1
|
Risteski P, Martinčić J, Jagrić M, Tintor E, Petelinec A, Tolić IM. Microtubule poleward flux as a target for modifying chromosome segregation errors. Proc Natl Acad Sci U S A 2024; 121:e2405015121. [PMID: 39541344 DOI: 10.1073/pnas.2405015121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer cells often display errors in chromosome segregation, some of which result from improper chromosome alignment at the spindle midplane. Chromosome alignment is facilitated by different rates of microtubule poleward flux between sister kinetochore fibers. However, the role of the poleward flux in supporting mitotic fidelity remains unknown. Here, we introduce the hypothesis that the finely tuned poleward flux safeguards against lagging chromosomes and micronuclei at mitotic exit by promoting chromosome alignment in metaphase. We used human untransformed RPE-1 cells depleted of KIF18A/kinesin-8 as a system with reduced mitotic fidelity, which we rescued by three mechanistically independent treatments, comprising low-dose taxol or codepletion of the spindle proteins HAUS8 or NuMA. The rescue of mitotic errors was due to shortening of the excessively long overlaps of antiparallel microtubules, serving as a platform for motor proteins that drive the flux, which in turn slowed down the overly fast flux and improved chromosome alignment. In contrast to the prevailing view, the rescue was not accompanied by reduction of overall microtubule growth rates. Instead, speckle microscopy revealed that the improved chromosome alignment in the rescue treatments was associated with slower growth and flux of kinetochore microtubules. In a similar manner, a low-dose taxol treatment rescued mitotic errors in a high-grade serous ovarian carcinoma cell line OVKATE. Collectively, our results highlight the potential of targeting microtubule poleward flux to modify chromosome instability and provide insight into the mechanism through which low doses of taxol rescue certain mitotic errors in cancer cells.
Collapse
Affiliation(s)
- Patrik Risteski
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Jelena Martinčić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Mihaela Jagrić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Erna Tintor
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ana Petelinec
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Iva M Tolić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10000, Croatia
| |
Collapse
|
2
|
Tang X, He Y, Tang Y, Chen K, Lin H, Liu B, Deng X. A kinetochore-associated kinesin-7 motor cooperates with BUB3.3 to regulate mitotic chromosome congression in Arabidopsis thaliana. NATURE PLANTS 2024:10.1038/s41477-024-01824-7. [PMID: 39414927 DOI: 10.1038/s41477-024-01824-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/20/2024] [Indexed: 10/18/2024]
Abstract
Faithful genome partition during cell division relies on proper congression of chromosomes to the spindle equator before sister chromatid segregation. Here we uncover a kinesin-7 motor, kinetochore-associated kinesin 1 (KAK1), that is required for mitotic chromosome congression in Arabidopsis. KAK1 associates dynamically with kinetochores throughout mitosis. Loss of KAK1 results in severe defects in chromosome congression at metaphase, yet segregation errors at anaphase are rarely observed. KAK1 specifically interacts with the spindle assembly checkpoint protein BUB3.3 and both proteins show interdependent kinetochore localization. Chromosome misalignment in BUB3.3-depleted plants can be rescued by artificial tethering of KAK1 to kinetochores but not vice versa, demonstrating that KAK1 acts downstream of BUB3.3 to orchestrate microtubule-based chromosome transport at kinetochores. Moreover, we show that KAK1's motor activity is essential for driving chromosome congression to the metaphase plate. Thus, our findings reveal that plants have repurposed BUB3.3 to interface with a specialized kinesin adapted to integrate proper chromosome congression and checkpoint control through a distinct kinetochore design.
Collapse
Affiliation(s)
- Xiaoya Tang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ying He
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yihang Tang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Keqi Chen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Honghui Lin
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Bo Liu
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA, USA
| | - Xingguang Deng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Chong MK, Rosas-Salvans M, Tran V, Dumont S. Chromosome size-dependent polar ejection force impairs mammalian mitotic error correction. J Cell Biol 2024; 223:e202310010. [PMID: 38727808 PMCID: PMC11090132 DOI: 10.1083/jcb.202310010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/28/2024] [Accepted: 04/29/2024] [Indexed: 05/15/2024] Open
Abstract
Accurate chromosome segregation requires sister kinetochores to biorient, attaching to opposite spindle poles. To this end, the mammalian kinetochore destabilizes incorrect attachments and stabilizes correct ones, but how it discriminates between these is not yet clear. Here, we test the model that kinetochore tension is the stabilizing cue and ask how chromosome size impacts that model. We live image PtK2 cells, with just 14 chromosomes, widely ranging in size, and find that long chromosomes align at the metaphase plate later than short chromosomes. Enriching for errors and imaging error correction live, we show that long chromosomes exhibit a specific delay in correcting attachments. Using chromokinesin overexpression and laser ablation to perturb polar ejection forces, we find that chromosome size and force on arms determine alignment order. Thus, we propose a model where increased force on long chromosomes can falsely stabilize incorrect attachments, delaying their biorientation. As such, long chromosomes may require compensatory mechanisms for correcting errors to avoid chromosomal instability.
Collapse
Affiliation(s)
- Megan K. Chong
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Miquel Rosas-Salvans
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vanna Tran
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sophie Dumont
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
4
|
Ghone DA, Evans EL, Bandini M, Stephenson KG, Sherer NM, Suzuki A. HIV-1 Vif disrupts phosphatase feedback regulation at the kinetochore, leading to a pronounced pseudo-metaphase arrest. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605839. [PMID: 39131328 PMCID: PMC11312601 DOI: 10.1101/2024.07.30.605839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The human immunodeficiency virus type 1 (HIV-1) Virion Infectivity Factor (Vif) targets and degrades cellular APOBEC3 proteins, key regulators of intrinsic and innate antiretroviral immune responses, thereby facilitating HIV-1 infection. While Vif's role in degrading APOBEC3G is well-studied, Vif is also known to cause cell cycle arrest but the detailed nature of Vif's effects on the cell cycle has yet to be delineated. In this study, we employed high-temporal single-cell live imaging and super-resolution microscopy to monitor individual cells during Vif-induced cell cycle arrest. Our findings reveal that Vif does not affect the G2/M boundary as previously thought. Instead, Vif triggers a unique and robust pseudo-metaphase arrest, which is markedly distinct from the mild prometaphase arrest induced by the HIV-1 accessory protein, Vpr, known for modulating the cell cycle. During Vif-mediated arrest, chromosomes align properly to form a metaphase plate but later disassemble, resulting in polar chromosomes. Notably, unlike Vpr, Vif significantly reduces the levels of both Phosphatase 1 (PP1) and 2 (PP2) at kinetochores, which are key regulators of chromosome-microtubule interactions. These results reveal a novel function of Vif in kinetochore regulation that governs the spatial organization of chromosomes during mitosis.
Collapse
Affiliation(s)
- Dhaval A. Ghone
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- These authors contributed equally
| | - Edward L. Evans
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- These authors contributed equally
- Present address: Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Madison Bandini
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kaelyn G. Stephenson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nathan M. Sherer
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Chong MK, Rosas-Salvans M, Tran V, Dumont S. Chromosome size-dependent polar ejection force impairs mammalian mitotic error correction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562637. [PMID: 37905080 PMCID: PMC10614862 DOI: 10.1101/2023.10.16.562637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Accurate chromosome segregation requires sister kinetochores to biorient, attaching to opposite spindle poles. To this end, the mammalian kinetochore destabilizes incorrect attachments and stabilizes correct ones, but how it discriminates between these is not yet clear. Here, we test the model that kinetochore tension is the stabilizing cue and ask how chromosome size impacts that model. We live image PtK2 cells, with just 14 chromosomes, widely ranging in size, and find that long chromosomes align at the metaphase plate later than short chromosomes. Enriching for errors and imaging error correction live, we show that long chromosomes exhibit a specific delay in correcting attachments. Using chromokinesin overexpression and laser ablation to perturb polar ejection forces, we find that chromosome size and force on arms determine alignment order. Thus, we propose a model where increased force on long chromosomes can falsely stabilize incorrect attachments, delaying their biorientation. As such, long chromosomes may require compensatory mechanisms for correcting errors to avoid chromosomal instability.
Collapse
Affiliation(s)
- Megan K. Chong
- Tetrad Graduate Program, UCSF, San Francisco, CA 94158, USA
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA 94158, USA
| | - Miquel Rosas-Salvans
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA 94158, USA
| | - Vanna Tran
- Tetrad Graduate Program, UCSF, San Francisco, CA 94158, USA
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA 94158, USA
| | - Sophie Dumont
- Tetrad Graduate Program, UCSF, San Francisco, CA 94158, USA
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA 94158, USA
- Department of Biochemistry & Biophysics, UCSF San Francisco 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
6
|
Valdez VA, Neahring L, Petry S, Dumont S. Mechanisms underlying spindle assembly and robustness. Nat Rev Mol Cell Biol 2023; 24:523-542. [PMID: 36977834 PMCID: PMC10642710 DOI: 10.1038/s41580-023-00584-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2023] [Indexed: 03/30/2023]
Abstract
The microtubule-based spindle orchestrates chromosome segregation during cell division. Following more than a century of study, many components and pathways contributing to spindle assembly have been described, but how the spindle robustly assembles remains incompletely understood. This process involves the self-organization of a large number of molecular parts - up to hundreds of thousands in vertebrate cells - whose local interactions give rise to a cellular-scale structure with emergent architecture, mechanics and function. In this Review, we discuss key concepts in our understanding of spindle assembly, focusing on recent advances and the new approaches that enabled them. We describe the pathways that generate the microtubule framework of the spindle by driving microtubule nucleation in a spatially controlled fashion and present recent insights regarding the organization of individual microtubules into structural modules. Finally, we discuss the emergent properties of the spindle that enable robust chromosome segregation.
Collapse
Affiliation(s)
| | - Lila Neahring
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA, USA
- Developmental & Stem Cell Biology Graduate Program, UCSF, San Francisco, CA, USA
| | - Sabine Petry
- Molecular Biology, Princeton University, Princeton, NJ, USA.
| | - Sophie Dumont
- Department of Bioengineering & Therapeutic Sciences, UCSF, San Francisco, CA, USA.
- Developmental & Stem Cell Biology Graduate Program, UCSF, San Francisco, CA, USA.
