Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zhikai Wang is active.

Publication


Featured researches published by Zhikai Wang.


Journal of Molecular Cell Biology | 2011

Aurora B kinase activation requires survivin priming phosphorylation by PLK1

Youjun Chu; Phil Yao; Wenwen Wang; Dongmei Wang; Zhikai Wang; Liangyu Zhang; Yuejia Huang; Yuwen Ke; Xia Ding; Xuebiao Yao

During cell division, chromosome segregation is orchestrated by the interaction of spindle microtubules with the centromere. Accurate attachment of spindle microtubules to kinetochore requires the chromosomal passenger of Aurora B kinase complex with borealin, INCENP and survivin (SUR). The current working model argues that SUR is responsible for docking Aurora B to the centromere whereas its precise role in Aurora B activation has been unclear. Here, we show that Aurora B kinase activation requires SUR priming phosphorylation at Ser20 which is catalyzed by polo-like kinase 1 (PLK1). Inhibition of PLK1 kinase activity or expression of non-phosphorylatable SUR mutant prevents Aurora B activation and correct spindle microtubule attachment. The PLK1-mediated regulation of Aurora B kinase activity was examined in real-time mitosis using fluorescence resonance energy transfer-based reporter and quantitative analysis of native Aurora B substrate phosphorylation. We reason that the PLK1-mediated priming phosphorylation is critical for orchestrating Aurora B activity in centromere which is essential for accurate chromosome segregation and faithful completion of cytokinesis.


Journal of Biological Chemistry | 2011

CENP-U Cooperates with Hec1 to Orchestrate Kinetochore-Microtubule Attachment

Shasha Hua; Zhikai Wang; Kai Jiang; Yuejia Huang; Tarsha Ward; Lingli Zhao; Zhen Dou; Xuebiao Yao

Mitosis is an orchestration of dynamic interaction between chromosomes and spindle microtubules by which genomic materials are equally distributed into two daughter cells. Previous studies showed that CENP-U is a constitutive centromere component essential for proper chromosome segregation. However, the precise molecular mechanism has remained elusive. Here, we identified CENP-U as a novel interacting partner of Hec1, an evolutionarily conserved kinetochore core component essential for chromosome plasticity. Suppression of CENP-U by shRNA resulted in mitotic defects with an impaired kinetochore-microtubule attachment. Interestingly, CENP-U not only binds microtubules directly but also displays a cooperative microtubule binding activity with Hec1 in vitro. Furthermore, we showed that CENP-U is a substrate of Aurora-B. Importantly, phosphorylation of CENP-U leads to reduced kinetochore-microtubule interaction, which contributes to the error-correcting function of Aurora-B. Taken together, our results indicate that CENP-U is a novel microtubule binding protein and plays an important role in kinetochore-microtubule attachment through its interaction with Hec1.


Proceedings of the National Academy of Sciences of the United States of America | 2012

EB1 acetylation by P300/CBP-associated factor (PCAF) ensures accurate kinetochore-microtubule interactions in mitosis

Peng Xia; Zhikai Wang; Xing Liu; Bing Wu; Juncheng Wang; Tarsha Ward; Liangyu Zhang; Xia Ding; Gary H. Gibbons; Yunyu Shi; Xuebiao Yao

In eukaryotes, microtubules are essential for cellular plasticity and dynamics. Here we show that P300/CBP-associated factor (PCAF), a kinetochore-associated acetyltransferase, acts as a negative modulator of microtubule stability through acetylation of EB1, a protein that controls the plus ends of microtubules. PCAF acetylates EB1 on K220 and disrupts the stability of a hydrophobic cavity on the dimerized EB1 C terminus, which was previously reported to interact with plus-end tracking proteins (TIPs) containing the SxIP motif. As determined with an EB1 acetyl-K220–specific antibody, K220 acetylation is dramatically increased in mitosis and localized to the spindle microtubule plus ends. Surprisingly, persistent acetylation of EB1 delays metaphase alignment, resulting in impaired checkpoint silencing. Consequently, suppression of Mad2 overrides mitotic arrest induced by persistent EB1 acetylation. Thus, our findings identify dynamic acetylation of EB1 as a molecular mechanism to orchestrate accurate kinetochore–microtubule interactions in mitosis. These results establish a previously uncharacterized regulatory mechanism governing localization of microtubule plus-end tracking proteins and thereby the plasticity and dynamics of cells.


