1
|
Yildiz A. Mechanism and regulation of kinesin motors. Nat Rev Mol Cell Biol 2025; 26:86-103. [PMID: 39394463 DOI: 10.1038/s41580-024-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
Kinesins are a diverse superfamily of microtubule-based motors that perform fundamental roles in intracellular transport, cytoskeletal dynamics and cell division. These motors share a characteristic motor domain that powers unidirectional motility and force generation along microtubules, and they possess unique tail domains that recruit accessory proteins and facilitate oligomerization, regulation and cargo recognition. The location, direction and timing of kinesin-driven processes are tightly regulated by various cofactors, adaptors, microtubule tracks and microtubule-associated proteins. This Review focuses on recent structural and functional studies that reveal how members of the kinesin superfamily use the energy of ATP hydrolysis to transport cargoes, depolymerize microtubules and regulate microtubule dynamics. I also survey how accessory proteins and post-translational modifications regulate the autoinhibition, cargo binding and motility of some of the best-studied kinesins. Despite much progress, the mechanism and regulation of kinesins are still emerging, and unresolved questions can now be tackled using newly developed approaches in biophysics and structural biology.
Collapse
Affiliation(s)
- Ahmet Yildiz
- Physics Department, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
2
|
Xie P. Modeling study of kinesin-13 MCAK microtubule depolymerase. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:339-354. [PMID: 39093405 DOI: 10.1007/s00249-024-01718-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Mitotic centromere-associated kinesin (MCAK) motor protein is a typical member of the kinesin-13 family, which can depolymerize microtubules from both plus and minus ends. A critical issue for the MCAK motor is how it performs the depolymerase activity. To address the issue, the pathway of the MCAK motor moving on microtubules and depolymerizing the microtubules is presented here. On the basis of the pathway, the dynamics of both the wild-type and mutant MCAK motors is studied theoretically, which include the full-length MCAK, the full-length MCAK with mutations in the α4-helix of the motor domain, the mutant full-length MCAK with a neutralized neck, the monomeric MCAK and the mutant monomeric MCAK with a neutralized neck. The studies show that a single dimeric MCAK motor can depolymerize microtubules in a processive manner, with either one tubulin or two tubulins being removed per times. The theoretical results are in agreement with the available experimental data. Moreover, predicted results are provided.
Collapse
Affiliation(s)
- Ping Xie
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
3
|
Gudivada IP, Amajala KC. Integrative Bioinformatics Analysis for Targeting Hub Genes in Hepatocellular Carcinoma Treatment. Curr Genomics 2024; 26:48-80. [PMID: 39911278 PMCID: PMC11793067 DOI: 10.2174/0113892029308243240709073945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/24/2024] [Accepted: 06/11/2024] [Indexed: 02/07/2025] Open
Abstract
Background The damage in the liver and hepatocytes is where the primary liver cancer begins, and this is referred to as Hepatocellular Carcinoma (HCC). One of the best methods for detecting changes in gene expression of hepatocellular carcinoma is through bioinformatics approaches. Objective This study aimed to identify potential drug target(s) hubs mediating HCC progression using computational approaches through gene expression and protein-protein interaction datasets. Methodology Four datasets related to HCC were acquired from the GEO database, and Differentially Expressed Genes (DEGs) were identified. Using Evenn, the common genes were chosen. Using the Fun Rich tool, functional associations among the genes were identified. Further, protein-protein interaction networks were predicted using STRING, and hub genes were identified using Cytoscape. The selected hub genes were subjected to GEPIA and Shiny GO analysis for survival analysis and functional enrichment studies for the identified hub genes. The up-regulating genes were further studied for immunohistopathological studies using HPA to identify gene/protein expression in normal vs HCC conditions. Drug Bank and Drug Gene Interaction Database were employed to find the reported drug status and targets. Finally, STITCH was performed to identify the functional association between the drugs and the identified hub genes. Results The GEO2R analysis for the considered datasets identified 735 upregulating and 284 downregulating DEGs. Functional gene associations were identified through the Fun Rich tool. Further, PPIN network analysis was performed using STRING. A comparative study was carried out between the experimental evidence and the other seven data evidence in STRING, revealing that most proteins in the network were involved in protein-protein interactions. Further, through Cytoscape plugins, the ranking of the genes was analyzed, and densely connected regions were identified, resulting in the selection of the top 20 hub genes involved in HCC pathogenesis. The identified hub genes were: KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Further, GEPIA and Shiny GO analyses provided insights into survival ratios and functional enrichment studied for the hub genes. The HPA database studies further found that upregulating genes were involved in changes in protein expression in Normal vs HCC tissues. These findings indicated that hub genes were certainly involved in the progression of HCC. STITCH database studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, could be used as leads to identify novel drugs, and identified hub genes could also be considered as potential and promising drug targets as they are involved in the gene-chemical interaction networks. Conclusion The present study involved various integrated bioinformatics approaches, analyzing gene expression and protein-protein interaction datasets, resulting in the identification of 20 top-ranked hubs involved in the progression of HCC. They are KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Gene-chemical interaction network studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, can be used as leads to identify novel drugs, and the identified hub genes can be promising drug targets. The current study underscores the significance of targeting these hub genes and utilizing existing molecules to generate new molecules to combat liver cancer effectively and can be further explored in terms of drug discovery research to develop treatments for HCC.
Collapse
Affiliation(s)
- Indu Priya Gudivada
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Krishna Chaitanya Amajala
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| |
Collapse
|
4
|
Lawrence EJ, Chatterjee S, Zanic M. More is different: Reconstituting complexity in microtubule regulation. J Biol Chem 2023; 299:105398. [PMID: 37898404 PMCID: PMC10694663 DOI: 10.1016/j.jbc.2023.105398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023] Open
Abstract
Microtubules are dynamic cytoskeletal filaments that undergo stochastic switching between phases of polymerization and depolymerization-a behavior known as dynamic instability. Many important cellular processes, including cell motility, chromosome segregation, and intracellular transport, require complex spatiotemporal regulation of microtubule dynamics. This coordinated regulation is achieved through the interactions of numerous microtubule-associated proteins (MAPs) with microtubule ends and lattices. Here, we review the recent advances in our understanding of microtubule regulation, focusing on results arising from biochemical in vitro reconstitution approaches using purified multiprotein ensembles. We discuss how the combinatory effects of MAPs affect both the dynamics of individual microtubule ends, as well as the stability and turnover of the microtubule lattice. In addition, we highlight new results demonstrating the roles of protein condensates in microtubule regulation. Our overall intent is to showcase how lessons learned from reconstitution approaches help unravel the regulatory mechanisms at play in complex cellular environments.
Collapse
Affiliation(s)
- Elizabeth J Lawrence
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Saptarshi Chatterjee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
5
|
Chen X, Portran D, Widmer LA, Stangier MM, Czub MP, Liakopoulos D, Stelling J, Steinmetz MO, Barral Y. The motor domain of the kinesin Kip2 promotes microtubule polymerization at microtubule tips. J Cell Biol 2023; 222:214052. [PMID: 37093124 PMCID: PMC10130750 DOI: 10.1083/jcb.202110126] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/01/2023] [Accepted: 03/22/2023] [Indexed: 04/25/2023] Open
Abstract
Kinesins are microtubule-dependent motor proteins, some of which moonlight as microtubule polymerases, such as the yeast protein Kip2. Here, we show that the CLIP-170 ortholog Bik1 stabilizes Kip2 at microtubule ends where the motor domain of Kip2 promotes microtubule polymerization. Live-cell imaging and mathematical estimation of Kip2 dynamics reveal that disrupting the Kip2-Bik1 interaction aborts Kip2 dwelling at microtubule ends and abrogates its microtubule polymerization activity. Structural modeling and biochemical experiments identify a patch of positively charged residues that enables the motor domain to bind free tubulin dimers alternatively to the microtubule shaft. Neutralizing this patch abolished the ability of Kip2 to promote microtubule growth both in vivo and in vitro without affecting its ability to walk along microtubules. Our studies suggest that Kip2 utilizes Bik1 as a cofactor to track microtubule tips, where its motor domain then recruits free tubulin and catalyzes microtubule assembly.
Collapse
Affiliation(s)
- Xiuzhen Chen
- Institute of Biochemistry, Eidgenössische Technische Hochschule Zürich , Zurich, Switzerland
| | - Didier Portran
- CRBM, Université de Montpellier , CNRS, Montpellier, France
| | - Lukas A Widmer
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Marcel M Stangier
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Mateusz P Czub
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Dimitris Liakopoulos
- CRBM, Université de Montpellier , CNRS, Montpellier, France
- Laboratory of Biology, University of Ioannina, Faculty of Medicine, Ioannina, Greece
| | - Jörg Stelling
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michel O Steinmetz
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- University of Basel, Biozentrum , Basel, Switzerland
| | - Yves Barral
- Institute of Biochemistry, Eidgenössische Technische Hochschule Zürich , Zurich, Switzerland
| |
Collapse
|
6
|
Fu G, Yan S, Khoo CJ, Chao VC, Liu Z, Mukhi M, Hervas R, Li XD, Ti SC. Integrated regulation of tubulin tyrosination and microtubule stability by human α-tubulin isotypes. Cell Rep 2023; 42:112653. [PMID: 37379209 DOI: 10.1016/j.celrep.2023.112653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 05/03/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
Tubulin isotypes are critical for the functions of cellular microtubules, which exhibit different stability and harbor various post-translational modifications. However, how tubulin isotypes determine the activities of regulators for microtubule stability and modifications remains unknown. Here, we show that human α4A-tubulin, a conserved genetically detyrosinated α-tubulin isotype, is a poor substrate for enzymatic tyrosination. To examine the stability of microtubules reconstituted with defined tubulin compositions, we develop a strategy to site-specifically label recombinant human tubulin for single-molecule TIRF microscopy-based in vitro assays. The incorporation of α4A-tubulin into the microtubule lattice stabilizes the polymers from passive and MCAK-stimulated depolymerization. Further characterization reveals that the compositions of α-tubulin isotypes and tyrosination/detyrosination states allow graded control for the microtubule binding and the depolymerization activities of MCAK. Together, our results uncover the tubulin isotype-dependent enzyme activity for an integrated regulation of α-tubulin tyrosination/detyrosination states and microtubule stability, two well-correlated features of cellular microtubules.