- Department of Biochemistry & Biophysics, UCSF, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
7
|
Pitayu-Nugroho L, Aubry M, Laband K, Geoffroy H, Ganeswaran T, Primadhanty A, Canman JC, Dumont J. Kinetochore component function in C. elegans oocytes revealed by 4D tracking of holocentric chromosomes. Nat Commun 2023; 14:4032. [PMID: 37419936 PMCID: PMC10329006 DOI: 10.1038/s41467-023-39702-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023] Open
Abstract
During cell division, chromosome congression to the spindle center, their orientation along the spindle long axis and alignment at the metaphase plate depend on interactions between spindle microtubules and kinetochores, and are pre-requisite for chromosome bi-orientation and accurate segregation. How these successive phases are controlled during oocyte meiosis remains elusive. Here we provide 4D live imaging during the first meiotic division in C. elegans oocytes with wild-type or disrupted kinetochore protein function. We show that, unlike in monocentric organisms, holocentric chromosome bi-orientation is not strictly required for accurate chromosome segregation. Instead, we propose a model in which initial kinetochore-localized BHC module (comprised of BUB-1Bub1, HCP-1/2CENP-F and CLS-2CLASP)-dependent pushing acts redundantly with Ndc80 complex-mediated pulling for accurate chromosome segregation in meiosis. In absence of both mechanisms, homologous chromosomes tend to co-segregate in anaphase, especially when initially mis-oriented. Our results highlight how different kinetochore components cooperate to promote accurate holocentric chromosome segregation in oocytes of C. elegans.
Collapse
Affiliation(s)
| | - Mélanie Aubry
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Kimberley Laband
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Hélène Geoffroy
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | | | | | - Julie C Canman
- Columbia University Irving Medical Center; Department of Pathology and Cell Biology, New York, NY, 10032, USA
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France.
| |
Collapse
|
8
|
Chen Y, Si H, Bao B, Li S, Teng D, Yan Y, Hu S, Xu Y, Du X. Integrated analysis of intestinal microbiota and host gene expression in colorectal cancer patients. J Med Microbiol 2022; 71. [PMID: 36136380 DOI: 10.1099/jmm.0.001596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction. Colorectal cancer (CRC) is one of the most common cancers and poses heavy burden on global health. The relationship between mucosal microbiome composition and colorectal gene expression are rarely studied. In this study, we integrated transcriptome data with microbiome data to investigate the relationship between them in colorectal cancer patients.Gap statement. Previous studies have identified the contribution of gut microbiota and DEGs to the pathogenesis of CRC, but the relationship between mucosal microbiome composition and colorectal gene expression are rarely studied.Aim. In this study, we integrated transcriptome data with microbiome data to investigate the relationship between mucosal microbiome composition and colorectal gene expression.Methodology. First, three independent CRC gene expression profiles (GSE184093, GSE156355 and GSE146587) from Gene Expression Omnibus (GEO) were used to identify differentially expressed genes (DEGs). Second, another dataset (GSE163366) was used to analyse gut mucosal microbiome differential abundance. GO (Gene Ontology) function and KEGG (Kyoto Encyclopaedia of Genes and Genomes) pathway enrichment analyses of the DEGs were performed. Protein-protein interactions (PPIs) of the DEGs were constructed. The Spearman correlation analysis was computed between host DEGs and gut microbiome abundance data.Results. A total of 1036 upregulated DEGs and 1194 downregulated DEGs between noncancerous tissues and cancerous tissues were identified based on the analysis. One significant module with a score 37.65 was selected out via MCODE including 41 upregulated DEGs, which are were mostly enriched in two pathways, including microtubule binding and tubulin binding. In particular, significant negative correlations are prevalent between Fusobacterium and the 41 DEGs with the correlation ranging between -0.54 and -0.35, and there commonly exist significant positive correlations between Blautia and the 41 DEGs with the correlation ranging between 0.42 and 0.54, indicating that Fusobacterium and Blautia are two of the most important microbes interacting with the gene regulation.Conclusion. Our results demonstrate significant correlation between some gut microbes and DEGs, providing a comprehensive bioinformatics analysis of them for future investigation into the molecular mechanisms and biomarkers.
Collapse
Affiliation(s)
- Yuhui Chen
- Chinese PLA medical school, Beijing, Haidian 100853, PR China.,Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Huiyan Si
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Baoshi Bao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Songyan Li
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Da Teng
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Yang Yan
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Shidong Hu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Yingxin Xu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| | - Xiaohui Du
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, PR China
| |
Collapse
|
9
|
A mitotic chromatin phase transition prevents perforation by microtubules. Nature 2022; 609:183-190. [PMID: 35922507 PMCID: PMC9433320 DOI: 10.1038/s41586-022-05027-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 06/27/2022] [Indexed: 12/20/2022]
Abstract
Dividing eukaryotic cells package extremely long chromosomal DNA molecules into discrete bodies to enable microtubule-mediated transport of one genome copy to each of the newly forming daughter cells1–3. Assembly of mitotic chromosomes involves DNA looping by condensin4–8 and chromatin compaction by global histone deacetylation9–13. Although condensin confers mechanical resistance to spindle pulling forces14–16, it is not known how histone deacetylation affects material properties and, as a consequence, segregation mechanics of mitotic chromosomes. Here we show how global histone deacetylation at the onset of mitosis induces a chromatin-intrinsic phase transition that endows chromosomes with the physical characteristics necessary for their precise movement during cell division. Deacetylation-mediated compaction of chromatin forms a structure dense in negative charge and allows mitotic chromosomes to resist perforation by microtubules as they are pushed to the metaphase plate. By contrast, hyperacetylated mitotic chromosomes lack a defined surface boundary, are frequently perforated by microtubules and are prone to missegregation. Our study highlights the different contributions of DNA loop formation and chromatin phase separation to genome segregation in dividing cells. Histone deacetylation at the onset of mitosis induces a chromatin-intrinsic phase transition that endows chromosomes with the physical characteristics necessary for their precise movement during cell division.
Collapse
|
10
|
Thompson AF, Blackburn PR, Arons NS, Stevens SN, Babovic-Vuksanovic D, Lian JB, Klee EW, Stumpff J. Pathogenic mutations in the chromokinesin KIF22 disrupt anaphase chromosome segregation. eLife 2022; 11:e78653. [PMID: 35730929 PMCID: PMC9302971 DOI: 10.7554/elife.78653] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/21/2022] [Indexed: 11/22/2022] Open
Abstract
The chromokinesin KIF22 generates forces that contribute to mitotic chromosome congression and alignment. Mutations in the α2 helix of the motor domain of KIF22 have been identified in patients with abnormal skeletal development, and we report the identification of a patient with a novel mutation in the KIF22 tail. We demonstrate that pathogenic mutations do not result in a loss of KIF22's functions in early mitosis. Instead, mutations disrupt chromosome segregation in anaphase, resulting in reduced proliferation, abnormal daughter cell nuclear morphology, and, in a subset of cells, cytokinesis failure. This phenotype could be explained by a failure of KIF22 to inactivate in anaphase. Consistent with this model, constitutive activation of the motor via a known site of phosphoregulation in the tail phenocopied the effects of pathogenic mutations. These results suggest that the motor domain α2 helix may be an important site for regulation of KIF22 activity at the metaphase to anaphase transition. In support of this conclusion, mimicking phosphorylation of α2 helix residue T158 also prevents inactivation of KIF22 in anaphase. These findings demonstrate the importance of both the head and tail of the motor in regulating the activity of KIF22 and offer insight into the cellular consequences of preventing KIF22 inactivation and disrupting force balance in anaphase.
Collapse
Affiliation(s)
- Alex F Thompson
- Department of Molecular Physiology and Biophysics, University of VermontBurlingtonUnited States
| | - Patrick R Blackburn
- Laboratory Medicine and Pathology, Mayo ClinicRochesterUnited States
- Pathology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Noah S Arons
- Department of Molecular Physiology and Biophysics, University of VermontBurlingtonUnited States
| | - Sarah N Stevens
- Department of Molecular Physiology and Biophysics, University of VermontBurlingtonUnited States
| | - Dusica Babovic-Vuksanovic
- Laboratory Medicine and Pathology, Mayo ClinicRochesterUnited States
- Clinical Genomics, Mayo ClinicRochesterUnited States
| | - Jane B Lian
- Department of Biochemistry, University of VermontBurlingtonUnited States
| | - Eric W Klee
- Biomedical Informatics, Mayo ClinicRochesterUnited States
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of VermontBurlingtonUnited States
| |
Collapse
|
11
|
Kif4A mediates resistance to neoadjuvant chemoradiotherapy in patients with advanced colorectal cancer via regulating DNA damage response. Acta Biochim Biophys Sin (Shanghai) 2022; 54:940-951. [PMID: 35882623 PMCID: PMC9828526 DOI: 10.3724/abbs.2022068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
More and more patients with advanced colorectal cancer (CRC) have benefited from surgical resection or ablation following neoadjuvant chemoradiotherapy (nCRT), but nCRT may be ineffective and have potential risks to some patients. Therefore, it is necessary to discover effective biomarkers for predicting the nCRT efficacy in CRC patients. Chromokinesin Kif4A plays a critical role in mitosis, DNA damage repair and tumorigenesis, but its relationship with nCRT efficacy in advanced CRC remains unclear. Here, we find that Kif4A expression in pretreated tumor tissue is positively correlated with poorer tumor regression after receiving nCRT ( P=0.005). Knockdown of endogenous Kif4A causes an increased sensitivity of CRC cells to chemotherapeutic drugs 5-fluorouracil (5-FU) and Cisplatin (DDP), while overexpression of Kif4A enhances resistance of CRC cells to the chemotherapeutic drugs. Furthermore, depending on its motor domain and tail domain, Kif4A regulates DNA damage response (DDR) induced by 5-FU or DDP treatment in CRC cells. In conclusion, we demonstrate that Kif4A may be a potential independent biomarker for predicting the nCRT efficacy in advanced CRC patients, and Kif4A regulates chemosensitivity of CRC cells through controlling DDR.
