Cancer Res Treat.  2019 Jul;51(3):1207-1221. 10.4143/crt.2018.460.

KIF11 Functions as an Oncogene and Is Associated with Poor Outcomes from Breast Cancer

Affiliations
  • 1Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, Xiamen, China.
  • 2Department of Breast Surgery, Zhuhai Maternity and Child Health Hospital, Zhuhai, China.
  • 3Department of Basic Medical Science, Medical College of Xiamen University, Xiamen, China.
  • 4Department of Radiation Oncology, Xiamen Cancer Hospital, the First Affiliated Hospital of Xiamen University, Xiamen, China. unowu12345@hotmail.com
  • 5Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China. hezhy@sysucc.org.cn

Abstract

PURPOSE
The study aimed to search and identify genes that were differentially expressed in breast cancer, and their roles in cancer growth and progression.
MATERIALS AND METHODS
The Gene Expression Omnibus (Oncomine) and The Cancer Genome Atlas databases (

Keyword

Breast neoplasms; KIF11; Survival; Oncogenes

MeSH Terms

AMP-Activated Protein Kinases
Animals
Apoptosis
Blotting, Western
Breast Neoplasms*
Breast*
Cadherins
Cell Survival
Gene Expression
Genome
Humans
In Vitro Techniques
Mice
Mice, Nude
Oncogenes*
Phosphorylation
Prognosis
Real-Time Polymerase Chain Reaction
Vimentin
Weights and Measures
AMP-Activated Protein Kinases
Cadherins
Vimentin

Figure

  • Fig. 1. Analysis of differentially expressed genes in patients with breast cancer. (A) Heat map of relative mRNA expression (normalized to tumor adjacent normal tissues) in patients with breast cancer, analyzed by the Oncomine database online tool. (B) Survival analyses of patients with breast cancer and different expression levels of LAGE3, KIF11, POP1, PARN, CCDC167, DDX39, NVL, and PPAPDC1A using the OncoLnc online survival analysis tool (http://www.oncolnc.org/).

  • Fig. 2. Relative expressions of KIF11 (A), POP1 (B), and PPAPDC1A (C) in 19 tumors.

  • Fig. 3. Expression analyses of KIF11, POP1, and PPAPDC1A in breast cancer based on The Cancer Genome Atlas. (A) The relative expression of KIF11, POP1, and PPAPDC1A in breast cancer versus non-paired tumor adjacent normal tissues. ****p < 0.0001. (B) The relative expressions of KIF11, POP1, and PPAPDC1A in breast cancer versus paired tumor adjacent normal tissues.

  • Fig. 4. Overall survival (OS) (A) and recurrence-free survival (RFS) (B) in patients with breast cancer by low and high KIF11, POP1, and PPAPDC1A expressions based on The Cancer Genome Atlas. HR, hazard ratio; CI, confidence interval.

  • Fig. 5. Analysis of KIF11 in breast cancer tissue and cells. (A) KIF11 expression in patients with breast cancer of grades I-1/II-A and II-B/III. (B) The relative background expressions of KIF11 in six breast cancer cell lines. (C) Both the BT549 and the MDA231 cell line were transduced with lentivirus (Lv003), siKIF11-1, or siKIF11-2 as negative controls, and the relative mRNA levels of KIF11 were detected by quantitative real-time polymerase chain reaction. (D) Protein levels of KIF11 in BT549 and MDA231 cells were detected by Western blotting. (E) Cell viability was evaluated by MTS in both BT549 and MDA231 cells. NC, cells transduced with empty Lv003 lentivirus; MOCK, cells without any treatment; siKIF11-2, cells transduced with siKIF11-2 lentivirus. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

  • Fig. 6. Downregulation of KIF11 influenced cell apoptosis, cell proliferation, cell migration, and invasion. (A) Cell apoptosis was determined by cytometry in KIF11-suppressed BT549 and MDA231 cells. (B) Cell proliferation of the KIF11-suppressed BT549 and MDA231 cells was assessed by clone forming assays. (C) Downregulation of KIF11 inhibited the cell migration and invasion capabilities of the BT549 and MDA231 cells. NC, normal control. *p < 0.05, **p < 0.01.

  • Fig. 7. Downregulation of KIF11 inhibited tumor growth in xenograft BALB/c nu/nu mice. (A) Appearance at 5-week post-implantation of BALB/c nu/nu mice xenografted with BT549 cells that had either normal (NC) or low KIF11 (siKIF) expression. (B) The tumor volume curve in each group between days 10 and 35 post-implantation. (C) The tumor weight in each group determined at 5 weeks after implantation. (D) Western blotting confirmed the downregulation of KIF11 in siKIF11 mice. (E) Representative image of hematoxylin and eosin showing the beneficial effect of KIF11 downregulation in breast tumors at 5-week post-implantation (magnification, 100×). GAPDH, glyceraldehyde 3-phosphate dehydrogenase. *p < 0.05, **p < 0.01, ****p < 0.0001.