Journal of Biological Chemistry | 2009

PRC1 Cooperates with CLASP1 to Organize Central Spindle Plasticity in Mitosis

Jing Liu; Zhikai Wang; Kai Jiang; Liangyu Zhang; Lingli Zhao; Shasha Hua; Feng Yan; Yong Yang; Dongmei Wang; Chuanhai Fu; Xia Ding; Zhen Guo; Xuebiao Yao

During cell division, chromosome segregation is governed by the interaction of spindle microtubules with the kinetochore. A dramatic remodeling of interpolar microtubules into an organized central spindle between the separating chromatids is required for the initiation and execution of cytokinesis. Central spindle organization requires mitotic kinesins, microtubule-bundling protein PRC1, and Aurora B kinase complex. However, the precise role of PRC1 in central spindle organization has remained elusive. Here we show that PRC1 recruits CLASP1 to the central spindle at early anaphase onset. CLASP1 belongs to a conserved microtubule-binding protein family that mediates the stabilization of overlapping microtubules of the central spindle. PRC1 physically interacts with CLASP1 and specifies its localization to the central spindle. Repression of CLASP1 leads to sister-chromatid bridges and depolymerization of spindle midzone microtubules. Disruption of PRC1-CLASP1 interaction by a membrane-permeable peptide abrogates accurate chromosome segregation, resulting in sister chromatid bridges. These findings reveal a key role for the PRC1-CLASP1 interaction in achieving a stable anti-parallel microtubule organization essential for faithful chromosome segregation. We propose that PRC1 forms a link between stabilization of CLASP1 association with central spindle microtubules and anti-parallel microtubule elongation.


Journal of Molecular Cell Biology | 2014

Aurora A orchestrates entosis by regulating a dynamic MCAK–TIP150 interaction

Peng Xia; Jinhua Zhou; Xiaoyu Song; Bing Wu; Xing Liu; Di Li; Shuyuan Zhang; Zhikai Wang; Huijuan Yu; Tarsha Ward; Jiancun Zhang; Yinmei Li; Xiaoning Wang; Yong Chen; Zhen Guo; Xuebiao Yao

Entosis, a cell-in-cell process, has been implicated in the formation of aneuploidy associated with an aberrant cell division control. Microtubule plus-end-tracking protein TIP150 facilitates the loading of MCAK onto the microtubule plus ends and orchestrates microtubule plus-end dynamics during cell division. Here we show that TIP150 cooperates with MCAK to govern entosis via a regulatory circuitry that involves Aurora A-mediated phosphorylation of MCAK. Our biochemical analyses show that MCAK forms an intra-molecular association, which is essential for TIP150 binding. Interestingly, Aurora A-mediated phosphorylation of MCAK modulates its intra-molecular association, which perturbs the MCAK-TIP150 interaction in vitro and inhibits entosis in vivo. To probe if MCAK-TIP150 interaction regulates microtubule plasticity to affect the mechanical properties of cells during entosis, we used an optical trap to measure the mechanical rigidity of live MCF7 cells. We find that the MCAK cooperates with TIP150 to promote microtubule dynamics and modulate the mechanical rigidity of the cells during entosis. Our results show that a dynamic interaction of MCAK-TIP150 orchestrated by Aurora A-mediated phosphorylation governs entosis via regulating microtubule plus-end dynamics and cell rigidity. These data reveal a previously unknown mechanism of Aurora A regulation in the control of microtubule plasticity during cell-in-cell processes.


Journal of Biological Chemistry | 2013

Regulation of a Dynamic Interaction between Two Microtubule-binding Proteins, EB1 and TIP150, by the Mitotic p300/CBP-associated Factor (PCAF) Orchestrates Kinetochore Microtubule Plasticity and Chromosome Stability during Mitosis

Tarsha Ward; Ming Wang; Xing Liu; Zhikai Wang; Peng Xia; Youjun Chu; Xiwei Wang; Lifang Liu; Kai Jiang; Huijuan Yu; Maomao Yan; Jianyu Wang; Donald L. Hill; Yuejia Huang; Tongge Zhu; Xuebiao Yao

Background: TIP150 is a unique microtubule plus-end tracking protein essential for mitotic regulation. Results: TIP150 cooperates with EB1 to mediate mitotic chromosome segregation and checkpoint control. Conclusion: The EB1-TIP150-PCAF interaction is essential for kinetochore-microtubule plus-end dynamics and chromosome plasticity in mitosis. Significance: EB1-TIP150 interaction orchestrated by PCAF governs chromosome dynamics during cell division. The microtubule cytoskeleton network orchestrates cellular dynamics and chromosome stability in mitosis. Although tubulin acetylation is essential for cellular plasticity, it has remained elusive how kinetochore microtubule plus-end dynamics are regulated by p300/CBP-associated factor (PCAF) acetylation in mitosis. Here, we demonstrate that the plus-end tracking protein, TIP150, regulates dynamic kinetochore-microtubule attachments by promoting the stability of spindle microtubule plus-ends. Suppression of TIP150 by siRNA results in metaphase alignment delays and perturbations in chromosome biorientation. TIP150 is a tetramer that binds an end-binding protein (EB1) dimer through the C-terminal domains, and overexpression of the C-terminal TIP150 or disruption of the TIP150-EB1 interface by a membrane-permeable peptide perturbs chromosome segregation. Acetylation of EB1-PCAF regulates the TIP150 interaction, and persistent acetylation perturbs EB1-TIP150 interaction and accurate metaphase alignment, resulting in spindle checkpoint activation. Suppression of the mitotic checkpoint serine/threonine protein kinase, BubR1, overrides mitotic arrest induced by impaired EB1-TIP150 interaction, but cells exhibit whole chromosome aneuploidy. Thus, the results identify a mechanism by which the TIP150-EB1 interaction governs kinetochore microtubule plus-end plasticity and establish that the temporal control of the TIP150-EB1 interaction by PCAF acetylation ensures chromosome stability in mitosis.


Scientific Reports | 2013

DDA3 associates with microtubule plus ends and orchestrates microtubule dynamics and directional cell migration

Liangyu Zhang; Hengyi Shao; Tongge Zhu; Peng Xia; Zhikai Wang; Lifang Liu; Maomao Yan; Donald L. Hill; Guowei Fang; Zhengjun Chen; Dongmei Wang; Xuebiao Yao

Cell motility and adhesion involve orchestrated interaction of microtubules (MTs) with their plus-end tracking proteins (+TIPs). However, the mechanisms underlying regulations of MT dynamics and directional cell migration are still elusive. Here, we show that DDA3-EB1 interaction orchestrates MT plus-end dynamics and facilitates directional cell migration. Biochemical characterizations reveal that DDA3 interacts with EB1 via its SxIP motif within the C-terminal Pro/Ser-rich region. Time-lapse and total internal reflection fluorescence (TIRF) microscopic assays demonstrate that DDA3 exhibits EB1-dependent, MT plus-end loading and tracking. The EB1-based loading of DDA3 is responsible for MT plus-ends stabilization at the cell cortex, which in turn orchestrates directional cell migration. Interestingly, the DDA3-EB1 interaction is potentially regulated by EB1 acetylation, which may account for physiological regulation underlying EGF-elicited cell migration. Thus, the EB1-based function of DDA3 links MT dynamics to directional cell migration.


Scientific Reports | 2015

Regulation of NDR1 activity by PLK1 ensures proper spindle orientation in mitosis

Maomao Yan; Lingluo Chu; Bo Qin; Zhikai Wang; Xing Liu; Changjiang Jin; Guanglan Zhang; Marta Gomez; Alexander Hergovich; Zhengjun Chen; Ping He; Xinjiao Gao; Xuebiao Yao

Accurate chromosome segregation during mitosis requires the physical separation of sister chromatids which depends on correct position of mitotic spindle relative to membrane cortex. Although recent work has identified the role of PLK1 in spindle orientation, the mechanisms underlying PLK1 signaling in spindle positioning and orientation have not been fully illustrated. Here, we identified a conserved signaling axis in which NDR1 kinase activity is regulated by PLK1 in mitosis. PLK1 phosphorylates NDR1 at three putative threonine residues (T7, T183 and T407) at mitotic entry, which elicits PLK1-dependent suppression of NDR1 activity and ensures correct spindle orientation in mitosis. Importantly, persistent expression of non-phosphorylatable NDR1 mutant perturbs spindle orientation. Mechanistically, PLK1-mediated phosphorylation protects the binding of Mob1 to NDR1 and subsequent NDR1 activation. These findings define a conserved signaling axis that integrates dynamic kinetochore-microtubule interaction and spindle orientation control to genomic stability maintenance.


Journal of Biological Chemistry | 2015

Mitotic Protein CSPP1 Interacts with CENP-H Protein to Coordinate Accurate Chromosome Oscillation in Mitosis.

Lijuan Zhu; Zhikai Wang; Wenwen Wang; Chunli Wang; Shasha Hua; Zeqi Su; Larry Brako; Minerva T. Garcia-Barrio; Mingliang Ye; Xuan Wei; Hanfa Zou; Xia Ding; Lifang Liu; Xing Liu; Xuebiao Yao

Background: Kinetochore microtubule dynamics orchestrate proper chromosome movement in mitosis. Results: CSPP1 is a novel kinetochore protein that regulates chromosome oscillation during cell division. Conclusion: CSPP1 associates with CENP-H to control chromosome oscillation by regulating kinetochore microtubule dynamics. Significance: Our findings provide insight into the molecular mechanism of chromosome movements. Mitotic chromosome segregation is orchestrated by the dynamic interaction of spindle microtubules with the kinetochores. During chromosome alignment, kinetochore-bound microtubules undergo dynamic cycles between growth and shrinkage, leading to an oscillatory movement of chromosomes along the spindle axis. Although kinetochore protein CENP-H serves as a molecular control of kinetochore-microtubule dynamics, the mechanistic link between CENP-H and kinetochore microtubules (kMT) has remained less characterized. Here, we show that CSPP1 is a kinetochore protein essential for accurate chromosome movements in mitosis. CSPP1 binds to CENP-H in vitro and in vivo. Suppression of CSPP1 perturbs proper mitotic progression and compromises the satisfaction of spindle assembly checkpoint. In addition, chromosome oscillation is greatly attenuated in CSPP1-depleted cells, similar to what was observed in the CENP-H-depleted cells. Importantly, CSPP1 depletion enhances velocity of kinetochore movement, and overexpression of CSPP1 decreases the speed, suggesting that CSPP1 promotes kMT stability during cell division. Specific perturbation of CENP-H/CSPP1 interaction using a membrane-permeable competing peptide resulted in a transient mitotic arrest and chromosome segregation defect. Based on these findings, we propose that CSPP1 cooperates with CENP-H on kinetochores to serve as a novel regulator of kMT dynamics for accurate chromosome segregation.


Journal of Biological Chemistry | 2016

The Microtubule Plus End Tracking Protein TIP150 Interacts with Cortactin to Steer Directional Cell Migration.

Gregory Adams; Jiajia Zhou; Wenwen Wang; Huihui Wu; Jie Quan; Yingying Liu; Peng Xia; Zhikai Wang; Shu Zhou; Jiying Jiang; Fei Mo; Xiaoxuan Zhuang; Kelwyn Thomas; Donald L. Hill; Felix O. Aikhionbare; Ping He; Xing Liu; Xia Ding; Xuebiao Yao

Cell migration is orchestrated by dynamic interactions of microtubules with the plasma membrane cortex. How these interactions facilitate these dynamic processes is still being actively investigated. TIP150 is a newly characterized microtubule plus end tracking protein essential for mitosis and entosis (Ward, T., Wang, M., Liu, X., Wang, Z., Xia, P., Chu, Y., Wang, X., Liu, L., Jiang, K., Yu, H., Yan, M., Wang, J., Hill, D. L., Huang, Y., Zhu, T., and Yao, X. (2013) Regulation of a dynamic interaction between two microtubule-binding proteins, EB1 and TIP150, by the mitotic p300/CBP-associated factor (PCAF) orchestrates kinetochore microtubule plasticity and chromosome stability during mitosis. J. Biol. Chem. 288, 15771–15785; Xia, P., Zhou, J., Song, X., Wu, B., Liu, X., Li, D., Zhang, S., Wang, Z., Yu, H., Ward, T., Zhang, J., Li, Y., Wang, X., Chen, Y., Guo, Z., and Yao, X. (2014) Aurora A orchestrates entosis by regulating a dynamic MCAK-TIP150 interaction. J. Mol. Cell Biol. 6, 240–254). Here we show that TIP150 links dynamic microtubules to steer cell migration by interacting with cortactin. Mechanistically, TIP150 binds to cortactin via its C-terminal tail. Interestingly, the C-terminal TIP150 proline-rich region (CT150) binds to the Src homology 3 domain of cortactin specifically, and such an interaction is negatively regulated by EGF-elicited tyrosine phosphorylation of cortactin. Importantly, suppression of TIP150 or overexpression of phospho-mimicking cortactin inhibits polarized cell migration. In addition, CT150 disrupts the biochemical interaction between TIP150 and cortactin in vitro, and perturbation of the TIP150-cortactin interaction in vivo using a membrane-permeable TAT-CT150 peptide results in an inhibition of directional cell migration. We reason that a dynamic TIP150-cortactin interaction orchestrates directional cell migration via coupling dynamic microtubule plus ends to the cortical cytoskeleton.

Collaboration


Dive into the Zhikai Wang's collaboration.

Top Co-Authors

Avatar

Xuebiao Yao

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Xing Liu

Morehouse School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Peng Xia

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar

Xia Ding

Beijing University of Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Liangyu Zhang

Morehouse School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tarsha Ward

Morehouse School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald L. Hill

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Maomao Yan

Morehouse School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Wenwen Wang

Morehouse School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge