General Information of Xenobiotics (ID: XEO00334)
Xenobiotics Name
Cisplatin
Xenobiotics Type
Pharmaceutical Agent(s)
Classification
Approved/Marketed Drug
Structure
<iframe style="width: 300px; height: 300px;" frameborder="0" src="https://embed.molview.org/v1/?mode=balls&cid=5702198"></iframe>
3D MOL 2D MOL
PubChem CID
5702198
DME(s) Modulated by This Xenobiotics
DME(s) Inhibited by This Xenobiotics
Aldo-keto reductase 1B1 (AKR1B1) DME Info Homo sapiens [1]
Aldo-keto reductase 1C2 (AKR1C2) DME Info Homo sapiens [2]
Aldehyde dehydrogenase 1 (ALDH1) DME Info Homo sapiens [3]
Aldehyde dehydrogenase 7 (ALDH7) DME Info Homo sapiens [4]
Alanyl aminopeptidase (ANPEP) DME Info Homo sapiens [5]
Arylsulfatase B (ARSB) DME Info Homo sapiens [4]
Bile acid-CoA thioesterase (BAAT) DME Info Homo sapiens [4]
Cytosolic branched aminotransferase (BCAT1) DME Info Homo sapiens [5]
Catalase (CAT) DME Info Homo sapiens [6]
Choline dehydrogenase (CHDH) DME Info Homo sapiens [4]
Cytochrome P450 1A1 (CYP1A1) DME Info Homo sapiens [7], [4]
Vitamin D(3) 24-hydroxylase (CYP24A1) DME Info Homo sapiens [4]
Retinoic acid 4-hydroxylase 26B1 (CYP26B1) DME Info Homo sapiens [4]
Cytochrome P450 3A4 (CYP3A4) DME Info Homo sapiens [8]
Delta(24)-sterol reductase (DHCR24) DME Info Homo sapiens [5]
Dihydrothymine dehydrogenase (DPYD) DME Info Homo sapiens [9]
Eosinophil peroxidase (EPX) DME Info Homo sapiens [5]
Hexosephosphate aminotransferase 2 (GFPT2) DME Info Homo sapiens [10]
L-glutamine amidohydrolase (GLS) DME Info Homo sapiens [4]
Glutathione reductase (GSR) DME Info Homo sapiens [11]
HMG-CoA reductase (HMGCR) DME Info Homo sapiens [5]
Iduronate 2-sulfatase (IDS) DME Info Homo sapiens [7]
Lactoperoxidase (LPO) DME Info Homo sapiens [5]
Methionine-tRNA ligase mitochondrial (MARS2) DME Info Homo sapiens [5]
S-adenosylmethionine synthase 2 (MAT2A) DME Info Homo sapiens [5]
Myeloperoxidase (MPO) DME Info Homo sapiens [5]
Methylsterol monooxygenase 1 (MSMO1) DME Info Homo sapiens [12], [5]
Peptide methionine sulfoxide reductase (MSRA) DME Info Homo sapiens [4], [5]
Mevalonate pyrophosphate decarboxylase (MPD) DME Info Homo sapiens [12], [4]
Cysteinyl-conjugate N-acetyltransferase (NAT8) DME Info Homo sapiens [4]
Deoxy-5'-nucleotidase 1 (NT5C) DME Info Homo sapiens [4]
Phosphodiesterase 5A (PDE5A) DME Info Homo sapiens [4]
Phosphodiesterase 7B (PDE7B) DME Info Homo sapiens [4]
Phosphoribosylformylglycinamidine synthase (PFAS) DME Info Homo sapiens [5]
Cytosolic phospholipase A2 (PLA2G4A) DME Info Homo sapiens [4]
Prostaglandin G/H synthase 2 (COX-2) DME Info Homo sapiens [13]
Selenocysteine lyase (SCLY) DME Info Homo sapiens [4]
Steroid 5-alpha-reductase 3 (SRD5A3) DME Info Homo sapiens [7]
Sulfotransferase 2B1 (SULT2B1) DME Info Homo sapiens [4]
Thioredoxin reductase TR1 (TXNRD1) DME Info Homo sapiens [14]
Thymidine phosphorylase (TYMP) DME Info Homo sapiens [9], [4]
UDP-glucuronosyltransferase 2A3 (UGT2A3) DME Info Homo sapiens [4]
Uridine 5'-monophosphate synthase (UMPS) DME Info Homo sapiens [9]
Uridine phosphorylase 1 (UPP1) DME Info Homo sapiens [4]
DME(s) Induced by This Xenobiotics
Acetylcholinesterase (ACHE) DME Info Homo sapiens [4]
Tartrate-resistant acid ATPase (ACP5) DME Info Homo sapiens [5]
Acetyl-CoA synthetase (ACSS2) DME Info Homo sapiens [5]
Adenosine aminohydrolase (ADA) DME Info Homo sapiens [4]
Hydroxyacid-oxoacid transhydrogenase (ADHFE1) DME Info Homo sapiens [4]
Small intestine reductase (AKR1B10) DME Info Homo sapiens [10], [4]
Farnesol dehydrogenase (AKR1B15) DME Info Homo sapiens [4]
Aldo-keto reductase 1C1 (AKR1C1) DME Info Homo sapiens [15], [4]
Aldo-keto reductase 1C3 (AKR1C3) DME Info Homo sapiens [15]
Aldo-keto reductase 1D1 (AKR1D1) DME Info Homo sapiens [4]
Succinic semialdehyde reductase (AKR7A2) DME Info Homo sapiens [5]
Aldehyde dehydrogenase 5 (ALDHX) DME Info Homo sapiens [4]
Succinate-semialdehyde dehydrogenase (ALDH5A1) DME Info Homo sapiens [4]
Arachidonate 5-lipoxygenase (ALOX5) DME Info Homo sapiens [4], [5]
Arylsulfatase A (ARSA) DME Info Homo sapiens [4]
Asparagine synthetase (ASNS) DME Info Homo sapiens [5]
Bleomycin hydrolase (BLMH) DME Info Homo sapiens [4]
Serine sulfhydrase (CBS) DME Info Homo sapiens [5]
Carboxylesterase 2 (CES2) DME Info Homo sapiens [4]
Microsomal cytochrome MCB5 (CYB5A) DME Info Homo sapiens [5]
Cytochrome P450 1B1 (CYP1B1) DME Info Homo sapiens [7], [5]
Cytochrome P450 2S1 (CYP2S1) DME Info Homo sapiens [4]
Lauric acid omega-hydroxylase (CYP4A11) DME Info Homo sapiens [16]
Leukotriene B4 omega-hydroxylase (CYP4F3) DME Info Homo sapiens [4]
Dicarbonyl/L-xylulose reductase (DCXR) DME Info Homo sapiens [4]
Dihydrofolate reductase (DHFR) DME Info Homo sapiens [5]
Thymidylate kinase (DTYMK) DME Info Homo sapiens [5]
Bifunctional epoxide hydrolase 2 (EPHX2) DME Info Homo sapiens [4]
Fatty acid desaturase 1 (FADS1) DME Info Homo sapiens [4], [5]
Fatty acid desaturase 2 (FADS2) DME Info Homo sapiens [5]
Friedreich ataxia protein (FXN) DME Info Homo sapiens [17]
Sodium/potassium-transporting ATPase gamma (FXYD2) DME Info Homo sapiens [4]
Gamma-Glu-X carboxypeptidase (GGH) DME Info Homo sapiens [5]
Cellular glutathione peroxidase (GPX1) DME Info Homo sapiens [4]
Glutathione S-transferase omega-1 (GSTO1) DME Info Homo sapiens [4]
Glutathione S-transferase pi (GSTP1) DME Info Homo sapiens [4]
N-acetyl-beta-glucosaminidase beta (HEXB) DME Info Homo sapiens [5]
Heparanase (HPSE) DME Info Homo sapiens [4]
Keto-steroid reductase (HSD17B7) DME Info Homo sapiens [4], [5]
Indoleamine 2,3-dioxygenase 1 (IDO1) DME Info Homo sapiens [4]
Alpha-L-iduronidase (IDUA) DME Info Homo sapiens [4]
Methionine adenosyltransferase II beta (MAT2B) DME Info Homo sapiens [4]
Microsomal glutathione S-transferase 2 (MGST2) DME Info Homo sapiens [4]
Metallothionein-1A (MT1A) DME Info Homo sapiens [18]
Metallothionein-2A (MT2A) DME Info Homo sapiens [7], [18]
Glutamine-dependent NAD(+) synthetase (NADSYN1) DME Info Homo sapiens [4]
NADH-ubiquinone oxidoreductase 30 kDa (NDUFS3) DME Info Homo sapiens [4]
Nitric oxide synthase inducible (NOS2) DME Info Homo sapiens [19]
Quinone reductase 1 (NQO1) DME Info Homo sapiens [20]
Purine nucleoside phosphorylase (PNP) DME Info Homo sapiens [4]
Pyridoxamine-phosphate oxidase (PNPO) DME Info Homo sapiens [4]
Proline dehydrogenase 1 (PRODH) DME Info Homo sapiens [4]
Prostaglandin G/H synthase 1 (COX-1) DME Info Homo sapiens [5]
Putrescine acetyltransferase (SSAT1) DME Info Homo sapiens [10], [5]
Superoxide dismutase 1 (SOD1) DME Info Homo sapiens [21]
Transglutaminase K (TGM1) DME Info Homo sapiens [4]
Transglutaminase X (TGM5) DME Info Homo sapiens [5]
Thiopurine methyltransferase (TPMT) DME Info Homo sapiens [5]
Thymidylate synthase (TYMS) DME Info Homo sapiens [9]
Xanthine dehydrogenase/oxidase (XDH) DME Info Homo sapiens [4]
References
1 Analysis of the in vitro synergistic effect of 5-fluorouracil and cisplatin on cervical carcinoma cells. Int J Gynecol Cancer. 2006 May-Jun;16(3):1321-9.
2 Oxidative stress mechanisms do not discriminate between genotoxic and nongenotoxic liver carcinogens. Chem Res Toxicol. 2015 Aug 17;28(8):1636-46.
3 Comparison of cellular and transcriptomic effects between electronic cigarette vapor and cigarette smoke in human bronchial epithelial cells. Toxicol In Vitro. 2017 Dec;45(Pt 3):417-425.
4 Activation of AIFM2 enhances apoptosis of human lung cancer cells undergoing toxicological stress. Toxicol Lett. 2016 Sep 6;258:227-236.
5 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.
6 Resveratrol loaded gelatin nanoparticles synergistically inhibits cell cycle progression and constitutive NF-kappaB activation, and induces apoptosis in non-small cell lung cancer cells. Biomed Pharmacother. 2015 Mar;70:274-82.
7 Transcriptomics hit the target: monitoring of ligand-activated and stress response pathways for chemical testing. Toxicol In Vitro. 2015 Dec 25;30(1 Pt A):7-18.
8 Proadifen sensitizes resistant ovarian adenocarcinoma cells to cisplatin. Toxicol Lett. 2016 Jan 22;243:56-66.
9 Influence of chemotherapeutic agents and cytokines on the expression of 5-fluorouracil-associated enzymes in human colon cancer cell lines. J Gastroenterol. 2006 Feb;41(2):140-50.
10 The thioxotriazole copper(II) complex A0 induces endoplasmic reticulum stress and paraptotic death in human cancer cells. J Biol Chem. 2009 Sep 4;284(36):24306-19.
11 Protective effects of emodin against cisplatin-induced oxidative stress in cultured human kidney (HEK 293) cells. J Appl Toxicol. 2013 Jul;33(7):626-30.
12 Characterisation of cisplatin-induced transcriptomics responses in primary mouse hepatocytes, HepG2 cells and mouse embryonic stem cells shows conservation of regulating transcription factor networks. Mutagenesis. 2014 Jan;29(1):17-26.
13 Arsenic sulfide combined with JQ1, chemotherapy agents, or celecoxib inhibit gastric and colon cancer cell growth. Drug Des Devel Ther. 2015 Oct 30;9:5851-62.
14 Substrate and inhibitor specificities differ between human cytosolic and mitochondrial thioredoxin reductases: Implications for development of specific inhibitors. Free Radic Biol Med. 2011 Mar 15;50(6):689-99.
15 Pathophysiological roles of aldo-keto reductases (AKR1C1 and AKR1C3) in development of cisplatin resistance in human colon cancers. Chem Biol Interact. 2013 Feb 25;202(1-3):234-42.
16 Cisplatin-mediated cytotoxicity through inducing CYP4A 11 expression in human renal tubular epithelial cells. J Toxicol Sci. 2015 Dec;40(6):895-900.
17 Pharmacological screening using an FXN-EGFP cellular genomic reporter assay for the therapy of Friedreich ataxia. PLoS One. 2013;8(2):e55940.
18 Expression pattern of cisplatin-induced metallothionein isoforms in squamous cell carcinoma. Anticancer Res. 2003 Jan-Feb;23(1A):299-303.
19 Astilbin ameliorates cisplatin-induced nephrotoxicity through reducing oxidative stress and inflammation. Food Chem Toxicol. 2018 Apr;114:227-236.
20 Deletion of NAD(P)H:quinone oxidoreductase 1 represses Mre11-Rad50-Nbs1 complex protein expression in cisplatin-induced nephrotoxicity. Toxicol Lett. 2016 Jan 22;243:22-30.
21 Mechanism of cisplatin proximal tubule toxicity revealed by integrating transcriptomics, proteomics, metabolomics and biokinetics. Toxicol In Vitro. 2015 Dec 25;30(1 Pt A):117-27.

If you find any error in data or bug in web service, please kindly report it to Dr. Yin and Dr. Li.