1
|
Zhu S, Xie P, Yang Y, Wang Y, Zhang C, Zhang Y, Si S, Zhang J, He J, Si H, Fang K, Ma B, Jiang X, Huang L, Li J, Min T, Zheng B, Da L, Lin D, Gao K, Li Y, Huang M, Qiao F, Huo H, Feng H, Zhao H, Chen Z, Xu Z, Xie J, Cao H, Liu J, Yao X, Xie W, Sun Y, Wu K, Xiong B, Hu P, Luo Z, Lin C. Maternal ELL3 loss-of-function leads to oocyte aneuploidy and early miscarriage. Nat Struct Mol Biol 2025:10.1038/s41594-024-01471-8. [PMID: 39820605 DOI: 10.1038/s41594-024-01471-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 12/09/2024] [Indexed: 01/19/2025]
Abstract
Up to an estimated 10% of women experience miscarriage in their lifetimes. Embryonic aneuploidy is a leading cause for miscarriage, infertility and congenital defects. Here we identify variants of ELL3, a gene encoding a transcription elongation factor, in couples who experienced consecutive early miscarriages due to embryonic aneuploidy. Maternal ELL3 knockout leads to mouse oocyte aneuploidy, subfertility and miscellaneous embryonic defects. Mechanistically, we find that ELL3 localizes to the spindle during meiosis, and that ELL3 depletion in both mouse and human oocytes increases the incidence of meiotic spindle abnormality. ELL3 coordinates with TPX2 to ensure the proper function of the microtubule motor KIF11. Live imaging analysis shows that ELL3 is paramount for promoting spindle assembly and driving chromosome movement. Together, our findings implicate maternal ELL3 deficiency in causing oocyte aneuploidy and early miscarriage.
Collapse
Affiliation(s)
- Shiqi Zhu
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Peng Xie
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yi Yang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Yan Wang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanxin Zhang
- Center of Reproductive Medicine, Shandong University, Jinan, China
| | - Yu Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shuhan Si
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Jin Zhang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Jingjing He
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Hao Si
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Ke Fang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Binbin Ma
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xu Jiang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Lindi Huang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Jiamin Li
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Tian Min
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Beihong Zheng
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujuan, China
| | - Lincui Da
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujuan, China
| | - Dianliang Lin
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujuan, China
| | - Kun Gao
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Yuanyuan Li
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Mingtao Huang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Fengchang Qiao
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Haiqin Huo
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Haoyang Feng
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Han Zhao
- Center of Reproductive Medicine, Shandong University, Jinan, China
| | - Zijiang Chen
- Center of Reproductive Medicine, Shandong University, Jinan, China
| | - Zhengfeng Xu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xie
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Hua Cao
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujuan, China
| | - Jin Liu
- School of Data Science, The Chinese University of Hong Kong, Shenzhen, China
| | - Xuebiao Yao
- Laboratory for Organelle Dynamics and Plasticity Control, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Yan Sun
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujuan, China.
| | - Keliang Wu
- Center of Reproductive Medicine, Shandong University, Jinan, China.
| | - Bo Xiong
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| | - Ping Hu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing, China.
| | - Zhuojuan Luo
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Shenzhen Research Institute, Southeast University, Shenzhen, China.
| | - Chengqi Lin
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Life Science and Technology, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Jones MH, Gergely ZR, Steckhahn D, Zhou B, Betterton MD. Kinesin-5/Cut7 C-terminal tail phosphorylation is essential for microtubule sliding force and bipolar mitotic spindle assembly. Curr Biol 2024; 34:4781-4793.e6. [PMID: 39413787 PMCID: PMC11550858 DOI: 10.1016/j.cub.2024.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 10/18/2024]
Abstract
Kinesin-5 motors play an essential role during mitotic spindle assembly in many organisms1,2,3,4,5,6,7,8,9,10,11: they crosslink antiparallel spindle microtubules, step toward plus ends, and slide the microtubules apart.12,13,14,15,16,17 This activity separates the spindle poles and chromosomes. Kinesin-5s are not only plus-end-directed but can walk or be carried toward MT minus ends,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34 where they show enhanced localization.3,5,7,27,29,32 The kinesin-5 C-terminal tail interacts with and regulates the motor, affecting structure, motility, and sliding force of purified kinesin-535,36,37 along with motility and spindle assembly in cells.27,38,39 The tail contains phosphorylation sites, particularly in the conserved BimC box.6,7,40,41,42,43,44 Nine mitotic tail phosphorylation sites were identified in the kinesin-5 motor of the fission yeast Schizosaccharomyces pombe,45,46,47,48 suggesting that multi-site phosphorylation may regulate kinesin-5s. Here, we show that mutating all nine sites to either alanine or glutamate causes temperature-sensitive lethality due to a failure of bipolar spindle assembly. We characterize kinesin-5 localization and sliding force in the spindle based on Cut7-dependent microtubule minus-end protrusions in cells lacking kinesin-14 motors.39,49,50,51,52 Imaging and computational modeling show that Cut7p simultaneously moves toward the minus ends of protrusion MTs and the plus ends of spindle midzone MTs. Phosphorylation mutants show dramatic decreases in protrusions and sliding force. Comparison to a model of force to create protrusions suggests that tail truncation and phosphorylation mutants decrease Cut7p sliding force similarly to tail-truncated human Eg5.36 Our results show that C-terminal tail phosphorylation is required for kinesin-5/Cut7 sliding force and bipolar spindle assembly in fission yeast.
Collapse
Affiliation(s)
- Michele H Jones
- Department of Physics, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA; Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA
| | - Zachary R Gergely
- Department of Physics, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA; Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA
| | - Daniel Steckhahn
- Department of Physics, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA
| | - Bojun Zhou
- Department of Physics, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA
| | - Meredith D Betterton
- Department of Physics, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA; Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Colorado Avenue, Boulder, CO 80309, USA.
| |
Collapse
|
3
|
Pena GE, Zhou X, Slevin L, Brownlee C, Heald R. Identification of a motif in TPX2 that regulates spindle architecture in Xenopus egg extracts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.579770. [PMID: 38370704 PMCID: PMC10871311 DOI: 10.1101/2024.02.10.579770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
A bipolar spindle composed of microtubules and many associated proteins functions to segregate chromosomes during cell division in all eukaryotes, yet spindle size and architecture varies dramatically across different species and cell types. Targeting protein for Xklp2 (TPX2) is one candidate factor for modulating spindle microtubule organization through its roles in branching microtubule nucleation, activation of the mitotic kinase Aurora A, and association with the kinesin-5 (Eg5) motor. Here we identify a conserved nuclear localization sequence (NLS) motif, 123 KKLK 126 in X. laevis TPX2, which regulates astral microtubule formation and spindle pole morphology in Xenopus egg extracts. Addition of recombinant TPX2 with this sequence mutated to AALA dramatically increased spontaneous formation of microtubule asters and recruitment of phosphorylated Aurora A, pericentrin, and Eg5 to meiotic spindle poles. We propose that TPX2 is a linchpin spindle assembly factor whose regulation contributes to the recruitment and activation of multiple microtubule polymerizing and organizing proteins, generating distinct spindle architectures.
Collapse
|
4
|
Qi X, Liu Y, Peng Y, Fu Y, Fu Y, Yin L, Li X. UHRF1 promotes spindle assembly and chromosome congression by catalyzing EG5 polyubiquitination. J Cell Biol 2023; 222:e202210093. [PMID: 37728657 PMCID: PMC10510743 DOI: 10.1083/jcb.202210093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 05/18/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023] Open
Abstract
UHRF1 is an epigenetic coordinator bridging DNA methylation and histone modifications. Additionally, UHRF1 regulates DNA replication and cell cycle, and its deletion induces G1/S or G2/M cell cycle arrest. The roles of UHRF1 in the regulation of G2/M transition remain poorly understood. UHRF1 depletion caused chromosome misalignment, thereby inducing cell cycle arrest at mitotic metaphase, and these cells exhibited the defects of spindle geometry, prominently manifested as shorter spindles. Mechanistically, UHRF1 protein directly interacts with EG5, a kinesin motor protein, during mitosis. Furthermore, UHRF1 induced EG5 polyubiquitination at the site of K1034 and further promoted the interaction of EG5 with spindle assembly factor TPX2, thereby ensuring accurate EG5 distribution to the spindles during metaphase. Our study clarifies a novel UHRF1 function as a nuclear protein catalyzing EG5 polyubiquitination for proper spindle architecture and faithful genomic transmission, which is independent of its roles in epigenetic regulation and DNA damage repair inside the nucleus. These findings revealed a previously unknown mechanism of UHRF1 in controlling mitotic spindle architecture and chromosome behavior and provided mechanistic evidence for UHRF1 deletion-mediated G2/M arrest.
