1
|
Fries B, Sekera E, Holbrook J, Hummon A. Multi-Omic Evaluation of PLK1 Inhibitor-Onvansertib-In Colorectal Cancer Spheroids. JOURNAL OF MASS SPECTROMETRY : JMS 2025; 60:e5137. [PMID: 40197665 PMCID: PMC11976698 DOI: 10.1002/jms.5137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Polo-like kinase 1 (Plk1) is a serine/threonine kinase involved in regulating the cell cycle. It is activated by aurora kinase B along with the cofactors Borealin, INCE, and survivin. Plk1 is involved in the development of resistances to chemotherapeutics such as doxorubicin, Taxol, and gemcitabine. It has been shown that patients with higher levels of Plk1 have lower survival rates. Onvansertib is a competitive ATP inhibitor for Plk1 in clinical trials for the treatment of tumors and has recently entered a trial for the treatment of KRAS mutant colorectal cancers (CRCs). In this study, we conducted an untargeted liquid chromatography-mass spectrometry (LC-MS) proteomics study as well as an untargeted lipidomics analysis of HCT 116 spheroids treated with onvansertib over a 72-h treatment time-course experiment. Mass spectrometry imaging (MSI) showed that onvansertib begins to accumulate most prominently after 12 h of treatment and continues to accumulate through 72 h. Proteomic results displayed alterations to cell cycle control proteins and an increasing abundance of aurora kinase B and Borealin. The proteomics data also showed alterations to many lipid metabolism enzymes. The MSI lipidomics data indicated alterations to phosphatidylcholine lipids, with many lipids increasing in abundance over time or increasing until 12 h of onvansertib treatment and decreasing after that time point. In summary, these results suggest that onvansertib is causing cells within the spheroid to halt at a certain phase of the cell cycle in accordance with previous literature. Our findings suggest the S phase is likely interrupted, with observed alterations in cell cycle control proteins and PC lipid abundance.
Collapse
Affiliation(s)
- Brian D. Fries
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
| | - Emily R. Sekera
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
| | | | - Amanda B. Hummon
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
2
|
Lakshmi S, Priya S. Molecular insights into epoxyazadiradione induced death in triple-negative breast cancer cells: A system biology approach. Gene 2024; 930:148814. [PMID: 39116958 DOI: 10.1016/j.gene.2024.148814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Epoxyazadiradione is an important limonoid with immense pharmacological potential. We have reported previously that epoxyazadiradione (EAD) induces apoptosis in triple negative breast cancer cells (MDA-MB 231) by modulating diverse cellular targets. Here, we identify the key genes/pathways responsible for this effect through next-generation sequencing of the transcriptome from EAD treated cells and integrated molecular data analysis using bioinformatics. In silico analysis indicated that EAD displayed favourable drug-like properties and could target multiple macromolecules relevant to TNBC. RNA sequencing revealed that EAD treatment results in the differential expression of 1838 genes in MDA-MB 231 cells, with 752 downregulated and 1086 upregulated. Gene set enrichment analysis of these genes suggested that EAD disrupts protein folding in the endoplasmic reticulum, triggering the unfolded protein response (UPR) and potentially leading to cell death. EAD also induced oxidative stress and DNA damage, downregulated pathways linked to metabolism, cell cycle progression, pro-survival signalling, cell adhesion, motility and inflammatory response. The identification of protein cluster and hub genes were also done. The validation of the identified hub genes gave an inverse correlation between their expression in EAD treated cells and TNBC patient samples. Thus, the identified hub genes could be explored as therapeutic or diagnostic markers for TNBC. Hence, EAD appears to be a promising therapeutic candidate for TNBC by targeting various hallmarks of cancer, including cell death resistance, uncontrolled proliferation and metastasis. To conclude, the identified pathways and validated targets for EAD will provide a roadmap for further in vivo studies and preclinical/clinical validation required for potential drug development.
Collapse
Affiliation(s)
- Sreerenjini Lakshmi
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695 019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sulochana Priya
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695 019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
3
|
Fesquet D, Rabeharivelo G, van Dijk J, Prigent C, Morin N, Rouquier S. CCDC69 maintains genome integrity by regulating KIF2C/MCAK depolymerase activity and the stability of the chromosomal passenger complex. Sci Rep 2024; 14:30401. [PMID: 39638803 PMCID: PMC11621681 DOI: 10.1038/s41598-024-81022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
Accurate genome inheritance during cell division relies on a complex chromosome segregation mechanism. This process occurs once all the kinetochores of sister chromatids are attached to microtubules emanating from the opposite poles of the mitotic spindle. To control the precision of this mechanism, the Chromosome Passenger Complex (CPC) actively identifies and corrects improper microtubule attachments. The depolymerase activity of the kinesin KIF2C/MCAK at the kinetochores is involved in this process. CCDC69 is a poorly characterized protein, primarily identified as a regulator of central spindle assembly, whose overexpression prompts rapid microtubule depolymerization. Here, we show that CCDC69 is a cell-cycle regulated protein belonging to the Microtubule-associated Tumor Suppressor (MTUS) superfamily, and even slight deregulation of its expression induces severe early mitotic phenotypes. Myristoylation anchors CCDC69 at the plasma membrane, thus protecting microtubule network integrity. We found that CCDC69 microtubule depolymerization activity relies on KIF2C, with a fraction of CCDC69 localizing to the centromere. Importantly, we demonstrated that CCDC69 regulates the stability of the CPC by safeguarding its members from degradation during mitosis. In summary, our findings underscore CCDC69's essential role as a mitotic regulator, which is crucial for maintaining the fidelity of chromosome segregation.
Collapse
Affiliation(s)
- Didier Fesquet
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Gabriel Rabeharivelo
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Juliette van Dijk
- CRBM CNRS UMR 5237, Equipe Centrosome Cil Et Pathologies, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Claude Prigent
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Nathalie Morin
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Sylvie Rouquier
- CRBM CNRS UMR 5237, Equipe Cycle Cellulaire, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France.
| |
Collapse
|
4
|
Dwivedi D, Meraldi P. Balancing Plk1 activity levels: The secret of synchrony between the cell and the centrosome cycle. Bioessays 2024; 46:e2400048. [PMID: 39128131 DOI: 10.1002/bies.202400048] [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: 02/29/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
The accuracy of cell division requires precise regulation of the cellular machinery governing DNA/genome duplication, ensuring its equal distribution among the daughter cells. The control of the centrosome cycle is crucial for the formation of a bipolar spindle, ensuring error-free segregation of the genome. The cell and centrosome cycles operate in close synchrony along similar principles. Both require a single duplication round in every cell cycle, and both are controlled by the activity of key protein kinases. Nevertheless, our comprehension of the precise cellular mechanisms and critical regulators synchronizing these two cycles remains poorly defined. Here, we present our hypothesis that the spatiotemporal regulation of a dynamic equilibrium of mitotic kinases activities forms a molecular clock that governs the synchronous progression of both the cell and the centrosome cycles.
Collapse
Affiliation(s)
- Devashish Dwivedi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
5
|
Bellah SF, Xiong F, Dou Z, Yang F, Liu X, Yao X, Gao X, Zhang L. PLK1 phosphorylation of ZW10 guides accurate chromosome segregation in mitosis. J Mol Cell Biol 2024; 16:mjae008. [PMID: 38402459 PMCID: PMC11328731 DOI: 10.1093/jmcb/mjae008] [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: 09/03/2023] [Revised: 11/09/2023] [Accepted: 02/23/2024] [Indexed: 02/26/2024] Open
Abstract
Stable transmission of genetic information during cell division requires faithful chromosome segregation. Mounting evidence has demonstrated that polo-like kinase 1 (PLK1) dynamics at kinetochores control correct kinetochore-microtubule attachments and subsequent silencing of the spindle assembly checkpoint. However, the mechanisms underlying PLK1-mediated silencing of the spindle checkpoint remain elusive. Here, we identified a regulatory mechanism by which PLK1-elicited zeste white 10 (ZW10) phosphorylation regulates spindle checkpoint silencing in mitosis. ZW10 is a cognate substrate of PLK1, and the phosphorylation of ZW10 at Ser12 enables dynamic ZW10-Zwint1 interactions. Inhibition of ZW10 phosphorylation resulted in misaligned chromosomes, while persistent expression of phospho-mimicking ZW10 mutant caused premature anaphase, in which sister chromatids entangled as cells entered anaphase. These findings reveal the previously uncharacterized PLK1-ZW10 interaction through which dynamic phosphorylation of ZW10 fine-tunes accurate chromosome segregation in mitosis.