Collapse
|
12
|
Vukušić K, Tolić IM. Polar Chromosomes-Challenges of a Risky Path. Cells 2022; 11:1531. [PMID: 35563837 PMCID: PMC9101661 DOI: 10.3390/cells11091531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/29/2022] Open
Abstract
The process of chromosome congression and alignment is at the core of mitotic fidelity. In this review, we discuss distinct spatial routes that the chromosomes take to align during prometaphase, which are characterized by distinct biomolecular requirements. Peripheral polar chromosomes are an intriguing case as their alignment depends on the activity of kinetochore motors, polar ejection forces, and a transition from lateral to end-on attachments to microtubules, all of which can result in the delayed alignment of these chromosomes. Due to their undesirable position close to and often behind the spindle pole, these chromosomes may be particularly prone to the formation of erroneous kinetochore-microtubule interactions, such as merotelic attachments. To prevent such errors, the cell employs intricate mechanisms to preposition the spindle poles with respect to chromosomes, ensure the formation of end-on attachments in restricted spindle regions, repair faulty attachments by error correction mechanisms, and delay segregation by the spindle assembly checkpoint. Despite this protective machinery, there are several ways in which polar chromosomes can fail in alignment, mis-segregate, and lead to aneuploidy. In agreement with this, polar chromosomes are present in certain tumors and may even be involved in the process of tumorigenesis.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | | |
Collapse
|
13
|
Thompson AF, Vandal S, Stumpff J. Quantifying Changes in Chromosome Position to Assess Chromokinesin Activity. Methods Mol Biol 2022; 2415:139-149. [PMID: 34972951 DOI: 10.1007/978-1-0716-1904-9_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The chromokinesin KIF22 (Kid, kinesin-10 family) is the primary generator of polar ejection forces, which contribute to chromosome positioning and alignment in mitotic cells. Assessment of KIF22 function requires quantitative comparison of relative polar ejection forces between experimental conditions. This is facilitated by the generation of monopolar spindles to reduce the impact of bioriented microtubule attachment at kinetochores on chromosome positions and increase the dependence of chromosome positions on chromokinesin activity. Radial profile plots measure the intensity of chromatin signal in concentric circles around the poles of monopolar cells and represent an expedient quantitative measure of relative polar ejection forces. As such, this assay can be used to measure changes in polar ejection forces resulting from chromokinesin depletion or perturbation.
Collapse
Affiliation(s)
- Alex F Thompson
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Sarah Vandal
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
14
|
KIF4A Regulates the Progression of Pancreatic Ductal Adenocarcinoma through Proliferation and Invasion. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8249293. [PMID: 34805404 PMCID: PMC8601854 DOI: 10.1155/2021/8249293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022]
Abstract
Background Pancreatic cancer is a malignant tumor of the digestive tract, which is difficult to diagnose and treat due to bad early diagnosis. We aimed to explore the role of kinesin superfamily 4A (KIF4A) in pancreatic ductal adenocarcinoma (PDAC). Methods We first used the bioinformatic website to screen the data of pancreatic cancer in TCGA, and KIF4A protein was detected among the 86 specimens of patients in our hospital combined with clinic-pathological characteristics and survival analysis. KIF4A loss-expression cell lines were established by RNA interference (RNAi). In addition, we performed in vitro cell assays to detect the changes in cell proliferation, migration, and invasion. The proteins involved in the proliferation and metastasis of cancer cells were also detected by western blot. The above results could be proved in vivo. Further, the correlation between KIF4A and CDC5L was analyzed by TCGA and IHC data. Results We first found a high expression of KIF4A in pancreatic cancer, suggesting a role of KIF4A in the development of pancreatic cancer. KIF4A was found to be differentially expressed (P < 0.05) among the 86 specimens of patients in our hospital and was significantly associated with PDAC TNM stages and tumor size. High KIF4A expression also significantly worsened overall survival (OS) and disease-free survival rate (DFS) (P < 0.05, respectively). In addition, cell proliferation, migration, and invasion were inhibited by the KIF4A-shRNA group compared with the control (P < 0.05, respectively). In the end, knockdown of KIF4A could inhibit tumor development and metastasis in vivo. Further, the positive correlation between KIF4A and CDC5L existed, and KIF4A might promote pancreatic cancer proliferation by affecting CDC5L expression. Conclusion In conclusion, the high expression level of KIF4A in PDAC was closely related to poor clinical and pathological status, lymphatic metastasis, and vascular invasion. KIF4A might be involved in promoting the development of PDAC in vitro and in vivo, which might be a new therapeutic target of PDAC.
Collapse
|
15
|
Iemura K, Yoshizaki Y, Kuniyasu K, Tanaka K. Attenuated Chromosome Oscillation as a Cause of Chromosomal Instability in Cancer Cells. Cancers (Basel) 2021; 13:cancers13184531. [PMID: 34572757 PMCID: PMC8470601 DOI: 10.3390/cancers13184531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chromosomal instability (CIN), a condition in which chromosome missegregation occurs at high rates, is widely seen in cancer cells. Causes of CIN in cancer cells are not fully understood. A recent report suggests that chromosome oscillation, an iterative chromosome motion typically seen in metaphase around the spindle equator, is attenuated in cancer cells, and is associated with CIN. Chromosome oscillation promotes the correction of erroneous kinetochore-microtubule attachments through phosphorylation of Hec1, a kinetochore protein that binds to microtubules, by Aurora A kinase residing on the spindle. In this review, we focused on this unappreciated link between chromosome oscillation and CIN. Abstract Chromosomal instability (CIN) is commonly seen in cancer cells, and related to tumor progression and poor prognosis. Among the causes of CIN, insufficient correction of erroneous kinetochore (KT)-microtubule (MT) attachments plays pivotal roles in various situations. In this review, we focused on the previously unappreciated role of chromosome oscillation in the correction of erroneous KT-MT attachments, and its relevance to the etiology of CIN. First, we provided an overview of the error correction mechanisms for KT-MT attachments, especially the role of Aurora kinases in error correction by phosphorylating Hec1, which connects MT to KT. Next, we explained chromosome oscillation and its underlying mechanisms. Then we introduced how chromosome oscillation is involved in the error correction of KT-MT attachments, based on recent findings. Chromosome oscillation has been shown to promote Hec1 phosphorylation by Aurora A which localizes to the spindle. Finally, we discussed the link between attenuated chromosome oscillation and CIN in cancer cells. This link underscores the role of chromosome dynamics in mitotic fidelity, and the mutual relationship between defective chromosome dynamics and CIN in cancer cells that can be a target for cancer therapy.
Collapse
|
16
|
Vukušić K, Tolić IM. Anaphase B: Long-standing models meet new concepts. Semin Cell Dev Biol 2021; 117:127-139. [PMID: 33849764 PMCID: PMC8406420 DOI: 10.1016/j.semcdb.2021.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022]
Abstract
Mitotic cell divisions ensure stable transmission of genetic information from a mother to daughter cells in a series of generations. To ensure this crucial task is accomplished, the cell forms a bipolar structure called the mitotic spindle that divides sister chromatids to the opposite sides of the dividing mother cell. After successful establishment of stable attachments of microtubules to chromosomes and inspection of connections between them, at the heart of mitosis, the cell starts the process of segregation. This spectacular moment in the life of a cell is termed anaphase, and it involves two distinct processes: depolymerization of microtubules bound to chromosomes, which is also known as anaphase A, and elongation of the spindle or anaphase B. Both processes ensure physical separation of disjointed sister chromatids. In this chapter, we review the mechanisms of anaphase B spindle elongation primarily in mammalian systems, combining different pioneering ideas and concepts with more recent findings that shed new light on the force generation and regulation of biochemical modules operating during spindle elongation. Finally, we present a comprehensive model of spindle elongation that includes structural, biophysical, and molecular aspects of anaphase B.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
17
|
Jang JK, Gladstein AC, Das A, Shapiro JG, Sisco ZL, McKim KS. Multiple pools of PP2A regulate spindle assembly, kinetochore attachments and cohesion in Drosophila oocytes. J Cell Sci 2021; 134:jcs254037. [PMID: 34297127 PMCID: PMC8325958 DOI: 10.1242/jcs.254037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 06/14/2021] [Indexed: 01/06/2023] Open
Abstract
Meiosis in female oocytes lacks centrosomes, the microtubule-organizing centers. In Drosophila oocytes, meiotic spindle assembly depends on the chromosomal passenger complex (CPC). To investigate the mechanisms that regulate Aurora B activity, we examined the role of protein phosphatase 2A (PP2A) in Drosophila oocyte meiosis. We found that both forms of PP2A, B55 and B56, antagonize the Aurora B spindle assembly function, suggesting that a balance between Aurora B and PP2A activity maintains the oocyte spindle during meiosis I. PP2A-B56, which has a B subunit encoded by two partially redundant paralogs, wdb and wrd, is also required for maintenance of sister chromatid cohesion, establishment of end-on microtubule attachments, and metaphase I arrest in oocytes. WDB recruitment to the centromeres depends on BUBR1, MEI-S332 and kinetochore protein SPC105R. Although BUBR1 stabilizes microtubule attachments in Drosophila oocytes, it is not required for cohesion maintenance during meiosis I. We propose at least three populations of PP2A-B56 regulate meiosis, two of which depend on SPC105R and a third that is associated with the spindle.
Collapse
Affiliation(s)
| | | | | | | | | | - Kim S. McKim
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
18
|
Ferreira LT, Maiato H. Prometaphase. Semin Cell Dev Biol 2021; 117:52-61. [PMID: 34127384 DOI: 10.1016/j.semcdb.2021.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 11/28/2022]
Abstract
The establishment of a metaphase plate in which all chromosomes are attached to mitotic spindle microtubules and aligned at the cell equator is required for faithful chromosome segregation in metazoans. The achievement of this configuration relies on the precise coordination between several concurrent mechanisms that start upon nuclear envelope breakdown, mediate chromosome capture at their kinetochores during mitotic spindle assembly and culminate with the congression of all chromosomes to the spindle equator. This period is called 'prometaphase'. Because the nature of chromosome capture by mitotic spindle microtubules is error prone, the cell is provided of error correction mechanisms that sense and correct most erroneous kinetochore-microtubule attachments before committing to separate sister chromatids in anaphase. In this review, aimed for newcomers in the field, more than providing an exhaustive mechanistic coverage of each and every concurrent mechanism taking place during prometaphase, we provide an integrative overview of these processes that ultimately promote the subsequent faithful segregation of chromosomes during mitosis.
Collapse
Affiliation(s)
- Luísa T Ferreira
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Helder Maiato
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
19
|
Barisic M, Rajendraprasad G. Mitotic poleward flux: Finding balance between microtubule dynamics and sliding. Bioessays 2021; 43:e2100079. [PMID: 34085708 DOI: 10.1002/bies.202100079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022]
Abstract
Continuous poleward motion of microtubules in metazoan mitotic spindles has been fascinating generations of cell biologists over the last several decades. In human cells, this so-called poleward flux was recently shown to be driven by the coordinated action of four mitotic kinesins. The sliding activities of kinesin-5/EG5 and kinesin-12/KIF15 are sequentially supported by kinesin-7/CENP-E at kinetochores and kinesin-4/KIF4A on chromosome arms, with the individual contributions peaking during prometaphase and metaphase, respectively. Although recent data elucidate the molecular mechanism underlying this cellular phenomenon, the functional roles of microtubule poleward flux during cell division remain largely elusive. Here, we discuss potential contribution of microtubule flux engine to various essential processes at different stages of mitosis.
Collapse
Affiliation(s)
- Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Girish Rajendraprasad
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center (DCRC), Copenhagen, Denmark
| |
Collapse
|
20
|
Abstract
Kinesins constitute a superfamily of ATP-driven microtubule motor enzymes that convert the chemical energy of ATP hydrolysis into mechanical work along microtubule tracks. Kinesins are found in all eukaryotic organisms and are essential to all eukaryotic cells, involved in diverse cellular functions such as microtubule dynamics and morphogenesis, chromosome segregation, spindle formation and elongation and transport of organelles. In this review, we explore recently reported functions of kinesins in eukaryotes and compare their specific cargoes in both plant and animal kingdoms to understand the possible roles of uncharacterized motors in a kingdom based on their reported functions in other kingdoms.
Collapse
Affiliation(s)
- Iftikhar Ali
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing, China
| | - Wei-Cai Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing, China.,The College of Advanced Agricultural Science, The University of Chinese Academy of Sciences , Beijing, China
| |
Collapse
|
21
|
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: 14] [Impact Index Per Article: 4.7] [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
|
22
|
Risteski P, Jagrić M, Pavin N, Tolić IM. Biomechanics of chromosome alignment at the spindle midplane. Curr Biol 2021; 31:R574-R585. [PMID: 34033791 DOI: 10.1016/j.cub.2021.03.082] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
During metaphase, chromosomes are aligned in a lineup at the equatorial plane of the spindle to ensure synchronous poleward movement of chromatids in anaphase and proper nuclear reformation at the end of mitosis. Chromosome alignment relies on microtubules, several types of motor protein and numerous other microtubule-associated and regulatory proteins. Because of the multitude of players involved, the mechanisms of chromosome alignment are still under debate. Here, we discuss the current models of alignment based on poleward pulling forces exerted onto sister kinetochores by kinetochore microtubules, which show length-dependent dynamics and undergo poleward flux, and polar ejection forces that push the chromosome arms away from the pole. We link these models with the recent ideas based on mechanical coupling between bridging and kinetochore microtubules, where sliding of bridging microtubules promotes overlap length-dependent sliding of kinetochore fibers and thus the alignment of sister kinetochores at the spindle equator. Finally, we discuss theoretical models of forces acting on chromosomes during metaphase.
Collapse
Affiliation(s)
- Patrik Risteski
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Mihaela Jagrić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Nenad Pavin
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička cesta 32, 10000 Zagreb, Croatia
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
23
|
Renda F, Khodjakov A. Role of spatial patterns and kinetochore architecture in spindle morphogenesis. Semin Cell Dev Biol 2021; 117:75-85. [PMID: 33836948 PMCID: PMC8762378 DOI: 10.1016/j.semcdb.2021.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/30/2022]
Abstract
Mitotic spindle is a self-assembling macromolecular machine responsible for the faithful segregation of chromosomes during cell division. Assembly of the spindle is believed to be governed by the 'Search & Capture' (S&C) principle in which dynamic microtubules explore space in search of kinetochores while the latter capture microtubules and thus connect chromosomes to the spindle. Due to the stochastic nature of the encounters between kinetochores and microtubules, the time required for incorporating all chromosomes into the spindle is profoundly affected by geometric constraints, such as the size and shape of kinetochores as well as their distribution in space at the onset of spindle assembly. In recent years, several molecular mechanisms that control these parameters have been discovered. It is now clear that stochastic S&C takes place in structured space, where components are optimally distributed and oriented to minimize steric hindrances. Nucleation of numerous non-centrosomal microtubules near kinetochores accelerates capture, while changes in the kinetochore architecture at various stages of spindle assembly promote proper connection of sister kinetochores to the opposite spindle poles. Here we discuss how the concerted action of multiple facilitating mechanisms ensure that the spindle assembles rapidly yet with a minimal number of errors.
Collapse
Affiliation(s)
- Fioranna Renda
- Biggs Laboratory, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12237, United States.
| | - Alexey Khodjakov
- Biggs Laboratory, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12237, United States; Rensselaer Polytechnic Institute, Troy, NY 12180, United States.
| |
Collapse
|
24
|
Sharp JA, Perea-Resa C, Wang W, Blower MD. Cell division requires RNA eviction from condensing chromosomes. J Cell Biol 2021; 219:211450. [PMID: 33053167 PMCID: PMC7549315 DOI: 10.1083/jcb.201910148] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022] Open
Abstract
During mitosis, the genome is transformed from a decondensed, transcriptionally active state to a highly condensed, transcriptionally inactive state. Mitotic chromosome reorganization is marked by the general attenuation of transcription on chromosome arms, yet how the cell regulates nuclear and chromatin-associated RNAs after chromosome condensation and nuclear envelope breakdown is unknown. SAF-A/hnRNPU is an abundant nuclear protein with RNA-to-DNA tethering activity, coordinated by two spatially distinct nucleic acid–binding domains. Here we show that RNA is evicted from prophase chromosomes through Aurora-B–dependent phosphorylation of the SAF-A DNA-binding domain; failure to execute this pathway leads to accumulation of SAF-A–RNA complexes on mitotic chromosomes, defects in metaphase chromosome alignment, and elevated rates of chromosome missegregation in anaphase. This work reveals a role for Aurora-B in removing chromatin-associated RNAs during prophase and demonstrates that Aurora-B–dependent relocalization of SAF-A during cell division contributes to the fidelity of chromosome segregation.
Collapse
Affiliation(s)
- Judith A Sharp
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA.,Department of Genetics, Harvard Medical School, Boston, MA
| | - Carlos Perea-Resa
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA.,Department of Genetics, Harvard Medical School, Boston, MA
| | - Wei Wang
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA.,Department of Genetics, Harvard Medical School, Boston, MA
| | - Michael D Blower
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA.,Department of Genetics, Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Shake It Off: The Elimination of Erroneous Kinetochore-Microtubule Attachments and Chromosome Oscillation. Int J Mol Sci 2021; 22:ijms22063174. [PMID: 33804687 PMCID: PMC8003821 DOI: 10.3390/ijms22063174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/18/2021] [Indexed: 01/17/2023] Open
Abstract
Cell proliferation and sexual reproduction require the faithful segregation of chromosomes. Chromosome segregation is driven by the interaction of chromosomes with the spindle, and the attachment of chromosomes to the proper spindle poles is essential. Initial attachments are frequently erroneous due to the random nature of the attachment process; however, erroneous attachments are selectively eliminated. Proper attachment generates greater tension at the kinetochore than erroneous attachments, and it is thought that attachment selection is dependent on this tension. However, studies of meiotic chromosome segregation suggest that attachment elimination cannot be solely attributed to tension, and the precise mechanism of selective elimination of erroneous attachments remains unclear. During attachment elimination, chromosomes oscillate between the spindle poles. A recent study on meiotic chromosome segregation in fission yeast has suggested that attachment elimination is coupled to chromosome oscillation. In this review, the possible contribution of chromosome oscillation in the elimination of erroneous attachment is discussed in light of the recent finding.
Collapse
|
26
|
Nehlig A, Seiler C, Steblyanko Y, Dingli F, Arras G, Loew D, Welburn J, Prigent C, Barisic M, Nahmias C. Reciprocal regulation of Aurora kinase A and ATIP3 in the control of metaphase spindle length. Cell Mol Life Sci 2021; 78:1765-1779. [PMID: 32789689 PMCID: PMC11072152 DOI: 10.1007/s00018-020-03614-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022]
Abstract
Maintaining the integrity of the mitotic spindle in metaphase is essential to ensure normal cell division. We show here that depletion of microtubule-associated protein ATIP3 reduces metaphase spindle length. Mass spectrometry analyses identified the microtubule minus-end depolymerizing kinesin Kif2A as an ATIP3 binding protein. We show that ATIP3 controls metaphase spindle length by interacting with Kif2A and its partner Dda3 in an Aurora kinase A-dependent manner. In the absence of ATIP3, Kif2A and Dda3 accumulate at spindle poles, which is consistent with reduced poleward microtubule flux and shortening of the spindle. ATIP3 silencing also limits Aurora A localization to the poles. Transfection of GFP-Aurora A, but not kinase-dead mutant, rescues the phenotype, indicating that ATIP3 maintains Aurora A activity on the poles to control Kif2A targeting and spindle size. Collectively, these data emphasize the pivotal role of Aurora kinase A and its mutual regulation with ATIP3 in controlling spindle length.
Collapse
Affiliation(s)
- Anne Nehlig
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France
| | - Cynthia Seiler
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France
| | - Yulia Steblyanko
- Cell Division Laboratory, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Florent Dingli
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Guillaume Arras
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Damarys Loew
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248, Paris Cedex 05, France
| | - Julie Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Claude Prigent
- Institut de Génétique et Développement de Rennes (IGDR), Unité CNRS, UMR 6290, Université de Rennes, 35043, Rennes, France
| | - Marin Barisic
- Cell Division Laboratory, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Clara Nahmias
- Inserm U981, Department of Molecular Medicine, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94800, Villejuif, France.
- LabEx LERMIT, Université Paris Saclay, 92296, Châtenay-Malabry, France.