  • Fig. 8. Representative Western blot showing the expressions of markers of epithelial-to-mesenchymal transition (EMT) and the associations of KIF11 with the classical cancer signaling pathways. (A) Markers of EMT (E-cadherin, N-cadherin, and vimentin) were measured by Western blot in both the siKIF11 or normal BT549 and MDA231 cells. (B) Multiple key signaling pathways components (ERK, AMPK, AKT, and CREB) were measured by western blot in both the siKIF11 or normal BT549 and MDA231 cells. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as the internal standard.


Reference

References

1. Zuo TT, Zheng RS, Zeng HM, Zhang SW, Chen WQ. Female breast cancer incidence and mortality in China, 2013. Thorac Cancer. 2017; 8:214–8.
Article
2. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016; 66:115–32.
Article
3. DeSantis CE, Ma J, Goding Sauer A, Newman LA, Jemal A. Breast cancer statistics, 2017, racial disparity in mortality by state. CA Cancer J Clin. 2017; 67:439–48.
Article
4. Mishra P, Tang W, Putluri V, Dorsey TH, Jin F, Wang F, et al. ADHFE1 is a breast cancer oncogene and induces metabolic reprogramming. J Clin Invest. 2018; 128:323–40.
Article
5. Chen Y, Shi L, Zhang L, Li R, Liang J, Yu W, et al. The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer. J Biol Chem. 2008; 283:17969–78.
Article
6. Traub F, Mengel M, Luck HJ, Kreipe HH, von Wasielewski R. Prognostic impact of Skp2 and p27 in human breast cancer. Breast Cancer Res Treat. 2006; 99:185–91.
Article
7. Marcotte R, Sayad A, Brown KR, Sanchez-Garcia F, Reimand J, Haider M, et al. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell. 2016; 164:293–309.
Article
8. Boehm JS, Zhao JJ, Yao J, Kim SY, Firestein R, Dunn IF, et al. Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell. 2007; 129:1065–79.
Article
9. Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007; 9:166–80.
Article
10. Roostalu J, Hentrich C, Bieling P, Telley IA, Schiebel E, Surrey T. Directional switching of the kinesin Cin8 through motor coupling. Science. 2011; 332:94–9.
Article
11. Ma HT, Erdal S, Huang S, Poon RY. Synergism between inhibitors of Aurora A and KIF11 overcomes KIF15-dependent drug resistance. Mol Oncol. 2014; 8:1404–18.
Article
12. Hu H, Xiao X, Li S, Jia X, Guo X, Zhang Q. KIF11 mutations are a common cause of autosomal dominant familial exudative vitreoretinopathy. Br J Ophthalmol. 2016; 100:278–83.
13. Rao FQ, Cai XB, Cheng FF, Cheng W, Fang XL, Li N, et al. Mutations in LRP5,FZD4, TSPAN12, NDP, ZNF408, or KIF11 genes account for 38.7% of Chinese patients with familial exudative vitreoretinopathy. Invest Ophthalmol Vis Sci. 2017; 58:2623–9.
14. Mears K, Bakall B, Harney LA, Penticoff JA, Stone EM. Autosomal dominant microcephaly associated with congenital lymphedema and chorioretinopathy due to a novel mutation in KIF11. JAMA Ophthalmol. 2015; 133:720–1.
15. Hu C, Zhang R, Wang C, Wang J, Ma X, Lu J, et al. PPARG, KCNJ11, CDKAL1, CDKN2A-CDKN2B, IDE-KIF11-HHEX, IGF2BP2 and SLC30A8 are associated with type 2 diabetes in a Chinese population. PLoS One. 2009; 4:e7643.
Article
16. Imai T, Oue N, Nishioka M, Mukai S, Oshima T, Sakamoto N, et al. Overexpression of KIF11 in gastric cancer with intestinal mucin phenotype. Pathobiology. 2017; 84:16–24.
Article
17. Wissing MD, De Morree ES, Dezentje VO, Buijs JT, De Krijger RR, Smit VT, et al. Nuclear Eg5 (kinesin spindle protein) expression predicts docetaxel response and prostate cancer aggressiveness. Oncotarget. 2014; 5:7357–67.
Article
18. Sun D, Lu J, Ding K, Bi D, Niu Z, Cao Q, et al. The expression of Eg5 predicts a poor outcome for patients with renal cell carcinoma. Med Oncol. 2013; 30:476.
Article
19. Liu L, Liu X, Mare M, Dumont AS, Zhang H, Yan D, et al. Overexpression of Eg5 correlates with high grade astrocytic neoplasm. J Neurooncol. 2016; 126:77–80.
Article
20. Lu M, Zhu H, Wang X, Zhang D, Xiong L, Xu L, et al. The prognostic role of Eg5 expression in laryngeal squamous cell carcinoma. Pathology. 2016; 48:214–8.
Article
21. Daigo K, Takano A, Thang PM, Yoshitake Y, Shinohara M, Tohnai I, et al. Characterization of KIF11 as a novel prognostic biomarker and therapeutic target for oral cancer. Int J Oncol. 2018; 52:155–65.
Article
22. Liu M, Wang X, Yang Y, Li D, Ren H, Zhu Q, et al. Ectopic expression of the microtubule-dependent motor protein Eg5 promotes pancreatic tumourigenesis. J Pathol. 2010; 221:221–8.
Article
23. Venere M, Horbinski C, Crish JF, Jin X, Vasanji A, Major J, et al. The mitotic kinesin KIF11 is a driver of invasion, proliferation, and self-renewal in glioblastoma. Sci Transl Med. 2015; 7:304ra143.
Article
24. Sun XD, Shi XJ, Sun XO, Luo YG, Wu XJ, Yao CF, et al. Dimethylenastron suppresses human pancreatic cancer cell migration and invasion in vitro via allosteric inhibition of mitotic kinesin Eg5. Acta Pharmacol Sin. 2011; 32:1543–8.
Article
25. Planas-Silva MD, Filatova IS. Estrogen-dependent regulation of Eg5 in breast cancer cells. Anticancer Drugs. 2007; 18:773–9.
Article
26. Guido BC, Ramos LM, Nolasco DO, Nobrega CC, Andrade BY, Pic-Taylor A, et al. Impact of kinesin Eg5 inhibition by 3,4-dihydropyrimidin-2(1H)-one derivatives on various breast cancer cell features. BMC Cancer. 2015; 15:283.
Article
27. De Iuliis F, Taglieri L, Salerno G, Giuffrida A, Milana B, Giantulli S, et al. The kinesin Eg5 inhibitor K858 induces apoptosis but also survivin-related chemoresistance in breast cancer cells. Invest New Drugs. 2016; 34:399–406.
Article
28. Creighton CJ, Chang JC, Rosen JM. Epithelial-mesenchymal transition (EMT) in tumor-initiating cells and its clinical implications in breast cancer. J Mammary Gland Biol Neoplasia. 2010; 15:253–60.
Article
29. Taglieri L, Rubinacci G, Giuffrida A, Carradori S, Scarpa S. The kinesin Eg5 inhibitor K858 induces apoptosis and reverses the malignant invasive phenotype in human glioblastoma cells. Invest New Drugs. 2018; 36:28–35.
Article
30. Wang TL, Rago C, Silliman N, Ptak J, Markowitz S, Willson JK, et al. Prevalence of somatic alterations in the colorectal cancer cell genome. Proc Natl Acad Sci U S A. 2002; 99:3076–80.
Article
31. Weng AP, Ferrando AA, Lee W, Morris JP 4th, Silverman LB, Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004; 306:269–71.
32. Armstrong L, Hughes O, Yung S, Hyslop L, Stewart R, Wappler I, et al. The role of PI3K/AKT, MAPK/ERK and NFkappabeta signalling in the maintenance of human embryonic stem cell pluripotency and viability highlighted by transcriptional profiling and functional analysis. Hum Mol Genet. 2006; 15:1894–913.
33. Huang B, Fu SJ, Fan WZ, Wang ZH, Chen ZB, Guo SJ, et al. PKCepsilon inhibits isolation and stemness of side population cells via the suppression of ABCB1 transporter and PI3K/Akt, MAPK/ERK signaling in renal cell carcinoma cell line 769P. Cancer Lett. 2016; 376:148–54.
34. Hu X, Zhai Y, Kong P, Cui H, Yan T, Yang J, et al. FAT1 prevents epithelial mesenchymal transition (EMT) via MAPK/ERK signaling pathway in esophageal squamous cell cancer. Cancer Lett. 2017; 397:83–93.
Article
35. Kim D, Kim S, Koh H, Yoon SO, Chung AS, Cho KS, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J. 2001; 15:1953–62.
36. Chang F, Lee JT, Navolanic PM, Steelman LS, Shelton JG, Blalock WL, et al. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy. Leukemia. 2003; 17:590–603.
Article
37. Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, et al. SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate specificity. Cell. 2006; 127:125–37.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr