General Information of Drug Off-Target (DOT) (ID: OTNQDS00)

DOT Name Kinesin-like protein KIFC1 (KIFC1)
Synonyms Kinesin-like protein 2; Kinesin-related protein HSET
Gene Name KIFC1
Related Disease
Adenocarcinoma ( )
Advanced cancer ( )
Anaplastic astrocytoma ( )
Breast neoplasm ( )
Endometrial carcinoma ( )
Epithelial ovarian cancer ( )
Esophageal squamous cell carcinoma ( )
Hepatocellular carcinoma ( )
Metastatic malignant neoplasm ( )
Neoplasm ( )
Nervous system disease ( )
Non-small-cell lung cancer ( )
Ovarian cancer ( )
Ovarian neoplasm ( )
Primary ciliary dyskinesia ( )
Prostate cancer ( )
Prostate carcinoma ( )
Seminoma ( )
Triple negative breast cancer ( )
Bladder cancer ( )
Breast cancer ( )
Breast carcinoma ( )
Clear cell renal carcinoma ( )
Gastric cancer ( )
Renal cell carcinoma ( )
Stomach cancer ( )
Urinary bladder cancer ( )
Urinary bladder neoplasm ( )
Meningioma ( )
UniProt ID
KIFC1_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
5WDH
Pfam ID
PF00225
Sequence
MDPQRSPLLEVKGNIELKRPLIKAPSQLPLSGSRLKRRPDQMEDGLEPEKKRTRGLGATT
KITTSHPRVPSLTTVPQTQGQTTAQKVSKKTGPRCSTAIATGLKNQKPVPAVPVQKSGTS
GVPPMAGGKKPSKRPAWDLKGQLCDLNAELKRCRERTQTLDQENQQLQDQLRDAQQQVKA
LGTERTTLEGHLAKVQAQAEQGQQELKNLRACVLELEERLSTQEGLVQELQKKQVELQEE
RRGLMSQLEEKERRLQTSEAALSSSQAEVASLRQETVAQAALLTEREERLHGLEMERRRL
HNQLQELKGNIRVFCRVRPVLPGEPTPPPGLLLFPSGPGGPSDPPTRLSLSRSDERRGTL
SGAPAPPTRHDFSFDRVFPPGSGQDEVFEEIAMLVQSALDGYPVCIFAYGQTGSGKTFTM
EGGPGGDPQLEGLIPRALRHLFSVAQELSGQGWTYSFVASYVEIYNETVRDLLATGTRKG
QGGECEIRRAGPGSEELTVTNARYVPVSCEKEVDALLHLARQNRAVARTAQNERSSRSHS
VFQLQISGEHSSRGLQCGAPLSLVDLAGSERLDPGLALGPGERERLRETQAINSSLSTLG
LVIMALSNKESHVPYRNSKLTYLLQNSLGGSAKMLMFVNISPLEENVSESLNSLRFASKV
NQCVIGTAQANRK
Function Minus end-directed microtubule-dependent motor required for bipolar spindle formation. May contribute to movement of early endocytic vesicles. Regulates cilium formation and structure.
KEGG Pathway
Motor proteins (hsa04814 )
Reactome Pathway
Kinesins (R-HSA-983189 )
COPI-dependent Golgi-to-ER retrograde traffic (R-HSA-6811434 )

Molecular Interaction Atlas (MIA) of This DOT

29 Disease(s) Related to This DOT
Disease Name Disease ID Evidence Level Mode of Inheritance REF
Adenocarcinoma DIS3IHTY Strong Biomarker [1]
Advanced cancer DISAT1Z9 Strong Biomarker [2]
Anaplastic astrocytoma DISSBE0K Strong Biomarker [3]
Breast neoplasm DISNGJLM Strong Biomarker [4]
Endometrial carcinoma DISXR5CY Strong Biomarker [5]
Epithelial ovarian cancer DIS56MH2 Strong Altered Expression [1]
Esophageal squamous cell carcinoma DIS5N2GV Strong Altered Expression [2]
Hepatocellular carcinoma DIS0J828 Strong Biomarker [6]
Metastatic malignant neoplasm DIS86UK6 Strong Altered Expression [7]
Neoplasm DISZKGEW Strong Altered Expression [6]
Nervous system disease DISJ7GGT Strong Biomarker [8]
Non-small-cell lung cancer DIS5Y6R9 Strong Altered Expression [6]
Ovarian cancer DISZJHAP Strong Biomarker [7]
Ovarian neoplasm DISEAFTY Strong Altered Expression [7]
Primary ciliary dyskinesia DISOBC7V Strong Biomarker [9]
Prostate cancer DISF190Y Strong Biomarker [10]
Prostate carcinoma DISMJPLE Strong Biomarker [10]
Seminoma DIS3J8LJ Strong Biomarker [11]
Triple negative breast cancer DISAMG6N Strong Biomarker [12]
Bladder cancer DISUHNM0 moderate Biomarker [13]
Breast cancer DIS7DPX1 moderate Altered Expression [2]
Breast carcinoma DIS2UE88 moderate Altered Expression [2]
Clear cell renal carcinoma DISBXRFJ moderate Biomarker [14]
Gastric cancer DISXGOUK moderate Biomarker [15]
Renal cell carcinoma DISQZ2X8 moderate Biomarker [14]
Stomach cancer DISKIJSX moderate Biomarker [15]
Urinary bladder cancer DISDV4T7 moderate Biomarker [13]
Urinary bladder neoplasm DIS7HACE moderate Biomarker [13]
Meningioma DISPT4TG Limited Biomarker [16]
------------------------------------------------------------------------------------
⏷ Show the Full List of 29 Disease(s)
Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
3 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 Kinesin-like protein KIFC1 (KIFC1). [17]
Arsenic DMTL2Y1 Approved Arsenic affects the methylation of Kinesin-like protein KIFC1 (KIFC1). [25]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the methylation of Kinesin-like protein KIFC1 (KIFC1). [45]
------------------------------------------------------------------------------------
27 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 Kinesin-like protein KIFC1 (KIFC1). [18]
Tretinoin DM49DUI Approved Tretinoin decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [19]
Acetaminophen DMUIE76 Approved Acetaminophen decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [20]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [21]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [22]
Estradiol DMUNTE3 Approved Estradiol increases the expression of Kinesin-like protein KIFC1 (KIFC1). [23]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [24]
Temozolomide DMKECZD Approved Temozolomide increases the expression of Kinesin-like protein KIFC1 (KIFC1). [26]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide affects the expression of Kinesin-like protein KIFC1 (KIFC1). [27]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [28]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [28]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Kinesin-like protein KIFC1 (KIFC1). [29]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [30]
Zoledronate DMIXC7G Approved Zoledronate decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [31]
Niclosamide DMJAGXQ Approved Niclosamide decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [32]
Bortezomib DMNO38U Approved Bortezomib decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [33]
Dasatinib DMJV2EK Approved Dasatinib decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [34]
Lucanthone DMZLBUO Approved Lucanthone decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [35]
Methamphetamine DMPM4SK Approved Methamphetamine decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [36]
Palbociclib DMD7L94 Approved Palbociclib decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [37]
Amiodarone DMUTEX3 Phase 2/3 Trial Amiodarone increases the expression of Kinesin-like protein KIFC1 (KIFC1). [38]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [40]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [41]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [42]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [43]
PJ34 DMXO6YH Preclinical PJ34 decreases the expression of Kinesin-like protein KIFC1 (KIFC1). [44]
Coumestrol DM40TBU Investigative Coumestrol increases the expression of Kinesin-like protein KIFC1 (KIFC1). [23]
------------------------------------------------------------------------------------
⏷ Show the Full List of 27 Drug(s)
1 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
DNCB DMDTVYC Phase 2 DNCB affects the binding of Kinesin-like protein KIFC1 (KIFC1). [39]
------------------------------------------------------------------------------------

References

1 A centrosome clustering protein, KIFC1, predicts aggressive disease course in serous ovarian adenocarcinomas.J Ovarian Res. 2016 Mar 18;9:17. doi: 10.1186/s13048-016-0224-0.
2 Prognostic and clinicopathological significance of kinesin family member C1 in various cancers: A meta-analysis.Medicine (Baltimore). 2019 Oct;98(40):e17346. doi: 10.1097/MD.0000000000017346.
3 Expression of targeting protein for Xenopus kinesin-like protein 2 is associated with progression of human malignant astrocytoma.Brain Res. 2010 Sep 17;1352:200-7. doi: 10.1016/j.brainres.2010.06.060. Epub 2010 Jun 30.
4 Effects of TPX2 gene on radiotherapy sensitization in breast cancer stem cells.Oncol Lett. 2017 Aug;14(2):1531-1535. doi: 10.3892/ol.2017.6277. Epub 2017 May 29.
5 MiR-29a-5p inhibits proliferation and invasion and induces apoptosis in endometrial carcinoma via targeting TPX2.Cell Cycle. 2018;17(10):1268-1278. doi: 10.1080/15384101.2018.1475829. Epub 2018 Jul 23.
6 KIFC1 regulated by miR-532-3p promotes epithelial-to-mesenchymal transition and metastasis of hepatocellular carcinoma via gankyrin/AKT signaling.Oncogene. 2019 Jan;38(3):406-420. doi: 10.1038/s41388-018-0440-8. Epub 2018 Aug 16.
7 KIFCI, a novel putative prognostic biomarker for ovarian adenocarcinomas: delineating protein interaction networks and signaling circuitries.J Ovarian Res. 2014 May 12;7:53. doi: 10.1186/1757-2215-7-53. eCollection 2014.
8 Mitotic Motor KIFC1 Is an Organizer of Microtubules in the Axon.J Neurosci. 2019 May 15;39(20):3792-3811. doi: 10.1523/JNEUROSCI.3099-18.2019. Epub 2019 Feb 25.
9 Genomic organization of the HSET locus and the possible association of HLA-linked genes with immotile cilia syndrome (ICS).Immunogenetics. 1999 Jul;49(7-8):644-52. doi: 10.1007/s002510050660.
10 KIFC1 Inhibitor CW069 Induces Apoptosis and Reverses Resistance to Docetaxel in Prostate Cancer.J Clin Med. 2019 Feb 9;8(2):225. doi: 10.3390/jcm8020225.
11 C-terminal kinesin motor KIFC1 participates in facilitating proper cell division of human seminoma.Oncotarget. 2017 May 24;8(37):61373-61384. doi: 10.18632/oncotarget.18139. eCollection 2017 Sep 22.
12 Multi-institutional study of nuclear KIFC1 as a biomarker of poor prognosis in African American women with triple-negative breast cancer.Sci Rep. 2017 Feb 20;7:42289. doi: 10.1038/srep42289.
13 Kinesin family member C1 accelerates bladder cancer cell proliferation and induces epithelial-mesenchymal transition via Akt/GSK3 signaling.Cancer Sci. 2019 Sep;110(9):2822-2833. doi: 10.1111/cas.14126. Epub 2019 Jul 23.
14 Kinesin Motor Protein KIFC1 Is a Target Protein of miR-338-3p and Is Associated With Poor Prognosis and Progression of Renal Cell Carcinoma.Oncol Res. 2018 Dec 27;27(1):125-137. doi: 10.3727/096504018X15213115046567. Epub 2018 Mar 21.
15 Overexpression of KIFC1 and its association with spheroid formation in esophageal squamous cell carcinoma.Pathol Res Pract. 2017 Nov;213(11):1388-1393. doi: 10.1016/j.prp.2017.09.009. Epub 2017 Sep 12.
16 Identification of KIF11 As a Novel Target in Meningioma.Cancers (Basel). 2019 Apr 15;11(4):545. doi: 10.3390/cancers11040545.