Collapse
Affiliation(s)
- Guoling Fu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Shan Yan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Chen Jing Khoo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Victor C Chao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Zheng Liu
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Mayur Mukhi
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Rubén Hervas
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Xiang David Li
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Shih-Chieh Ti
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
7
|
Smith JC, Husted S, Pilrose J, Ems-McClung SC, Stout JR, Carpenter RL, Walczak CE. MCAK Inhibitors Induce Aneuploidy in Triple-Negative Breast Cancer Models. Cancers (Basel) 2023; 15:3309. [PMID: 37444419 PMCID: PMC10340532 DOI: 10.3390/cancers15133309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Standard of care for triple-negative breast cancer (TNBC) involves the use of microtubule poisons such as paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies are common. Unfortunately, patients often relapse with drug-resistant tumors. Identifying agents against targets that limit aneuploidy may be a valuable approach for therapeutic development. One potential target is the microtubule depolymerizing kinesin, MCAK, which limits aneuploidy by regulating microtubule dynamics during mitosis. Using publicly available datasets, we found that MCAK is upregulated in triple-negative breast cancer and is associated with poorer prognoses. Knockdown of MCAK in tumor-derived cell lines caused a two- to five-fold reduction in the IC50 for paclitaxel, without affecting normal cells. Using FRET and image-based assays, we screened compounds from the ChemBridge 50 k library and discovered three putative MCAK inhibitors. These compounds reproduced the aneuploidy-inducing phenotype of MCAK loss, reduced clonogenic survival of TNBC cells regardless of taxane-resistance, and the most potent of the three, C4, sensitized TNBC cells to paclitaxel. Collectively, our work shows promise that MCAK may serve as both a biomarker of prognosis and as a therapeutic target.
Collapse
Affiliation(s)
- John C. Smith
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Stefan Husted
- LabCorp Drug Development Indianapolis, Indianapolis, IN 46214, USA;
| | - Jay Pilrose
- Catalent Pharma Solutions Bloomington, Bloomington, IN 47403, USA;
| | - Stephanie C. Ems-McClung
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Jane R. Stout
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Richard L. Carpenter
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Claire E. Walczak
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| |
Collapse
|
8
|
Smith JC, Husted S, Pilrose J, Ems-McClung SC, Stout JR, Carpenter RL, Walczak CE. MCAK Inhibitors Induce Aneuploidy in Triple Negative Breast Cancer Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543118. [PMID: 37397990 PMCID: PMC10312595 DOI: 10.1101/2023.05.31.543118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Standard of care for triple negative breast cancer (TNBC) involves the use of microtubule poisons like paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies are common. Unfortunately, patients often relapse with drug resistant tumors. Identifying agents against targets that limit aneuploidy may be a valuable approach for therapeutic development. One potential target is the microtubule depolymerizing kinesin, MCAK, which limits aneuploidy by regulating microtubule dynamics during mitosis. Using publicly available datasets, we found that MCAK is upregulated in triple negative breast cancer and is associated with poorer prognoses. Knockdown of MCAK in tumor-derived cell lines caused a two- to five-fold reduction in the IC 50 for paclitaxel, without affecting normal cells. Using FRET and image-based assays, we screened compounds from the ChemBridge 50k library and discovered three putative MCAK inhibitors. These compounds reproduced the aneuploidy-inducing phenotype of MCAK loss, reduced clonogenic survival of TNBC cells regardless of taxane-resistance, and the most potent of the three, C4, sensitized TNBC cells to paclitaxel. Collectively, our work shows promise that MCAK may serve as both a biomarker of prognosis and as a therapeutic target. Simple Summary Triple negative breast cancer (TNBC) is the most lethal breast cancer subtype with few treatment options available. Standard of care for TNBC involves the use of taxanes, which are initially effective, but dose limiting toxicities are common, and patients often relapse with resistant tumors. Specific drugs that produce taxane-like effects may be able to improve patient quality of life and prognosis. In this study we identify three novel inhibitors of the Kinesin-13 MCAK. MCAK inhibition induces aneuploidy; similar to cells treated with taxanes. We demonstrate that MCAK is upregulated in TNBC and is associated with poorer prognoses. These MCAK inhibitors reduce the clonogenic survival of TNBC cells, and the most potent of the three inhibitors, C4, sensitizes TNBC cells to taxanes, similar to the effects of MCAK knockdown. This work will expand the field of precision medicine to include aneuploidy-inducing drugs that have the potential to improve patient outcomes.
Collapse
|
9
|
Clarke MN, Marsoner T, Adell MAY, Ravichandran MC, Campbell CS. Adaptation to high rates of chromosomal instability and aneuploidy through multiple pathways in budding yeast. EMBO J 2023; 42:e111500. [PMID: 36530167 PMCID: PMC10106982 DOI: 10.15252/embj.2022111500] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Both an increased frequency of chromosome missegregation (chromosomal instability, CIN) and the presence of an abnormal complement of chromosomes (aneuploidy) are hallmarks of cancer. To better understand how cells are able to adapt to high levels of chromosomal instability, we previously examined yeast cells that were deleted of the gene BIR1, a member of the chromosomal passenger complex (CPC). We found bir1Δ cells quickly adapted by acquiring specific combinations of beneficial aneuploidies. In this study, we monitored these yeast strains for longer periods of time to determine how cells adapt to high levels of both CIN and aneuploidy in the long term. We identify suppressor mutations that mitigate the chromosome missegregation phenotype. The mutated proteins fall into four main categories: outer kinetochore subunits, the SCFCdc4 ubiquitin ligase complex, the mitotic kinase Mps1, and the CPC itself. The identified suppressor mutations functioned by reducing chromosomal instability rather than alleviating the negative effects of aneuploidy. Following the accumulation of suppressor point mutations, the number of beneficial aneuploidies decreased. These experiments demonstrate a time line of adaptation to high rates of CIN.
Collapse
Affiliation(s)
- Matthew N Clarke
- Department of Chromosome Biology, Max Perutz Labs, Vienna Biocenter (VBC)University of ViennaViennaAustria
| | - Theodor Marsoner
- Department of Chromosome Biology, Max Perutz Labs, Vienna Biocenter (VBC)University of ViennaViennaAustria
| | - Manuel Alonso Y Adell
- Department of Chromosome Biology, Max Perutz Labs, Vienna Biocenter (VBC)University of ViennaViennaAustria
| | - Madhwesh C Ravichandran
- Department of Chromosome Biology, Max Perutz Labs, Vienna Biocenter (VBC)University of ViennaViennaAustria
| | - Christopher S Campbell
- Department of Chromosome Biology, Max Perutz Labs, Vienna Biocenter (VBC)University of ViennaViennaAustria
| |
Collapse
|
10
|
McHugh T, Welburn JPI. Potent microtubule-depolymerizing activity of a mitotic Kif18b-MCAK-EB network. J Cell Sci 2023; 136:275263. [PMID: 35502670 DOI: 10.1242/jcs.260144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
The precise regulation of microtubule length during mitosis is essential to assemble and position the mitotic spindle and segregate chromosomes. The kinesin-13 Kif2C or MCAK acts as a potent microtubule depolymerase that diffuses short distances on microtubules, whereas the kinesin-8 Kif18b is a processive motor with weak depolymerase activity. However, the individual activities of these factors cannot explain the dramatic increase in microtubule dynamics in mitosis. Using in vitro reconstitution and single-molecule imaging, we demonstrate that Kif18b, MCAK and the plus-end tracking protein EB3 (also known as MAPRE3) act in an integrated manner to potently promote microtubule depolymerization at very low concentrations. We find that Kif18b can transport EB3 and MCAK and promotes their accumulation to microtubule plus ends through multivalent weak interactions. Together, our work defines the mechanistic basis for a cooperative Kif18b-MCAK-EB network at microtubule plus ends, that acts to efficiently shorten and regulate microtubules in mitosis, essential for correct chromosome segregation.
Collapse
Affiliation(s)
- Toni McHugh
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| |
Collapse
|
11
|
KIF2C Facilitates Tumor Growth and Metastasis in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:cancers15051502. [PMID: 36900292 PMCID: PMC10000478 DOI: 10.3390/cancers15051502] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with a poor prognosis. For PDAC, an increase in the survival time of patients and a reduction mortality have not yet successfully been achieved. In many research works, Kinesin family member 2C (KIF2C) is highly expressed in several tumors. Nevertheless, the role of KIF2C in pancreatic cancer is unknown. In this study, we found that KIF2C expression is significantly upregulated in human PDAC tissues and cell lines such as ASPC-1 and MIA-PaCa2. Moreover, KIF2C upregulation is associated with a poor prognosis when combining the expression of KIF2C with clinical information. Through cell functional assays and the construction of animal models, we showed that KIF2C promotes PDAC cell proliferation, migration, invasion, and metastasis, both in vitro and in vivo. Finally, the results of sequencing showed that the overexpression of KIF2C causes a decrease in some proinflammatory factors and chemokines. The cell cycle detection indicated that the pancreatic cancer cells in the overexpressed group had abnormal proliferation in the G2 and S phases. These results revealed the potential of KIF2C as a therapeutic target for the treatment of PDAC.
Collapse
|
12
|
Atomic force microscopy reveals distinct protofilament-scale structural dynamics in depolymerizing microtubule arrays. Proc Natl Acad Sci U S A 2022; 119:2115708119. [PMID: 35101922 PMCID: PMC8812519 DOI: 10.1073/pnas.2115708119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 12/01/2022] Open
Abstract
One cannot help but marvel at the precise organization of microtubule polymers in cellular structures such as the axoneme and the spindle. However, our understanding of the biochemical mechanisms that sculpt these arrays comes largely from in vitro experiments with a small number (one or two) of microtubules. This is somewhat akin to studying the architecture of multilane highways by studying one-lane streets. Here, we directly visualize depolymerizing microtubule arrays at individual microtubule and protofilament resolution using atomic force microscopy. Our results reveal differences in microtubule depolymerase activity and provide insights into how these differences in enzymatic activity on the nanometer scale can result in the differential remodeling of multimicrotubule arrays on the micron-length scale. The dynamic reorganization of microtubule-based cellular structures, such as the spindle and the axoneme, fundamentally depends on the dynamics of individual polymers within multimicrotubule arrays. A major class of enzymes implicated in both the complete demolition and fine size control of microtubule-based arrays are depolymerizing kinesins. How different depolymerases differently remodel microtubule arrays is poorly understood. A major technical challenge in addressing this question is that existing optical or electron-microscopy methods lack the spatial-temporal resolution to observe the dynamics of individual microtubules within larger arrays. Here, we use atomic force microscopy (AFM) to image depolymerizing arrays at single-microtubule and protofilament resolution. We discover previously unseen modes of microtubule array destabilization by conserved depolymerases. We find that the kinesin-13 MCAK mediates asynchronous protofilament depolymerization and lattice-defect propagation, whereas the kinesin-8 Kip3p promotes synchronous protofilament depolymerization. Unexpectedly, MCAK can depolymerize the highly stable axonemal doublets, but Kip3p cannot. We propose that distinct protofilament-level activities underlie the functional dichotomy of depolymerases, resulting in either large-scale destabilization or length regulation of microtubule arrays. Our work establishes AFM as a powerful strategy to visualize microtubule dynamics within arrays and reveals how nanometer-scale substrate specificity leads to differential remodeling of micron-scale cytoskeletal structures.
