Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Sumoylation of Kif18A plays a role in regulating mitotic progression

Authors: Feikun Yang, Yan Chen, Wei Dai

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Kif18A, the kinesin-8 motor protein, plays an essential role in regulating alignment of bi-oriented chromosomes at the midzone during mitosis. Kinesin proteins, including Kif18A, are often deregulated in many types of cancers and are thought to play a critical role in cancer progression. However, little is known about the post-translational modifications of Kif18A and their effects on its biological activity.

Methods

Kif18A was identified to be a SUMO2 acceptor by using Ni-IDA resin to precipitate proteins from cells stably expressing His6-SUMO2. To identify the potential lysine residues, multi-site directed mutagenesis together with transient transfection and Ni-IDA pull-down assay were carried out. The confocal time-lapse imaging and immunofluorescent staining were used to study the roles of SUMO2 modification on Kif18A’s activity during the cell cycle.

Results

Kif18A is covalently modified by SUMO2 during the cell cycle, and its sumoylation peaks at metaphase and then rapidly decreases upon anaphase onset. Mutational analysis identifies multiple lysine residues (K148, K442, K533, K660 and K683) as potential SUMO acceptors. The functional studies reveal that sumoylation of Kif18A has little effect on protein stability and subcellular localization. However, compared with the wild-type control, ectopic expression of SUMO-resistant mutants of Kif18A results in a significant delay of mitotic exit. Confocal microscopy shows that cells expressing SUMO-resistant Kif18A display a compromised dissociation of BubR1 from kinetochores after anaphase onset.

