General Information of Drug Off-Target (DOT) (ID: OTZ3HWTO)

DOT Name Complement decay-accelerating factor (CD55)
Synonyms CD antigen CD55
Gene Name CD55
Related Disease
Protein-losing enteropathy ( )
UniProt ID
DAF_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
1H03; 1H04; 1H2P; 1H2Q; 1M11; 1NWV; 1OJV; 1OJW; 1OJY; 1OK1; 1OK2; 1OK3; 1OK9; 1UOT; 1UPN; 2C8I; 2QZD; 2QZF; 2QZH; 3IYP; 3J24; 5FOA; 6ILJ; 6ILK; 6LA5; 7C9W; 7DO4; 7VY5; 7VY6; 8B8R
Pfam ID
PF00084
Sequence
MTVARPSVPAALPLLGELPRLLLLVLLCLPAVWGDCGLPPDVPNAQPALEGRTSFPEDTV
ITYKCEESFVKIPGEKDSVICLKGSQWSDIEEFCNRSCEVPTRLNSASLKQPYITQNYFP
VGTVVEYECRPGYRREPSLSPKLTCLQNLKWSTAVEFCKKKSCPNPGEIRNGQIDVPGGI
LFGATISFSCNTGYKLFGSTSSFCLISGSSVQWSDPLPECREIYCPAPPQIDNGIIQGER
DHYGYRQSVTYACNKGFTMIGEHSIYCTVNNDEGEWSGPPPECRGKSLTSKVPPTVQKPT
TVNVPTTEVSPTSQKTTTKTTTPNAQATRSTPVSRTTKHFHETTPNKGSGTTSGTTRLLS
GHTCFTLTGLLGTLVTMGLLT
Function
This protein recognizes C4b and C3b fragments that condense with cell-surface hydroxyl or amino groups when nascent C4b and C3b are locally generated during C4 and c3 activation. Interaction of daf with cell-associated C4b and C3b polypeptides interferes with their ability to catalyze the conversion of C2 and factor B to enzymatically active C2a and Bb and thereby prevents the formation of C4b2a and C3bBb, the amplification convertases of the complement cascade. Inhibits complement activation by destabilizing and preventing the formation of C3 and C5 convertases, which prevents complement damage ; (Microbial infection) Acts as a receptor for Coxsackievirus A21, coxsackieviruses B1, B3 and B5; (Microbial infection) Acts as a receptor for Human enterovirus 70 and D68 (Probable); (Microbial infection) Acts as a receptor for Human echoviruses 6, 7, 11, 12, 20 and 21.
Tissue Specificity
Expressed on the plasma membranes of all cell types that are in intimate contact with plasma complement proteins. It is also found on the surfaces of epithelial cells lining extracellular compartments, and variants of the molecule are present in body fluids and in extracellular matrix.
KEGG Pathway
Complement and coagulation cascades (hsa04610 )
Hematopoietic cell lineage (hsa04640 )
Viral myocarditis (hsa05416 )
Reactome Pathway
Neutrophil degranulation (R-HSA-6798695 )
COPI-mediated anterograde transport (R-HSA-6807878 )
Regulation of Complement cascade (R-HSA-977606 )
Class B/2 (Secretin family receptors) (R-HSA-373080 )