Collapse
|
13
|
Motor usage imprints microtubule stability along the shaft. Dev Cell 2021; 57:5-18.e8. [PMID: 34883065 DOI: 10.1016/j.devcel.2021.11.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/27/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Tubulin dimers assemble into dynamic microtubules, which are used by molecular motors as tracks for intracellular transport. Organization and dynamics of the microtubule network are commonly thought to be regulated at the polymer ends, where tubulin dimers can be added or removed. Here, we show that molecular motors running on microtubules cause exchange of dimers along the shaft in vitro and in cells. These sites of dimer exchange act as rescue sites where depolymerizing microtubules stop shrinking and start re-growing. Consequently, the average length of microtubules increases depending on how frequently they are used as motor tracks. An increase of motor activity densifies the cellular microtubule network and enhances cell polarity. Running motors leave marks in the shaft, serving as traces of microtubule usage to organize the polarity landscape of the cell.
Collapse
|
14
|
KIF2C Is a Novel Prognostic Biomarker and Correlated with Immune Infiltration in Endometrial Cancer. Stem Cells Int 2021; 2021:1434856. [PMID: 34650608 PMCID: PMC8510809 DOI: 10.1155/2021/1434856] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/24/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022] Open
Abstract
Endometrial cancer (EC) is commonly diagnosed cancer in women, and the prognosis of advanced types of EC is extremely poor. Kinesin family member 2C (KIF2C) has been reported as an oncogene in cancers. However, its pathophysiological roles and the correlation with tumor-infiltrating lymphocytes in EC remain unclear. The mRNA and protein levels of KIF2C in EC tissues were detected by qRT-PCR, Western blot (WB), and IHC. CCK8, Transwell, and colony formation assay were applied to assess the effects of KIF2C on cell proliferation, migration, and invasion. Cell apoptosis and cell cycle were analyzed by flow cytometry. The antitumor effect was further validated in the nude mouse xenograft cancer model and humanized mouse model. KIF2C expression was higher in EC. Knockdown of KIF2C prolonged the G1 phases and inhibited EC cell proliferation, migration, and invasion in vitro. Bioinformatics analysis indicated that KIF2C is negatively correlated with the infiltration level of CD8+ T cells but positively with the poor prognosis of EC patients. The apoptosis of CD8+ T cell was inhibited after the knockdown of KIF2C and was further inhibited when it is combined with anti-PD1. Conversely, compared to the knockdown of KIF2C expression alone, the combination of anti-PD1 further promoted the apoptosis of Ishikawa and RL95-2 cells. Moreover, the knockdown of KIF2C inhibited the expression of Ki-67 and the growth of tumors in the nude mouse xenograft cancer model. Our study found that the antitumor efficacy was further evaluated by the combination of anti-PD1 and KIF2C knockdown in a humanized mouse model. This study indicated that KIF2C is a novel prognostic biomarker that determines cancer progression and also a target for the therapy of EC and correlated with tumor immune cells infiltration in EC.
Collapse
|
15
|
Schweiggert J, Habeck G, Hess S, Mikus F, Beloshistov R, Meese K, Hata S, Knobeloch K, Melchior F. SCF Fbxw5 targets kinesin-13 proteins to facilitate ciliogenesis. EMBO J 2021; 40:e107735. [PMID: 34368969 PMCID: PMC8441365 DOI: 10.15252/embj.2021107735] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 11/23/2022] Open
Abstract
Microtubule depolymerases of the kinesin-13 family play important roles in various cellular processes and are frequently overexpressed in different cancer types. Despite the importance of their correct abundance, remarkably little is known about how their levels are regulated in cells. Using comprehensive screening on protein microarrays, we identified 161 candidate substrates of the multi-subunit ubiquitin E3 ligase SCFFbxw5 , including the kinesin-13 member Kif2c/MCAK. In vitro reconstitution assays demonstrate that MCAK and its closely related orthologs Kif2a and Kif2b become efficiently polyubiquitylated by neddylated SCFFbxw5 and Cdc34, without requiring preceding modifications. In cells, SCFFbxw5 targets MCAK for proteasomal degradation predominantly during G2 . While this seems largely dispensable for mitotic progression, loss of Fbxw5 leads to increased MCAK levels at basal bodies and impairs ciliogenesis in the following G1 /G0 , which can be rescued by concomitant knockdown of MCAK, Kif2a or Kif2b. We thus propose a novel regulatory event of ciliogenesis that begins already within the G2 phase of the preceding cell cycle.
Collapse
Affiliation(s)
- Jörg Schweiggert
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Gregor Habeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Sandra Hess
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Felix Mikus
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Roman Beloshistov
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Klaus Meese
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Shoji Hata
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | | | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| |
Collapse
|
16
|
Barisic M, Rajendraprasad G, Steblyanko Y. The metaphase spindle at steady state - Mechanism and functions of microtubule poleward flux. Semin Cell Dev Biol 2021; 117:99-117. [PMID: 34053864 DOI: 10.1016/j.semcdb.2021.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 11/24/2022]
Abstract
The mitotic spindle is a bipolar cellular structure, built from tubulin polymers, called microtubules, and interacting proteins. This macromolecular machine orchestrates chromosome segregation, thereby ensuring accurate distribution of genetic material into the two daughter cells during cell division. Powered by GTP hydrolysis upon tubulin polymerization, the microtubule ends exhibit a metastable behavior known as the dynamic instability, during which they stochastically switch between the growth and shrinkage phases. In the context of the mitotic spindle, dynamic instability is furthermore regulated by microtubule-associated proteins and motor proteins, which enables the spindle to undergo profound changes during mitosis. This highly dynamic behavior is essential for chromosome capture and congression in prometaphase, as well as for chromosome alignment to the spindle equator in metaphase and their segregation in anaphase. In this review we focus on the mechanisms underlying microtubule dynamics and sliding and their importance for the maintenance of shape, structure and dynamics of the metaphase spindle. We discuss how these spindle properties are related to the phenomenon of microtubule poleward flux, highlighting its highly cooperative molecular basis and role in keeping the metaphase spindle at a steady state.
Collapse
Affiliation(s)
- Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| | - Girish Rajendraprasad
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Yulia Steblyanko
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Strandboulevarden 49, 2100 Copenhagen, Denmark
| |
Collapse
|
17
|
Minagawa M, Shirato M, Toya M, Sato M. Dual Impact of a Benzimidazole Resistant β-Tubulin on Microtubule Behavior in Fission Yeast. Cells 2021; 10:1042. [PMID: 33925026 PMCID: PMC8145593 DOI: 10.3390/cells10051042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
The cytoskeleton microtubule consists of polymerized αβ-tubulin dimers and plays essential roles in many cellular events. Reagents that inhibit microtubule behaviors have been developed as antifungal, antiparasitic, and anticancer drugs. Benzimidazole compounds, including thiabendazole (TBZ), carbendazim (MBC), and nocodazole, are prevailing microtubule poisons that target β-tubulin and inhibit microtubule polymerization. The molecular basis, however, as to how the drug acts on β-tubulin remains controversial. Here, we characterize the S. pombe β-tubulin mutant nda3-TB101, which was previously isolated as a mutant resistance to benzimidazole. The mutation site tyrosine at position 50 is located in the interface of two lateral β-tubulin proteins and at the gate of a putative binging pocket for benzimidazole. Our observation revealed two properties of the mutant tubulin. First, the dynamics of cellular microtubules comprising the mutant β-tubulin were stabilized in the absence of benzimidazole. Second, the mutant protein reduced the affinity to benzimidazole in vitro. We therefore conclude that the mutant β-tubulin Nda3-TB101 exerts a dual effect on microtubule behaviors: the mutant β-tubulin stabilizes microtubules and is insensitive to benzimidazole drugs. This notion fine-tunes the current elusive molecular model regarding binding of benzimidazole to β-tubulin.
Collapse
Affiliation(s)
- Mamika Minagawa
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
| | - Minamo Shirato
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
| | - Mika Toya
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
- Faculty of Science and Engineering, Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Masamitsu Sato
- Laboratory of Cytoskeletal Logistics, Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan; (M.M.); (M.S.); (M.T.)
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Institute for Medical-Oriented Structural Biology, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
18
|
McIntosh JR. Anaphase A. Semin Cell Dev Biol 2021; 117:118-126. [PMID: 33781672 DOI: 10.1016/j.semcdb.2021.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
Anaphase A is the motion of recently separated chromosomes to the spindle pole they face. It is accompanied by the shortening of kinetochore-attached microtubules. The requisite tubulin depolymerization may occur at kinetochores, at poles, or both, depending on the species and/or the time in mitosis. These depolymerization events are local and suggest that cells regulate microtubule dynamics in specific places, presumably by the localization of relevant enzymes and microtubule-associated proteins to specific loci, such as pericentriolar material and outer kinetochores. Motor enzymes can contribute to anaphase A, both by altering microtubule stability and by pushing or pulling microtubules through the cell. The generation of force on chromosomes requires couplings that can both withstand the considerable force that spindles can generate and simultaneously permit tubulin addition and loss. This chapter reviews literature on the molecules that regulate anaphase microtubule dynamics, couple dynamic microtubules to kinetochores and poles, and generate forces for microtubule and chromosome motion.
Collapse
Affiliation(s)
- J Richard McIntosh
- Dept. of Molecular, Cellular, and Developmental Biology University of Colorado, Boulder, CO 80309-0347, USA.
| |
Collapse
|
19
|
Sperm Methylome Profiling Can Discern Fertility Levels in the Porcine Biomedical Model. Int J Mol Sci 2021; 22:ijms22052679. [PMID: 33800945 PMCID: PMC7961483 DOI: 10.3390/ijms22052679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
A combined Genotyping By Sequencing (GBS) and methylated DNA immunoprecipitation (MeDIP) protocol was used to identify—in parallel—genetic variation (Genomic-Wide Association Studies (GWAS) and epigenetic differences of Differentially Methylated Regions (DMR) in the genome of spermatozoa from the porcine animal model. Breeding boars with good semen quality (n = 11) and specific and well-documented differences in fertility (farrowing rate, FR) and prolificacy (litter size, LS) (n = 7) in artificial insemination programs, using combined FR and LS, were categorized as High Fertile (HF, n = 4) or Low Fertile (LF, n = 3), and boars with Unknown Fertility (UF, n = 4) were tested for eventual epigenetical similarity with those fertility-proven. We identified 165,944 Single Nucleotide Polymorphisms (SNPs) that explained 14–15% of variance among selection lines. Between HF and LF individuals (n = 7, 4 HF and 3 LF), we identified 169 SNPs with p ≤ 0.00015, which explained 58% of the variance. For the epigenetic analyses, we considered fertility and period of ejaculate collection (late-summer and mid-autumn). Approximately three times more DMRs were observed in HF than in LF boars across these periods. Interestingly, UF boars were clearly clustered with one of the other HF or LF groups. The highest differences in DMRs between HF and LF experimental groups across the pig genome were located in the chr 3, 9, 13, and 16, with most DMRs being hypermethylated in LF boars. In both HF and LF boars, DMRs were mostly hypermethylated in late-summer compared to mid-autumn. Three overlaps were detected between SNPs (p ≤ 0.0005, n = 1318) and CpG sites within DMRs. In conclusion, fertility levels in breeding males including FR and LS can be discerned using methylome analyses. The findings in this biomedical animal model ought to be applied besides sire selection for andrological diagnosis of idiopathic sub/infertility.