Collapse
Affiliation(s)
- Xuli Qi
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuchong Peng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuxin Fu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongming Fu
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Linglong Yin
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
5
|
Gergely ZR, Jones MH, Zhou B, Cash C, McIntosh JR, Betterton MD. Distinct regions of the kinesin-5 C-terminal tail are essential for mitotic spindle midzone localization and sliding force. Proc Natl Acad Sci U S A 2023; 120:e2306480120. [PMID: 37725645 PMCID: PMC10523502 DOI: 10.1073/pnas.2306480120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 09/21/2023] Open
Abstract
Kinesin-5 motor proteins play essential roles during mitosis in most organisms. Their tetrameric structure and plus-end-directed motility allow them to bind to and move along antiparallel microtubules, thereby pushing spindle poles apart to assemble a bipolar spindle. Recent work has shown that the C-terminal tail is particularly important to kinesin-5 function: The tail affects motor domain structure, ATP hydrolysis, motility, clustering, and sliding force measured for purified motors, as well as motility, clustering, and spindle assembly in cells. Because previous work has focused on presence or absence of the entire tail, the functionally important regions of the tail remain to be identified. We have therefore characterized a series of kinesin-5/Cut7 tail truncation alleles in fission yeast. Partial truncation causes mitotic defects and temperature-sensitive growth, while further truncation that removes the conserved BimC motif is lethal. We compared the sliding force generated by cut7 mutants using a kinesin-14 mutant background in which some microtubules detach from the spindle poles and are pushed into the nuclear envelope. These Cut7-driven protrusions decreased as more of the tail was truncated, and the most severe truncations produced no observable protrusions. Our observations suggest that the C-terminal tail of Cut7p contributes to both sliding force and midzone localization. In the context of sequential tail truncation, the BimC motif and adjacent C-terminal amino acids are particularly important for sliding force. In addition, moderate tail truncation increases midzone localization, but further truncation of residues N-terminal to the BimC motif decreases midzone localization.
Collapse
Affiliation(s)
- Zachary R Gergely
- Department of Physics, University of Colorado, Boulder, CO 80309
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Michele H Jones
- Department of Physics, University of Colorado, Boulder, CO 80309
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Bojun Zhou
- Department of Physics, University of Colorado, Boulder, CO 80309
| | - Cai Cash
- Department of Physics, University of Colorado, Boulder, CO 80309
| | - J Richard McIntosh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Meredith D Betterton
- Department of Physics, University of Colorado, Boulder, CO 80309
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309
| |
Collapse
|
6
|
Gergely Z, Jones MH, Zhou B, Cash C, McIntosh R, Betterton M. Distinct regions of the kinesin-5 C-terminal tail are essential for mitotic spindle midzone localization and sliding force. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538972. [PMID: 37205432 PMCID: PMC10187184 DOI: 10.1101/2023.05.01.538972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Kinesin-5 motor proteins play essential roles during mitosis in most organisms. Their tetrameric structure and plus-end-directed motility allow them to bind to and move along antiparallel microtubules, thereby pushing spindle poles apart to assemble a bipolar spindle. Recent work has shown that the C-terminal tail is particularly important to kinesin-5 function: the tail affects motor domain structure, ATP hydrolysis, motility, clustering, and sliding force measured for purified motors, as well as motility, clustering, and spindle assembly in cells. Because previous work has focused on presence or absence of the entire tail, the functionally important regions of the tail remain to be identified. We have therefore characterized a series of kinesin-5/Cut7 tail truncation alleles in fission yeast. Partial truncation causes mitotic defects and temperature-sensitive growth, while further truncation that removes the conserved BimC motif is lethal. We compared the sliding force generated by cut7 mutants using a kinesin-14 mutant background in which some microtubules detach from the spindle poles and are pushed into the nuclear envelope. These Cut7-driven protrusions decreased as more of the tail was truncated, and the most severe truncations produced no observable protrusions. Our observations suggest that the C-terminal tail of Cut7p contributes to both sliding force and midzone localization. In the context of sequential tail truncation, the BimC motif and adjacent C-terminal amino acids are particularly important for sliding force. In addition, moderate tail truncation increases midzone localization, but further truncation of residues N terminal to the BimC motif decreases midzone localization.
Collapse
|
7
|
Gergely ZR, Ansari S, Jones MH, Zhou B, Cash C, McIntosh R, Betterton MD. The kinesin-5 protein Cut7 moves bidirectionally on fission yeast spindles with activity that increases in anaphase. J Cell Sci 2023; 136:jcs260474. [PMID: 36655493 PMCID: PMC10112985 DOI: 10.1242/jcs.260474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Kinesin-5 motors are essential to separate mitotic spindle poles and assemble a bipolar spindle in many organisms. These motors crosslink and slide apart antiparallel microtubules via microtubule plus-end-directed motility. However, kinesin-5 localization is enhanced away from antiparallel overlaps. Increasing evidence suggests this localization occurs due to bidirectional motility or trafficking. The purified fission-yeast kinesin-5 protein Cut7 moves bidirectionally, but bidirectionality has not been shown in cells, and the function of the minus-end-directed movement is unknown. Here, we characterized the motility of Cut7 on bipolar and monopolar spindles and observed movement toward both plus- and minus-ends of microtubules. Notably, the activity of the motor increased at anaphase B onset. Perturbations to microtubule dynamics only modestly changed Cut7 movement, whereas Cut7 mutation reduced movement. These results suggest that the directed motility of Cut7 contributes to the movement of the motor. Comparison of the Cut7 mutant and human Eg5 (also known as KIF11) localization suggest a new hypothesis for the function of minus-end-directed motility and spindle-pole localization of kinesin-5s.
Collapse
Affiliation(s)
- Zachary R. Gergely
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Saad Ansari
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Michele H. Jones
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Bojun Zhou
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Cai Cash
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Richard McIntosh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80305, USA
| | - Meredith D. Betterton
- Department of Physics, University of Colorado Boulder, Boulder, CO 80305, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80305, USA
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| |
Collapse
|
8
|
Luo Y, Liu W, Zhu Y, Tian Y, Wu K, Ji L, Ding L, Zhang W, Gao T, Liu X, Zhao J. KIF11 as a potential cancer prognostic marker promotes tumorigenesis in children with Wilms tumor. Pediatr Hematol Oncol 2022; 39:145-157. [PMID: 34378481 DOI: 10.1080/08880018.2021.1953655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Emerging evidence suggests that KIF11 could play a pivotal role in cancer cell proliferation; however, its biological functions and molecular mechanisms in Wilms tumor (WT) cells are largely unknown. The aim of this study was to evaluate the clinical significance and therapeutic potential of KIF11 proteins in WT. KIF11 expression in WT tissues and adjacent nontumor tissues was determined using qRT-PCR, Western blotting, immunohistochemistry (IHC) and bioinformatics. The function of KIF11 protein was determined by its correlation with tumor cell growth, angiogenesis, and apoptosis using IHC and lentiviral vector-mediated KIF11 depletion. KIF11 expression was upregulated in WT tissues and was associated with WT clinical outcomes. Tumor KIF11 expression was significantly associated with the Ki67 proliferation index. CCK-8, flow-cytometric analysis, and Western blotting revealed that KIF11 knockdown significantly inhibited WT cell growth. Functional studies have indicated that increased KIF11 expression is significantly correlated with vascular endothelial growth factor (VEGF) expression and intratumoral microvessel density. We further confirmed that downregulated expression of KIF11 promoted cell apoptosis and significantly increased Bcl-2 and Bax expression. Our findings demonstrate that KIF11 plays a role in promoting the development of human WT and can serve as a potential molecular marker for the treatment of WT.
Collapse
Affiliation(s)
- Yishu Luo
- School of Medicine, Nantong University, Nantong, China
| | - Wei Liu
- Department of General Surgery, Yancheng Third People's Hospital, Yancheng, China
| | - Yinmei Zhu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongshen Tian
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Ke Wu
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Linghua Ji
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Li Ding
- School of Medicine, Nantong University, Nantong, China
| | - Wenwen Zhang
- Department of Radiation Oncology, Nantong Third People's Hospital, Nantong, China
| | - Tingting Gao
- Department of General Surgery, Shanghai children's Hospital, Shanghai, China
| | - Xiaoqin Liu
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jun Zhao
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
9
|
Hwang W, Toda T, Yukawa M. Complementation of fission yeast kinesin-5/Cut7 with human Eg5 provides a versatile platform for screening of anticancer compounds. Biosci Biotechnol Biochem 2022; 86:254-259. [PMID: 34864879 DOI: 10.1093/bbb/zbab212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 01/04/2023]
Abstract
Kinesin-5 family proteins are essential for bipolar spindle assembly to ensure mitotic fidelity. Here, we demonstrate evolutionary functional conservation of kinesin-5 between human and fission yeast. Human Eg5 expressed in the nucleus replaces fission yeast counterpart Cut7. Intriguingly, Eg5 overproduction results in cytotoxicity. This phenotype provides a useful platform for the development of novel kinesin-5 inhibitors as anticancer drugs.
Collapse
Affiliation(s)
- Woosang Hwang
- Laboratory of Molecular and Chemical Cell Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Takashi Toda
- Laboratory of Molecular and Chemical Cell Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Japan
| | - Masashi Yukawa
- Laboratory of Molecular and Chemical Cell Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
10
|
Various effects of two types of kinesin-5 inhibitors on mitosis and cell proliferation. Biochem Pharmacol 2021; 193:114789. [PMID: 34582773 DOI: 10.1016/j.bcp.2021.114789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/21/2022]
Abstract
Kinesin-5 has received considerable attention as a new target for mitosis. Various small-molecule compounds targeting kinesin-5 have been developed in the last few decades. However, the differences in the cellular effects of kinesin-5 inhibitors remain poorly understood. Here, we used two different kinesin-5 inhibitors, biphenyl-type PVZB1194 and S-trityl-L-cysteine-type PVEI0021, to examine their effects on molecular events involving kinesin-5. Our biochemical study of kinesin-5 protein-protein interactions showed that PVZB1194-treated kinesin-5 interacted with TPX2 microtubule nucleation factor, Aurora-A kinase, receptor for hyaluronan-mediated motility, and γ-tubulin, as did untreated mitotic kinesin-5. However, PVEI0021 prevented kinesin-5 from binding to these proteins. In mitotic HeLa cells recovered from nocodazole inhibition, kinesin-5 colocalized with these binding proteins, along with microtubules nucleated near kinetochores. By acting on kinesin-5 interactions with chromatin-associated microtubules, PVZB1194, rather than PVEI0021, not only affected the formation of dispersed microtubule clusters but also enhanced the stability of microtubules. In addition, screening for mitotic inhibitors working synergistically with the kinesin-5 inhibitors revealed that paclitaxel synergistically inhibited HeLa cell proliferation only with PVZB1194. In contrast, the Aurora-A inhibitor MLN8237 exerted a synergistic anti-cell proliferation effect when combined with either inhibitor. Together, these results have provided a better understanding of the molecular action of kinesin-5 inhibitors and indicate their usefulness as molecular tools for the study of mitosis and the development of anticancer agents.