Conclusions

Our studies reveal that sumoylation functions as an unidentified form of post-translational modification that regulates Kif18A activity during mitotic progression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Mayr MI, Hummer S, Bormann J, Gruner T, Adio S, Woehlke G, et al. The human kinesin Kif18A is a motile microtubule depolymerase essential for chromosome congression. Curr Biol. 2007;17:488–98.CrossRefPubMed Mayr MI, Hummer S, Bormann J, Gruner T, Adio S, Woehlke G, et al. The human kinesin Kif18A is a motile microtubule depolymerase essential for chromosome congression. Curr Biol. 2007;17:488–98.CrossRefPubMed
2.
go back to reference Du Y, English CA, Ohi R. The kinesin-8Kif18A dampens microtubule plus-end dynamics. Curr Biol. 2010;20:374–80.CrossRefPubMed Du Y, English CA, Ohi R. The kinesin-8Kif18A dampens microtubule plus-end dynamics. Curr Biol. 2010;20:374–80.CrossRefPubMed
3.
go back to reference Stumpff J, Du Y, English CA, Maliga Z, Wagenbach M, Asbury CL, et al. A tethering mechanism controls the processivity and kinetochore-microtubule plus-end enrichment of the kinesin-8 Kif18A. Mol Cell. 2011;43:764–75.CrossRefPubMedPubMedCentral Stumpff J, Du Y, English CA, Maliga Z, Wagenbach M, Asbury CL, et al. A tethering mechanism controls the processivity and kinetochore-microtubule plus-end enrichment of the kinesin-8 Kif18A. Mol Cell. 2011;43:764–75.CrossRefPubMedPubMedCentral
4.
go back to reference Stumpff J, von Dassow G, Wagenbach M, Asbury C, Wordeman L. The kinesin-8 motor Kif18A suppresses kinetochore movements to control mitotic chromosome alignment. Dev Cell. 2008;14:252–62.CrossRefPubMedPubMedCentral Stumpff J, von Dassow G, Wagenbach M, Asbury C, Wordeman L. The kinesin-8 motor Kif18A suppresses kinetochore movements to control mitotic chromosome alignment. Dev Cell. 2008;14:252–62.CrossRefPubMedPubMedCentral
6.
go back to reference Huang Y, Yao Y, Xu HZ, Wang ZG, Lu L, Dai W. Defects in chromosome congression and mitotic progression in KIF18A-deficient cells are partly mediated through impaired functions of CENP-E. Cell Cycle. 2009;8:2643–9.CrossRefPubMedPubMedCentral Huang Y, Yao Y, Xu HZ, Wang ZG, Lu L, Dai W. Defects in chromosome congression and mitotic progression in KIF18A-deficient cells are partly mediated through impaired functions of CENP-E. Cell Cycle. 2009;8:2643–9.CrossRefPubMedPubMedCentral
7.
go back to reference Liu XS, Zhao XD, Wang X, Yao YX, Zhang LL, Shu RZ, et al. Germinal cell aplasia in Kif18a mutant male mice Due to impaired chromosome congression and dysregulated BubR1 and CENP-E. Genes Cancer. 2010;1:26–39.CrossRefPubMedPubMedCentral Liu XS, Zhao XD, Wang X, Yao YX, Zhang LL, Shu RZ, et al. Germinal cell aplasia in Kif18a mutant male mice Due to impaired chromosome congression and dysregulated BubR1 and CENP-E. Genes Cancer. 2010;1:26–39.CrossRefPubMedPubMedCentral
8.
go back to reference Yu Y, Feng YM. The role of kinesin family proteins in tumorigenesis and progression: potential biomarkers and molecular targets for cancer therapy. Cancer. 2010;116:5150–60.CrossRefPubMed Yu Y, Feng YM. The role of kinesin family proteins in tumorigenesis and progression: potential biomarkers and molecular targets for cancer therapy. Cancer. 2010;116:5150–60.CrossRefPubMed
10.
go back to reference Zou JX, Duan Z, Wang J, Sokolov A, Xu J, Chen CZ, et al. Kinesin family deregulation coordinated by bromodomain protein ANCCA and histone methyltransferase MLL for breast cancer cell growth, survival and tamoxifen resistance. Mol Cancer Res. 2014;2:539–49.CrossRef Zou JX, Duan Z, Wang J, Sokolov A, Xu J, Chen CZ, et al. Kinesin family deregulation coordinated by bromodomain protein ANCCA and histone methyltransferase MLL for breast cancer cell growth, survival and tamoxifen resistance. Mol Cancer Res. 2014;2:539–49.CrossRef
11.
go back to reference Zhang C, Zhu C, Chen H, Li L, Guo L, Jiang W, et al. Kif18A is involved in human breast carcinogenesis. Carcinogenesis. 2010;31:1676–84.CrossRefPubMed Zhang C, Zhu C, Chen H, Li L, Guo L, Jiang W, et al. Kif18A is involved in human breast carcinogenesis. Carcinogenesis. 2010;31:1676–84.CrossRefPubMed