Collapse
|
20
|
Vemu A, Szczesna E, Roll-Mecak A. In Vitro Reconstitution Assays of Microtubule Amplification and Lattice Repair by the Microtubule-Severing Enzymes Katanin and Spastin. Methods Mol Biol 2021; 2101:27-38. [PMID: 31879896 DOI: 10.1007/978-1-0716-0219-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Microtubules are non-covalent dynamic polymers essential for the life of all eukaryotic cells. Their dynamic behavior is regulated by a large array of cellular effectors. In vitro microtubule assays have been instrumental in dissecting the mechanism of microtubule-associated proteins. In this chapter, we focus on microtubule-severing enzymes katanin and spastin. They are AAA ATPases that generate internal breaks in microtubules by extracting tubulin dimers out of the microtubule lattice. We present protocols for TIRF microscopy-based assays that were instrumental in proving that these enzymes not only sever microtubules but also remodel the microtubule lattice by promoting the exchange of lattice GDP-tubulin with GTP-tubulin from the soluble pool. This activity can modulate microtubule dynamics and support microtubule-dependent microtubule amplification in the absence of a nucleating factor.
Collapse
Affiliation(s)
- Annapurna Vemu
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ewa Szczesna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA. .,Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
21
|
Wang Z, Chu KC, Tsao HK, Sheng YJ. Preferred penetration of active nano-rods into narrow channels and their clustering. Phys Chem Chem Phys 2021; 23:16234-16241. [PMID: 34308947 DOI: 10.1039/d1cp01065d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In a channel connected to a reservoir, passive particles prefer staying in the reservoir than the channel due to the entropic effect, as the size of the particles is comparable to that of the channel. Self-propelled rods can exhibit out-of-equilibrium phenomena, and their partition behavior may differ from that of passive rods due to their persistent swimming ability. In this work, the distribution of active nano-rods between the nanoscale channel and reservoir is explored using dissipative particle dynamics. The ratio of the nano-rod concentration in the slit to that in the reservoir, defined as the partition ratio Ψ, is a function of active force, channel width, and rod length. Although passive nano-rods prefer staying in bulk (Ψ < 1), active rods can overcome the entropic barrier and show favorable partition toward narrow channels (Ψ > 1). As the slit width decreases to about the rod's width, active rods entering the slit behave like a quasi-two-dimensional system dynamically. At sufficiently high concentrations and Peclet numbers, nano-rods tend to align and move together in the same direction for a certain time. The distribution (PM) of the cluster size (M) follows a power law, PM ∝ M-2, for small clusters.
Collapse
Affiliation(s)
- Zhengjia Wang
- Condensed Matter Science and Technology Institute, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | | | | | | |
Collapse
|
22
|
Singh D, Schmidt N, Müller F, Bange T, Bird AW. Destabilization of Long Astral Microtubules via Cdk1-Dependent Removal of GTSE1 from Their Plus Ends Facilitates Prometaphase Spindle Orientation. Curr Biol 2020; 31:766-781.e8. [PMID: 33333009 DOI: 10.1016/j.cub.2020.11.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/25/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
The precise regulation of microtubule dynamics over time and space in dividing cells is critical for several mitotic mechanisms that ultimately enable cell proliferation, tissue organization, and development. Astral microtubules, which extend from the centrosome toward the cell cortex, must be present for the mitotic spindle to properly orient, as well as for the faithful execution of anaphase and cytokinesis. However, little is understood about how the dynamic properties of astral microtubules are regulated spatiotemporally, or the contribution of astral microtubule dynamics to spindle positioning. The mitotic regulator Cdk1-CyclinB promotes destabilization of centrosomal microtubules and increased microtubule dynamics as cells enter mitosis, but how Cdk1 activity modulates astral microtubule stability, and whether it impacts spindle positioning, is unknown. Here, we uncover a mechanism revealing that Cdk1 destabilizes astral microtubules in prometaphase and thereby influences spindle reorientation. Phosphorylation of the EB1-dependent microtubule plus-end tracking protein GTSE1 by Cdk1 in early mitosis abolishes its interaction with EB1 and recruitment to microtubule plus ends. Loss of Cdk1 activity, or mutation of phosphorylation sites in GTSE1, induces recruitment of GTSE1 to growing microtubule plus ends in mitosis. This decreases the catastrophe frequency of astral microtubules and causes an increase in the number of long astral microtubules reaching the cell cortex, which restrains the ability of cells to reorient spindles along the long cellular axis in early mitosis. Astral microtubules thus must not only be present but also dynamic to allow the spindle to reorient, a state assisted by selective destabilization of long astral microtubules via Cdk1.
Collapse
Affiliation(s)
- Divya Singh
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Franziska Müller
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Tanja Bange
- Department for Systems Chronobiology, Institute of Medical Psychology, LMU Munich, Goethestrasse 31/ I, 80336 Munich, Germany
| | - Alexander W Bird
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.
| |
Collapse
|
23
|
Rondelet A, Lin YC, Singh D, Porfetye AT, Thakur HC, Hecker A, Brinkert P, Schmidt N, Bendre S, Müller F, Mazul L, Widlund PO, Bange T, Hiller M, Vetter IR, Bird AW. Clathrin's adaptor interaction sites are repurposed to stabilize microtubules during mitosis. J Cell Biol 2020; 219:133599. [PMID: 31932847 PMCID: PMC7041688 DOI: 10.1083/jcb.201907083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/31/2019] [Accepted: 11/24/2019] [Indexed: 11/22/2022] Open
Abstract
Clathrin ensures mitotic spindle stability and efficient chromosome alignment, independently of its vesicle trafficking function. Although clathrin localizes to the mitotic spindle and kinetochore fiber microtubule bundles, the mechanisms by which clathrin stabilizes microtubules are unclear. We show that clathrin adaptor interaction sites on clathrin heavy chain (CHC) are repurposed during mitosis to directly recruit the microtubule-stabilizing protein GTSE1 to the spindle. Structural analyses reveal that these sites interact directly with clathrin-box motifs on GTSE1. Disruption of this interaction releases GTSE1 from spindles, causing defects in chromosome alignment. Surprisingly, this disruption destabilizes astral microtubules, but not kinetochore-microtubule attachments, and chromosome alignment defects are due to a failure of chromosome congression independent of kinetochore-microtubule attachment stability. GTSE1 recruited to the spindle by clathrin stabilizes microtubules by inhibiting the microtubule depolymerase MCAK. This work uncovers a novel role of clathrin adaptor-type interactions to stabilize nonkinetochore fiber microtubules to support chromosome congression, defining for the first time a repurposing of this endocytic interaction mechanism during mitosis.
Collapse
Affiliation(s)
- Arnaud Rondelet
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Yu-Chih Lin
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Divya Singh
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Harish C Thakur
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andreas Hecker
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Pia Brinkert
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Shweta Bendre
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Lisa Mazul
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Per O Widlund
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tanja Bange
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.,Center for Systems Biology, Dresden, Germany
| | - Ingrid R Vetter
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | |
Collapse
|
24
|
Roll-Mecak A. The Tubulin Code in Microtubule Dynamics and Information Encoding. Dev Cell 2020; 54:7-20. [PMID: 32634400 PMCID: PMC11042690 DOI: 10.1016/j.devcel.2020.06.008] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/08/2020] [Accepted: 06/03/2020] [Indexed: 01/05/2023]
Abstract
Microtubules are non-covalent mesoscale polymers central to the eukaryotic cytoskeleton. Microtubule structure, dynamics, and mechanics are modulated by a cell's choice of tubulin isoforms and post-translational modifications, a "tubulin code," which is thought to support the diverse morphology and dynamics of microtubule arrays across various cell types, cell cycle, and developmental stages. We give a brief historical overview of research into tubulin diversity and highlight recent progress toward uncovering the mechanistic underpinnings of the tubulin code. As a large number of essential pathways converge upon the microtubule cytoskeleton, understanding how cells utilize tubulin diversity is crucial to understanding cellular physiology and disease.
Collapse
Affiliation(s)
- Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Kixmoeller K, Allu PK, Black BE. The centromere comes into focus: from CENP-A nucleosomes to kinetochore connections with the spindle. Open Biol 2020; 10:200051. [PMID: 32516549 PMCID: PMC7333888 DOI: 10.1098/rsob.200051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic chromosome segregation relies upon specific connections from DNA to the microtubule-based spindle that forms at cell division. The chromosomal locus that directs this process is the centromere, where a structure called the kinetochore forms upon entry into mitosis. Recent crystallography and single-particle electron microscopy have provided unprecedented high-resolution views of the molecular complexes involved in this process. The centromere is epigenetically specified by nucleosomes harbouring a histone H3 variant, CENP-A, and we review recent progress on how it differentiates centromeric chromatin from the rest of the chromosome, the biochemical pathway that mediates its assembly and how two non-histone components of the centromere specifically recognize CENP-A nucleosomes. The core centromeric nucleosome complex (CCNC) is required to recruit a 16-subunit complex termed the constitutive centromere associated network (CCAN), and we highlight recent structures reported of the budding yeast CCAN. Finally, the structures of multiple modular sub-complexes of the kinetochore have been solved at near-atomic resolution, providing insight into how connections are made to the CCAN on one end and to the spindle microtubules on the other. One can now build molecular models from the DNA through to the physical connections to microtubules.
Collapse
Affiliation(s)
- Kathryn Kixmoeller
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Graduate Program in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Praveen Kumar Allu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ben E Black
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Graduate Program in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Yang C, Li Q, Chen X, Zhang Z, Mou Z, Ye F, Jin S, Jun X, Tang F, Jiang H. Circular RNA circRGNEF promotes bladder cancer progression via miR-548/KIF2C axis regulation. Aging (Albany NY) 2020; 12:6865-6879. [PMID: 32305958 PMCID: PMC7202505 DOI: 10.18632/aging.103047] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/09/2020] [Indexed: 01/27/2023]
Abstract
Circular RNAs (circRNAs) play an important role in bladder cancer (BC). Though circRNA involvement in BC has been reported, the underlying regulatory mechanisms are unknown. In this study, we performed EdU, CCK8, colony formation and Transwell assays to establish the role of circRGNEF in BC cell migration, proliferation, and invasion. We used bioinformatics and luciferase reporter experiments to investigate the regulatory mechanism. Nude mice xenografts and live imaging were used to explore the role of circRGNEF in tumor metastasis and growth. Expression profile analysis of human circRNAs in BC revealed that circRGNEF was upregulated significantly. High circRGNEF expression was correlated with aggressive BC phenotypes. The downregulation of circRGNEF suppressed BC cell metastasis and proliferation by targeting the miR-548/KIF2C axis in vitro and in vivo; these results were verified with luciferase reporter assays. Our results show that miR-548 downregulation or KIF2C overexpression restored BC cell proliferation, migration, and invasion following silencing of circRGNEF. KIF2C overexpression reversed miR-548-induced cell invasion and migration as well as growth inhibition in vitro. In summary, the data illustrate that circRGNEF suppresses BC progression by functioning as a miR-548 sponge to enhance KIF2C expression. Therefore, circRGNEF might be a candidate BC treatment target.
Collapse
Affiliation(s)
- Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai 200032, China
| | - Qiong Li
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xinan Chen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zheyu Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zezhong Mou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shengming Jin
- Shanghai Cancer Center, Fudan University, Shanghai 200040, China
| | - Xiang Jun
- Department of Urinary Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Feng Tang
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|
27
|
She ZY, Zhong N, Yu KW, Xiao Y, Wei YL, Lin Y, Li YL, Lu MH. Kinesin-5 Eg5 is essential for spindle assembly and chromosome alignment of mouse spermatocytes. Cell Div 2020; 15:6. [PMID: 32165913 PMCID: PMC7060529 DOI: 10.1186/s13008-020-00063-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/29/2020] [Indexed: 11/10/2022] Open
Abstract
Background Microtubule organization is essential for bipolar spindle assembly and chromosome segregation, which contribute to genome stability. Kinesin-5 Eg5 is known to be a crucial regulator in centrosome separation and spindle assembly in mammalian somatic cells, however, the functions and mechanisms of Eg5 in male meiotic cell division remain largely unknown. Results In this study, we have found that Eg5 proteins are expressed in mouse spermatogonia, spermatocytes and spermatids. After Eg5 inhibition by specific inhibitors Monastrol, STLC and Dimethylenastron, the meiotic spindles of dividing spermatocytes show spindle collapse and the defects in bipolar spindle formation. We demonstrate that Eg5 regulates spindle bipolarity and the maintenance of meiotic spindles in meiosis. Eg5 inhibition leads to monopolar spindles, spindle abnormalities and chromosome misalignment in cultured GC-2 spd cells. Furthermore, Eg5 inhibition results in the decrease of the spermatids and the abnormalities in mature sperms. Conclusions Our results have revealed an important role of kinesin-5 Eg5 in male meiosis and the maintenance of male fertility. We demonstrate that Eg5 is crucial for bipolar spindle assembly and chromosome alignment in dividing spermatocytes. Our data provide insights into the functions of Eg5 in meiotic spindle assembly of dividing spermatocytes.
Collapse
Affiliation(s)
- Zhen-Yu She
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China.,Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122 Fujian China
| | - Ning Zhong
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| | - Kai-Wei Yu
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| | - Yu Xiao
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001 Fujian China.,4Fujian Provincial Children's Hospital, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, 350001 Fujian China
| | - Yang Lin
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| | - Yue-Ling Li
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| | - Ming-Hui Lu
- 1Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122 Fujian China
| |
Collapse
|
28
|
Wang Z, Si T, Hao J, Guan Y, Qin F, Yang B, Cao W. Defect dynamics in clusters of self-propelled rods in circular confinement. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2019; 42:150. [PMID: 31773335 DOI: 10.1140/epje/i2019-11911-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/22/2019] [Indexed: 06/10/2023]
Abstract
Rod-shaped active micro/nano-particles, such as bacterial and bipolar metallic micro/nano-motors, demonstrate novel collective phenomena far from the equilibrium state compared to passive particles. We apply a simulation approach --dissipative particle dynamics (DPD)-- to explore the collectively ordered states of self-propelled rods (SPRs). The SPRs are confined in a finite circular zone and repel each other when two rods touch each other. It is found that for a long enough rods system, the global vortex patterns, dynamic pattern oscillation between hedgehog pattern and vortex pattern, and hedgehog patterns are observed successively with increasing active force Fa. For the vortex pattern, the total interaction energy between the rods U is linear with active force Fa, i.e., U ∼ Fa . While the relation U ∼ Fa2 is obtained for the hedgehog structure. It is observed that a new hedgehog pattern with one defect core is created by two ejections of polar cluster in opposite directions from the original hedgehog pattern, and then merges into one through the diffusion of the two aggregates, i.e., the creation and annihilation of topological charges.
Collapse
Affiliation(s)
- Zhengjia Wang
- Condensed Matter Science and Technology Institute, School of Instrumentation Science and Engineering, Harbin Institute of Technology, 150080, Harbin, P.R. China
| | - Tieyan Si
- School of Physics, Harbin Institute of Technology, 150080, Harbin, P.R. China
| | - Junhua Hao
- Department of Physics, Tianjin University Renai College, 301636, Tianjin, P.R. China.
| | - Yu Guan
- Amur State University, 675004, Blagoveshchensk, Russia
| | - Feng Qin
- Condensed Matter Science and Technology Institute, School of Instrumentation Science and Engineering, Harbin Institute of Technology, 150080, Harbin, P.R. China
| | - Bin Yang
- Condensed Matter Science and Technology Institute, School of Instrumentation Science and Engineering, Harbin Institute of Technology, 150080, Harbin, P.R. China
| | - Wenwu Cao
- Condensed Matter Science and Technology Institute, School of Instrumentation Science and Engineering, Harbin Institute of Technology, 150080, Harbin, P.R. China
| |
Collapse
|
29
|
Zeeshan M, Shilliday F, Liu T, Abel S, Mourier T, Ferguson DJP, Rea E, Stanway RR, Roques M, Williams D, Daniel E, Brady D, Roberts AJ, Holder AA, Pain A, Le Roch KG, Moores CA, Tewari R. Plasmodium kinesin-8X associates with mitotic spindles and is essential for oocyst development during parasite proliferation and transmission. PLoS Pathog 2019; 15:e1008048. [PMID: 31600347 PMCID: PMC6786531 DOI: 10.1371/journal.ppat.1008048] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022] Open
Abstract
Kinesin-8 proteins are microtubule motors that are often involved in regulation of mitotic spindle length and chromosome alignment. They move towards the plus ends of spindle microtubules and regulate the dynamics of these ends due, at least in some species, to their microtubule depolymerization activity. Plasmodium spp. exhibit an atypical endomitotic cell division in which chromosome condensation and spindle dynamics in the different proliferative stages are not well understood. Genome-wide shared orthology analysis of Plasmodium spp. revealed the presence of two kinesin-8 motor proteins, kinesin-8X and kinesin-8B. Here we studied the biochemical properties of kinesin-8X and its role in parasite proliferation. In vitro, kinesin-8X has motility and depolymerization activities like other kinesin-8 motors. To understand the role of Plasmodium kinesin-8X in cell division, we used fluorescence-tagging and live cell imaging to define its location, and gene targeting to analyse its function, during all proliferative stages of the rodent malaria parasite P. berghei life cycle. The results revealed a spatio-temporal involvement of kinesin-8X in spindle dynamics and an association with both mitotic and meiotic spindles and the putative microtubule organising centre (MTOC). Deletion of the kinesin-8X gene revealed a defect in oocyst development, confirmed by ultrastructural studies, suggesting that this protein is required for oocyst development and sporogony. Transcriptome analysis of Δkinesin-8X gametocytes revealed modulated expression of genes involved mainly in microtubule-based processes, chromosome organisation and the regulation of gene expression, supporting a role for kinesin-8X in cell division. Kinesin-8X is thus required for parasite proliferation within the mosquito and for transmission to the vertebrate host.
Collapse
Affiliation(s)
- Mohammad Zeeshan
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Fiona Shilliday
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, United Kingdom
| | - Tianyang Liu
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, United Kingdom
| | - Steven Abel
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Tobias Mourier
- Biological Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Jeddah, Kingdom of Saudi Arabia
| | - David J. P. Ferguson
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Gipsy Lane, Oxford, United Kingdom
| | - Edward Rea
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Desiree Williams
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Emilie Daniel
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Declan Brady
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Anthony J. Roberts
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, United Kingdom
| | - Anthony A. Holder
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Arnab Pain
- Biological Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Jeddah, Kingdom of Saudi Arabia
- Research Center for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Kita-ku, Sapporo, Japan
| | - Karine G. Le Roch
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Carolyn A. Moores
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, United Kingdom
| | - Rita Tewari
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
30
|
Strothman C, Farmer V, Arpağ G, Rodgers N, Podolski M, Norris S, Ohi R, Zanic M. Microtubule minus-end stability is dictated by the tubulin off-rate. J Cell Biol 2019; 218:2841-2853. [PMID: 31420452 PMCID: PMC6719460 DOI: 10.1083/jcb.201905019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/11/2019] [Accepted: 07/23/2019] [Indexed: 12/25/2022] Open
Abstract
Dynamic organization of microtubule minus ends is vital for the formation and maintenance of acentrosomal microtubule arrays. In vitro, both microtubule ends switch between phases of assembly and disassembly, a behavior called dynamic instability. Although minus ends grow slower, their lifetimes are similar to those of plus ends. The mechanisms underlying these distinct dynamics remain unknown. Here, we use an in vitro reconstitution approach to investigate minus-end dynamics. We find that minus-end lifetimes are not defined by the mean size of the protective GTP-tubulin cap. Rather, we conclude that the distinct tubulin off-rate is the primary determinant of the difference between plus- and minus-end dynamics. Further, our results show that the minus-end-directed kinesin-14 HSET/KIFC1 suppresses tubulin off-rate to specifically suppress minus-end catastrophe. HSET maintains its protective minus-end activity even when challenged by a known microtubule depolymerase, kinesin-13 MCAK. Our results provide novel insight into the mechanisms of minus-end dynamics, essential for our understanding of microtubule minus-end regulation in cells.
Collapse
Affiliation(s)
- Claire Strothman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Veronica Farmer
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Göker Arpağ
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Nicole Rodgers
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Marija Podolski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Stephen Norris
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
- Department of Biochemistry, Vanderbilt University, Nashville, TN
| |
Collapse
|
31
|
Persico V, Callaini G, Riparbelli MG. The Microtubule-Depolymerizing Kinesin-13 Klp10A Is Enriched in the Transition Zone of the Ciliary Structures of Drosophila melanogaster. Front Cell Dev Biol 2019; 7:173. [PMID: 31497602 PMCID: PMC6713071 DOI: 10.3389/fcell.2019.00173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
The precursor of the flagellar axoneme is already present in the primary spermatocytes of Drosophila melanogaster. During spermatogenesis each primary spermatocyte shows a centriole pair that moves to the cell membrane and organizes an axoneme-based structure, the cilium-like region (CLR). The CLRs persist through the meiotic divisions and are inherited by young spermatids. During spermatid differentiation the ciliary caps elongate giving rise to the sperm axoneme. Mutations in Klp10A, a kinesin-13 of Drosophila, results in defects of centriole/CLR organization in spermatocytes and of ciliary cap assembly in elongating spermatids. Reduced Klp10A expression also results in strong structural defects of sensory type I neurons. We show, here, that this protein displays a peculiar localization during male gametogenesis. The Klp10A signal is first detected at the distal ends of the centrioles when they dock to the plasma membrane of young primary spermatocytes. At the onset of the first meiotic prometaphase, when the CLRs reach their full size, Klp10A is enriched in a distinct narrow area at the distal end of the centrioles and persists in elongating spermatids at the base of the ciliary cap. We conclude that Klp10A could be a core component of the ciliary transition zone in Drosophila.
Collapse
Affiliation(s)
| | - Giuliano Callaini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | |
Collapse
|
32
|
Letort G, Bennabi I, Dmitrieff S, Nedelec F, Verlhac MH, Terret ME. A computational model of the early stages of acentriolar meiotic spindle assembly. Mol Biol Cell 2019; 30:863-875. [PMID: 30650011 PMCID: PMC6589792 DOI: 10.1091/mbc.e18-10-0644] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 11/11/2022] Open
Abstract
The mitotic spindle is an ensemble of microtubules responsible for the repartition of the chromosomal content between the two daughter cells during division. In metazoans, spindle assembly is a gradual process involving dynamic microtubules and recruitment of numerous associated proteins and motors. During mitosis, centrosomes organize and nucleate the majority of spindle microtubules. In contrast, oocytes lack canonical centrosomes but are still able to form bipolar spindles, starting from an initial ball that self-organizes in several hours. Interfering with early steps of meiotic spindle assembly can lead to erroneous chromosome segregation. Although not fully elucidated, this process is known to rely on antagonistic activities of plus end- and minus end-directed motors. We developed a model of early meiotic spindle assembly in mouse oocytes, including key factors such as microtubule dynamics and chromosome movement. We explored how the balance between plus end- and minus end-directed motors, as well as the influence of microtubule nucleation, impacts spindle morphology. In a refined model, we added spatial regulation of microtubule stability and minus-end clustering. We could reproduce the features of early stages of spindle assembly from 12 different experimental perturbations and predict eight additional perturbations. With its ability to characterize and predict chromosome individualization, this model can help deepen our understanding of spindle assembly.
Collapse
Affiliation(s)
- Gaelle Letort
- CIRB, Collège de France, UMR7241/U1050, F-75005 Paris, France
| | - Isma Bennabi
- CIRB, Collège de France, UMR7241/U1050, F-75005 Paris, France
| | - Serge Dmitrieff
- Institut Jacques Monod, UMR7592 and Université Paris-Diderot, F-75205 Paris, France
| | - François Nedelec
- Centre de Recherche Interdisciplinaire, F-75004 Paris, France
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | | |
Collapse
|
33
|
Parts list for a microtubule depolymerising kinesin. Biochem Soc Trans 2018; 46:1665-1672. [PMID: 30467119 PMCID: PMC6299235 DOI: 10.1042/bst20180350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
The Kinesin superfamily is a large group of molecular motors that use the turnover of ATP to regulate their interaction with the microtubule cytoskeleton. The coupled relationship between nucleotide turnover and microtubule binding is harnessed in various ways by these motors allowing them to carry out a variety of cellular functions. The Kinesin-13 family is a group of specialist microtubule depolymerising motors. Members of this family use their microtubule destabilising activity to regulate processes such as chromosome segregation, maintenance of cilia and neuronal development. Here, we describe the current understanding of the structure of this family of kinesins and the role different parts of these proteins play in their microtubule depolymerisation activity and in the wider function of this family of kinesins.
Collapse
|
34
|
Abstract
Microtubules are dynamic polymers of αβ-tubulin that are essential for intracellular organization, organelle trafficking and chromosome segregation. Microtubule growth and shrinkage occur via addition and loss of αβ-tubulin subunits, which are biochemical processes. Dynamic microtubules can also engage in mechanical processes, such as exerting forces by pushing or pulling against a load. Recent advances at the intersection of biochemistry and mechanics have revealed the existence of multiple conformations of αβ-tubulin subunits and their central role in dictating the mechanisms of microtubule dynamics and force generation. It has become apparent that microtubule-associated proteins (MAPs) selectively target specific tubulin conformations to regulate microtubule dynamics, and mechanical forces can also influence microtubule dynamics by altering the balance of tubulin conformations. Importantly, the conformational states of tubulin dimers are likely to be coupled throughout the lattice: the conformation of one dimer can influence the conformation of its nearest neighbours, and this effect can propagate over longer distances. This coupling provides a long-range mechanism by which MAPs and forces can modulate microtubule growth and shrinkage. These findings provide evidence that the interplay between biochemistry and mechanics is essential for the cellular functions of microtubules.
Collapse
Affiliation(s)
- Gary J Brouhard
- Department of Biology, McGill University, Montréal, Quebec, Canada.
| | - Luke M Rice
- Department of Biophysics, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
35
|
Mechanism of Catalytic Microtubule Depolymerization via KIF2-Tubulin Transitional Conformation. Cell Rep 2018; 20:2626-2638. [PMID: 28903043 DOI: 10.1016/j.celrep.2017.08.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/31/2017] [Accepted: 08/18/2017] [Indexed: 11/23/2022] Open
Abstract
Microtubules (MTs) are dynamic structures that are fundamental for cell morphogenesis and motility. MT-associated motors work efficiently to perform their functions. Unlike other motile kinesins, KIF2 catalytically depolymerizes MTs from the peeled protofilament end during ATP hydrolysis. However, the detailed mechanism by which KIF2 drives processive MT depolymerization remains unknown. To elucidate the catalytic mechanism, the transitional KIF2-tubulin complex during MT depolymerization was analyzed through multiple methods, including atomic force microscopy, size-exclusion chromatography, multi-angle light scattering, small-angle X-ray scattering, analytical ultracentrifugation, and mass spectrometry. The analyses outlined the conformation in which one KIF2core domain binds tightly to two tubulin dimers in the middle pre-hydrolysis state during ATP hydrolysis, a process critical for catalytic MT depolymerization. The X-ray crystallographic structure of the KIF2core domain displays the activated conformation that sustains the large KIF2-tubulin 1:2 complex.
Collapse
|
36
|
Wang Z, Chen YF, Chen HY, Sheng YJ, Tsao HK. Mechanical pressure, surface excess, and polar order of a dilute rod-like nanoswimmer suspension: role of swimmer-wall interactions. SOFT MATTER 2018; 14:2906-2914. [PMID: 29589848 DOI: 10.1039/c7sm02372c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The mechanical pressure, surface excess, and polar order of a dilute rod-like nanoswimmer suspension confined by two parallel plates are explored by dissipative particle dynamics. The accumulation and preferred orientation of swimmers near the walls are distinctly shown through the density and polar order distributions for various active force, Fa, values and rod lengths. As Fa is increased, it is interesting to observe that there exists a maximum of the polar order, revealing that the dominant mechanism of the swimmer behavior can be altered by the coupling between the active force and the rod-wall interaction. As a result, the influences of the active force on the swim pressure Π(w)a contributed by the swimmers directly and the surface excess Γ* can be classified into two scaling regimes, natural rotation (weak propulsion) and forced rotation (strong propulsion). Π(w)a and Γ* are proportional to Fa2 in the former regime but become proportional to Fa in the latter regime. For all rod-wall repulsions, the swim pressure of active rods in confined systems Π(w)a always differs from that in unbounded systems Π(b)a which is simply proportional to Fa2 associated with the active diffusivity. That is, unlike thermal equilibrium systems, Π(w)a is not a state function because of the presence of the wall-torque.
Collapse
Affiliation(s)
- Zhengjia Wang
- Condensed Matter Science and Technology Institute, School of Science, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | | | | | | | | |
Collapse
|
37
|
Duan H, Zhang X, Wang FX, Cai MY, Ma GW, Yang H, Fu JH, Tan ZH, Fu XY, Ma QL, Wang XY, Lin P. KIF-2C expression is correlated with poor prognosis of operable esophageal squamous cell carcinoma male patients. Oncotarget 2018; 7:80493-80507. [PMID: 27563815 PMCID: PMC5348336 DOI: 10.18632/oncotarget.11492] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 08/09/2016] [Indexed: 01/23/2023] Open
Abstract
To determine the prognostic significance of Kinesin family member 2C (KIF-2C) expression in patients with operable esophageal squamous cell carcinoma (ESCC), we conducted an immunohistochemical analysis of KIF-2C expression in 415 surgically resected primary tumor tissues and 40 adjacent non-cancerous tissues from patients with operable ESCC. The median duration of postoperative follow-up was 76.0 months. Higher KIF-2C expression was associated with significantly increased risks of higher pathologic tumor (pT) status (P=0.038) and poorer tumor differentiation (P=0.022). For the entire cohort, KIF-2C expression was not an independent factor significantly associated with overall survival (OS) (P=0.097) or disease-free survival (DFS) (P=0.152). In female patients, KIF-2C expression had no effect on OS (P=0.880) and DFS (P=0.864). However, OS (hazard ratio (HR)=1.480, P=0.013) and DFS (HR=1.418, P=0.024) were worse for male patients with high KIF-2C expression compared with male patients with low KIF-2C expression. Moreover, the OS and DFS of male patients with high KIF-2C expression were also significantly shorter compared with female patients with low KIF-2C expression (P=0.022, P=0.029) and female patients with high KIF-2C expression (P=0.014, P=0.018). Based on these findings, KIF-2C expression in tumor tissues promises to serve as an independent prognostic marker for male, but not female, patients with operable ESCC. Prognosis was worse for male patients with high KIF-2C expression compared with patients with the same pathologic tumor-node-metastasis (pTNM) stage.
Collapse
Affiliation(s)
- Hao Duan
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xu Zhang
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China.,Guangdong Esophageal Cancer Research Institute, Guangzhou 510060, Guangdong Province, China
| | - Fei-Xiang Wang
- Department of Thoracic Oncology, Cancer Center of Guangzhou Medical University, Guangzhou 510095, Guangdong Province, China
| | - Mu-Yan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Guo-Wei Ma
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hong Yang
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China.,Guangdong Esophageal Cancer Research Institute, Guangzhou 510060, Guangdong Province, China
| | - Jian-Hua Fu
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China.,Guangdong Esophageal Cancer Research Institute, Guangzhou 510060, Guangdong Province, China
| | - Zi-Hui Tan
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xia-Yu Fu
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Qi-Long Ma
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xin-Ye Wang
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Peng Lin
- Department of Thoracic Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong Province, China.,Guangdong Esophageal Cancer Research Institute, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
38
|
Decker F, Oriola D, Dalton B, Brugués J. Autocatalytic microtubule nucleation determines the size and mass of Xenopus laevis egg extract spindles. eLife 2018; 7:31149. [PMID: 29323637 PMCID: PMC5814149 DOI: 10.7554/elife.31149] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/09/2018] [Indexed: 01/27/2023] Open
Abstract
Regulation of size and growth is a fundamental problem in biology. A prominent example is the formation of the mitotic spindle, where protein concentration gradients around chromosomes are thought to regulate spindle growth by controlling microtubule nucleation. Previous evidence suggests that microtubules nucleate throughout the spindle structure. However, the mechanisms underlying microtubule nucleation and its spatial regulation are still unclear. Here, we developed an assay based on laser ablation to directly probe microtubule nucleation events in Xenopus laevis egg extracts. Combining this method with theory and quantitative microscopy, we show that the size of a spindle is controlled by autocatalytic growth of microtubules, driven by microtubule-stimulated microtubule nucleation. The autocatalytic activity of this nucleation system is spatially regulated by the limiting amounts of active microtubule nucleators, which decrease with distance from the chromosomes. This mechanism provides an upper limit to spindle size even when resources are not limiting.
Collapse
Affiliation(s)
- Franziska Decker
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany,Center for Systems Biology DresdenDresdenGermany,Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| | - David Oriola
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany,Center for Systems Biology DresdenDresdenGermany,Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| | - Benjamin Dalton
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany,Center for Systems Biology DresdenDresdenGermany,Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| | - Jan Brugués
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany,Center for Systems Biology DresdenDresdenGermany,Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| |
Collapse
|
39
|
Ogawa T, Hirokawa N. Multiple analyses of protein dynamics in solution. Biophys Rev 2017; 10:299-306. [PMID: 29204883 DOI: 10.1007/s12551-017-0354-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022] Open
Abstract
The need for accurate description of protein behavior in solution has gained importance in various fields, including biophysics, biochemistry, structural biology, drug discovery, and antibody drugs. To achieve the desired accuracy, multiple precise analyses should be performed on the target molecule, compared, and effectively combined. This review focuses on the combination of multiple analyses in solution: size-exclusion chromatography (SEC), multi-angle light scattering (MALS), small-angle X-ray scattering (SAXS), analytical ultracentrifugation (AUC), and their complementary methods, such as atomic force microscopy (AFM) and mass spectrometry (MS). We also discuss the comparison between the determined molar mass value of not only the standard proteins, but of a target molecule tubulin and its depolymerizing protein, KIF2, as an example. The comparison of the estimated molar mass value from the different methods provides additional information about the target molecule, because the value reflects the dynamically changing states of the target molecule in solution. The combination and integration of multiple methods will permit a deeper understanding of protein dynamics in solution.
Collapse
Affiliation(s)
- Tadayuki Ogawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
40
|
Structural basis of small molecule ATPase inhibition of a human mitotic kinesin motor protein. Sci Rep 2017; 7:15121. [PMID: 29123223 PMCID: PMC5680195 DOI: 10.1038/s41598-017-14754-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/13/2017] [Indexed: 11/08/2022] Open
Abstract
Kinesin microtubule motor proteins play essential roles in division, including attaching chromosomes to spindles and crosslinking microtubules for spindle assembly. Human kinesin-14 KIFC1 is unique in that cancer cells with amplified centrosomes are dependent on the motor for viable division because of its ability to cluster centrosomes and form bipolar spindles, but it is not required for division in almost all normal cells. Screens for small molecule inhibitors of KIFC1 have yielded several candidates for further development, but obtaining structural data to determine their sites of binding has been difficult. Here we compare a previously unreported KIFC1 crystal structure with new structures of two closely related kinesin-14 proteins, Ncd and KIFC3, to determine the potential binding site of a known KIFC1 ATPase inhibitor, AZ82. We analyze the previously identified kinesin inhibitor binding sites and identify features of AZ82 that favor binding to one of the sites, the α4/α6 site. This selectivity can be explained by unique structural features of the KIFC1 α4/α6 binding site. These features may help improve the drug-like properties of AZ82 and other specific KIFC1 inhibitors.
Collapse
|
41
|
Barsegov V, Ross JL, Dima RI. Dynamics of microtubules: highlights of recent computational and experimental investigations. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2017; 29:433003. [PMID: 28812545 DOI: 10.1088/1361-648x/aa8670] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Microtubules are found in most eukaryotic cells, with homologs in eubacteria and archea, and they have functional roles in mitosis, cell motility, intracellular transport, and the maintenance of cell shape. Numerous efforts have been expended over the last two decades to characterize the interactions between microtubules and the wide variety of microtubule associated proteins that control their dynamic behavior in cells resulting in microtubules being assembled and disassembled where and when they are required by the cell. We present the main findings regarding microtubule polymerization and depolymerization and review recent work about the molecular motors that modulate microtubule dynamics by inducing either microtubule depolymerization or severing. We also discuss the main experimental and computational approaches used to quantify the thermodynamics and mechanics of microtubule filaments.
Collapse
Affiliation(s)
- Valeri Barsegov
- Department of Chemistry, University of Massachusetts, Lowell, MA 01854, United States of America
| | | | | |
Collapse
|
42
|
Vemu A, Atherton J, Spector JO, Moores CA, Roll-Mecak A. Tubulin isoform composition tunes microtubule dynamics. Mol Biol Cell 2017; 28:3564-3572. [PMID: 29021343 PMCID: PMC5706985 DOI: 10.1091/mbc.e17-02-0124] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 09/25/2017] [Accepted: 10/03/2017] [Indexed: 12/30/2022] Open
Abstract
We report the cryo-EM structure and dynamic parameters for unmodified α1B/βI+βIVb microtubules. These microtubules display markedly different dynamics compared to heterogeneous brain microtubules, and their dynamic parameters can be proportionally tuned by the addition of a recombinant neuronal tubulin isoform with different dynamic properties. Microtubules polymerize and depolymerize stochastically, a behavior essential for cell division, motility, and differentiation. While many studies advanced our understanding of how microtubule-associated proteins tune microtubule dynamics in trans, we have yet to understand how tubulin genetic diversity regulates microtubule functions. The majority of in vitro dynamics studies are performed with tubulin purified from brain tissue. This preparation is not representative of tubulin found in many cell types. Here we report the 4.2-Å cryo-electron microscopy (cryo-EM) structure and in vitro dynamics parameters of α1B/βI+βIVb microtubules assembled from tubulin purified from a human embryonic kidney cell line with isoform composition characteristic of fibroblasts and many immortalized cell lines. We find that these microtubules grow faster and transition to depolymerization less frequently compared with brain microtubules. Cryo-EM reveals that the dynamic ends of α1B/βI+βIVb microtubules are less tapered and that these tubulin heterodimers display lower curvatures. Interestingly, analysis of EB1 distributions at dynamic ends suggests no differences in GTP cap sizes. Last, we show that the addition of recombinant α1A/βIII tubulin, a neuronal isotype overexpressed in many tumors, proportionally tunes the dynamics of α1B/βI+βIVb microtubules. Our study is an important step toward understanding how tubulin isoform composition tunes microtubule dynamics.
Collapse
Affiliation(s)
- Annapurna Vemu
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Lung and Blood Institute, Bethesda, MD 20892
| | - Joseph Atherton
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London WC1E 7HX, United Kingdom
| | - Jeffrey O Spector
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Lung and Blood Institute, Bethesda, MD 20892
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London WC1E 7HX, United Kingdom
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Lung and Blood Institute, Bethesda, MD 20892 .,Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892
| |
Collapse
|
43
|
Feng S, Song Y, Shen M, Xie S, Li W, Lu Y, Yang Y, Ou G, Zhou J, Wang F, Liu W, Yan X, Liang X, Zhou T. Microtubule-binding protein FOR20 promotes microtubule depolymerization and cell migration. Cell Discov 2017; 3:17032. [PMID: 28884019 PMCID: PMC5583970 DOI: 10.1038/celldisc.2017.32] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022] Open
Abstract
Microtubules are highly dynamic filaments assembled from αβ-tubulin heterodimers and play important roles in many cellular processes, including cell division and migration. Microtubule dynamics is tightly regulated by microtubule-associated proteins (MAPs) that function by binding to microtubules or free tubulin dimers. Here, we report that FOR20 (FOP-related protein of 20 kDa), a conserved protein critical for ciliogenesis and cell cycle progression, is a previously uncharacterized MAP that facilitates microtubule depolymerization and promotes cell migration. FOR20 not only directly binds to microtubules but also regulates microtubule dynamics in vitro by decreasing the microtubule growth rate and increasing the depolymerization rate and catastrophe frequency. In the in vitro microtubule dynamics assays, FOR20 appears to preferentially interact with free tubulin dimers over microtubules. Depletion of FOR20 inhibits microtubule depolymerization and promotes microtubule regrowth after the nocodazole treatment in HeLa cells. In addition, FOR20 knockdown significantly inhibits both individual and collective migration of mammalian cells. Taken together, these data suggest that FOR20 functions as a MAP to promote microtubule depolymerization and cell migration.
Collapse
Affiliation(s)
- Sijie Feng
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| | - Yinlong Song
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.,Max-Planck Partner Group, School of Life Sciences, Tsinghua University, Beijing, Tianjing 300073, China
| | - Minhong Shen
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China
| | - Shanshan Xie
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| | - Wenjing Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Lu
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| | - Yuehong Yang
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jun Zhou
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fudi Wang
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China
| | - Wei Liu
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China
| | - Xiaoyi Yan
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| | - Xin Liang
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.,Max-Planck Partner Group, School of Life Sciences, Tsinghua University, Beijing, Tianjing 300073, China
| | - Tianhua Zhou
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Yuhangtang Road, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, Hangzhou 310058, China
| |
Collapse
|
44
|
Kuan HS, Betterton MD. Motor Protein Accumulation on Antiparallel Microtubule Overlaps. Biophys J 2017; 110:2034-43. [PMID: 27166811 DOI: 10.1016/j.bpj.2016.03.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/04/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022] Open
Abstract
Biopolymers serve as one-dimensional tracks on which motor proteins move to perform their biological roles. Motor protein phenomena have inspired theoretical models of one-dimensional transport, crowding, and jamming. Experiments studying the motion of Xklp1 motors on reconstituted antiparallel microtubule overlaps demonstrated that motors recruited to the overlap walk toward the plus end of individual microtubules and frequently switch between filaments. We study a model of this system that couples the totally asymmetric simple exclusion process for motor motion with switches between antiparallel filaments and binding kinetics. We determine steady-state motor density profiles for fixed-length overlaps using exact and approximate solutions of the continuum differential equations and compare to kinetic Monte Carlo simulations. Overlap motor density profiles and motor trajectories resemble experimental measurements. The phase diagram of the model is similar to the single-filament case for low switching rate, while for high switching rate we find a new (to our knowledge) low density-high density-low density-high density phase. The overlap center region, far from the overlap ends, has a constant motor density as one would naïvely expect. However, rather than following a simple binding equilibrium, the center motor density depends on total overlap length, motor speed, and motor switching rate. The size of the crowded boundary layer near the overlap ends is also dependent on the overlap length and switching rate in addition to the motor speed and bulk concentration. The antiparallel microtubule overlap geometry may offer a previously unrecognized mechanism for biological regulation of protein concentration and consequent activity.
Collapse
Affiliation(s)
- Hui-Shun Kuan
- Department of Physics, University of Colorado at Boulder, Boulder, Colorado; Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, Colorado
| | | |
Collapse
|
45
|
Mooney P, Sulerud T, Pelletier J, Dilsaver M, Tomschik M, Geisler C, Gatlin JC. Tau-based fluorescent protein fusions to visualize microtubules. Cytoskeleton (Hoboken) 2017; 74:221-232. [PMID: 28407416 PMCID: PMC5592782 DOI: 10.1002/cm.21368] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 01/10/2023]
Abstract
The ability to visualize cytoskeletal proteins and their dynamics in living cells has been critically important in advancing our understanding of numerous cellular processes, including actin- and microtubule (MT)-dependent phenomena such as cell motility, cell division, and mitosis. Here, we describe a novel set of fluorescent protein (FP) fusions designed specifically to visualize MTs in living systems using fluorescence microscopy. Each fusion contains a FP module linked in frame to a modified phospho-deficient version of the MT-binding domain of Tau (mTMBD). We found that expressed and purified constructs containing a single mTMBD decorated Xenopus egg extract spindles more homogenously than similar constructs containing the MT-binding domain of Ensconsin, suggesting that the binding affinity of mTMBD is minimally affected by localized signaling gradients generated during mitosis. Furthermore, MT dynamics were not grossly perturbed by the presence of Tau-based FP fusions. Interestingly, the addition of a second mTMBD to the opposite terminus of our construct caused dramatic changes to the spatial localization of probes within spindles. These results support the use of Tau-based FP fusions as minimally perturbing tools to accurately visualize MTs in living systems.
Collapse
Affiliation(s)
- Paul Mooney
- Department of Molecular Biology, University of Wyoming, Laramie, WY,
82071, USA
- Molecular & Cellular Life Sciences Program, University of
Wyoming, Laramie, WY, 82071, USA
- Cell Organization and Division Group, Marine Biological
Laboratories, Woods Hole, MA, 02543, USA
| | - Taylor Sulerud
- Department of Molecular Biology, University of Wyoming, Laramie, WY,
82071, USA
- Molecular & Cellular Life Sciences Program, University of
Wyoming, Laramie, WY, 82071, USA
- Cell Organization and Division Group, Marine Biological
Laboratories, Woods Hole, MA, 02543, USA
| | - James Pelletier
- Cell Organization and Division Group, Marine Biological
Laboratories, Woods Hole, MA, 02543, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA,
02115, USA
| | - Matthew Dilsaver
- Department of Molecular Biology, University of Wyoming, Laramie, WY,
82071, USA
| | - Miroslav Tomschik
- Department of Molecular Biology, University of Wyoming, Laramie, WY,
82071, USA
| | | | - Jesse C. Gatlin
- Department of Molecular Biology, University of Wyoming, Laramie, WY,
82071, USA
- Molecular & Cellular Life Sciences Program, University of
Wyoming, Laramie, WY, 82071, USA
- Cell Organization and Division Group, Marine Biological
Laboratories, Woods Hole, MA, 02543, USA
| |
Collapse
|
46
|
Bermudez JG, Chen H, Einstein LC, Good MC. Probing the biology of cell boundary conditions through confinement of Xenopus cell-free cytoplasmic extracts. Genesis 2017; 55. [PMID: 28132422 DOI: 10.1002/dvg.23013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 11/11/2022]
Abstract
Cell-free cytoplasmic extracts prepared from Xenopus eggs and embryos have for decades provided a biochemical system with which to interrogate complex cell biological processes in vitro. Recently, the application of microfabrication and microfluidic strategies in biology has narrowed the gap between in vitro and in vivo studies by enabling formation of cell-size compartments containing functional cytoplasm. These approaches provide numerous advantages over traditional biochemical experiments performed in a test tube. Most notably, the cell-free cytoplasm is confined using a two- or three-dimensional boundary, which mimics the natural configuration of a cell. This strategy enables characterization of the spatial organization of a cell, and the role that boundaries play in regulating intracellular assembly and function. In this review, we describe the marriage of Xenopus cell-free cytoplasm and confinement technologies to generate synthetic cell-like systems, the recent biological insights they have enabled, and the promise they hold for future scientific discovery.
Collapse
Affiliation(s)
- Jessica G Bermudez
- Department of Bioengineering, University of Pennsylvania, 421 Curie Blvd, 1151 BRB II/III, Philadelphia, Pennsylvania, 19104
| | - Hui Chen
- Department of Cell and Developmental Biology, University of Pennsylvania, 421 Curie Blvd, 1151 BRB II/III, Philadelphia, Pennsylvania, 19104
| | - Lily C Einstein
- Department of Cell and Developmental Biology, University of Pennsylvania, 421 Curie Blvd, 1151 BRB II/III, Philadelphia, Pennsylvania, 19104
| | - Matthew C Good
- Department of Bioengineering, University of Pennsylvania, 421 Curie Blvd, 1151 BRB II/III, Philadelphia, Pennsylvania, 19104.,Department of Cell and Developmental Biology, University of Pennsylvania, 421 Curie Blvd, 1151 BRB II/III, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
47
|
Gigant E, Stefanutti M, Laband K, Gluszek-Kustusz A, Edwards F, Lacroix B, Maton G, Canman JC, Welburn JPI, Dumont J. Inhibition of ectopic microtubule assembly by the kinesin-13 KLP-7 prevents chromosome segregation and cytokinesis defects in oocytes. Development 2017; 144:1674-1686. [PMID: 28289130 DOI: 10.1242/dev.147504] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/07/2017] [Indexed: 01/02/2023]
Abstract
In most species, oocytes lack centrosomes. Accurate meiotic spindle assembly and chromosome segregation - essential to prevent miscarriage or developmental defects - thus occur through atypical mechanisms that are not well characterized. Using quantitative in vitro and in vivo functional assays in the C. elegans oocyte, we provide novel evidence that the kinesin-13 KLP-7 promotes destabilization of the whole cellular microtubule network. By counteracting ectopic microtubule assembly and disorganization of the microtubule network, this function is strictly required for spindle organization, chromosome segregation and cytokinesis in meiotic cells. Strikingly, when centrosome activity was experimentally reduced, the absence of KLP-7 or the mammalian kinesin-13 protein MCAK (KIF2C) also resulted in ectopic microtubule asters during mitosis in C. elegans zygotes or HeLa cells, respectively. Our results highlight the general function of kinesin-13 microtubule depolymerases in preventing ectopic, spontaneous microtubule assembly when centrosome activity is defective or absent, which would otherwise lead to spindle microtubule disorganization and aneuploidy.
Collapse
Affiliation(s)
- Emmanuelle Gigant
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Marine Stefanutti
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Kimberley Laband
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Agata Gluszek-Kustusz
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Frances Edwards
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Benjamin Lacroix
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Gilliane Maton
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Julie C Canman
- Columbia University, Department of Pathology and Cell Biology, New York, NY 10032, USA
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Julien Dumont
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| |
Collapse
|
48
|
Mechanisms of Chromosome Congression during Mitosis. BIOLOGY 2017; 6:biology6010013. [PMID: 28218637 PMCID: PMC5372006 DOI: 10.3390/biology6010013] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/07/2017] [Accepted: 01/28/2017] [Indexed: 12/13/2022]
Abstract
Chromosome congression during prometaphase culminates with the establishment of a metaphase plate, a hallmark of mitosis in metazoans. Classical views resulting from more than 100 years of research on this topic have attempted to explain chromosome congression based on the balance between opposing pulling and/or pushing forces that reach an equilibrium near the spindle equator. However, in mammalian cells, chromosome bi-orientation and force balance at kinetochores are not required for chromosome congression, whereas the mechanisms of chromosome congression are not necessarily involved in the maintenance of chromosome alignment after congression. Thus, chromosome congression and maintenance of alignment are determined by different principles. Moreover, it is now clear that not all chromosomes use the same mechanism for congressing to the spindle equator. Those chromosomes that are favorably positioned between both poles when the nuclear envelope breaks down use the so-called "direct congression" pathway in which chromosomes align after bi-orientation and the establishment of end-on kinetochore-microtubule attachments. This favors the balanced action of kinetochore pulling forces and polar ejection forces along chromosome arms that drive chromosome oscillatory movements during and after congression. The other pathway, which we call "peripheral congression", is independent of end-on kinetochore microtubule-attachments and relies on the dominant and coordinated action of the kinetochore motors Dynein and Centromere Protein E (CENP-E) that mediate the lateral transport of peripheral chromosomes along microtubules, first towards the poles and subsequently towards the equator. How the opposite polarities of kinetochore motors are regulated in space and time to drive congression of peripheral chromosomes only now starts to be understood. This appears to be regulated by position-dependent phosphorylation of both Dynein and CENP-E and by spindle microtubule diversity by means of tubulin post-translational modifications. This so-called "tubulin code" might work as a navigation system that selectively guides kinetochore motors with opposite polarities along specific spindle microtubule populations, ultimately leading to the congression of peripheral chromosomes. We propose an integrated model of chromosome congression in mammalian cells that depends essentially on the following parameters: (1) chromosome position relative to the spindle poles after nuclear envelope breakdown; (2) establishment of stable end-on kinetochore-microtubule attachments and bi-orientation; (3) coordination between kinetochore- and arm-associated motors; and (4) spatial signatures associated with post-translational modifications of specific spindle microtubule populations. The physiological consequences of abnormal chromosome congression, as well as the therapeutic potential of inhibiting chromosome congression are also discussed.
Collapse
|
49
|
Ma DD, Wang DH, Yang WX. Kinesins in spermatogenesis†. Biol Reprod 2017; 96:267-276. [DOI: 10.1095/biolreprod.116.144113] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/20/2016] [Accepted: 12/26/2016] [Indexed: 11/01/2022] Open
|
50
|
Ganguly A, DeMott L, Dixit R. Function of the Arabidopsis kinesin-4, FRA1, requires abundant processive motility. J Cell Sci 2017; 130:1232-1238. [DOI: 10.1242/jcs.196857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/14/2017] [Indexed: 01/26/2023] Open
Abstract
Processivity is important for kinesins that mediate intracellular transport. Structure-function analyses of N-terminal kinesins have identified several non-motor regions that affect processivity in vitro. However, whether these structural elements affect kinesin processivity and function in vivo is not known. Here, we used an Arabidopsis kinesin-4, called Fragile Fiber1 (FRA1), which is thought to mediate vesicle transport to test whether mutations that alter processivity in vitro behave similarly in vivo and whether processivity is important for FRA1’s function. We generated several FRA1 mutants that differed in their run lengths in vitro and then transformed them into the fra1-5 mutant for complementation and in vivo motility analyses. Our data show that the behavior of processivity mutants in vivo can differ dramatically from in vitro properties, underscoring the need to extend structure-function analyses of kinesins in vivo. In addition, we found that high density of processive motility is necessary for FRA1’s physiological function.
Collapse
Affiliation(s)
- Anindya Ganguly
- Biology Department, Washington University in St. Louis, MO 63130, USA
| | - Logan DeMott
- Biology Department, Washington University in St. Louis, MO 63130, USA
| | - Ram Dixit
- Biology Department, Washington University in St. Louis, MO 63130, USA
| |
Collapse
|