Collapse
|
11
|
Vukušić K, Tolić IM. Anaphase B: Long-standing models meet new concepts. Semin Cell Dev Biol 2021; 117:127-139. [PMID: 33849764 PMCID: PMC8406420 DOI: 10.1016/j.semcdb.2021.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022]
Abstract
Mitotic cell divisions ensure stable transmission of genetic information from a mother to daughter cells in a series of generations. To ensure this crucial task is accomplished, the cell forms a bipolar structure called the mitotic spindle that divides sister chromatids to the opposite sides of the dividing mother cell. After successful establishment of stable attachments of microtubules to chromosomes and inspection of connections between them, at the heart of mitosis, the cell starts the process of segregation. This spectacular moment in the life of a cell is termed anaphase, and it involves two distinct processes: depolymerization of microtubules bound to chromosomes, which is also known as anaphase A, and elongation of the spindle or anaphase B. Both processes ensure physical separation of disjointed sister chromatids. In this chapter, we review the mechanisms of anaphase B spindle elongation primarily in mammalian systems, combining different pioneering ideas and concepts with more recent findings that shed new light on the force generation and regulation of biochemical modules operating during spindle elongation. Finally, we present a comprehensive model of spindle elongation that includes structural, biophysical, and molecular aspects of anaphase B.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
12
|
Pandey H, Popov M, Goldstein-Levitin A, Gheber L. Mechanisms by Which Kinesin-5 Motors Perform Their Multiple Intracellular Functions. Int J Mol Sci 2021; 22:6420. [PMID: 34203964 PMCID: PMC8232732 DOI: 10.3390/ijms22126420] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
Bipolar kinesin-5 motor proteins perform multiple intracellular functions, mainly during mitotic cell division. Their specialized structural characteristics enable these motors to perform their essential functions by crosslinking and sliding apart antiparallel microtubules (MTs). In this review, we discuss the specialized structural features of kinesin-5 motors, and the mechanisms by which these features relate to kinesin-5 functions and motile properties. In addition, we discuss the multiple roles of the kinesin-5 motors in dividing as well as in non-dividing cells, and examine their roles in pathogenetic conditions. We describe the recently discovered bidirectional motility in fungi kinesin-5 motors, and discuss its possible physiological relevance. Finally, we also focus on the multiple mechanisms of regulation of these unique motor proteins.
Collapse
Affiliation(s)
| | | | | | - Larisa Gheber
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel; (H.P.); (M.P.); (A.G.-L.)
| |
Collapse
|
13
|
Vukušić K, Ponjavić I, Buđa R, Risteski P, Tolić IM. Microtubule-sliding modules based on kinesins EG5 and PRC1-dependent KIF4A drive human spindle elongation. Dev Cell 2021; 56:1253-1267.e10. [PMID: 33910056 PMCID: PMC8098747 DOI: 10.1016/j.devcel.2021.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/03/2021] [Accepted: 04/05/2021] [Indexed: 12/13/2022]
Abstract
Proper chromosome segregation into two future daughter cells requires the mitotic spindle to elongate in anaphase. However, although some candidate proteins are implicated in this process, the molecular mechanism that drives spindle elongation in human cells is unknown. Using combined depletion and inactivation assays together with CRISPR technology to explore redundancy between multiple targets, we discovered that the force-generating mechanism of spindle elongation consists of EG5/kinesin-5 together with the PRC1-dependent motor KIF4A/kinesin-4, with contribution from kinesin-6 and kinesin-8. Disruption of EG5 and KIF4A leads to total failure of chromosome segregation due to blocked spindle elongation, despite poleward chromosome motion. Tubulin photoactivation, stimulated emission depletion (STED), and expansion microscopy show that perturbation of both proteins impairs midzone microtubule sliding without affecting microtubule stability. Thus, two mechanistically distinct sliding modules, one based on a self-sustained and the other on a crosslinker-assisted motor, power the mechanism that drives spindle elongation in human cells.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Ivana Ponjavić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Renata Buđa
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Patrik Risteski
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
14
|
Fang CT, Kuo HH, Hsu SC, Yih LH. HSP70 regulates Eg5 distribution within the mitotic spindle and modulates the cytotoxicity of Eg5 inhibitors. Cell Death Dis 2020; 11:715. [PMID: 32873777 PMCID: PMC7462862 DOI: 10.1038/s41419-020-02919-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
The heat shock protein 70 (HSP70) is a conserved molecular chaperone and proteostasis regulator that protects cells from pharmacological stress and promotes drug resistance in cancer cells. In this study, we found that HSP70 may promote resistance to anticancer drugs that target the mitotic kinesin, Eg5, which is essential for assembly and maintenance of the mitotic spindle and cell proliferation. Our data show that loss of HSP70 activity enhances Eg5 inhibitor-induced cytotoxicity and spindle abnormalities. Furthermore, HSP70 colocalizes with Eg5 in the mitotic spindle, and inhibition of HSP70 disrupts this colocalization. Inhibition or depletion of HSP70 also causes Eg5 to accumulate at the spindle pole, altering microtubule dynamics and leading to chromosome misalignment. Using ground state depletion microscopy followed by individual molecule return (GSDIM), we found that HSP70 inhibition reduces the size of Eg5 ensembles and prevents their localization to the inter-polar region of the spindle. In addition, bis(maleimido)hexane-mediated protein-protein crosslinking and proximity ligation assays revealed that HSP70 inhibition deregulates the interaction between Eg5 tetramers and TPX2 at the spindle pole, leading to their accumulation in high-molecular-weight complexes. Finally, we showed that the passive substrate-binding activity of HSP70 is required for appropriate Eg5 distribution and function. Together, our results show that HSP70 substrate-binding activity may regulate proper assembly of Eg5 ensembles and Eg5-TPX2 complexes to modulate mitotic distribution/function of Eg5. Thus, HSP70 inhibition may sensitize cancer cells to Eg5 inhibitor-induced cytotoxicity.
Collapse
Affiliation(s)
- Chieh-Ting Fang
- Department of Life Science, National Taiwan University, Taipei, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hsiao-Hui Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shao-Chun Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Ling-Huei Yih
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
15
|
Mann BJ, Wadsworth P. Kinesin-5 Regulation and Function in Mitosis. Trends Cell Biol 2019; 29:66-79. [DOI: 10.1016/j.tcb.2018.08.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/30/2018] [Accepted: 08/17/2018] [Indexed: 11/16/2022]
|
16
|
Mann BJ, Wadsworth P. Distribution of Eg5 and TPX2 in mitosis: Insight from CRISPR tagged cells. Cytoskeleton (Hoboken) 2018; 75:508-521. [DOI: 10.1002/cm.21486] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/11/2018] [Accepted: 07/30/2018] [Indexed: 11/07/2022]
Affiliation(s)
- B. J. Mann
- Department of Biology, Program in Molecular and Cellular Biology University of Massachusetts Amherst Massachusetts
| | - P. Wadsworth
- Department of Biology, Program in Molecular and Cellular Biology University of Massachusetts Amherst Massachusetts
| |
Collapse
|
17
|
Hsu WH, Wang WJ, Lin WY, Huang YM, Lai CC, Liao JC, Chen HC. Adducin-1 is essential for spindle pole integrity through its interaction with TPX2. EMBO Rep 2018; 19:embr.201745607. [PMID: 29925526 PMCID: PMC6073210 DOI: 10.15252/embr.201745607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/21/2018] [Accepted: 05/28/2018] [Indexed: 11/25/2022] Open
Abstract
Bipolar spindle assembly is necessary to ensure the proper progression of cell division. Loss of spindle pole integrity leads to multipolar spindles and aberrant chromosomal segregation. However, the mechanism underlying the maintenance of spindle pole integrity remains unclear. In this study, we show that the actin‐binding protein adducin‐1 (ADD1) is phosphorylated at S726 during mitosis. S726‐phosphorylated ADD1 localizes to centrosomes, wherein it organizes into a rosette‐like structure at the pericentriolar material. ADD1 depletion causes centriole splitting and therefore results in multipolar spindles during mitosis, which can be restored by re‐expression of ADD1 and the phosphomimetic S726D mutant but not by the S726A mutant. Moreover, the phosphorylation of ADD1 at S726 is crucial for its interaction with TPX2, which is essential for spindle pole integrity. Together, our findings unveil a novel function of ADD1 in maintaining spindle pole integrity through its interaction with TPX2.
Collapse
Affiliation(s)
- Wen-Hsin Hsu
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Yi Lin
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Min Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Chen Lai
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Jung-Chi Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Hong-Chen Chen
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan .,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
18
|
Chen H, Connell M, Mei L, Reid GSD, Maxwell CA. The nonmotor adaptor HMMR dampens Eg5-mediated forces to preserve the kinetics and integrity of chromosome segregation. Mol Biol Cell 2018; 29:786-796. [PMID: 29386294 PMCID: PMC5905292 DOI: 10.1091/mbc.e17-08-0531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The nonmotor adaptor protein HMMR maintains the kinetics and integrity of chromosome segregation by promoting TPX2-Eg5 complexes that dampen Eg5-mediated forces and support K-fiber stability, kinetochore–microtubule attachments, and inter-kinetochore tension. HMMR is needed to prevent the generation of aneuploid progeny cells. Mitotic spindle assembly and organization require forces generated by motor proteins. The activity of these motors is regulated by nonmotor adaptor proteins. However, there are limited studies reporting the functional importance of adaptors on the balance of motor forces and the promotion of faithful and timely cell division. Here we show that genomic deletion or small interfering RNA silencing of the nonmotor adaptor Hmmr/HMMR disturbs spindle microtubule organization and bipolar chromosome–kinetochore attachments with a consequent elevated occurrence of aneuploidy. Rescue experiments show a conserved motif in HMMR is required to generate interkinetochore tension and promote anaphase entry. This motif bears high homology with the kinesin Kif15 and is known to interact with TPX2, a spindle assembly factor. We find that HMMR is required to dampen kinesin Eg5-mediated forces through localizing TPX2 and promoting the formation of inhibitory TPX2-Eg5 complexes. In HMMR-silenced cells, K-fiber stability is reduced while the frequency of unattached chromosomes and the time needed for chromosome segregation are both increased. These defects can be alleviated in HMMR-silenced cells with chemical inhibition of Eg5 but not through the silencing of Kif15. Together, our findings indicate that HMMR balances Eg5-mediated forces to preserve the kinetics and integrity of chromosome segregation.
Collapse
Affiliation(s)
- Helen Chen
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Lin Mei
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Gregor S D Reid
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital, Vancouver, BC V5Z 4H4, Canada
| | - Christopher A Maxwell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
19
|
Eibes S, Gallisà-Suñé N, Rosas-Salvans M, Martínez-Delgado P, Vernos I, Roig J. Nek9 Phosphorylation Defines a New Role for TPX2 in Eg5-Dependent Centrosome Separation before Nuclear Envelope Breakdown. Curr Biol 2017; 28:121-129.e4. [PMID: 29276125 DOI: 10.1016/j.cub.2017.11.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 11/18/2022]
Abstract
Centrosomes [1, 2] play a central role during spindle assembly in most animal cells [3]. In early mitosis, they organize two symmetrical microtubule arrays that upon separation define the two poles of the forming spindle. Centrosome separation is tightly regulated [4, 5], occurring through partially redundant mechanisms that rely on the action of microtubule-based dynein and kinesin motors and the actomyosin system [6]. While centrosomes can separate in prophase or in prometaphase after nuclear envelope breakdown (NEBD), prophase centrosome separation optimizes spindle assembly and minimizes the occurrence of abnormal chromosome attachments that could end in aneuploidy [7, 8]. Prophase centrosome separation relies on the activity of Eg5/KIF11, a mitotic kinesin [9] that accumulates around centrosomes in early mitosis under the control of CDK1 and the Nek9/Nek6/7 kinase module [10-17]. Here, we show that Eg5 localization and centrosome separation in prophase depend on the nuclear microtubule-associated protein TPX2 [18], a pool of which localizes to the centrosomes before NEBD. This localization involves RHAMM/HMMR [19] and the kinase Nek9 [20], which phosphorylates TPX2 nuclear localization signal (NLS) preventing its interaction with importin and nuclear import. The pool of centrosomal TPX2 in prophase has a critical role for both microtubule aster organization and Eg5 localization, and thereby for centrosome separation. Our results uncover an unsuspected role for TPX2 before NEBD and define a novel regulatory mechanism for centrosome separation in prophase. They furthermore suggest NLS phosphorylation as a novel regulatory mechanism for spindle assembly factors controlled by the importin/Ran system.
Collapse
Affiliation(s)
- Susana Eibes
- Molecular Biology Institute of Barcelona (IBMB-CSIC) and Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Núria Gallisà-Suñé
- Molecular Biology Institute of Barcelona (IBMB-CSIC) and Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Miquel Rosas-Salvans
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain
| | - Paula Martínez-Delgado
- Molecular Biology Institute of Barcelona (IBMB-CSIC) and Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Isabelle Vernos
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Joan Roig
- Molecular Biology Institute of Barcelona (IBMB-CSIC) and Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain.
| |
Collapse
|
20
|
Ly T, Whigham A, Clarke R, Brenes-Murillo AJ, Estes B, Madhessian D, Lundberg E, Wadsworth P, Lamond AI. Proteomic analysis of cell cycle progression in asynchronous cultures, including mitotic subphases, using PRIMMUS. eLife 2017; 6:e27574. [PMID: 29052541 PMCID: PMC5650473 DOI: 10.7554/elife.27574] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Abstract
The temporal regulation of protein abundance and post-translational modifications is a key feature of cell division. Recently, we analysed gene expression and protein abundance changes during interphase under minimally perturbed conditions (Ly et al., 2014, 2015). Here, we show that by using specific intracellular immunolabelling protocols, FACS separation of interphase and mitotic cells, including mitotic subphases, can be combined with proteomic analysis by mass spectrometry. Using this PRIMMUS (PRoteomic analysis of Intracellular iMMUnolabelled cell Subsets) approach, we now compare protein abundance and phosphorylation changes in interphase and mitotic fractions from asynchronously growing human cells. We identify a set of 115 phosphorylation sites increased during G2, termed 'early risers'. This set includes phosphorylation of S738 on TPX2, which we show is important for TPX2 function and mitotic progression. Further, we use PRIMMUS to provide the first a proteome-wide analysis of protein abundance remodeling between prophase, prometaphase and anaphase.
Collapse
Affiliation(s)
- Tony Ly
- Centre for Gene Regulation and Expression, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
- Wellcome Centre for Cell BiologyUniversity of EdinburghEdinburghUnited Kingdom
| | - Arlene Whigham
- CAST Flow Cytometry Facility, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Rosemary Clarke
- CAST Flow Cytometry Facility, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Alejandro J Brenes-Murillo
- Centre for Gene Regulation and Expression, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Brett Estes
- Department of BiologyUniversity of MassachusettsMassachusettsUnited States
- Program in Molecular and Cellular BiologyUniversity of MassachusettsMassachusettsUnited States
| | - Diana Madhessian
- Science for Life LaboratoryRoyal Institute of TechnologyStockholmSweden
| | - Emma Lundberg
- Science for Life LaboratoryRoyal Institute of TechnologyStockholmSweden
| | - Patricia Wadsworth
- Department of BiologyUniversity of MassachusettsMassachusettsUnited States
- Program in Molecular and Cellular BiologyUniversity of MassachusettsMassachusettsUnited States
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| |
Collapse
|
21
|
Bickel KG, Mann BJ, Waitzman JS, Poor TA, Rice SE, Wadsworth P. Src family kinase phosphorylation of the motor domain of the human kinesin-5, Eg5. Cytoskeleton (Hoboken) 2017. [PMID: 28646493 DOI: 10.1002/cm.21380] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Spindle formation in mammalian cells requires precise spatial and temporal regulation of the kinesin-5, Eg5, which generates outward force to establish spindle bipolarity. Our results demonstrate that Eg5 is phosphorylated in cultured cells by Src family kinases (SFKs) at three sites in the motor head: Y125, Y211, and Y231. Mutation of these sites diminishes motor activity in vitro, and replacement of endogenous Eg5 with phosphomimetic Y211 in LLC-Pk1 cells results in monopolar spindles, consistent with loss of Eg5 activity. Cells treated with SFK inhibitors show defects in spindle formation, similar to those in cells expressing the nonphosphorylatable Y211 mutant, and distinct from inhibition of other mitotic kinases. We propose that this phosphoregulatory mechanism tunes Eg5 enzymatic activity for optimal spindle morphology.
Collapse
Affiliation(s)
- Kathleen G Bickel
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611
| | - Barbara J Mann
- Department of Biology, University of Massachusetts, Amherst, Massachusetts, 01003
| | - Joshua S Waitzman
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611
| | - Taylor A Poor
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611
| | - Sarah E Rice
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611
| | - Patricia Wadsworth
- Department of Biology, University of Massachusetts, Amherst, Massachusetts, 01003
| |
Collapse
|
22
|
Jin Q, Huang F, Wang X, Zhu H, Xian Y, Li J, Zhang S, Ni Q. High Eg5 expression predicts poor prognosis in breast cancer. Oncotarget 2017; 8:62208-62216. [PMID: 28977938 PMCID: PMC5617498 DOI: 10.18632/oncotarget.19215] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/12/2017] [Indexed: 12/18/2022] Open
Abstract
Eg5 is a motor protein belonging to the kinesin-5 family and has been suggested to exert important function in tumors. In this study, we determined the mRNA and protein expression levels of Eg5 in cancerous and non-cancerous breast tissue by quantitative real-time polymerase chain reaction (qRT-PCR) and tissue microarray immunohistochemistry analysis (TMA-IHC) respectively. The results of 20 fresh-frozen BC samples demonstrated that Eg5 mRNA levels were significantly higher in BC tissues compared with corresponding non-cancerous tissue (p = 0.0009). TMA-IHC analysis in 127 BC tissues revealed that Eg5 expression obviously correlated with clinicopathologial parameters, including tumor grade (p = 0.004), ER status (p = 0.030), Ki67 status (p = 0.005), molecular classification (p = 0.026), N stage (p = 0.015), and TNM stage (p = 0.001). Kaplan-Meier survival curve indicated that high Eg5 expression (p = 0.012), Ki67 status (p = 0.014) and TNM stage (p = 0.026) were independent factors to predict poor prognosis for patients with breast cancer. Our data suggest that Eg5 is not only overexpressed in BC, it may be also served as a potential prognostic marker.
Collapse
Affiliation(s)
- Qin Jin
- Department of Pathlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Fang Huang
- Department of Pathlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xudong Wang
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Huijun Zhu
- Department of Pathlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yun Xian
- Health Insurance Office, Nantong University, Nantong 226001, Jiangsu, China
| | - Jieying Li
- Department of Pathlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Shu Zhang
- Department of Pathlogy, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Qichao Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
23
|
Ross JL. The Dark Matter of Biology. Biophys J 2017; 111:909-16. [PMID: 27602719 PMCID: PMC5018137 DOI: 10.1016/j.bpj.2016.07.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/06/2016] [Accepted: 07/26/2016] [Indexed: 02/04/2023] Open
Abstract
The inside of the cell is full of important, yet invisible species of molecules and proteins that interact weakly but couple together to have huge and important effects in many biological processes. Such “dark matter” inside cells remains mostly hidden, because our tools were developed to investigate strongly interacting species and folded proteins. Example dark-matter species include intrinsically disordered proteins, posttranslational states, ion species, and rare, transient, and weak interactions undetectable by biochemical assays. The dark matter of biology is likely to have multiple, vital roles to regulate signaling, rates of reactions, water structure and viscosity, crowding, and other cellular activities. We need to create new tools to image, detect, and understand these dark-matter species if we are to truly understand fundamental physical principles of biology.
Collapse
Affiliation(s)
- Jennifer L Ross
- Department of Physics, University of Massachusetts Amherst, Amherst, Massachusetts.
| |
Collapse
|
24
|
Li X, Zhu Y, Cao Y, Wang Q, Du J, Tian J, Liang Y, Ma W. LIM kinase activity is required for microtubule organising centre positioning in mouse oocyte meiosis. Reprod Fertil Dev 2017; 29:791-804. [DOI: 10.1071/rd15406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 11/27/2015] [Indexed: 12/23/2022] Open
Abstract
LIM kinase 1 (LIMK1) activity is essential for cell migration and cell cycle progression. Little is known about LIMK1 expression and function in mammalian oocytes. In the present study we assessed LIMK1 protein expression, subcellular distribution and function during mouse oocyte meiosis. Western blot analysis revealed high and stable expression of LIMK1 from the germinal vesicle (GV) to MII stage. In contrast, activated LIMK1 (i.e. LIMK1 phosphorylated at threonine 508 (pLIMK1Thr508)) was only detected after GV breakdown, with levels increasing gradually to peak at MI and MII. Immunofluorescence showed pLIMK1Thr508 was colocalised with the microtubule organising centre (MTOC) components pericentrin and γ-tubulin at the spindle poles. A direct interaction between γ-tubulin and pLIMK1Thr508 was confirmed by co-immunoprecipitation. LIMK inhibition with 1 μM BMS3 damaged MTOC protein localisation to spindle poles, undermined the formation and positioning of functional MTOC and thus disrupted spindle formation and chromosome alignment. These effects were phenocopied by microinjection of LIMK1 antibody into mouse oocytes. In summary, the data demonstrate that LIMK activity is essential for MTOC organisation and distribution and so bipolar spindle formation and maintenance in mouse oocytes.
Collapse
|
25
|
Mann BJ, Balchand SK, Wadsworth P. Regulation of Kif15 localization and motility by the C-terminus of TPX2 and microtubule dynamics. Mol Biol Cell 2016; 28:65-75. [PMID: 27852894 PMCID: PMC5221630 DOI: 10.1091/mbc.e16-06-0476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
Mitotic motor proteins generate force to establish and maintain spindle bipolarity, but how they are temporally and spatially regulated in vivo is unclear. Prior work demonstrated that a microtubule-associated protein, TPX2, targets kinesin-5 and kinesin-12 motors to spindle microtubules. The C-terminal domain of TPX2 contributes to the localization and motility of the kinesin-5, Eg5, but it is not known whether this domain regulates kinesin-12, Kif15. We found that the C-terminal domain of TPX2 contributes to the localization of Kif15 to spindle microtubules in cells and suppresses motor walking in vitro. Kif15 and Eg5 are partially redundant motors, and overexpressed Kif15 can drive spindle formation in the absence of Eg5 activity. Kif15-dependent bipolar spindle formation in vivo requires the C-terminal domain of TPX2. In the spindle, fluorescent puncta of GFP-Kif15 move toward the equatorial region at a rate equivalent to microtubule growth. Reduction of microtubule growth with paclitaxel suppresses GFP-Kif15 motility, demonstrating that dynamic microtubules contribute to Kif15 behavior. Our results show that the C-terminal region of TPX2 regulates Kif15 in vitro, contributes to motor localization in cells, and is required for Kif15 force generation in vivo and further reveal that dynamic microtubules contribute to Kif15 behavior in vivo.
Collapse
Affiliation(s)
- Barbara J Mann
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Sai K Balchand
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Patricia Wadsworth
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
26
|
Takshak A, Roy T, Tandaiya P, Kunwar A. Effect of fuel concentration and force on collective transport by a team of dynein motors. Protein Sci 2016; 26:186-197. [PMID: 27727483 DOI: 10.1002/pro.3065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 09/17/2016] [Accepted: 10/09/2016] [Indexed: 12/20/2022]
Abstract
Motor proteins are essential components of intracellular transport inside eukaryotic cells. These protein molecules use chemical energy obtained from hydrolysis of ATP to produce mechanical forces required for transporting cargos inside cells, from one location to another, in a directed manner. Of these motors, cytoplasmic dynein is structurally more complex than other motor proteins involved in intracellular transport, as it shows force and fuel (ATP) concentration dependent step-size. Cytoplasmic dynein motors are known to work in a team during cargo transport and force generation. Here, we use a complete Monte-Carlo model of single dynein constrained by in vitro experiments, which includes the effect of both force and ATP on stepping as well as detachment of motors under force. We then use our complete Monte-Carlo model of single dynein motor to understand collective cargo transport by a team of dynein motors, such as dependence of cargo travel distance and velocity on applied force and fuel concentration. In our model, cargos pulled by a team of dynein motors do not detach rapidly under higher forces, confirming the experimental observation of longer persistence time of dynein team on microtubule under higher forces.
Collapse
Affiliation(s)
- Anjneya Takshak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Tanushree Roy
- Centre for Research in Nanotechnology and Science, Indian Institute of Technology Bombay, Mumbai, India
| | - Parag Tandaiya
- Department of Mechanical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ambarish Kunwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
27
|
Fu J, Bian M, Xin G, Deng Z, Luo J, Guo X, Chen H, Wang Y, Jiang Q, Zhang C. TPX2 phosphorylation maintains metaphase spindle length by regulating microtubule flux. J Cell Biol 2016; 210:373-83. [PMID: 26240182 PMCID: PMC4523612 DOI: 10.1083/jcb.201412109] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
TPX2 is phosphorylated by Aurora A and is essential for normal microtubule flux on the metaphase spindle. A steady-state metaphase spindle maintains constant length, although the microtubules undergo intensive dynamics. Tubulin dimers are incorporated at plus ends of spindle microtubules while they are removed from the minus ends, resulting in poleward movement. Such microtubule flux is regulated by the microtubule rescue factors CLASPs at kinetochores and depolymerizing protein Kif2a at the poles, along with other regulators of microtubule dynamics. How microtubule polymerization and depolymerization are coordinated remains unclear. Here we show that TPX2, a microtubule-bundling protein and activator of Aurora A, plays an important role. TPX2 was phosphorylated by Aurora A during mitosis. Its phospho-null mutant caused short metaphase spindles coupled with low microtubule flux rate. Interestingly, phosphorylation of TPX2 regulated its interaction with CLASP1 but not Kif2a. The effect of its mutant in shortening the spindle could be rescued by codepletion of CLASP1 and Kif2a that abolished microtubule flux. Together we propose that Aurora A–dependent TPX2 phosphorylation controls mitotic spindle length through regulating microtubule flux.
Collapse
Affiliation(s)
- Jingyan Fu
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Minglei Bian
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangwei Xin
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Zhaoxuan Deng
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jia Luo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Xiao Guo
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hao Chen
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yao Wang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing 100871, China The State Key Laboratory of Bio-membrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
28
|
Grenfell AW, Strzelecka M, Crowder ME, Helmke KJ, Schlaitz AL, Heald R. A versatile multivariate image analysis pipeline reveals features of Xenopus extract spindles. J Cell Biol 2016; 213:127-36. [PMID: 27044897 PMCID: PMC4828689 DOI: 10.1083/jcb.201509079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 03/07/2016] [Indexed: 01/28/2023] Open
Abstract
The authors describe automated image and data analysis tools that reveal architectural principles of the Xenopus egg extract spindle, allow for rapid, unbiased assessment of spindle phenotypes, and can be adapted to analyze other subcellular structures such as nuclei. Imaging datasets are rich in quantitative information. However, few cell biologists possess the tools necessary to analyze them. Here, we present a large dataset of Xenopus extract spindle images together with an analysis pipeline designed to assess spindle morphology across a range of experimental conditions. Our analysis of different spindle types illustrates how kinetochore microtubules amplify spindle microtubule density. Extract mixing experiments reveal that some spindle features titrate, while others undergo switch-like transitions, and multivariate analysis shows the pleiotropic morphological effects of modulating the levels of TPX2, a key spindle assembly factor. We also apply our pipeline to analyze nuclear morphology in human cell culture, showing the general utility of the segmentation approach. Our analyses provide new insight into the diversity of spindle types and suggest areas for future study. The approaches outlined can be applied by other researchers studying spindle morphology and adapted with minimal modification to other experimental systems.
Collapse
Affiliation(s)
- Andrew W Grenfell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Magdalena Strzelecka
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Marina E Crowder
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Kara J Helmke
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Anne-Lore Schlaitz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
29
|
Reid TA, Schuster BM, Mann BJ, Balchand SK, Plooster M, McClellan M, Coombes CE, Wadsworth P, Gardner MK. Suppression of microtubule assembly kinetics by the mitotic protein TPX2. J Cell Sci 2016; 129:1319-28. [PMID: 26869224 DOI: 10.1242/jcs.178806] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023] Open
Abstract
TPX2 is a widely conserved microtubule-associated protein that is required for mitotic spindle formation and function. Previous studies have demonstrated that TPX2 is required for the nucleation of microtubules around chromosomes; however, the molecular mechanism by which TPX2 promotes microtubule nucleation remains a mystery. In this study, we found that TPX2 acts to suppress tubulin subunit off-rates during microtubule assembly and disassembly, thus allowing for the support of unprecedentedly slow rates of plus-end microtubule growth, and also leading to a dramatically reduced microtubule shortening rate. These changes in microtubule dynamics can be explained in computational simulations by a moderate increase in tubulin-tubulin bond strength upon TPX2 association with the microtubule lattice, which in turn acts to reduce the departure rate of tubulin subunits from the microtubule ends. Thus, the direct suppression of tubulin subunit off-rates by TPX2 during microtubule growth and shortening could provide a molecular mechanism to explain the nucleation of new microtubules in the presence of TPX2.
Collapse
Affiliation(s)
- Taylor A Reid
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Breanna M Schuster
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Barbara J Mann
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Melissa Plooster
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Courtney E Coombes
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pat Wadsworth
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
30
|
Overlap microtubules link sister k-fibres and balance the forces on bi-oriented kinetochores. Nat Commun 2016; 7:10298. [PMID: 26728792 PMCID: PMC4728446 DOI: 10.1038/ncomms10298] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/26/2015] [Indexed: 02/04/2023] Open
Abstract
During metaphase, forces on kinetochores are exerted by k-fibres, bundles of microtubules that end at the kinetochore. Interestingly, non-kinetochore microtubules have been observed between sister kinetochores, but their function is unknown. Here we show by laser-cutting of a k-fibre in HeLa and PtK1 cells that a bundle of non-kinetochore microtubules, which we term ‘bridging fibre', bridges sister k-fibres and balances the interkinetochore tension. We found PRC1 and EB3 in the bridging fibre, suggesting that it consists of antiparallel dynamic microtubules. By using a theoretical model that includes a bridging fibre, we show that the forces at the pole and at the kinetochore depend on the bridging fibre thickness. Moreover, our theory and experiments show larger relaxation of the interkinetochore distance for cuts closer to kinetochores. We conclude that the bridging fibre, by linking sister k-fibres, withstands the tension between sister kinetochores and enables the spindle to obtain a curved shape. During metaphase, k-fibre microtubules exert force on kinetochores, but there are also non-kinetochore microtubules close to kinetochores without a known function. Here the authors show that these microtubules, which they call bridging fibres, balance interkinetochore tension by bridging sister k-fibres.
Collapse
|
31
|
Shimamoto Y, Forth S, Kapoor TM. Measuring Pushing and Braking Forces Generated by Ensembles of Kinesin-5 Crosslinking Two Microtubules. Dev Cell 2016; 34:669-81. [PMID: 26418296 DOI: 10.1016/j.devcel.2015.08.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/07/2015] [Accepted: 08/26/2015] [Indexed: 01/10/2023]
Abstract
The proper organization of the microtubule-based mitotic spindle is proposed to depend on nanometer-sized motor proteins generating forces that scale with a micron-sized geometric feature, such as microtubule overlap length. However, it is unclear whether such regulation can be achieved by any mitotic motor protein. Here, we employ an optical-trap- and total internal reflection fluorescence (TIRF)-based assay to show that ensembles of kinesin-5, a conserved mitotic motor protein, can push apart overlapping antiparallel microtubules to generate a force whose magnitude scales with filament overlap length. We also find that kinesin-5 can produce overlap-length-dependent "brake-like" resistance against relative microtubule sliding in both parallel and antiparallel geometries, an activity that has been suggested by cell biological studies but had not been directly measured. Together, these findings, along with numerical simulations, reveal how a motor protein can function as an analog converter, "reading" simple geometric and dynamic features in cytoskeletal networks to produce regulated force outputs.
Collapse
Affiliation(s)
- Yuta Shimamoto
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065, USA; JST PRESTO, The Rockefeller University, New York, NY 10065, USA
| | - Scott Forth
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
32
|
Reboutier D, Benaud C, Prigent C. Aurora A's Functions During Mitotic Exit: The Guess Who Game. Front Oncol 2015; 5:290. [PMID: 26734572 PMCID: PMC4685928 DOI: 10.3389/fonc.2015.00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/07/2015] [Indexed: 11/24/2022] Open
Abstract
Until recently, the knowledge of Aurora A kinase functions during mitosis was limited to pre-metaphase events, particularly centrosome maturation, G2/M transition, and mitotic spindle assembly. However, an involvement of Aurora A in post-metaphase events was also suspected, but not clearly demonstrated due to the technical difficulty to perform the appropriate experiments. Recent developments of both an analog-specific version of Aurora A and small molecule inhibitors have led to the first demonstration that Aurora A is required for the early steps of cytokinesis. As in pre-metaphase, Aurora A plays diverse functions during anaphase, essentially participating in astral microtubules dynamics and central spindle assembly and functioning. The present review describes the experimental systems used to decipher new functions of Aurora A during late mitosis and situate these functions into the context of cytokinesis mechanisms.
Collapse
Affiliation(s)
- David Reboutier
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Christelle Benaud
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Claude Prigent
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| |
Collapse
|
33
|
Balchand SK, Mann BJ, Titus J, Ross JL, Wadsworth P. TPX2 Inhibits Eg5 by Interactions with Both Motor and Microtubule. J Biol Chem 2015; 290:17367-79. [PMID: 26018074 DOI: 10.1074/jbc.m114.612903] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Indexed: 12/27/2022] Open
Abstract
The microtubule-associated protein, TPX2, regulates the activity of the mitotic kinesin, Eg5, but the mechanism of regulation is not established. Using total internal reflection fluorescence microscopy, we observed that Eg5, in extracts of mammalian cells expressing Eg5-EGFP, moved processively toward the microtubule plus-end at an average velocity of 14 nm/s. TPX2 bound to microtubules with an apparent dissociation constant of ∼ 200 nm, and microtubule binding was not dependent on the C-terminal tails of tubulin. Using single molecule assays, we found that full-length TPX2 dramatically reduced Eg5 velocity, whereas truncated TPX2, which lacks the domain that is required for the interaction with Eg5, was a less effective inhibitor at the same concentration. To determine the region(s) of Eg5 that is required for interaction with TPX2, we performed microtubule gliding assays. Dimeric, but not monomeric, Eg5 was differentially inhibited by full-length and truncated TPX2, demonstrating that dimerization or residues in the neck region are important for the interaction of TPX2 with Eg5. These results show that both microtubule binding and interaction with Eg5 contribute to motor inhibition by TPX2 and demonstrate the utility of mammalian cell extracts for biophysical assays.
Collapse
Affiliation(s)
- Sai K Balchand
- the Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Barbara J Mann
- the Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Janel Titus
- the Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Jennifer L Ross
- the Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 Physics and
| | - Patricia Wadsworth
- the Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 From the Departments of Biology and
| |
Collapse
|
34
|
Helmke KJ, Heald R. TPX2 levels modulate meiotic spindle size and architecture in Xenopus egg extracts. ACTA ACUST UNITED AC 2014; 206:385-93. [PMID: 25070954 PMCID: PMC4121975 DOI: 10.1083/jcb.201401014] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
TPX2 levels modulate spindle architecture through Eg5, partitioning microtubules between a tiled, antiparallel array that promotes spindle expansion and a cross-linked, parallel architecture that concentrates microtubules at spindle poles. The spindle segregates chromosomes in dividing eukaryotic cells, and its assembly pathway and morphology vary across organisms and cell types. We investigated mechanisms underlying differences between meiotic spindles formed in egg extracts of two frog species. Small Xenopus tropicalis spindles resisted inhibition of two factors essential for assembly of the larger Xenopus laevis spindles: RanGTP, which functions in chromatin-driven spindle assembly, and the kinesin-5 motor Eg5, which drives antiparallel microtubule (MT) sliding. This suggested a role for the MT-associated protein TPX2 (targeting factor for Xenopus kinesin-like protein 2), which is regulated by Ran and binds Eg5. Indeed, TPX2 was threefold more abundant in X. tropicalis extracts, and elevated TPX2 levels in X. laevis extracts reduced spindle length and sensitivity to Ran and Eg5 inhibition. Higher TPX2 levels recruited Eg5 to the poles, where MT density increased. We propose that TPX2 levels modulate spindle architecture through Eg5, partitioning MTs between a tiled, antiparallel array that promotes spindle expansion and a cross-linked, parallel architecture that concentrates MTs at spindle poles.
Collapse
Affiliation(s)
- Kara J Helmke
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
35
|
Song L, Craney A, Rape M. Microtubule-dependent regulation of mitotic protein degradation. Mol Cell 2014; 53:179-92. [PMID: 24462202 DOI: 10.1016/j.molcel.2013.12.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/18/2013] [Accepted: 12/20/2013] [Indexed: 01/10/2023]
Abstract
Accurate cell division depends on tightly regulated ubiquitylation events catalyzed by the anaphase-promoting complex (APC/C). Among its many substrates, the APC/C triggers the degradation of proteins that stabilize the mitotic spindle, and loss or accumulation of such spindle assembly factors can result in aneuploidy and cancer. Although critical for cell division, it has remained poorly understood how the timing of spindle assembly factor degradation is established during mitosis. Here, we report that active spindle assembly factors are protected from APC/C-dependent degradation by microtubules. In contrast, those molecules that are not bound to microtubules are highly susceptible to proteolysis and turned over immediately after APC/C activation. The correct timing of spindle assembly factor degradation, as achieved by this regulatory circuit, is required for accurate spindle structure and function. We propose that the localized stabilization of APC/C substrates provides a mechanism for the selective disposal of cell-cycle regulators that have fulfilled their mitotic roles.
Collapse
Affiliation(s)
- Ling Song
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Allison Craney
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Rape
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
36
|
Rogers-Nieman GM, Dinu CZ. Therapeutic applications of carbon nanotubes: opportunities and challenges. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 6:327-37. [PMID: 24715535 DOI: 10.1002/wnan.1268] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 02/21/2014] [Accepted: 02/27/2014] [Indexed: 11/11/2022]
Abstract
UNLABELLED Based on their physicochemical properties that allow efficient functionalization with biomolecules and cellular membrane translocation, as well as on their applications in Raman and near-infrared fluorescence imaging, carbon nanotubes (CNTs) have been proposed as viable candidates for developing therapeutic platforms that ensure targeting of tumor cells without affecting healthy cells. This article reviews the research on toxicological effects of CNTs on host cells, as well as their pharmacological profiles on cancer cells. The potential impact of this approach is discussed along with some potential pitfalls that will need to be overcome when therapeutic implementation CNTs are considered. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The authors declare no competing financial interest.
Collapse
|
37
|
Collins E, Mann BJ, Wadsworth P. Eg5 restricts anaphase B spindle elongation in mammalian cells. Cytoskeleton (Hoboken) 2013; 71:136-44. [PMID: 24285623 DOI: 10.1002/cm.21158] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/29/2013] [Accepted: 11/11/2013] [Indexed: 01/31/2023]
Abstract
During anaphase, overlapping antiparallel microtubules in the spindle interzone elongate and contribute to chromosome segregation. Kinesin-5 family members are required for spindle elongation in some cells, but in other cases they restrict elongation acting like a brake. To determine how kinesin-5 contributes to spindle elongation in mammalian cells, we treated LLC-Pk1 epithelial cells with small molecule inhibitors of the mammalian kinesin-5, Eg5, at anaphase onset and measured the rate and extent of spindle pole separation using multidimensional tracking of centrosomes in cells expressing GFP-γ-tubulin. Centrosome separation was biphasic, with an initial fast phase followed by a slower phase. Treatment with the small molecule inhibitor, STLC, which weakens the interaction of Eg5 with microtubules, resulted in an increase in the rate of centrosome separation. Conversely, treatment with FCPT, which induces a rigor-like interaction of Eg5 with microtubules, reduced the rate of spindle elongation. In control cells, GFP-Eg5 was localized to spindle microtubules and accumulated in the interzone as anaphase progressed. Spindle fluorescence of GFP-Eg5 was decreased following treatment with STLC and increased in cells treated with FCPT. In anaphase cells, cortical dynein increases and rocking motion of spindle poles was detected consistent with the possibility that dynein mediates spindle elongation. In summary, our results demonstrate that Eg5 is not required for spindle elongation, and in fact, restricts the rate of spindle elongation in mammalian cells.
Collapse
Affiliation(s)
- Elizabeth Collins
- Department of Biology University of Massachusetts Amherst, Amherst, Massachusetts
| | | | | |
Collapse
|
38
|
Waitzman JS, Rice SE. Mechanism and regulation of kinesin-5, an essential motor for the mitotic spindle. Biol Cell 2013; 106:1-12. [PMID: 24125467 DOI: 10.1111/boc.201300054] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/09/2013] [Indexed: 11/27/2022]
Abstract
Mitotic cell division is the most fundamental task of all living cells. Cells have intricate and tightly regulated machinery to ensure that mitosis occurs with appropriate frequency and high fidelity. A core element of this machinery is the kinesin-5 motor protein, which plays essential roles in spindle formation and maintenance. In this review, we discuss how the structural and mechanical properties of kinesin-5 motors uniquely suit them to their mitotic role. We describe some of the small molecule inhibitors and regulatory proteins that act on kinesin-5, and discuss how these regulators may influence the process of cell division. Finally, we touch on some more recently described functions of kinesin-5 motors in non-dividing cells. Throughout, we highlight a number of open questions that impede our understanding of both this motor's function and the potential utility of kinesin-5 inhibitors.
Collapse
Affiliation(s)
- Joshua S Waitzman
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, U.S.A
| | | |
Collapse
|
39
|
Kinesin-5: cross-bridging mechanism to targeted clinical therapy. Gene 2013; 531:133-49. [PMID: 23954229 DOI: 10.1016/j.gene.2013.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022]
Abstract
Kinesin motor proteins comprise an ATPase superfamily that works hand in hand with microtubules in every eukaryote. The mitotic kinesins, by virtue of their potential therapeutic role in cancerous cells, have been a major focus of research for the past 28 years since the discovery of the canonical Kinesin-1 heavy chain. Perhaps the simplest player in mitotic spindle assembly, Kinesin-5 (also known as Kif11, Eg5, or kinesin spindle protein, KSP) is a plus-end-directed motor localized to interpolar spindle microtubules and to the spindle poles. Comprised of a homotetramer complex, its function primarily is to slide anti-parallel microtubules apart from one another. Based on multi-faceted analyses of this motor from numerous laboratories over the years, we have learned a great deal about the function of this motor at the atomic level for catalysis and as an integrated element of the cytoskeleton. These data have, in turn, informed the function of motile kinesins on the whole, as well as spearheaded integrative models of the mitotic apparatus in particular and regulation of the microtubule cytoskeleton in general. We review what is known about how this nanomotor works, its place inside the cytoskeleton of cells, and its small-molecule inhibitors that provide a toolbox for understanding motor function and for anticancer treatment in the clinic.
Collapse
|
40
|
Salemi JD, McGilvray PT, Maresca TJ. Development of a Drosophila cell-based error correction assay. Front Oncol 2013; 3:187. [PMID: 23888285 PMCID: PMC3719216 DOI: 10.3389/fonc.2013.00187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/04/2013] [Indexed: 12/13/2022] Open
Abstract
Accurate transmission of the genome through cell division requires microtubules from opposing spindle poles to interact with protein super-structures called kinetochores that assemble on each sister chromatid. Most kinetochores establish erroneous attachments that are destabilized through a process called error correction. Failure to correct improper kinetochore-microtubule (kt-MT) interactions before anaphase onset results in chromosomal instability (CIN), which has been implicated in tumorigenesis and tumor adaptation. Thus, it is important to characterize the molecular basis of error correction to better comprehend how CIN occurs and how it can be modulated. An error correction assay has been previously developed in cultured mammalian cells in which incorrect kt-MT attachments are created through the induction of monopolar spindle assembly via chemical inhibition of kinesin-5. Error correction is then monitored following inhibitor wash out. Implementing the error correction assay in Drosophila melanogaster S2 cells would be valuable because kt-MT attachments are easily visualized and the cells are highly amenable to RNAi and high-throughput screening. However, Drosophila kinesin-5 (Klp61F) is unaffected by available small molecule inhibitors. To overcome this limitation, we have rendered S2 cells susceptible to kinesin-5 inhibitors by functionally replacing Klp61F with human kinesin-5 (Eg5). Eg5 expression rescued the assembly of monopolar spindles typically caused by Klp61F depletion. Eg5-mediated bipoles collapsed into monopoles due, in part, to kinesin-14 (Ncd) activity when treated with the kinesin-5 inhibitor S-trityl-L-cysteine (STLC). Furthermore, bipolar spindles reassembled and error correction was observed after STLC wash out. Importantly, error correction in Eg5-expressing S2 cells was dependent on the well-established error correction kinase Aurora B. This system provides a powerful new cell-based platform for studying error correction and CIN.
Collapse
Affiliation(s)
- Jeffrey D Salemi
- Biology Department, University of Massachusetts , Amherst, MA , USA
| | | | | |
Collapse
|
41
|
Stender AS, Marchuk K, Liu C, Sander S, Meyer MW, Smith EA, Neupane B, Wang G, Li J, Cheng JX, Huang B, Fang N. Single cell optical imaging and spectroscopy. Chem Rev 2013; 113:2469-527. [PMID: 23410134 PMCID: PMC3624028 DOI: 10.1021/cr300336e] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Anthony S. Stender
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Kyle Marchuk
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Chang Liu
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Suzanne Sander
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Matthew W. Meyer
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Emily A. Smith
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| | - Bhanu Neupane
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Gufeng Wang
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Junjie Li
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Ji-Xin Cheng
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Bo Huang
- Department of Pharmaceutical Chemistry and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
| | - Ning Fang
- Department of Chemistry, Iowa State University and Ames Laboratory, U. S. Department of Energy, Ames, IA 50011, USA
| |
Collapse
|
42
|
Interaction of NuMA protein with the kinesin Eg5: its possible role in bipolar spindle assembly and chromosome alignment. Biochem J 2013; 451:195-204. [DOI: 10.1042/bj20121447] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bipolar spindle assembly in mitotic cells is a prerequisite to ensure correct alignment of chromosomes for their segregation to each daughter cell; spindle microtubules are tethered at plus ends to chromosomes and focused at minus ends to either of the two spindle poles. NuMA (nuclear mitotic apparatus protein) is present solely in the nucleus in interphase cells, but relocalizes during mitosis to the spindle poles to play a crucial role in spindle assembly via focusing spindle microtubules to each pole. In the present study we show that the kinesin-5 family motor Eg5 is a protein that directly interacts with NuMA, using a proteomics approach and various binding assays both in vivo and in vitro. During mitosis Eg5 appears to interact with NuMA in the vicinity of the spindle poles, whereas the interaction does not occur in interphase cells, where Eg5 is distributed throughout the cytoplasm but NuMA exclusively localizes to the nucleus. Slight, but significant, depletion of Eg5 in HeLa cells by RNA interference results in formation of less-focused spindle poles with misaligned chromosomes in metaphase; these phenotypes are similar to those induced by depletion of NuMA. Since NuMA is less accumulated at the spindle poles in Eg5-depleted cells, Eg5 probably contributes to spindle assembly via regulating NuMA localization. Furthermore, depletion of cytoplasmic dynein induces mislocalization of NuMA and phenotypes similar to those observed in NuMA-depleted cells, without affecting Eg5 localization to the spindles. Thus dynein appears to control NuMA function in conjunction with Eg5.
Collapse
|
43
|
Yang G. Bioimage informatics for understanding spatiotemporal dynamics of cellular processes. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 5:367-80. [PMID: 23408597 DOI: 10.1002/wsbm.1214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The inner environment of the cell is highly dynamic and heterogeneous yet exquisitely organized. Successful completion of cellular processes within this environment depends on the right molecules or molecular complexes to function at the right place at the right time. Understanding spatiotemporal behaviors of cellular processes is therefore essential to understanding their molecular mechanisms at the systems level. These behaviors are usually visualized and recorded using imaging techniques. However, to infer from them systems-level molecular mechanisms, computational analysis and understanding of recorded image data is crucial, not only for acquiring quantitative behavior measurements but also for comprehending complex interactions among the molecules or molecular complexes involved. The technology of computational analysis and understanding of biological images is often referred to simply as bioimage informatics. This article introduces fundamentals of bioimage informatics for understanding spatiotemporal dynamics of cellular processes and reviews recent advances on this topic. Basic bioimage informatics concepts and techniques for characterizing spatiotemporal cell dynamics are introduced first. Studies on specific cellular processes such as cell migration and signal transduction are then used as examples to analyze and summarize recent advances, with the focus on transforming quantitative measurements of spatiotemporal cellular behaviors into knowledge of underlying molecular mechanisms. Despite the advances made, substantial technological challenges remain, especially in representation of spatiotemporal cellular behaviors and inference of systems-level molecular mechanisms. These challenges are briefly discussed. Overall, understanding spatiotemporal cell dynamics will provide critical insights into how specific cellular processes as well as the entire inner cellular environment are dynamically organized and regulated.
Collapse
Affiliation(s)
- Ge Yang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Goulet A, Moores C. New insights into the mechanism of force generation by kinesin-5 molecular motors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:419-66. [PMID: 23809441 DOI: 10.1016/b978-0-12-407696-9.00008-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Kinesin-5 motors are members of a superfamily of microtubule-dependent ATPases and are widely conserved among eukaryotes. Kinesin-5s typically form homotetramers with pairs of motor domains located at either end of a dumbbell-shaped molecule. This quaternary structure enables cross-linking and ATP-driven sliding of pairs of microtubules, although the exact molecular mechanism of this activity is still unclear. Kinesin-5 function has been characterized in greatest detail in cell division, although a number of interphase roles have also been defined. The kinesin-5 ATPase is tuned for slow microtubule sliding rather than cellular transport and-in vertebrates-can be inhibited specifically by allosteric small molecules currently in cancer clinical trials. The biophysical and structural basis of kinesin-5 mechanochemistry is being elucidated and has provided further insight into kinesin-5 activities. However, it is likely that the precise mechanism of these important motors has evolved according to functional context and regulation in individual organisms.
Collapse
Affiliation(s)
- Adeline Goulet
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| | | |
Collapse
|
45
|
The role of NEDD1 phosphorylation by Aurora A in chromosomal microtubule nucleation and spindle function. Curr Biol 2012; 23:143-9. [PMID: 23273898 DOI: 10.1016/j.cub.2012.11.046] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 09/28/2012] [Accepted: 11/21/2012] [Indexed: 01/05/2023]
Abstract
Chromatin directs de novo microtubule (MT) nucleation in dividing cells by generating a gradient of GTP-bound Ran protein (RanGTP) that controls the activity of a number of spindle assembly factors (SAFs). It is now well established that these MTs are essential for the assembly of a functional bipolar spindle. Although it has been shown that RanGTP-dependent MT nucleation requires γ-tubulin and a number of RanGTP-regulated proteins, the mechanism involved is still poorly understood. We previously showed that the mitotic kinase Aurora A, which is activated in a RanGTP-dependent manner in mitotic cells, has a role in this pathway. Here we show that Aurora A interacts with and phosphorylates the γTURC adaptor protein NEDD1 at a single residue, Ser405. Ser405 phosphorylation is not required for centrosomal MT nucleation but is critical for MT nucleation in the vicinity of the chromosomes in mitotic cells. Moreover, it is essential for RanGTP aster formation and chromatin-driven MT assembly in Xenopus egg extracts. Our data suggest that one important function of Aurora A in mitotic cells is to promote MT nucleation around the chromatin by phosphorylating NEDD1, and thereby to promote functional spindle assembly.
Collapse
|
46
|
Eichwald C, Arnoldi F, Laimbacher AS, Schraner EM, Fraefel C, Wild P, Burrone OR, Ackermann M. Rotavirus viroplasm fusion and perinuclear localization are dynamic processes requiring stabilized microtubules. PLoS One 2012; 7:e47947. [PMID: 23110139 PMCID: PMC3479128 DOI: 10.1371/journal.pone.0047947] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022] Open
Abstract
Rotavirus viroplasms are cytosolic, electron-dense inclusions corresponding to the viral machinery of replication responsible for viral template transcription, dsRNA genome segments replication and assembly of new viral cores. We have previously observed that, over time, those viroplasms increase in size and decrease in number. Therefore, we hypothesized that this process was dependent on the cellular microtubular network and its associated dynamic components. Here, we present evidence demonstrating that viroplasms are dynamic structures, which, in the course of an ongoing infection, move towards the perinuclear region of the cell, where they fuse among each other, thereby gaining considerably in size and, simultaneouly, explaining the decrease in numbers. On the viral side, this process seems to depend on VP2 for movement and on NSP2 for fusion. On the cellular side, both the temporal transition and the maintenance of the viroplasms are dependent on the microtubular network, its stabilization by acetylation, and, surprisingly, on a kinesin motor of the kinesin-5 family, Eg5. Thus, we provide for the first time deeper insights into the dynamics of rotavirus replication, which can explain the behavior of viroplasms in the infected cell.
Collapse
|
47
|
Collins ES, Balchand SK, Faraci JL, Wadsworth P, Lee WL. Cell cycle-regulated cortical dynein/dynactin promotes symmetric cell division by differential pole motion in anaphase. Mol Biol Cell 2012; 23:3380-90. [PMID: 22809624 PMCID: PMC3431930 DOI: 10.1091/mbc.e12-02-0109] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Evidence is presented for dynamic cortical association of dynein and dynactin in mammalian cells and its regulation by Plk1, astral microtubules, and the cell cycle. The asymmetric spindle positioning in LLC-Pk1 cells and its correction by dynein and dynactin provide a new system for analysis of spindle position and symmetric cell division. In cultured mammalian cells, how dynein/dynactin contributes to spindle positioning is poorly understood. To assess the role of cortical dynein/dynactin in this process, we generated mammalian cell lines expressing localization and affinity purification (LAP)–tagged dynein/dynactin subunits from bacterial artificial chromosomes and observed asymmetric cortical localization of dynein and dynactin during mitosis. In cells with asymmetrically positioned spindles, dynein and dynactin were both enriched at the cortex distal to the spindle. NuMA, an upstream targeting factor, localized asymmetrically along the cell cortex in a manner similar to dynein and dynactin. During spindle motion toward the distal cortex, dynein and dynactin were locally diminished and subsequently enriched at the new distal cortex. At anaphase onset, we observed a transient increase in cortical dynein, followed by a reduction in telophase. Spindle motion frequently resulted in cells entering anaphase with an asymmetrically positioned spindle. These cells gave rise to symmetric daughter cells by dynein-dependent differential spindle pole motion in anaphase. Our results demonstrate that cortical dynein and dynactin dynamically associate with the cell cortex in a cell cycle–regulated manner and are required to correct spindle mispositioning in LLC-Pk1 epithelial cells.
Collapse
|