17 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.
18 Inter-laboratory comparison of human renal proximal tubule (HK-2) transcriptome alterations due to Cyclosporine A exposure and medium exhaustion. Toxicol In Vitro. 2009 Apr;23(3):486-99.
19 Transcriptional and Metabolic Dissection of ATRA-Induced Granulocytic Differentiation in NB4 Acute Promyelocytic Leukemia Cells. Cells. 2020 Nov 5;9(11):2423. doi: 10.3390/cells9112423.
20 Multiple microRNAs function as self-protective modules in acetaminophen-induced hepatotoxicity in humans. Arch Toxicol. 2018 Feb;92(2):845-858.
21 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.
22 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
23 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
24 Quantitative proteomics reveals a broad-spectrum antiviral property of ivermectin, benefiting for COVID-19 treatment. J Cell Physiol. 2021 Apr;236(4):2959-2975. doi: 10.1002/jcp.30055. Epub 2020 Sep 22.
25 Prenatal arsenic exposure and the epigenome: identifying sites of 5-methylcytosine alterations that predict functional changes in gene expression in newborn cord blood and subsequent birth outcomes. Toxicol Sci. 2015 Jan;143(1):97-106. doi: 10.1093/toxsci/kfu210. Epub 2014 Oct 10.
26 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.
27 Minimal peroxide exposure of neuronal cells induces multifaceted adaptive responses. PLoS One. 2010 Dec 17;5(12):e14352. doi: 10.1371/journal.pone.0014352.
28 Effects of 1alpha,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011 May 18;10:58.
29 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.
30 Methotrexate modulates folate phenotype and inflammatory profile in EA.hy 926 cells. Eur J Pharmacol. 2014 Jun 5;732:60-7.
31 Interleukin-19 as a translational indicator of renal injury. Arch Toxicol. 2015 Jan;89(1):101-6.
32 Computational discovery of niclosamide ethanolamine, a repurposed drug candidate that reduces growth of hepatocellular carcinoma cells initro and in mice by inhibiting cell division cycle 37 signaling. Gastroenterology. 2017 Jun;152(8):2022-2036.
33 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.
34 Dasatinib reverses cancer-associated fibroblasts (CAFs) from primary lung carcinomas to a phenotype comparable to that of normal fibroblasts. Mol Cancer. 2010 Jun 27;9:168.
35 Lucanthone is a novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem. 2011 Feb 25;286(8):6602-13.
36 Methamphetamine alters the normal progression by inducing cell cycle arrest in astrocytes. PLoS One. 2014 Oct 7;9(10):e109603.
37 Cdk4/6 inhibition induces epithelial-mesenchymal transition and enhances invasiveness in pancreatic cancer cells. Mol Cancer Ther. 2012 Oct;11(10):2138-48. doi: 10.1158/1535-7163.MCT-12-0562. Epub 2012 Aug 6.
38 Identification by automated screening of a small molecule that selectively eliminates neural stem cells derived from hESCs but not dopamine neurons. PLoS One. 2009 Sep 23;4(9):e7155.
39 Proteomic analysis of the cellular response to a potent sensitiser unveils the dynamics of haptenation in living cells. Toxicology. 2020 Dec 1;445:152603. doi: 10.1016/j.tox.2020.152603. Epub 2020 Sep 28.
40 Transcriptional signature of human macrophages exposed to the environmental contaminant benzo(a)pyrene. Toxicol Sci. 2010 Apr;114(2):247-59.
41 Inhibition of BRD4 attenuates tumor cell self-renewal and suppresses stem cell signaling in MYC driven medulloblastoma. Oncotarget. 2014 May 15;5(9):2355-71.
42 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
43 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.
44 KIFC1 is a novel potential therapeutic target for breast cancer. Cancer Biol Ther. 2015;16(9):1316-22. doi: 10.1080/15384047.2015.1070980. Epub 2015 Jul 15.
45 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.