12.
go back to reference Nagahara M, Nishida N, Iwatsuki M, Ishimaru S, Mimori K, Tanaka F, et al. Kinesin 18A expression: clinical relevance to colorectal cancer progression. Int J Cancer. 2011;129:2543–52.CrossRefPubMed Nagahara M, Nishida N, Iwatsuki M, Ishimaru S, Mimori K, Tanaka F, et al. Kinesin 18A expression: clinical relevance to colorectal cancer progression. Int J Cancer. 2011;129:2543–52.CrossRefPubMed
13.
go back to reference Zhu H, Xu W, Zhang H, Liu J, Xu H, Lu S, et al. Targeted deletion of Kif18a protects from colitis-associated colorectal (CAC) tumors in mice through impairing Akt phosphorylation. Biochem Biophys Res Commun. 2013;438:97–102.CrossRefPubMed Zhu H, Xu W, Zhang H, Liu J, Xu H, Lu S, et al. Targeted deletion of Kif18a protects from colitis-associated colorectal (CAC) tumors in mice through impairing Akt phosphorylation. Biochem Biophys Res Commun. 2013;438:97–102.CrossRefPubMed
14.
go back to reference Rucksaken R, Khoontawad J, Roytrakul S, Pinlaor P, Hiraku Y, Wongkham C, et al. Proteomic analysis to identify plasma orosomucoid 2 and kinesin 18A as potential biomarkers of cholangiocarcinoma. Cancer Biomark. 2012;12:81–95.PubMed Rucksaken R, Khoontawad J, Roytrakul S, Pinlaor P, Hiraku Y, Wongkham C, et al. Proteomic analysis to identify plasma orosomucoid 2 and kinesin 18A as potential biomarkers of cholangiocarcinoma. Cancer Biomark. 2012;12:81–95.PubMed
15.
go back to reference Tooker BC, Newman LS, Bowler RP, Karjalainen A, Oksa P, Vainio H, et al. Proteomic detection of cancer in asbestosis patients using SELDI-TOF discovered serum protein biomarkers. Biomarkers. 2011;16:181–91.CrossRefPubMed Tooker BC, Newman LS, Bowler RP, Karjalainen A, Oksa P, Vainio H, et al. Proteomic detection of cancer in asbestosis patients using SELDI-TOF discovered serum protein biomarkers. Biomarkers. 2011;16:181–91.CrossRefPubMed
16.
go back to reference Vagnoni A, Rodriguez L, Manser C, De Vos KJ, Miller CC. Phosphorylation of kinesin light chain 1 at serine 460 modulates binding and trafficking of calsyntenin-1. J Cell Sci. 2011;124:1032–42.CrossRefPubMedPubMedCentral Vagnoni A, Rodriguez L, Manser C, De Vos KJ, Miller CC. Phosphorylation of kinesin light chain 1 at serine 460 modulates binding and trafficking of calsyntenin-1. J Cell Sci. 2011;124:1032–42.CrossRefPubMedPubMedCentral
17.
go back to reference Knowlton AL, Vorozhko VV, Lan W, Gorbsky GJ, Stukenberg PT. ICIS and Aurora B coregulate the microtubule depolymerase Kif2a. Curr Biol. 2009;19:758–63.CrossRefPubMedPubMedCentral Knowlton AL, Vorozhko VV, Lan W, Gorbsky GJ, Stukenberg PT. ICIS and Aurora B coregulate the microtubule depolymerase Kif2a. Curr Biol. 2009;19:758–63.CrossRefPubMedPubMedCentral
18.
go back to reference Uehara R, Tsukada Y, Kamasaki T, Poser I, Yoda K, Gerlich DW, et al. Aurora B and Kif2A control microtubule length for assembly of a functional central spindle during anaphase. J Cell Biol. 2013;202:623–36.CrossRefPubMedPubMedCentral Uehara R, Tsukada Y, Kamasaki T, Poser I, Yoda K, Gerlich DW, et al. Aurora B and Kif2A control microtubule length for assembly of a functional central spindle during anaphase. J Cell Biol. 2013;202:623–36.CrossRefPubMedPubMedCentral
19.
go back to reference Zhang XD, Goeres J, Zhang H, Yen TJ, Porter AC, Matunis MJ. SUMO-2/3 modification and binding regulate the association of CENP-E with kinetochores and progression through mitosis. Mol Cell. 2008;29:729–41.CrossRefPubMedPubMedCentral Zhang XD, Goeres J, Zhang H, Yen TJ, Porter AC, Matunis MJ. SUMO-2/3 modification and binding regulate the association of CENP-E with kinetochores and progression through mitosis. Mol Cell. 2008;29:729–41.CrossRefPubMedPubMedCentral
20.
go back to reference Mayr MI. Functional characterization of the mitotic kinesin-like protein Kif18A, doctorate. Konstanz, Germany: University of Konstanz, Department of Biology and Konstanz Research School Chemical Biology, University of Konstanz; 2010. Mayr MI. Functional characterization of the mitotic kinesin-like protein Kif18A, doctorate. Konstanz, Germany: University of Konstanz, Department of Biology and Konstanz Research School Chemical Biology, University of Konstanz; 2010.
21.
go back to reference Sedgwick GG, Hayward DG, Di Fiore B, Pardo M, Yu L, Pines J, et al. Mechanisms controlling the temporal degradation of Nek2A and Kif18A by the APC/C-Cdc20 complex. EMBO J. 2013;32:303–14.CrossRefPubMedPubMedCentral Sedgwick GG, Hayward DG, Di Fiore B, Pardo M, Yu L, Pines J, et al. Mechanisms controlling the temporal degradation of Nek2A and Kif18A by the APC/C-Cdc20 complex. EMBO J. 2013;32:303–14.CrossRefPubMedPubMedCentral
22.
go back to reference Singh SA, Winter D, Kirchner M, Chauhan R, Ahmed S, Ozlu N, et al. Co-regulation proteomics reveals substrates and mechanisms of APC/C-dependent degradation. EMBO J. 2014;33:385–99.CrossRefPubMedPubMedCentral Singh SA, Winter D, Kirchner M, Chauhan R, Ahmed S, Ozlu N, et al. Co-regulation proteomics reveals substrates and mechanisms of APC/C-dependent degradation. EMBO J. 2014;33:385–99.CrossRefPubMedPubMedCentral
23.
24.
25.
go back to reference Tatham MH, Rodriguez MS, Xirodimas DP, Hay RT. Detection of protein SUMOylation in vivo. Nat Protoc. 2009;4:1363–71.CrossRefPubMed Tatham MH, Rodriguez MS, Xirodimas DP, Hay RT. Detection of protein SUMOylation in vivo. Nat Protoc. 2009;4:1363–71.CrossRefPubMed
26.
go back to reference Masuda N, Shimodaira T, Shiu SJ, Tokai-Nishizumi N, Yamamoto T, Ohsugi M. Microtubule stabilization triggers the plus-end accumulation of Kif18A/kinesin-8. Cell Struct Funct. 2011;36:261–7.CrossRefPubMed Masuda N, Shimodaira T, Shiu SJ, Tokai-Nishizumi N, Yamamoto T, Ohsugi M. Microtubule stabilization triggers the plus-end accumulation of Kif18A/kinesin-8. Cell Struct Funct. 2011;36:261–7.CrossRefPubMed
27.
go back to reference Mukhopadhyay D, Dasso M. Modification in reverse: the SUMO proteases. Trends Biochem Sci. 2007;32:286–95.CrossRefPubMed Mukhopadhyay D, Dasso M. Modification in reverse: the SUMO proteases. Trends Biochem Sci. 2007;32:286–95.CrossRefPubMed
28.
go back to reference Flotho A, Melchior F. Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem. 2013;82:357–85.CrossRefPubMed Flotho A, Melchior F. Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem. 2013;82:357–85.CrossRefPubMed
29.
go back to reference Bassi C, Ho J, Srikumar T, Dowling RJ, Gorrini C, Miller SJ, et al. Nuclear PTEN controls DNA repair and sensitivity to genotoxic stress. Science. 2013;341:395–9.CrossRefPubMed Bassi C, Ho J, Srikumar T, Dowling RJ, Gorrini C, Miller SJ, et al. Nuclear PTEN controls DNA repair and sensitivity to genotoxic stress. Science. 2013;341:395–9.CrossRefPubMed
30.
go back to reference Wohlschlegel JA, Johnson ES, Reed SI, Yates 3rd JR. Global analysis of protein sumoylation in Saccharomyces cerevisiae. J Biol Chem. 2004;279:45662–8.CrossRefPubMed Wohlschlegel JA, Johnson ES, Reed SI, Yates 3rd JR. Global analysis of protein sumoylation in Saccharomyces cerevisiae. J Biol Chem. 2004;279:45662–8.CrossRefPubMed
31.
go back to reference Denison C, Rudner AD, Gerber SA, Bakalarski CE, Moazed D, Gygi SP. A proteomic strategy for gaining insights into protein sumoylation in yeast. Mol Cell Proteomics. 2005;4:246–54.CrossRefPubMed Denison C, Rudner AD, Gerber SA, Bakalarski CE, Moazed D, Gygi SP. A proteomic strategy for gaining insights into protein sumoylation in yeast. Mol Cell Proteomics. 2005;4:246–54.CrossRefPubMed
32.
go back to reference Montpetit B, Hazbun TR, Fields S, Hieter P. Sumoylation of the budding yeast kinetochore protein Ndc10 is required for Ndc10 spindle localization and regulation of anaphase spindle elongation. J Cell Biol. 2006;174:653–63.CrossRefPubMedPubMedCentral Montpetit B, Hazbun TR, Fields S, Hieter P. Sumoylation of the budding yeast kinetochore protein Ndc10 is required for Ndc10 spindle localization and regulation of anaphase spindle elongation. J Cell Biol. 2006;174:653–63.CrossRefPubMedPubMedCentral
33.
34.
go back to reference Yang F, Hu L, Chen C, Yu J, O’Connell CB, Khodjakov A, et al. BubR1 is modified by sumoylation during mitotic progression. J Biol Chem. 2012;287:4875–82.CrossRefPubMed Yang F, Hu L, Chen C, Yu J, O’Connell CB, Khodjakov A, et al. BubR1 is modified by sumoylation during mitotic progression. J Biol Chem. 2012;287:4875–82.CrossRefPubMed