- Institut Gustave Roussy, Inserm, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, Université Paris-Saclay, 94800, Villejuif, France.
| |
Collapse
|
27
|
Jagrić M, Risteski P, Martinčić J, Milas A, Tolić IM. Optogenetic control of PRC1 reveals its role in chromosome alignment on the spindle by overlap length-dependent forces. eLife 2021; 10:61170. [PMID: 33480356 PMCID: PMC7924949 DOI: 10.7554/elife.61170] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/21/2021] [Indexed: 12/27/2022] Open
Abstract
During metaphase, chromosome position at the spindle equator is regulated by the forces exerted by kinetochore microtubules and polar ejection forces. However, the role of forces arising from mechanical coupling of sister kinetochore fibers with bridging fibers in chromosome alignment is unknown. Here, we develop an optogenetic approach for acute removal of PRC1 to partially disassemble bridging fibers and show that they promote chromosome alignment. Tracking of the plus-end protein EB3 revealed longer antiparallel overlaps of bridging microtubules upon PRC1 removal, which was accompanied by misaligned and lagging kinetochores. Kif4A/kinesin-4 and Kif18A/kinesin-8 were found within the bridging fiber and largely lost upon PRC1 removal, suggesting that these proteins regulate the overlap length of bridging microtubules. We propose that PRC1-mediated crosslinking of bridging microtubules and recruitment of kinesins to the bridging fiber promote chromosome alignment by overlap length-dependent forces transmitted to the associated kinetochore fibers. Before cells divide to create copies of themselves, they need to duplicate their genetic material. To help split their DNA evenly, they build a machine called the mitotic spindle. The mitotic spindle is made of fine, tube-like structures called microtubules, which catch the chromosomes containing the genetic information and line them up at the center of the spindle. Microtubules push and pull the chromosomes by elongating or shortening their tips. But it remains unclear how the microtubules know when the chromosomes have reached center point. One way to find out is to remove proteins that accumulate in the middle of the spindle during division, such as the protein PRC1, which helps to assemble a subset of microtubules called bridging fibers, and the proteins Kif4A and Kif18A, which work like molecular rulers, shortening long microtubules. Usually, scientists would delete one of these proteins to see what impact this has. However, these experiments take days, giving the cell enough time to adapt and thus making it difficult to study the role of each of the proteins. Here, Jagrić, Risteski, Martinčić et al. used light to manipulate proteins at the exact moment of chromosome alignment and to move PRC1 from the spindle to the cell membrane. Consequently, Kif4A and Kif18A were removed from the spindle center. This caused the bridging fibers, which overlap with the microtubules that connect to the chromosomes, to become thinner. Jagrić et al. discovered that without the molecular ruler proteins, the bridging fibers were also too long. This increased the overlap between the microtubules in the center of the spindle, causing the chromosomes to migrate away from the center. This suggests that the alignment of chromosomes in the middle of the spindle depends on the bridging microtubules, which need to be of a certain length to effectively move and keep the chromosomes at the center. Thus, forces that move the chromosomes are generated both at the tips of the microtubules and along the wall of microtubules. These results might inspire other researchers to reassess the role of bridging fibers in cell division. The optogenetic technique described here could also help to determine the parts other proteins have to play. Ultimately, this might allow researchers to identify all the proteins needed to align the chromosomes.
Collapse
Affiliation(s)
- Mihaela Jagrić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Patrik Risteski
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Jelena Martinčić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ana Milas
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
28
|
Steblyanko Y, Rajendraprasad G, Osswald M, Eibes S, Jacome A, Geley S, Pereira AJ, Maiato H, Barisic M. Microtubule poleward flux in human cells is driven by the coordinated action of four kinesins. EMBO J 2020; 39:e105432. [PMID: 33073400 PMCID: PMC7705458 DOI: 10.15252/embj.2020105432] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Mitotic spindle microtubules (MTs) undergo continuous poleward flux, whose driving force and function in humans remain unclear. Here, we combined loss-of-function screenings with analysis of MT-dynamics in human cells to investigate the molecular mechanisms underlying MT-flux. We report that kinesin-7/CENP-E at kinetochores (KTs) is the predominant driver of MT-flux in early prometaphase, while kinesin-4/KIF4A on chromosome arms facilitates MT-flux during late prometaphase and metaphase. Both these activities work in coordination with kinesin-5/EG5 and kinesin-12/KIF15, and our data suggest that the MT-flux driving force is transmitted from non-KT-MTs to KT-MTs by the MT couplers HSET and NuMA. Additionally, we found that the MT-flux rate correlates with spindle length, and this correlation depends on the establishment of stable end-on KT-MT attachments. Strikingly, we find that MT-flux is required to regulate spindle length by counteracting kinesin 13/MCAK-dependent MT-depolymerization. Thus, our study unveils the long-sought mechanism of MT-flux in human cells as relying on the coordinated action of four kinesins to compensate for MT-depolymerization and regulate spindle length.
Collapse
Affiliation(s)
| | | | - Mariana Osswald
- i3S ‐ Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC ‐ Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Susana Eibes
- Danish Cancer Society Research Center (DCRC)CopenhagenDenmark
| | - Ariana Jacome
- i3S ‐ Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC ‐ Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Stephan Geley
- Institute of PathophysiologyBiocenterMedical University of InnsbruckInnsbruckAustria
| | - António J Pereira
- i3S ‐ Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC ‐ Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Helder Maiato
- i3S ‐ Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC ‐ Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
- Experimental Biology UnitDepartment of BiomedicineFaculdade de MedicinaUniversidade do PortoPortoPortugal
| | - Marin Barisic
- Danish Cancer Society Research Center (DCRC)CopenhagenDenmark
- Department of Cellular and Molecular MedicineFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
29
|
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: 13] [Impact Index Per Article: 3.3] [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
|
30
|
Warecki B, Sullivan W. Mechanisms driving acentric chromosome transmission. Chromosome Res 2020; 28:229-246. [PMID: 32712740 DOI: 10.1007/s10577-020-09636-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023]
Abstract
The kinetochore-microtubule association is a core, conserved event that drives chromosome transmission during mitosis. Failure to establish this association on even a single chromosome results in aneuploidy leading to cell death or the development of cancer. However, although many chromosomes lacking centromeres, termed acentrics, fail to segregate, studies in a number of systems reveal robust alternative mechanisms that can drive segregation and successful poleward transport of acentrics. In contrast to the canonical mechanism that relies on end-on microtubule attachments to kinetochores, mechanisms of acentric transmission largely fall into three categories: direct attachments to other chromosomes, kinetochore-independent lateral attachments to microtubules, and long-range tether-based attachments. Here, we review these "non-canonical" methods of acentric chromosome transmission. Just as the discovery and exploration of cell cycle checkpoints provided insight into both the origins of cancer and new therapies, identifying mechanisms and structures specifically involved in acentric segregation may have a significant impact on basic and applied cancer research.
Collapse
Affiliation(s)
- Brandt Warecki
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - William Sullivan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
31
|
Wang X, Garvanska DH, Nasa I, Ueki Y, Zhang G, Kettenbach AN, Peti W, Nilsson J, Page R. A dynamic charge-charge interaction modulates PP2A:B56 substrate recruitment. eLife 2020; 9:55966. [PMID: 32195664 PMCID: PMC7108865 DOI: 10.7554/elife.55966] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/14/2020] [Indexed: 12/16/2022] Open
Abstract
The recruitment of substrates by the ser/thr protein phosphatase 2A (PP2A) is poorly understood, limiting our understanding of PP2A-regulated signaling. Recently, the first PP2A:B56 consensus binding motif, LxxIxE, was identified. However, most validated LxxIxE motifs bind PP2A:B56 with micromolar affinities, suggesting that additional motifs exist to enhance PP2A:B56 binding. Here, we report the requirement of a positively charged motif in a subset of PP2A:B56 interactors, including KIF4A, to facilitate B56 binding via dynamic, electrostatic interactions. Using molecular and cellular experiments, we show that a conserved, negatively charged groove on B56 mediates dynamic binding. We also discovered that this positively charged motif, in addition to facilitating KIF4A dephosphorylation, is essential for condensin I binding, a function distinct and exclusive from PP2A-B56 binding. Together, these results reveal how dynamic, charge-charge interactions fine-tune the interactions mediated by specific motifs, providing a new framework for understanding how PP2A regulation drives cellular signaling.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, United States
| | - Dimitriya H Garvanska
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Isha Nasa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Yumi Ueki
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gang Zhang
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, United States.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Medical Center Drive, Lebanon, United States
| | - Wolfgang Peti
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, United States
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Page
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, United States
| |
Collapse
|
32
|
Wu S, Mipam T, Xu C, Zhao W, Shah MA, Yi C, Luo H, Cai X, Zhong J. Testis transcriptome profiling identified genes involved in spermatogenic arrest of cattleyak. PLoS One 2020; 15:e0229503. [PMID: 32092127 PMCID: PMC7039509 DOI: 10.1371/journal.pone.0229503] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background Cattleyak are the hybrid offspring between cattle and yak and combine yak hardiness with cattle productivity. Much attempt has been made to examine the mechanisms of male sterility caused by spermatogenic arrest, but yet there is no research systematically and precisely elucidated testis gene expression profiling between cattleyak and yak. Methods To explore the higher resolution comparative transcriptome map between the testes of yak and cattleyak, and further analyze the mRNA expression dynamics of spermatogenic arrest in cattleyak. We characterized the comparative transcriptome profile from the testes of yak and cattleyak using high-throughput sequencing. Then we used quantitative analysis to validate several differentially expressed genes (DEGs) in testicular tissue and spermatogenic cells. Results Testis transcriptome profiling identified 6477 DEGs (2919 upregulated and 3558 downregulated) between cattleyak and yak. Further analysis revealed that the marker genes and apoptosis regulatory genes for undifferentiated spermatogonia were upregulated, while the genes for differentiation maintenance were downregulated in cattleyak. A majority of DEGs associated with mitotic checkpoint, and cell cycle progression were downregulated in cattleyak during spermatogonial mitosis. Furthermore, almost all DEGs related to synaptonemal complex assembly, and meiotic progression presented no sign of expression in cattleyak. Even worse, dozens of genes involved in acrosome formation, and flagellar development were dominantly downregulated in cattleyak. Conclusion DEGs indicated that spermatogenic arrest of cattleyak may originate from the differentiation stage of spermatogonial stem cells and be aggravated during spermatogonial mitosis and spermatocyte meiosis, which contributes to the scarcely presented sperms in cattleyak.
Collapse
Affiliation(s)
- Shixin Wu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - TserangDonko Mipam
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Chuanfei Xu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Mujahid Ali Shah
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Chuanping Yi
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Hui Luo
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- * E-mail: (XC); (JZ)
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, China
- * E-mail: (XC); (JZ)
| |
Collapse
|
33
|
Keating L, Touati SA, Wassmann K. A PP2A-B56-Centered View on Metaphase-to-Anaphase Transition in Mouse Oocyte Meiosis I. Cells 2020; 9:E390. [PMID: 32046180 PMCID: PMC7072534 DOI: 10.3390/cells9020390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Meiosis is required to reduce to haploid the diploid genome content of a cell, generating gametes-oocytes and sperm-with the correct number of chromosomes. To achieve this goal, two specialized cell divisions without intermediate S-phase are executed in a time-controlled manner. In mammalian female meiosis, these divisions are error-prone. Human oocytes have an exceptionally high error rate that further increases with age, with significant consequences for human fertility. To understand why errors in chromosome segregation occur at such high rates in oocytes, it is essential to understand the molecular players at work controlling these divisions. In this review, we look at the interplay of kinase and phosphatase activities at the transition from metaphase-to-anaphase for correct segregation of chromosomes. We focus on the activity of PP2A-B56, a key phosphatase for anaphase onset in both mitosis and meiosis. We start by introducing multiple roles PP2A-B56 occupies for progression through mitosis, before laying out whether or not the same principles may apply to the first meiotic division in oocytes, and describing the known meiosis-specific roles of PP2A-B56 and discrepancies with mitotic cell cycle regulation.
Collapse
Affiliation(s)
- Leonor Keating
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Sandra A. Touati
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Katja Wassmann
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
34
|
Dudka D, Castrogiovanni C, Liaudet N, Vassal H, Meraldi P. Spindle-Length-Dependent HURP Localization Allows Centrosomes to Control Kinetochore-Fiber Plus-End Dynamics. Curr Biol 2019; 29:3563-3578.e6. [PMID: 31668617 DOI: 10.1016/j.cub.2019.08.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/23/2019] [Accepted: 08/22/2019] [Indexed: 11/25/2022]
Abstract
During mitosis, centrosomes affect the length of kinetochore fibers (k-fibers) and the stability of kinetochore-microtubule attachments, implying that they regulate k-fiber dynamics. However, the exact cellular and molecular mechanisms of this regulation remain unknown. Here, we created human cells with only one centrosome to investigate these mechanisms. Such cells formed asymmetric bipolar spindles that resulted in asymmetric cell divisions. K-fibers in the acentrosomal half-spindles were shorter, more stable, and had a reduced poleward microtubule flux at minus ends and more frequent pausing events at their plus ends. This indicates that centrosomes regulate k-fiber dynamics both locally at minus ends and far away at plus ends. At the molecular level, we find that the microtubule-stabilizing protein HURP is enriched on the k-fiber plus ends in the acentrosomal half-spindles of cells with only one centrosome. HURP depletion rebalances k-fiber stability and plus-end dynamics in such cells and improves spindle and cell division symmetry. Our data from 3 different cell lines indicate that HURP accumulates on k-fibers inversely proportionally to half-spindle length. We therefore propose that centrosomes regulate k-fiber plus ends indirectly via length-dependent accumulation of HURP.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland
| | - Cédric Castrogiovanni
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland
| | - Nicolas Liaudet
- Bioimaging Facility, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland
| | - Hélène Vassal
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland; National Institute of Applied Sciences, Villeurbanne 69621, France
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland; Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.
| |
Collapse
|
35
|
Abstract
During the cell cycle it is critical that the duplicated DNA faithfully segregates to give rise to two genetically identical daughter cells. An even distribution of the genome during mitosis is mediated by mitotic spindle microtubules, assisted by, among others, motor proteins of the kinesin superfamily. Chromokinesins are members of the kinesin superfamily that harbour a specific DNA-binding domain. The best characterized chromokinesins belong to the kinesin-4/Kif4 and kinesin-10/Kif22 families, respectively. Functional analysis of chromokinesins in several model systems revealed their involvement in chromosome arm orientation and oscillations. This is consistent with their originally proposed role in the generation of polar ejection forces that assist chromosome congression to the spindle equator. Kinesin-12/Kif15 members comprise a third family of chromokinesins, but their role remains less understood. Noteworthy, all chromokinesins exhibit chromosome-independent localization on spindle microtubules, and recent works have significantly extended the portfolio of mitotic processes in which chromokinesins play a role, from error correction and DNA compaction, to the regulation of spindle microtubule dynamics.
Collapse
|
36
|
Vukušić K, Buđa R, Tolić IM. Force-generating mechanisms of anaphase in human cells. J Cell Sci 2019; 132:132/18/jcs231985. [DOI: 10.1242/jcs.231985] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT
What forces drive chromosome segregation remains one of the most challenging questions in cell division. Even though the duration of anaphase is short, it is of utmost importance for genome fidelity that no mistakes are made. Seminal studies in model organisms have revealed different mechanisms operating during chromosome segregation in anaphase, but the translation of these mechanisms to human cells is not straightforward. Recent work has shown that kinetochore fiber depolymerization during anaphase A is largely motor independent, whereas spindle elongation during anaphase B is coupled to sliding of interpolar microtubules in human cells. In this Review, we discuss the current knowledge on the mechanisms of force generation by kinetochore, interpolar and astral microtubules. By combining results from numerous studies, we propose a comprehensive picture of the role of individual force-producing and -regulating proteins. Finally, by linking key concepts of anaphase to most recent data, we summarize the contribution of all proposed mechanisms to chromosome segregation and argue that sliding of interpolar microtubules and depolymerization at the kinetochore are the main drivers of chromosome segregation during early anaphase in human cells.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Renata Buđa
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Iva M. Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
37
|
Yan X, Guo ZX, Liu XP, Feng YJ, Zhao YJ, Liu TZ, Li S. Four novel biomarkers for bladder cancer identified by weighted gene coexpression network analysis. J Cell Physiol 2019; 234:19073-19087. [PMID: 30927274 DOI: 10.1002/jcp.28546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Bladder cancer (BC) is one of the most malignancies in terms of incidence and recurrence worldwide. The aim of this study is to find out novel and prognostic biomarkers for patients with BC. First, we identified 258 differentially expressed genes by using GSE19915 from Gene Expression Omnibus database. Second, a total of 33 modules were identified by constructing a coexpression network by using weighted gene coexpression network analysis and yellow module was regarded as the key module. Furthermore, by constructing protein-protein interaction networks, we preliminarily picked out 13 genes. Among them, four hub genes (CCNB1, KIF4A, TPX2, and TRIP13) were eventually identified by using five different methods (survival analysis, one-way analysis of variance, the Spearman correlation analysis, receiver operating characteristic curve, and expression value comparison), which were significantly correlated with the prognosis of BC. The validation of transcriptional and translational levels made sense (based on Oncomine and The Human Protein Atlas database). Moreover, functional enrichment analysis suggested that all the hub genes played crucial roles in chromosome segregation, sister chromatid segregation, nuclear chromosome segregation, mitotic nuclear division, nuclear division, and organelle fission during cell mitosis. In addition, three of the hub genes (KIF4A, TPX2, and TRIP13) might be potential targets of cancer drugs according to the results of the genetical alteration. In conclusion, this study indicates that four hub genes have great predictive value for the prognosis of BC, and may contribute to the exploration of the further and more in-depth research of BC.
Collapse
Affiliation(s)
- Xin Yan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zi-Xin Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao-Ping Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Jia Feng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying-Jie Zhao
- Department of Urology, Zunyi Medical University, Zunyi, China
| | - Tong-Zu Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sheng Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| |
Collapse
|
38
|
Poser E, Caous R, Gruneberg U, Barr FA. Aurora A promotes chromosome congression by activating the condensin-dependent pool of KIF4A. J Cell Biol 2019; 219:e201905194. [PMID: 31881080 PMCID: PMC7041678 DOI: 10.1083/jcb.201905194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/08/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Aurora kinases create phosphorylation gradients within the spindle during prometaphase and anaphase, thereby locally regulating factors that promote spindle organization, chromosome condensation and movement, and cytokinesis. We show that one such factor is the kinesin KIF4A, which is present along the chromosome axes throughout mitosis and the central spindle in anaphase. These two pools of KIF4A depend on condensin I and PRC1, respectively. Previous work has shown KIF4A is activated by Aurora B at the anaphase central spindle. However, whether or not chromosome-associated KIF4A bound to condensin I is regulated by Aurora kinases remain unclear. To determine the roles of the two different pools of KIF4A, we generated specific point mutants that are unable to interact with either condensin I or PRC1 or are deficient for Aurora kinase regulation. By analyzing these mutants, we show that Aurora A phosphorylates the condensin I-dependent pool of KIF4A and thus actively promotes chromosome congression from the spindle poles to the metaphase plate.
Collapse
Affiliation(s)
- Elena Poser
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Renaud Caous
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Li ZY, Wang ZX, Li CC. Kinesin family member 20B regulates tongue cancer progression by promoting cell proliferation. Mol Med Rep 2019; 19:2202-2210. [PMID: 30664160 PMCID: PMC6390006 DOI: 10.3892/mmr.2019.9851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/12/2018] [Indexed: 01/06/2023] Open
Abstract
Oral cancer refers to the malignant tumors that occur in the oral cavity, of which 80% are squamous cell carcinomas. The incidence of oral cancer accounts for ~5% of the incidence of systemic malignancies, with rapid progression, extensive infiltration and poor prognosis. In the present study, Kinesin family member (KIF)20B, a member of Kinesin-6 family, was identified as a potential biomarker which could promote cancer progression. A total of 82 patients were recruited and KIF20B expression levels were investigated by immunohistochemistry, and were divided into high and low groups based on the median of KIF20B expression levels. The clinicopathological features and survival-associated data of the two groups were analyzed and the results were provided as a table and by a Kaplan-Meier plot, respectively. Additionally, KIF20B was successfully silenced in two tongue cancer cell lines, CAL-27 and TCA-8113. MTT and colony formation assay were performed to determine the changes of cell proliferation in knocked down-KIF20B cell lines. In addition, proliferation-associated proteins Ki67 and PCNA were investigated, by western blotting. In animal experiments, subcutaneous tumor formation was performed with control cells and cells with knocked down KIF20B, to determine the inhibitory effect of KIF20B in vivo. Firstly, it was found that there was significantly high expression levels of KIF20B in tongue cancer patients (P<0.05). Patients with high expression of KIF20B had poorer clinicopathological results including tumor differentiation level, lymph node metastasis and clinical stages. The overall survival and relapse-free survival of high-expression group were also poor. Secondly, after successful establishment of cells with knocked down KIF20B, this resulted in a notable reduction in cell proliferation in vitro. Subsequent western blotting further confirmed that Ki67 and PCNA expression levels had a significant decline. Finally, it was demonstrated that knocking down KIF20B could inhibit tumor volume growth in vivo. In conclusion, the high level of KIF20B in oral squamous cell carcinoma was significantly associated with poor clinicopathological features and survival. KIF20B might promote cancer development through enhancing cell proliferation in vitro, and might be a potential biomarker of oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Zhang-Yi Li
- Department of Stomatology, The Fifth Central Hospital of Tianjin, Tianjin Medical University, Tanggu, Tianjin 300450, P.R. China
| | - Zhi-Xing Wang
- Department of Stomatology, The Fifth Central Hospital of Tianjin, Tianjin Medical University, Tanggu, Tianjin 300450, P.R. China
| | - Chang-Chun Li
- Department of Stomatology, The Fifth Central Hospital of Tianjin, Tianjin Medical University, Tanggu, Tianjin 300450, P.R. China
| |
Collapse
|
40
|
Courthéoux T, Reboutier D, Vazeille T, Cremet JY, Benaud C, Vernos I, Prigent C. Microtubule nucleation during central spindle assembly requires NEDD1 phosphorylation on Serine 405 by Aurora A. J Cell Sci 2019; 132:jcs.231118. [DOI: 10.1242/jcs.231118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
During mitosis, the cell sequentially constructs two microtubule-based spindles to ensure faithful segregation of chromosomes. A bipolar spindle first pulls apart the sister chromatids, then a central spindle further separates them away. Although the assembly of the first spindle is well described, the assembly of the second remains poorly understood. We report here that the inhibition of Aurora A leads to an absence of the central spindle due to a lack of nucleation of microtubules in the midzone. In the absence of Aurora A, the HURP and NEDD1 proteins that are involved in nucleation of microtubules fail to concentrate in the midzone. HURP is an effector of RanGTP and NEDD1 serves as an anchor for the γTURC. Interestingly, Aurora A already phosphorylates them during assembly of the bipolar spindle. We show here that the expression of a NEDD1 isoform mimicking Aurora A phosphorylation is sufficient to restore microtubule nucleation in the midzone in a context of Aurora A inhibition. These results reveal a new control mechanism of nucleation of microtubules by Aurora A during assembly of the central spindle.
Collapse
Affiliation(s)
- Thibault Courthéoux
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - David Reboutier
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Thibaut Vazeille
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jean-Yves Cremet
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Christelle Benaud
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| | - Isabelle Vernos
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Claude Prigent
- Univ. Rennes, CNRS, Institut de Génétique et de Développement de Rennes (IGDR), UMR6290, Equipe labellisée Ligue 2014, F35000 Rennes, France
| |
Collapse
|
41
|
Delayed Chromosome Alignment to the Spindle Equator Increases the Rate of Chromosome Missegregation in Cancer Cell Lines. Biomolecules 2018; 9:biom9010010. [PMID: 30597919 PMCID: PMC6359495 DOI: 10.3390/biom9010010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022] Open
Abstract
For appropriate chromosome segregation, kinetochores on sister chromatids have to attach to microtubules from opposite spindle poles (bi-orientation). Chromosome alignment at the spindle equator, referred to as congression, can occur through the attachment of kinetochores to the lateral surface of spindle microtubules, facilitating bi-orientation establishment. However, the contribution of this phenomenon to mitotic fidelity has not been clarified yet. Here, we addressed whether delayed chromosome alignment to the spindle equator increases the rate of chromosome missegregation. Cancer cell lines depleted of Kid, a chromokinesin involved in chromosome congression, showed chromosome alignment with a slight delay, and increased frequency of lagging chromosomes. Delayed chromosome alignment concomitant with an increased rate of lagging chromosomes was also seen in cells depleted of kinesin family member 4A (KIF4A), another chromokinesin. Cells that underwent chromosome missegregation took relatively longer time to align chromosomes in both control and Kid/KIF4A-depleted cells. Tracking of late-aligning chromosomes showed that they exhibit a higher rate of lagging chromosomes. Intriguingly, the metaphase of cells that underwent chromosome missegregation was shortened, and delaying anaphase onset ameliorated the increased chromosome missegregation. These data suggest that late-aligning chromosomes do not have sufficient time to establish bi-orientation, leading to chromosome missegregation. Our data imply that delayed chromosome alignment is not only a consequence, but also a cause of defective bi-orientation establishment, which can lead to chromosomal instability in cells without severe mitotic defects.
Collapse
|
42
|
Olziersky AM, Smith CA, Burroughs N, McAinsh AD, Meraldi P. Mitotic live-cell imaging at different timescales. Methods Cell Biol 2018; 145:1-27. [PMID: 29957199 DOI: 10.1016/bs.mcb.2018.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitosis is a highly dynamic and choreographed process in which chromosomes are captured by the mitotic spindle and physically segregated into the two daughter cells to ensure faithful transmission of the genetic material. Live-cell fluorescence microscopy enables these dynamics to be analyzed over diverse temporal scales. Here we present the methodologies to study chromosome segregation at three timescales: we first show how automated tracking of kinetochores enables investigation of mitotic spindle and chromosome dynamics in the seconds-to-minutes timescale; next we highlight how new DNA live dyes allow the study of chromosome segregation over a period of several hours in any cell line; finally, we demonstrate how image sequences acquired over several days can reveal the fate of whole cell populations over several consecutive cell divisions.
Collapse
Affiliation(s)
- Anna-Maria Olziersky
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Chris A Smith
- Centre for Mechanochemical Cell Biology & Division of Biomedical Science, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Nigel Burroughs
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
| | - Andrew D McAinsh
- Centre for Mechanochemical Cell Biology & Division of Biomedical Science, Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
43
|
The multiple functions of kinesin-4 family motor protein KIF4 and its clinical potential. Gene 2018; 678:90-99. [DOI: 10.1016/j.gene.2018.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
|
44
|
Pan LN, Zhang Y, Zhu CJ, Dong ZX. Kinesin KIF4A is associated with chemotherapeutic drug resistance by regulating intracellular trafficking of lung resistance-related protein. J Zhejiang Univ Sci B 2018; 18:1046-1054. [PMID: 29204984 DOI: 10.1631/jzus.b1700129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Multidrug resistance (MDR) is the major impediment to cancer chemotherapy. The expression of lung resistance-related protein (LRP), a non-ATP-binding cassette (ABC) transporter, is high in tumor cells, resulting in their resistance to a variety of cytotoxic drugs. However, the function of LRP in tumor drug resistance is not yet explicit. Our previous studies had shown that Kinesin KIF4A was overexpressed in cisplatin (DDP)-resistant human lung adenocarcinoma cells (A549/DDP cells) compared with A549 cells. The expression of KIF4A in A549 or A549/DDP cells significantly affects cisplatin resistance but the detailed mechanisms remain unclear. Here, we performed co-immunoprecipitation experiments to show that the tail domain of KIF4A interacted with the N-terminal of LRP. Immunofluorescence images showed that both the ability of binding to LRP and the motility of KIF4A were essential for the dispersed cytoplasm distribution of LRP. Altogether, our results shed light on a potential mechanism in that motor protein KIF4A promotes drug resistance of lung adenocarcinoma cells through transporting LRP-based vaults along microtubules towards the cell membrane. Thus KIF4A might be a cisplatin resistance-associated protein and serves as a potential target for chemotherapeutic drug resistance in lung cancer.
Collapse
Affiliation(s)
- Li-Na Pan
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin 300387, China.,Key Laboratory of Molecular and Cellular Systems Biology, Tianjin Normal University, Tianjin 300387, China
| | - Yuan Zhang
- Department of Cancer Research Institute, Cancer Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Chang-Jun Zhu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin 300387, China.,Key Laboratory of Molecular and Cellular Systems Biology, Tianjin Normal University, Tianjin 300387, China
| | - Zhi-Xiong Dong
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin 300387, China.,Key Laboratory of Molecular and Cellular Systems Biology, Tianjin Normal University, Tianjin 300387, China
| |
Collapse
|
45
|
Ye AA, Verma V, Maresca TJ. NOD is a plus end-directed motor that binds EB1 via a new microtubule tip localization sequence. J Cell Biol 2018; 217:3007-3017. [PMID: 29899040 PMCID: PMC6122986 DOI: 10.1083/jcb.201708109] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 03/14/2018] [Accepted: 05/25/2018] [Indexed: 02/08/2023] Open
Abstract
The mechanism by which the Drosophila chromokinesin NOD promotes chromosome congression is unknown. Ye et al. demonstrate that NOD generates force by two mechanisms: plus end–directed motility and microtubule plus-tip tracking via interaction with EB1 through a newly identified motif. Chromosome congression, the process of positioning chromosomes in the midspindle, promotes the stable transmission of the genome to daughter cells during cell division. Congression is typically facilitated by DNA-associated, microtubule (MT) plus end–directed motors called chromokinesins. The Drosophila melanogaster chromokinesin NOD contributes to congression, but the means by which it does so are unknown in large part because NOD has been classified as a nonmotile, orphan kinesin. It has been postulated that NOD promotes congression, not by conventional plus end–directed motility, but by harnessing polymerization forces by end-tracking on growing MT plus ends via a mechanism that is also uncertain. Here, for the first time, it is demonstrated that NOD possesses MT plus end–directed motility. Furthermore, NOD directly binds EB1 through unconventional EB1-interaction motifs that are similar to a newly characterized MT tip localization sequence. We propose NOD produces congression forces by MT plus end–directed motility and tip-tracking on polymerizing MT plus ends via association with EB1.
Collapse
Affiliation(s)
- Anna A Ye
- Biology Department, University of Massachusetts, Amherst, Amherst, MA.,Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Amherst, MA
| | - Vikash Verma
- Biology Department, University of Massachusetts, Amherst, Amherst, MA
| | - Thomas J Maresca
- Biology Department, University of Massachusetts, Amherst, Amherst, MA .,Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Amherst, MA
| |
Collapse
|
46
|
Drpic D, Almeida AC, Aguiar P, Renda F, Damas J, Lewin HA, Larkin DM, Khodjakov A, Maiato H. Chromosome Segregation Is Biased by Kinetochore Size. Curr Biol 2018; 28:1344-1356.e5. [PMID: 29706521 PMCID: PMC5954971 DOI: 10.1016/j.cub.2018.03.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 02/02/2018] [Accepted: 03/13/2018] [Indexed: 11/30/2022]
Abstract
Chromosome missegregation during mitosis or meiosis is a hallmark of cancer and the main cause of prenatal death in humans. The gain or loss of specific chromosomes is thought to be random, with cell viability being essentially determined by selection. Several established pathways including centrosome amplification, sister-chromatid cohesion defects, or a compromised spindle assembly checkpoint can lead to chromosome missegregation. However, how specific intrinsic features of the kinetochore—the critical chromosomal interface with spindle microtubules—impact chromosome segregation remains poorly understood. Here we used the unique cytological attributes of female Indian muntjac, the mammal with the lowest known chromosome number (2n = 6), to characterize and track individual chromosomes with distinct kinetochore size throughout mitosis. We show that centromere and kinetochore functional layers scale proportionally with centromere size. Measurement of intra-kinetochore distances, serial-section electron microscopy, and RNAi against key kinetochore proteins confirmed a standard structural and functional organization of the Indian muntjac kinetochores and revealed that microtubule binding capacity scales with kinetochore size. Surprisingly, we found that chromosome segregation in this species is not random. Chromosomes with larger kinetochores bi-oriented more efficiently and showed a 2-fold bias to congress to the equator in a motor-independent manner. Despite robust correction mechanisms during unperturbed mitosis, chromosomes with larger kinetochores were also strongly biased to establish erroneous merotelic attachments and missegregate during anaphase. This bias was impervious to the experimental attenuation of polar ejection forces on chromosome arms by RNAi against the chromokinesin Kif4a. Thus, kinetochore size is an important determinant of chromosome segregation fidelity. Centromere/kinetochore functional layers scale proportionally with centromere size Kinetochore microtubule binding capacity scales with kinetochore size Chromosome congression and bi-orientation are biased by kinetochore size Error formation leading to chromosome missegregation is biased by kinetochore size
Collapse
Affiliation(s)
- Danica Drpic
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Ana C Almeida
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Paulo Aguiar
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto Nacional de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Fioranna Renda
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Joana Damas
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
| | - Harris A Lewin
- Department of Evolution and Ecology, University of California, Davis, Davis, CA 95616, USA
| | - Denis M Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London NW1 0TU, UK
| | - Alexey Khodjakov
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA; Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Helder Maiato
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
47
|
Klemm AH, Bosilj A, Gluncˇic M, Pavin N, Tolic IM. Metaphase kinetochore movements are regulated by kinesin-8 motors and microtubule dynamic instability. Mol Biol Cell 2018; 29:1332-1345. [PMID: 29851559 PMCID: PMC5994901 DOI: 10.1091/mbc.e17-11-0667] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
During metaphase, sister chromatids are connected to microtubules extending from the opposite spindle poles via kinetochores to protein complexes on the chromosome. Kinetochores congress to the equatorial plane of the spindle and oscillate around it, with kinesin-8 motors restricting these movements. Yet, the physical mechanism underlying kinetochore movements is unclear. We show that kinetochore movements in the fission yeast Schizosaccharomyces pombe are regulated by kinesin-8-promoted microtubule catastrophe, force-induced rescue, and microtubule dynamic instability. A candidate screen showed that among the selected motors only kinesin-8 motors Klp5/Klp6 are required for kinetochore centering. Kinesin-8 accumulates at the end of microtubules, where it promotes catastrophe. Laser ablation of the spindle resulted in kinetochore movement toward the intact spindle pole in wild-type and klp5Δ cells, suggesting that kinetochore movement is driven by pulling forces. Our theoretical model with Langevin description of microtubule dynamic instability shows that kinesin-8 motors are required for kinetochore centering, whereas sensitivity of rescue to force is necessary for the generation of oscillations. We found that irregular kinetochore movements occur for a broader range of parameters than regular oscillations. Thus, our work provides an explanation for how regulation of microtubule dynamic instability contributes to kinetochore congression and the accompanying movements around the spindle center.
Collapse
Affiliation(s)
- Anna H Klemm
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Agneza Bosilj
- Department of Physics, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Matko Gluncˇic
- Department of Physics, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Nenad Pavin
- Department of Physics, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Iva M Tolic
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.,Division of Molecular Biology, Rud¯er Boškovic´ Institute, 10000 Zagreb, Croatia
| |
Collapse
|
48
|
Ben-Shimon L, Paul VD, David-Kadoch G, Volpe M, Stümpfig M, Bill E, Mühlenhoff U, Lill R, Ben-Aroya S. Fe-S cluster coordination of the chromokinesin KIF4A alters its sub-cellular localization during mitosis. J Cell Sci 2018; 131:jcs.211433. [DOI: 10.1242/jcs.211433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/11/2018] [Indexed: 11/20/2022] Open
Abstract
Fe-S clusters act as co-factors of proteins with diverse functions, e.g. in DNA repair. Down-regulation of the cytosolic iron-sulfur protein assembly (CIA) machinery promotes genomic instability by the inactivation of multiple DNA repair pathways. Furthermore, CIA deficiencies are associated with so far unexplained mitotic defects. Here, we show that CIA2B and MMS19, constituents of the CIA targeting complex involved in facilitating Fe-S cluster insertion into cytosolic and nuclear target proteins, co-localize with components of the mitotic machinery. Down-regulation of CIA2B and MMS19 impairs the mitotic cycle. We identify the chromokinesin KIF4A as a mitotic component involved in these effects. KIF4A binds a Fe-S cluster in vitro through its conserved cysteine-rich domain. We demonstrate in vivo that this domain is required for the mitosis-related KIF4A localization and for the mitotic defects associated with KIF4A knockout. KIF4A is the first identified mitotic component carrying such a post-translational modification. These findings suggest that the lack of Fe-S clusters in KIF4A upon down-regulation of the CIA targeting complex contributes to the mitotic defects.
Collapse
Affiliation(s)
- Lilach Ben-Shimon
- The Nano Center, Building 206 room B-840, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Viktoria D. Paul
- Institut für Zytobiologie und Zytopathologie, Philipps-Universität Marburg, Robert-Koch-Strasse 6, 35032 Marburg, Germany
- LOEWE Zentrum für Synthetische Mikrobiologie SynMikro, Hans-Meerwein-Strasse, 35043 Marburg, Germany
| | - Galit David-Kadoch
- The Nano Center, Building 206 room B-840, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Marina Volpe
- The Nano Center, Building 206 room B-840, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Martin Stümpfig
- Institut für Zytobiologie und Zytopathologie, Philipps-Universität Marburg, Robert-Koch-Strasse 6, 35032 Marburg, Germany
| | - Eckhard Bill
- Max-Planck-Institut für Chemische Energiekonversion, Stiftstrasse 34-36, 45470 Mülheim-Ruhr, Germany
| | - Ulrich Mühlenhoff
- Institut für Zytobiologie und Zytopathologie, Philipps-Universität Marburg, Robert-Koch-Strasse 6, 35032 Marburg, Germany
| | - Roland Lill
- Institut für Zytobiologie und Zytopathologie, Philipps-Universität Marburg, Robert-Koch-Strasse 6, 35032 Marburg, Germany
- LOEWE Zentrum für Synthetische Mikrobiologie SynMikro, Hans-Meerwein-Strasse, 35043 Marburg, Germany
| | - Shay Ben-Aroya
- The Nano Center, Building 206 room B-840, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| |
Collapse
|
49
|
Sld5 Ensures Centrosomal Resistance to Congression Forces by Preserving Centriolar Satellites. Mol Cell Biol 2017; 38:MCB.00371-17. [PMID: 29061732 DOI: 10.1128/mcb.00371-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/11/2017] [Indexed: 11/20/2022] Open
Abstract
The migration of chromosomes during mitosis is mediated primarily by kinesins that bind to the chromosomes and move along the microtubules, exerting pulling and pushing forces on the centrosomes. We report that a DNA replication protein, Sld5, localizes to the centrosomes, resisting the microtubular pulling forces experienced during chromosome congression. In the absence of Sld5, centriolar satellites, which normally cluster around the centrosomes, are dissipated throughout the cytoplasm, resulting in the loss of their known function of recruiting the centrosomal protein, pericentrin. We observed that Sld5-deficient centrosomes lacking pericentrin were unable to endure the CENP-E- and Kid-mediated microtubular forces that converge on the centrosomes during chromosome congression, resulting in monocentriolar and acentriolar spindle poles. The minus-end-directed kinesin-14 motor protein, HSET, sustains the traction forces that mediate centrosomal fragmentation in Sld5-depleted cells. Thus, we report that a DNA replication protein has an as yet unknown function of ensuring spindle pole resistance to traction forces exerted during chromosome congression.
Collapse
|
50
|
Matsumoto Y, Saito M, Saito K, Kanke Y, Watanabe Y, Onozawa H, Hayase S, Sakamoto W, Ishigame T, Momma T, Kumamoto K, Ohki S, Takenoshita S. Enhanced expression of KIF4A in colorectal cancer is associated with lymph node metastasis. Oncol Lett 2017; 15:2188-2194. [PMID: 29434924 PMCID: PMC5776904 DOI: 10.3892/ol.2017.7555] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 01/19/2017] [Indexed: 01/08/2023] Open
Abstract
Kinesin family member 4A (KIF4A) is a member of the kinesin 4 subfamily of kinesin-related proteins and serves an important role in cell division. The expression levels of KIF4A have been investigated in numerous types of cancer, including cervical, lung, oral, and breast cancer, and are established to be associated with poor patient prognosis. However, the role of KIF4A, as well as its expression in colorectal cancer (CRC), remains to be elucidated. Therefore, the current study investigated KIF4A expression levels in patients with CRC and demonstrated that its levels were increased in tumor tissues compared with non-tumor tissues. To investigate the functional role of KIF4A, KIF4A was knocked down in CRC cells and cell viability was evaluated. CRC cells with KIF4A knockdown exhibited lower cell proliferation compared with control cells. In addition, KIF4A expression levels, as determined by immunohistochemistry, were compared with the expression of Ki-67, but no significant associations were observed in the patients with CRC. Therefore, KIF4A was found to be upregulated in patients with CRC and downregulation of KIF4A reduced cell proliferation in CRC cells. These results suggest that KIF4A may be a potential therapeutic target, which may improve the outcomes of patients with CRC.
Collapse
Affiliation(s)
- Yoshiko Matsumoto
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Motonobu Saito
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Katsuharu Saito
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yasuyuki Kanke
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yohei Watanabe
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hisashi Onozawa
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Suguru Hayase
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Wataru Sakamoto
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Teruhide Ishigame
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Tomoyuki Momma
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kensuke Kumamoto
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shinji Ohki
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Seiichi Takenoshita
- Department of Organ Regulatory Surgery, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|