Molecular Interaction Atlas (MIA) of This DOT

1 Disease(s) Related to This DOT
Disease Name Disease ID Evidence Level Mode of Inheritance REF
Protein-losing enteropathy DISM3WQO Strong Autosomal recessive [1]
------------------------------------------------------------------------------------
Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Drug Response of 3 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Etoposide DMNH3PG Approved Complement decay-accelerating factor (CD55) affects the response to substance of Etoposide. [36]
Mitoxantrone DMM39BF Approved Complement decay-accelerating factor (CD55) affects the response to substance of Mitoxantrone. [36]
Methamphetamine DMPM4SK Approved Complement decay-accelerating factor (CD55) affects the response to substance of Methamphetamine. [37]
------------------------------------------------------------------------------------
3 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 Complement decay-accelerating factor (CD55). [2]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene increases the methylation of Complement decay-accelerating factor (CD55). [26]
Bisphenol A DM2ZLD7 Investigative Bisphenol A increases the methylation of Complement decay-accelerating factor (CD55). [31]
------------------------------------------------------------------------------------
32 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Ciclosporin DMAZJFX Approved Ciclosporin increases the expression of Complement decay-accelerating factor (CD55). [3]
Tretinoin DM49DUI Approved Tretinoin increases the expression of Complement decay-accelerating factor (CD55). [4]
Acetaminophen DMUIE76 Approved Acetaminophen increases the expression of Complement decay-accelerating factor (CD55). [5]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of Complement decay-accelerating factor (CD55). [6]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate increases the expression of Complement decay-accelerating factor (CD55). [7]
Cisplatin DMRHGI9 Approved Cisplatin increases the expression of Complement decay-accelerating factor (CD55). [8]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of Complement decay-accelerating factor (CD55). [9]
Quercetin DM3NC4M Approved Quercetin increases the expression of Complement decay-accelerating factor (CD55). [10]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Complement decay-accelerating factor (CD55). [11]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Complement decay-accelerating factor (CD55). [12]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Complement decay-accelerating factor (CD55). [13]
Methotrexate DM2TEOL Approved Methotrexate increases the expression of Complement decay-accelerating factor (CD55). [14]
Marinol DM70IK5 Approved Marinol increases the expression of Complement decay-accelerating factor (CD55). [15]
Zoledronate DMIXC7G Approved Zoledronate increases the expression of Complement decay-accelerating factor (CD55). [16]
Phenobarbital DMXZOCG Approved Phenobarbital affects the expression of Complement decay-accelerating factor (CD55). [17]
Panobinostat DM58WKG Approved Panobinostat increases the expression of Complement decay-accelerating factor (CD55). [18]
Demecolcine DMCZQGK Approved Demecolcine increases the expression of Complement decay-accelerating factor (CD55). [19]
Niclosamide DMJAGXQ Approved Niclosamide increases the expression of Complement decay-accelerating factor (CD55). [20]
Cannabidiol DM0659E Approved Cannabidiol increases the expression of Complement decay-accelerating factor (CD55). [21]
Troglitazone DM3VFPD Approved Troglitazone increases the expression of Complement decay-accelerating factor (CD55). [22]
Piroxicam DMTK234 Approved Piroxicam increases the expression of Complement decay-accelerating factor (CD55). [23]
Urethane DM7NSI0 Phase 4 Urethane increases the expression of Complement decay-accelerating factor (CD55). [24]
Curcumin DMQPH29 Phase 3 Curcumin decreases the expression of Complement decay-accelerating factor (CD55). [25]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide increases the expression of Complement decay-accelerating factor (CD55). [27]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 increases the expression of Complement decay-accelerating factor (CD55). [28]
Torcetrapib DMDHYM7 Discontinued in Phase 2 Torcetrapib increases the expression of Complement decay-accelerating factor (CD55). [29]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN increases the expression of Complement decay-accelerating factor (CD55). [30]
Coumestrol DM40TBU Investigative Coumestrol decreases the expression of Complement decay-accelerating factor (CD55). [9]
Paraquat DMR8O3X Investigative Paraquat increases the expression of Complement decay-accelerating factor (CD55). [32]
Phencyclidine DMQBEYX Investigative Phencyclidine increases the expression of Complement decay-accelerating factor (CD55). [33]
Forskolin DM6ITNG Investigative Forskolin increases the expression of Complement decay-accelerating factor (CD55). [34]
AM251 DMTAWHL Investigative AM251 increases the expression of Complement decay-accelerating factor (CD55). [35]
------------------------------------------------------------------------------------
⏷ Show the Full List of 32 Drug(s)

References

1 CD55 Deficiency, Early-Onset Protein-Losing Enteropathy, and Thrombosis. N Engl J Med. 2017 Jul 6;377(1):52-61. doi: 10.1056/NEJMoa1615887. Epub 2017 Jun 28.
2 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.
3 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.
4 Effect of retinoic acid on gene expression in human conjunctival epithelium: secretory phospholipase A2 mediates retinoic acid induction of MUC16. Invest Ophthalmol Vis Sci. 2005 Nov;46(11):4050-61.
5 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.
6 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.
7 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
8 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.
9 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
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 A comprehensive analysis of Wnt/beta-catenin signaling pathway-related genes and crosstalk pathways in the treatment of As2O3 in renal cancer. Ren Fail. 2018 Nov;40(1):331-339.
12 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.
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-mediated activation of an AMPK-CREB-dependent pathway: a novel mechanism for vascular protection in chronic systemic inflammation. Ann Rheum Dis. 2016 Feb;75(2):439-48. doi: 10.1136/annrheumdis-2014-206305. Epub 2015 Jan 9.
15 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.
16 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.
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 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.
19 Characterization of formaldehyde's genotoxic mode of action by gene expression analysis in TK6 cells. Arch Toxicol. 2013 Nov;87(11):1999-2012.
20 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.
21 Cannabidiol Modulates the Immunophenotype and Inhibits the Activation of the Inflammasome in Human Gingival Mesenchymal Stem Cells. Front Physiol. 2016 Nov 24;7:559. doi: 10.3389/fphys.2016.00559. eCollection 2016.
22 Effects of ciglitazone and troglitazone on the proliferation of human stomach cancer cells. World J Gastroenterol. 2009 Jan 21;15(3):310-20.
23 Apoptosis induced by piroxicam plus cisplatin combined treatment is triggered by p21 in mesothelioma. PLoS One. 2011;6(8):e23569.
24 Ethyl carbamate induces cell death through its effects on multiple metabolic pathways. Chem Biol Interact. 2017 Nov 1;277:21-32.
25 Curcumin downregulates the inflammatory cytokines CXCL1 and -2 in breast cancer cells via NFkappaB. Carcinogenesis. 2008 Apr;29(4):779-89.
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 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
28 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.
29 Clarifying off-target effects for torcetrapib using network pharmacology and reverse docking approach. BMC Syst Biol. 2012 Dec 10;6:152.
30 DON shares a similar mode of action as the ribotoxic stress inducer anisomycin while TBTO shares ER stress patterns with the ER stress inducer thapsigargin based on comparative gene expression profiling in Jurkat T cells. Toxicol Lett. 2014 Jan 30;224(3):395-406. doi: 10.1016/j.toxlet.2013.11.005. Epub 2013 Nov 15.
31 DNA methylome-wide alterations associated with estrogen receptor-dependent effects of bisphenols in breast cancer. Clin Epigenetics. 2019 Oct 10;11(1):138. doi: 10.1186/s13148-019-0725-y.
32 An in vitro strategy using multiple human induced pluripotent stem cell-derived models to assess the toxicity of chemicals: A case study on paraquat. Toxicol In Vitro. 2022 Jun;81:105333. doi: 10.1016/j.tiv.2022.105333. Epub 2022 Feb 16.
33 Differential response of Mono Mac 6, BEAS-2B, and Jurkat cells to indoor dust. Environ Health Perspect. 2007 Sep;115(9):1325-32.
34 Selective aryl hydrocarbon receptor modulator-mediated repression of CD55 expression induced by cytokine exposure. J Pharmacol Exp Ther. 2012 Aug;342(2):345-55. doi: 10.1124/jpet.112.193482. Epub 2012 May 2.
35 Cannabinoid derivatives induce cell death in pancreatic MIA PaCa-2 cells via a receptor-independent mechanism. FEBS Lett. 2006 Mar 20;580(7):1733-9.
36 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.
37 Genome-wide association for methamphetamine dependence: convergent results from 2 samples. Arch Gen Psychiatry. 2008 Mar;65(3):345-55. doi: 10.1001/archpsyc.65.3.345.