35.
go back to reference Garcia MA, Koonrugsa N, Toda T. Spindle-kinetochore attachment requires the combined action of Kin I-like Klp5/6 and Alp14/Dis1-MAPs in fission yeast. EMBO J. 2002;21:6015–24.CrossRefPubMedPubMedCentral Garcia MA, Koonrugsa N, Toda T. Spindle-kinetochore attachment requires the combined action of Kin I-like Klp5/6 and Alp14/Dis1-MAPs in fission yeast. EMBO J. 2002;21:6015–24.CrossRefPubMedPubMedCentral
36.
go back to reference Gupta Jr ML, Carvalho P, Roof DM, Pellman D. Plus end-specific depolymerase activity of Kip3, a kinesin-8 protein, explains its role in positioning the yeast mitotic spindle. Nat Cell Biol. 2006;8:913–23.CrossRefPubMed Gupta Jr ML, Carvalho P, Roof DM, Pellman D. Plus end-specific depolymerase activity of Kip3, a kinesin-8 protein, explains its role in positioning the yeast mitotic spindle. Nat Cell Biol. 2006;8:913–23.CrossRefPubMed
37.
go back to reference Rao CV, Yamada HY, Yao Y, Dai W. Enhanced genomic instabilities caused by deregulated microtubule dynamics and chromosome segregation: a perspective from genetic studies in mice. Carcinogenesis. 2009;30:1469–74.CrossRefPubMedPubMedCentral Rao CV, Yamada HY, Yao Y, Dai W. Enhanced genomic instabilities caused by deregulated microtubule dynamics and chromosome segregation: a perspective from genetic studies in mice. Carcinogenesis. 2009;30:1469–74.CrossRefPubMedPubMedCentral
38.
go back to reference Stumpff J, Wagenbach M, Franck A, Asbury CL, Wordeman L. Kif18A and chromokinesins confine centromere movements via microtubule growth suppression and spatial control of kinetochore tension. Dev Cell. 2012;22:1017–29.CrossRefPubMedPubMedCentral Stumpff J, Wagenbach M, Franck A, Asbury CL, Wordeman L. Kif18A and chromokinesins confine centromere movements via microtubule growth suppression and spatial control of kinetochore tension. Dev Cell. 2012;22:1017–29.CrossRefPubMedPubMedCentral
39.
go back to reference Muller S, Matunis MJ, Dejean A. Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus. EMBO J. 1998;17:61–70.CrossRefPubMedPubMedCentral Muller S, Matunis MJ, Dejean A. Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus. EMBO J. 1998;17:61–70.CrossRefPubMedPubMedCentral
40.
go back to reference Gostissa M, Hengstermann A, Fogal V, Sandy P, Schwarz SE, Scheffner M, et al. Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. EMBO J. 1999;18:6462–71.CrossRefPubMedPubMedCentral Gostissa M, Hengstermann A, Fogal V, Sandy P, Schwarz SE, Scheffner M, et al. Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. EMBO J. 1999;18:6462–71.CrossRefPubMedPubMedCentral
41.
go back to reference Rodriguez MS, Desterro JM, Lain S, Midgley CA, Lane DP, Hay RT. SUMO-1 modification activates the transcriptional response of p53. EMBO J. 1999;18:6455–61.CrossRefPubMedPubMedCentral Rodriguez MS, Desterro JM, Lain S, Midgley CA, Lane DP, Hay RT. SUMO-1 modification activates the transcriptional response of p53. EMBO J. 1999;18:6455–61.CrossRefPubMedPubMedCentral
42.
go back to reference Buschmann T, Fuchs SY, Lee CG, Pan ZQ, Ronai Z. SUMO-1 modification of Mdm2 prevents its self-ubiquitination and increases Mdm2 ability to ubiquitinate p53. Cell. 2000;101:753–62.CrossRefPubMed Buschmann T, Fuchs SY, Lee CG, Pan ZQ, Ronai Z. SUMO-1 modification of Mdm2 prevents its self-ubiquitination and increases Mdm2 ability to ubiquitinate p53. Cell. 2000;101:753–62.CrossRefPubMed
43.
go back to reference Muller S, Berger M, Lehembre F, Seeler JS, Haupt Y, Dejean A. c-Jun and p53 activity is modulated by SUMO-1 modification. J Biol Chem. 2000;275:13321–9.CrossRefPubMed Muller S, Berger M, Lehembre F, Seeler JS, Haupt Y, Dejean A. c-Jun and p53 activity is modulated by SUMO-1 modification. J Biol Chem. 2000;275:13321–9.CrossRefPubMed
Metadata
Title
Sumoylation of Kif18A plays a role in regulating mitotic progression
Authors
Feikun Yang
Yan Chen
Wei Dai
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1226-9

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine