Transcription factor Jun (JUN). [38]
BAICALEIN DM4C7E6 Phase 2 BAICALEIN affects the binding of Transcription factor Jun (JUN). [78]
------------------------------------------------------------------------------------

References

1 Human embryonic stem cell-derived test systems for developmental neurotoxicity: a transcriptomics approach. Arch Toxicol. 2013 Jan;87(1):123-43.
2 Cyclosporine A--induced oxidative stress in human renal mesangial cells: a role for ERK 1/2 MAPK signaling. Toxicol Sci. 2012 Mar;126(1):101-13.
3 Repression of hepatocyte nuclear factor 4 alpha by AP-1 underlies dyslipidemia associated with retinoic acid. J Lipid Res. 2019 Apr;60(4):794-804. doi: 10.1194/jlr.M088880. Epub 2019 Feb 1.
4 Gene expression data from acetaminophen-induced toxicity in human hepatic in vitro systems and clinical liver samples. Data Brief. 2016 Mar 26;7:1052-1057. doi: 10.1016/j.dib.2016.03.069. eCollection 2016 Jun.
5 pRb2/p130 decreases sensitivity to apoptosis induced by camptothecin and doxorubicin but not by taxol. Clin Cancer Res. 2004 Dec 1;10(23):8085-93. doi: 10.1158/1078-0432.CCR-04-0996.
6 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
7 Activation of AIFM2 enhances apoptosis of human lung cancer cells undergoing toxicological stress. Toxicol Lett. 2016 Sep 6;258:227-236.
8 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
9 LncRNA UCA1 attenuates autophagy-dependent cell death through blocking autophagic flux under arsenic stress. Toxicol Lett. 2018 Mar 1;284:195-204. doi: 10.1016/j.toxlet.2017.12.009. Epub 2017 Dec 15.
10 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.
11 Synergistic antiproliferative effect of arsenic trioxide combined with bortezomib in HL60 cell line and primary blasts from patients affected by myeloproliferative disorders. Cancer Genet Cytogenet. 2010 Jun;199(2):110-20. doi: 10.1016/j.cancergencyto.2010.02.010.
12 Oxidative stress modulates theophylline effects on steroid responsiveness. Biochem Biophys Res Commun. 2008 Dec 19;377(3):797-802.
13 Regulation of keratinocyte proliferation and differentiation by all-trans-retinoic acid, 9-cis-retinoic acid and 1,25-dihydroxy vitamin D3. Arch Dermatol Res. 1996 Nov;288(12):729-38. doi: 10.1007/BF02505289.
14 Definition of transcriptome-based indices for quantitative characterization of chemically disturbed stem cell development: introduction of the STOP-Toxukn and STOP-Toxukk tests. Arch Toxicol. 2017 Feb;91(2):839-864.
15 Drug-induced endoplasmic reticulum and oxidative stress responses independently sensitize toward TNF-mediated hepatotoxicity. Toxicol Sci. 2014 Jul;140(1):144-59. doi: 10.1093/toxsci/kfu072. Epub 2014 Apr 20.
16 [Regulation mechanism of autophagy-related protein LC3 by c-Jun in methotrexate resistant human choriocarcinoma JEG-3 cells]. Zhonghua Fu Chan Ke Za Zhi. 2014 Jul;49(7):531-5.
17 Acute hypersensitivity of pluripotent testicular cancer-derived embryonal carcinoma to low-dose 5-aza deoxycytidine is associated with global DNA Damage-associated p53 activation, anti-pluripotency and DNA demethylation. PLoS One. 2012;7(12):e53003. doi: 10.1371/journal.pone.0053003. Epub 2012 Dec 27.
18 Single-cell Transcriptome Mapping Identifies Common and Cell-type Specific Genes Affected by Acute Delta9-tetrahydrocannabinol in Humans. Sci Rep. 2020 Feb 26;10(1):3450. doi: 10.1038/s41598-020-59827-1.
19 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.
20 Gene expression after treatment with hydrogen peroxide, menadione, or t-butyl hydroperoxide in breast cancer cells. Cancer Res. 2002 Nov 1;62(21):6246-54.
21 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.
22 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.
23 Gene expression profiles with activation of the estrogen receptor alpha-selective estrogen receptor modulator complex in breast cancer cells expressing wild-type estrogen receptor. Cancer Res. 2002 Aug 1;62(15):4419-26.
24 Characterization of formaldehyde's genotoxic mode of action by gene expression analysis in TK6 cells. Arch Toxicol. 2013 Nov;87(11):1999-2012.
25 Niclosamide enhances ROS-mediated cell death through c-Jun activation. Biomed Pharmacother. 2014 Jun;68(5):619-24. doi: 10.1016/j.biopha.2014.03.018. Epub 2014 Mar 28.
26 Inhibition of autophagic flux differently modulates cannabidiol-induced death in 2D and 3D glioblastoma cell cultures. Sci Rep. 2020 Feb 14;10(1):2687. doi: 10.1038/s41598-020-59468-4.
27 Dual function of troglitazone in ICAM-1 gene expression in human vascular endothelium. Biochem Biophys Res Commun. 2001 Apr 6;282(3):717-22. doi: 10.1006/bbrc.2001.4628.
28 p38 MAPK/PP2Ac/TTP pathway on the connection of TNF- and caspases activation on hydroquinone-induced apoptosis. Carcinogenesis. 2013 Apr;34(4):818-27. doi: 10.1093/carcin/bgs409. Epub 2013 Jan 3.
29 A transcriptomics-based in vitro assay for predicting chemical genotoxicity in vivo. Carcinogenesis. 2012 Jul;33(7):1421-9.
30 Granulocyte-macrophage colony-stimulating factor/interleukin-3 fusion protein (pIXY 321) enhances high-dose Ara-C-induced programmed cell death or apoptosis in human myeloid leukemia cells. Blood. 1992 Dec 1;80(11):2883-90.
31 DNA array analysis of the effects of aspirin on colon cancer cells: involvement of Rac1. Carcinogenesis. 2004 Jul;25(7):1293-8.
32 In vitro and in vivo irinotecan-induced changes in expression profiles of cell cycle and apoptosis-associated genes in acute myeloid leukemia cells. Mol Cancer Ther. 2005 Jun;4(6):885-900.
33 Functional role of beta-adrenergic receptors in the mitogenic action of nicotine on gastric cancer cells. Toxicol Sci. 2007 Mar;96(1):21-9.
34 Mechanisms of indomethacin-induced alterations in the choline phospholipid metabolism of breast cancer cells. Neoplasia. 2006 Sep;8(9):758-71.
35 Repurposing L-menthol for systems medicine and cancer therapeutics? L-menthol induces apoptosis through caspase 10 and by suppressing HSP90. OMICS. 2016 Jan;20(1):53-64.
36 Regulation of expression and activity of multidrug resistance proteins MRP2 and MDR1 by estrogenic compounds in Caco-2 cells. Role in prevention of xenobiotic-induced cytotoxicity. Toxicology. 2014 Jun 5;320:46-55. doi: 10.1016/j.tox.2014.03.007. Epub 2014 Mar 28.
37 Effect of acute and chronic psychostimulant drugs on redox status, AP-1 activation and pro-enkephalin mRNA in the human astrocyte-like U373 MG cells. Neuropharmacology. 2005 Apr;48(5):673-84. doi: 10.1016/j.neuropharm.2004.12.010.
38 Simvastatin suppresses tissue factor expression and increases fibrinolytic activity in tumor necrosis factor-alpha-activated human peritoneal mesothelial cells. Kidney Int. 2003 Jun;63(6):2065-74. doi: 10.1046/j.1523-1755.2003.t01-2-00004.x.
39 Human three prime exonuclease TREX1 is induced by genotoxic stress and involved in protection of glioma and melanoma cells to anticancer drugs. Biochim Biophys Acta. 2013 Aug;1833(8):1832-43. doi: 10.1016/j.bbamcr.2013.03.029. Epub 2013 Apr 8.
40 Pharmacotoxicology of clinically-relevant concentrations of obeticholic acid in an organotypic human hepatocyte system. Toxicol In Vitro. 2017 Mar;39:93-103.
41 High-dose mitoxantrone induces programmed cell death or apoptosis in human myeloid leukemia cells. Blood. 1993 Nov 15;82(10):3133-40.
42 Nocodazole-induced p53-dependent c-Jun N-terminal kinase activation reduces apoptosis in human colon carcinoma HCT116 cells. J Biol Chem. 2002 Nov 15;277(46):43648-58. doi: 10.1074/jbc.M203214200. Epub 2002 Sep 6.
43 Triggering of transient receptor potential vanilloid type 1 (TRPV1) by capsaicin induces Fas/CD95-mediated apoptosis of urothelial cancer cells in an ATM-dependent manner. Carcinogenesis. 2009 Aug;30(8):1320-9. doi: 10.1093/carcin/bgp138. Epub 2009 Jun 5.
44 Methamphetamine induces AP-1 and NF-kappaB binding and transactivation in human brain endothelial cells. J Neurosci Res. 2001 Nov 15;66(4):583-91. doi: 10.1002/jnr.1248.
45 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.
46 Polyunsaturated fatty acids synergize with lipid droplet binding thalidomide analogs to induce oxidative stress in cancer cells. Lipids Health Dis. 2010 Jun 2;9:56. doi: 10.1186/1476-511X-9-56.
47 Effects of organochlorine insecticides on MAP kinase pathways in human HaCaT keratinocytes: key role of reactive oxygen species. Toxicol Sci. 2005 Aug;86(2):444-52. doi: 10.1093/toxsci/kfi192. Epub 2005 May 11.
48 Induction of DNA damage-inducible gene GADD45beta contributes to sorafenib-induced apoptosis in hepatocellular carcinoma cells. Cancer Res. 2010 Nov 15;70(22):9309-18. doi: 10.1158/0008-5472.CAN-10-1033. Epub 2010 Nov 9.
49 Fluoxetine inhibits the extracellular signal regulated kinase pathway and suppresses growth of cancer cells. Cancer Biol Ther. 2008 Oct;7(10):1685-93. doi: 10.4161/cbt.7.10.6664. Epub 2008 Oct 22.
50 Sertraline induces endoplasmic reticulum stress in hepatic cells. Toxicology. 2014 Aug 1;322:78-88. doi: 10.1016/j.tox.2014.05.007. Epub 2014 May 24.
51 The HIV protease inhibitor ritonavir synergizes with butyrate for induction of apoptotic cell death and mediates expression of heme oxygenase-1 in DLD-1 colon carcinoma cells. Br J Pharmacol. 2004 Dec;143(7):890-8. doi: 10.1038/sj.bjp.0706023. Epub 2004 Oct 25.
52 Involvement of mitochondrial dysfunction in nefazodone-induced hepatotoxicity. Food Chem Toxicol. 2016 Aug;94:148-58. doi: 10.1016/j.fct.2016.06.001. Epub 2016 Jun 8.
53 All-trans retinoic acid potentiates Taxotere-induced cell death mediated by Jun N-terminal kinase in breast cancer cells. Oncogene. 2004 Jan 15;23(2):426-33. doi: 10.1038/sj.onc.1207040.
54 Targeting iron homeostasis induces cellular differentiation and synergizes with differentiating agents in acute myeloid leukemia. J Exp Med. 2010 Apr 12;207(4):731-50. doi: 10.1084/jem.20091488. Epub 2010 Apr 5.
55 Parkin protects human dopaminergic neuroblastoma cells against dopamine-induced apoptosis. Hum Mol Genet. 2004 Aug 15;13(16):1745-54. doi: 10.1093/hmg/ddh180. Epub 2004 Jun 15.
56 Anticancer effects of cantharidin in A431 human skin cancer (Epidermoid carcinoma) cells in vitro and in vivo. Environ Toxicol. 2017 Mar;32(3):723-738. doi: 10.1002/tox.22273. Epub 2016 Apr 25.
57 Endoplasmic reticulum stress-mediated apoptosis in imatinib-resistant leukemic K562-r cells triggered by AMN107 combined with arsenic trioxide. Exp Biol Med (Maywood). 2013 Aug 1;238(8):932-42. doi: 10.1177/1535370213492689. Epub 2013 Jul 24.
58 Dehydroepiandrosterone Activation of G-protein-coupled Estrogen Receptor Rapidly Stimulates MicroRNA-21 Transcription in Human Hepatocellular Carcinoma Cells. J Biol Chem. 2015 Jun 19;290(25):15799-15811. doi: 10.1074/jbc.M115.641167. Epub 2015 May 11.
59 Inhibition of AP-1 transcription activator induces myc-dependent apoptosis in HL60 cells. J Cell Biochem. 2004 Apr 1;91(5):973-86. doi: 10.1002/jcb.10768.
60 Efavirenz and 8-hydroxyefavirenz induce cell death via a JNK- and BimEL-dependent mechanism in primary human hepatocytes. Toxicol Appl Pharmacol. 2011 Dec 1;257(2):227-34. doi: 10.1016/j.taap.2011.09.008. Epub 2011 Sep 19.
61 Hydroxychloroquine-inhibited dengue virus is associated with host defense machinery. J Interferon Cytokine Res. 2015 Mar;35(3):143-56. doi: 10.1089/jir.2014.0038. Epub 2014 Oct 16.
62 Nitrogen mustard prevents transport of Fra-1 into the nucleus to promote c-Fos- and FosB-dependent IL-8 induction in injured mouse epidermis. Toxicol Lett. 2020 Feb 1;319:256-263. doi: 10.1016/j.toxlet.2019.10.006. Epub 2019 Oct 19.
63 Intracellular mobility and nuclear trafficking of the stress-activated kinase JNK1 are impeded by hyperosmotic stress. Biochim Biophys Acta. 2014 Feb;1843(2):253-64. doi: 10.1016/j.bbamcr.2013.10.017. Epub 2013 Nov 1.
64 Role of the redox protein thioredoxin in cytoprotective mechanism evoked by (-)-deprenyl. Mol Pharmacol. 2005 Nov;68(5):1408-14. doi: 10.1124/mol.105.012302. Epub 2005 Aug 12.
65 Ethyl carbamate induces cell death through its effects on multiple metabolic pathways. Chem Biol Interact. 2017 Nov 1;277:21-32.
66 Soy isoflavones exert differential effects on androgen responsive genes in LNCaP human prostate cancer cells. J Nutr. 2007 Apr;137(4):964-72.
67 Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer Manag Res. 2009 Apr 3;1:25-37.
68 Resveratrol-induced modification of polyamine metabolism is accompanied by induction of c-Fos. Carcinogenesis. 2003 Mar;24(3):469-74. doi: 10.1093/carcin/24.3.469.
69 Gene expression profiling identifies activating transcription factor 3 as a novel contributor to the proapoptotic effect of curcumin. Mol Cancer Ther. 2005 Feb;4(2):233-41.
70 Identification of retinoid-modulated proteins in squamous carcinoma cells using high-throughput immunoblotting. Cancer Res. 2004 Apr 1;64(7):2439-48. doi: 10.1158/0008-5472.can-03-2643.
71 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.
72 Anticarcinogenic effect of indole-3-carbinol (I3C) on human hepatocellular carcinoma SNU449 cells. Hum Exp Toxicol. 2019 Jan;38(1):136-147. doi: 10.1177/0960327118785235. Epub 2018 Jul 11.
73 Phytoestrogens activate estrogen receptor beta1 and estrogenic responses in human breast and bone cancer cell lines. Mol Nutr Food Res. 2007 Feb;51(2):171-7. doi: 10.1002/mnfr.200600091.
74 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.
75 MIP-1beta, a novel biomarker for in vitro sensitization test using human monocytic cell line. Toxicol In Vitro. 2006 Aug;20(5):736-42.
76 Estrogenic GPR30 signalling induces proliferation and migration of breast cancer cells through CTGF. EMBO J. 2009 Mar 4;28(5):523-32.
77 Quercetin inhibits inducible ICAM-1 expression in human endothelial cells through the JNK pathway. Am J Physiol. 1999 Sep;277(3):C403-11. doi: 10.1152/ajpcell.1999.277.3.C403.
78 Mechanism analysis of Buyang Huanwu decoction in treating atherosclerosis based on network pharmacology and in?vitro experiments. Chem Biol Drug Des. 2024 Jan;103(1):e14447. doi: 10.1111/cbdd.14447.
79 Induction of apoptosis by the Anthocyanidins through regulation of Bcl-2 gene and activation of c-Jun N-terminal kinase cascade in hepatoma cells. J Agric Food Chem. 2005 Mar 9;53(5):1740-9. doi: 10.1021/jf048955e.
80 Nitric oxide-donating aspirin inhibits colon cancer cell growth via mitogen-activated protein kinase activation. J Pharmacol Exp Ther. 2006 Jan;316(1):25-34. doi: 10.1124/jpet.105.091363. Epub 2005 Sep 16.
81 Antroquinonol from Antrodia Camphorata suppresses breast tumor migration/invasion through inhibiting ERK-AP-1- and AKT-NF-B-dependent MMP-9 and epithelial-mesenchymal transition expressions. Food Chem Toxicol. 2015 Apr;78:33-41. doi: 10.1016/j.fct.2015.01.012. Epub 2015 Feb 2.
82 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.
83 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.
84 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
85 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.
86 Tetrandrine inhibits human brain glioblastoma multiforme GBM 8401 cancer cell migration and invasion in vitro. Environ Toxicol. 2019 Apr;34(4):364-374. doi: 10.1002/tox.22691. Epub 2018 Dec 13.
87 Induction of apoptosis in human leukemia cells by the tyrosine kinase inhibitor adaphostin proceeds through a RAF-1/MEK/ERK- and AKT-dependent process. Oncogene. 2004 Feb 19;23(7):1364-76. doi: 10.1038/sj.onc.1207248.
88 Clarifying off-target effects for torcetrapib using network pharmacology and reverse docking approach. BMC Syst Biol. 2012 Dec 10;6:152.
89 Esterase inhibitors diminish the modulation of gene expression by butyric acid derivative, pivaloyloxymethyl butyrate (AN-9). Isr J Med Sci. 1996 Dec;32(12):1186-91.
90 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.
91 The chemopreventive effect of taxifolin is exerted through ARE-dependent gene regulation. Biol Pharm Bull. 2007 Jun;30(6):1074-9.
92 Bisphenol A stimulates the epithelial mesenchymal transition of estrogen negative breast cancer cells via FOXA1 signals. Arch Biochem Biophys. 2015 Nov 1;585:10-16. doi: 10.1016/j.abb.2015.09.006. Epub 2015 Sep 9.
93 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.
94 ADReCS-Target: target profiles for aiding drug safety research and application. Nucleic Acids Res. 2018 Jan 4;46(D1):D911-D917. doi: 10.1093/nar/gkx899.