Collapse
Affiliation(s)
- Sm Faysal Bellah
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Fangyuan Xiong
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xinjiao Gao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Liangyu Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230027, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| |
Collapse
|
6
|
Kulkarni H, Dagar N, Gaikwad AB. Targeting polo-like kinase 1 to treat kidney diseases. Cell Biochem Funct 2024; 42:e4099. [PMID: 39016459 DOI: 10.1002/cbf.4099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
Globally, ∼850 million individuals suffer from some form of kidney disease. This staggering figure underscores the importance of continued research and innovation in the field of nephrology to develop effective treatments and improve overall global kidney health. In current research, the polo-like kinase (Plk) family has emerged as a group of highly conserved enzyme kinases vital for proper cell cycle regulation. Plks are defined by their N-terminal kinase domain and C-terminal polo-box domain, which regulate their catalytic activity, subcellular localization, and substrate recognition. Among the Plk family members, Plk1 has garnered significant attention due to its pivotal role in regulating multiple mitotic processes, particularly in the kidneys. It is a crucial serine-threonine (Ser-Thr) kinase involved in cell division and genomic stability. In this review, we delve into the types and functions of Plks, focusing on Plk1's significance in processes such as cell proliferation, spindle assembly, and DNA damage repair. The review also underscores Plk1's vital contributions to maintaining kidney homeostasis, elucidating its involvement in nuclear envelope breakdown, anaphase-promoting complex/cyclosome activation, and the regulation of mRNA translation machinery. Furthermore, the review discusses how Plk1 contributes to the development and progression of kidney diseases, emphasizing its overexpression in conditions such as acute kidney injury, chronic kidney disease, and so forth. It also highlights the importance of exploring Plk1 modulators as targeted therapies for kidney diseases in future. This review will help in understanding the role of Plk1 in kidney disease development, paving the way for the discovery and development of novel therapeutic approaches to manage kidney diseases effectively.
Collapse
Affiliation(s)
- Hrushikesh Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Neha Dagar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| |
Collapse
|
7
|
Zhang B, Ayra-Pardo C, Liu X, Song M, Li D, Kan Y. siRNA-Mediated BmAurora B Depletion Impedes the Formation of Holocentric Square Spindles in Silkworm Metaphase BmN4 Cells. INSECTS 2024; 15:72. [PMID: 38276821 PMCID: PMC10817069 DOI: 10.3390/insects15010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Silkworm ovary-derived BmN4 cells rely on chromatin-induced spindle assembly to form microtubule-based square mitotic spindles that ensure accurate segregation of holocentric chromosomes during cell division. The chromosome passenger protein Aurora B regulates chromosomal condensation and segregation, spindle assembly checkpoint activation, and cytokinesis; however, its role in holocentric organisms needs further clarification. This study examined the architecture and dynamics of spindle microtubules during prophase and metaphase in BmN4 cells and those with siRNA-mediated BmAurora B knockdown using immunofluorescence labeling. Anti-α-tubulin and anti-γ-tubulin antibodies revealed faint γ-tubulin signals colocalized with α-tubulin in early prophase during nuclear membrane rupture, which intensified as prophase progressed. At this stage, bright regions of α-tubulin around and on the nuclear membrane surrounding the chromatin suggested the start of microtubules assembling in the microtubule-organizing centers (MTOCs). In metaphase, fewer but larger γ-tubulin foci were detected on both sides of the chromosomes. This resulted in a distinctive multipolar square spindle with holocentric chromosomes aligned at the metaphase plate. siRNA-mediated BmAurora B knockdown significantly reduced the γ-tubulin foci during prophase, impacting microtubule nucleation and spindle structure in metaphase. Spatiotemporal BmAurora B expression analysis provided new insights into the regulation of this mitotic kinase in silkworm larval gonads during gametogenesis. Our results suggest that BmAurora B is crucial for the formation of multipolar square spindles in holocentric insects, possibly through the activation of γ-tubulin ring complexes in multiple centrosome-like MTOCs.
Collapse
Affiliation(s)
- Bing Zhang
- Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan International Joint Laboratory of Insect Biology, College of Life Science and Agricultural Engineering, Nanyang Normal University, 1638 Wolong Road, Nanyang 473061, China; (X.L.); (M.S.); (D.L.)
| | - Camilo Ayra-Pardo
- CIIMAR–Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões, University of Porto, Avda. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal;
| | - Xiaoning Liu
- Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan International Joint Laboratory of Insect Biology, College of Life Science and Agricultural Engineering, Nanyang Normal University, 1638 Wolong Road, Nanyang 473061, China; (X.L.); (M.S.); (D.L.)
| | - Meiting Song
- Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan International Joint Laboratory of Insect Biology, College of Life Science and Agricultural Engineering, Nanyang Normal University, 1638 Wolong Road, Nanyang 473061, China; (X.L.); (M.S.); (D.L.)
| | - Dandan Li
- Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan International Joint Laboratory of Insect Biology, College of Life Science and Agricultural Engineering, Nanyang Normal University, 1638 Wolong Road, Nanyang 473061, China; (X.L.); (M.S.); (D.L.)
| | - Yunchao Kan
- Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan International Joint Laboratory of Insect Biology, College of Life Science and Agricultural Engineering, Nanyang Normal University, 1638 Wolong Road, Nanyang 473061, China; (X.L.); (M.S.); (D.L.)
- School of Life Science and Technology, Henan Institute of Science and Technology, 90 East of Hualan Avenue, Xinxiang 453003, China
| |
Collapse
|
8
|
Chen YZ, Zimyanin V, Redemann S. Loss of the mitochondrial protein SPD-3 elevates PLK-1 levels and dysregulates mitotic events. Life Sci Alliance 2023; 6:e202302011. [PMID: 37684042 PMCID: PMC10488725 DOI: 10.26508/lsa.202302011] [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: 02/23/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
In metazoans, Polo-like kinase (PLK1) controls several mitotic events including nuclear envelope breakdown, centrosome maturation, spindle assembly and progression through mitosis. Here we show that a mutation in the mitochondria-localized protein SPD-3 affects mitotic events by inducing elevated levels of PLK-1 in early Caenorhabditis elegans embryos. SPD-3 mutant embryos contain abnormally positioned mitotic chromosomes, show a delay in anaphase onset and asymmetrically disassemble the nuclear lamina. We found that more PLK-1 accumulated on centrosomes, nuclear envelope, nucleoplasm, and chromatin before NEBD, suggesting that PLK-1 overexpression is responsible for some of the observed mitotic phenotypes. In agreement with this, the chromosome positioning defects of the spd-3(oj35) mutant could be rescued by reducing PLK-1 levels. Our data suggests that the mitochondrial SPD-3 protein affects chromosome positioning and nuclear envelope integrity by up-regulating the endogenous levels of PLK-1 during early embryogenesis in C. elegans This finding suggests a novel link between mitochondria and nuclear envelope dynamics and chromosome positioning by increasing the amount of a key mitotic regulator, PLK-1, providing a novel link between mitochondria and mitosis.
Collapse
Affiliation(s)
- Yu-Zen Chen
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Vitaly Zimyanin
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Stefanie Redemann
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Taylor SJP, Bel Borja L, Soubigou F, Houston J, Cheerambathur DK, Pelisch F. BUB-1 and CENP-C recruit PLK-1 to control chromosome alignment and segregation during meiosis I in C. elegans oocytes. eLife 2023; 12:e84057. [PMID: 37067150 PMCID: PMC10156168 DOI: 10.7554/elife.84057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 04/14/2023] [Indexed: 04/18/2023] Open
Abstract
Phosphorylation is a key post-translational modification that is utilised in many biological processes for the rapid and reversible regulation of protein localisation and activity. Polo-like kinase 1 (PLK-1) is essential for both mitotic and meiotic cell divisions, with key functions being conserved in eukaryotes. The roles and regulation of PLK-1 during mitosis have been well characterised. However, the discrete roles and regulation of PLK-1 during meiosis have remained obscure. Here, we used Caenorhabditis elegans oocytes to show that PLK-1 plays distinct roles in meiotic spindle assembly and/or stability, chromosome alignment and segregation, and polar body extrusion during meiosis I. Furthermore, by a combination of live imaging and biochemical analysis we identified the chromosomal recruitment mechanisms of PLK-1 during C. elegans oocyte meiosis. The spindle assembly checkpoint kinase BUB-1 directly recruits PLK-1 to the kinetochore and midbivalent while the chromosome arm population of PLK-1 depends on a direct interaction with the centromeric-associated protein CENP-CHCP-4. We found that perturbing both BUB-1 and CENP-CHCP-4 recruitment of PLK-1 leads to severe meiotic defects, resulting in highly aneuploid oocytes. Overall, our results shed light on the roles played by PLK-1 during oocyte meiosis and provide a mechanistic understanding of PLK-1 targeting to meiotic chromosomes.
Collapse
Affiliation(s)
- Samuel JP Taylor
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Laura Bel Borja
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Flavie Soubigou
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Jack Houston
- Ludwig Institute for Cancer Research, San Diego BranchLa JollaUnited States
| | - Dhanya K Cheerambathur
- Wellcome Centre for Cell Biology & Institute of Cell Biology, School of Biological Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Federico Pelisch
- Centre for Gene Regulation and Expression, Sir James Black Centre, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
10
|
CCAR2 controls mitotic progression through spatiotemporal regulation of Aurora B. Cell Death Dis 2022; 13:534. [PMID: 35672287 PMCID: PMC9174277 DOI: 10.1038/s41419-022-04990-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 01/21/2023]
Abstract
CCAR2 (cell cycle and apoptosis regulator 2) is a multifaceted protein involved in cell survival and death following cytotoxic stress. However, little is known about the physiological functions of CCAR2 in regulating cell proliferation in the absence of external stimuli. The present study shows that CCAR2-deficient cells possess multilobulated nuclei, suggesting a defect in cell division. In particular, the duration of mitotic phase was perturbed. This disturbance of mitotic progression resulted from premature loss of cohesion with the centromere, and inactivation of the spindle assembly checkpoint during prometaphase and metaphase. It resulted in the formation of lagging chromosomes during anaphase, leading ultimately to the activation of the abscission checkpoint to halt cytokinesis. The CCAR2-dependent mitotic progression was related to spatiotemporal regulation of active Aurora B. In conclusion, the results suggest that CCAR2 governs mitotic events, including proper chromosome segregation and cytokinetic division, to maintain chromosomal stability.
Collapse
|
11
|
Park EA, Kim J, Shin MY, Park SJ. Kinesin-13, a Motor Protein, is Regulated by Polo-like Kinase in Giardia lamblia. THE KOREAN JOURNAL OF PARASITOLOGY 2022; 60:163-172. [PMID: 35772734 PMCID: PMC9256289 DOI: 10.3347/kjp.2022.60.3.163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
Kinesin-13 (Kin-13), a depolymerizer of microtubule (MT), has been known to affect the length of Giardia. Giardia Kin-13 (GlKin-13) was localized to axoneme, flagellar tips, and centrosomes, where phosphorylated forms of Giardia polo-like kinase (GlPLK) were distributed. We observed the interaction between GlKin-13 and GlPLK via co-immunoprecipitation using transgenic Giardia cells expressing Myc-tagged GlKin-13, hemagglutinin-tagged GlPLK, and in vitro-synthesized GlKin-13 and GlPLK proteins. In vitro-synthesized GlPLK was demonstrated to auto-phosphorylate and phosphorylate GlKin-13 upon incubation with [γ-32P]ATP. Morpholino-mediated depletion of both GlKin-13 and GlPLK caused an extension of flagella and a decreased volume of median bodies in Giardia trophozoites. Our results suggest that GlPLK plays a pertinent role in formation of flagella and median bodies by modulating MT depolymerizing activity of GlKin-13.
Collapse
|
12
|
Zhang Y, Song C, Wang L, Jiang H, Zhai Y, Wang Y, Fang J, Zhang G. Zombies Never Die: The Double Life Bub1 Lives in Mitosis. Front Cell Dev Biol 2022; 10:870745. [PMID: 35646932 PMCID: PMC9136299 DOI: 10.3389/fcell.2022.870745] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
When eukaryotic cells enter mitosis, dispersed chromosomes move to the cell center along microtubules to form a metaphase plate which facilitates the accurate chromosome segregation. Meanwhile, kinetochores not stably attached by microtubules activate the spindle assembly checkpoint and generate a wait signal to delay the initiation of anaphase. These events are highly coordinated. Disruption of the coordination will cause severe problems like chromosome gain or loss. Bub1, a conserved serine/threonine kinase, plays important roles in mitosis. After extensive studies in the last three decades, the role of Bub1 on checkpoint has achieved a comprehensive understanding; its role on chromosome alignment also starts to emerge. In this review, we summarize the latest development of Bub1 on supporting the two mitotic events. The essentiality of Bub1 in higher eukaryotic cells is also discussed. At the end, some undissolved questions are raised for future study.
Collapse
Affiliation(s)
- Yuqing Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunlin Song
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Wang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujing Zhai
- School of Public Health, Qingdao University, Qingdao, China
| | - Ying Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Jing Fang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Jing Fang, ; Gang Zhang,
| | - Gang Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Jing Fang, ; Gang Zhang,
| |
Collapse
|
13
|
Barbosa J, Sunkel CE, Conde C. The Role of Mitotic Kinases and the RZZ Complex in Kinetochore-Microtubule Attachments: Doing the Right Link. Front Cell Dev Biol 2022; 10:787294. [PMID: 35155423 PMCID: PMC8832123 DOI: 10.3389/fcell.2022.787294] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022] Open
Abstract
During mitosis, the interaction of kinetochores (KTs) with microtubules (MTs) drives chromosome congression to the spindle equator and supports the segregation of sister chromatids. Faithful genome partition critically relies on the ability of chromosomes to establish and maintain proper amphitelic end-on attachments, a configuration in which sister KTs are connected to robust MT fibers emanating from opposite spindle poles. Because the capture of spindle MTs by KTs is error prone, cells use mechanisms that sense and correct inaccurate KT-MT interactions before committing to segregate sister chromatids in anaphase. If left unresolved, these errors can result in the unequal distribution of chromosomes and lead to aneuploidy, a hallmark of cancer. In this review, we provide an overview of the molecular strategies that monitor the formation and fine-tuning of KT-MT attachments. We describe the complex network of proteins that operates at the KT-MT interface and discuss how AURORA B and PLK1 coordinate several concurrent events so that the stability of KT-MT attachments is precisely modulated throughout mitotic progression. We also outline updated knowledge on how the RZZ complex is regulated to ensure the formation of end-on attachments and the fidelity of mitosis.
Collapse
Affiliation(s)
- João Barbosa
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Claudio E. Sunkel
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
14
|
Gao Z, Jia H, Yu F, Guo H, Li B. KIF2C promotes the proliferation of hepatocellular carcinoma cells in vitro and in vivo. Exp Ther Med 2021; 22:1094. [PMID: 34504548 PMCID: PMC8383772 DOI: 10.3892/etm.2021.10528] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies with high mortality and morbidity rates. In recent years, HCC targeted therapy has gained increasing attention. Due to the heterogeneity and high metastasis of HCC, more effective therapeutic targets are needed. Kinesin family member 2C (KIF2C), also known as mitotic centromere-associated kinesin, is a microtubule-based motor protein which is involved in a variety of important cellular processes, such as mitosis. The effects of KIF2C on cancer progression and development have been widely studied; however, its potential effects on HCC remains unclear. In the present study, high expression of KIF2C in human HCC tissues was demonstrated using The Cancer Genome Atlas database and immunohistochemistry assays. KIF2C expression was associated with HCC prognosis, including overall survival and disease-free survival. KIF2C expression was also associated with clinical pathological characteristics including the number of tumor nodes (P=0.015) and tumor size (P=0.009). KIF2C knockdown inhibited the proliferation of HCC cells in vitro, confirmed by MTT and colony formation assays, and suppressed tumor growth in mice which was confirmed by a xenograft mouse model. Together, the results suggested that KIF2C may serve as a promising therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Zhenya Gao
- Department of Clinical Medicine, School of Medicine, Xuchang University, Xuchang, Henan 461000, P.R. China
| | - Huanxia Jia
- Department of Clinical Medicine, School of Medicine, Xuchang University, Xuchang, Henan 461000, P.R. China
| | - Fang Yu
- Department of Clinical Medicine, School of Medicine, Xuchang University, Xuchang, Henan 461000, P.R. China
| | - Hongfang Guo
- Department of Clinical Medicine, School of Medicine, Xuchang University, Xuchang, Henan 461000, P.R. China
| | - Baoyu Li
- Department of General Surgery, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
15
|
Rasamizafy SF, Delsert C, Rabeharivelo G, Cau J, Morin N, van Dijk J. Mitotic Acetylation of Microtubules Promotes Centrosomal PLK1 Recruitment and Is Required to Maintain Bipolar Spindle Homeostasis. Cells 2021; 10:1859. [PMID: 34440628 PMCID: PMC8394630 DOI: 10.3390/cells10081859] [Citation(s) in RCA: 6] [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: 07/06/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Tubulin post-translational modifications regulate microtubule properties and functions. Mitotic spindle microtubules are highly modified. While tubulin detyrosination promotes proper mitotic progression by recruiting specific microtubule-associated proteins motors, tubulin acetylation that occurs on specific microtubule subsets during mitosis is less well understood. Here, we show that siRNA-mediated depletion of the tubulin acetyltransferase ATAT1 in epithelial cells leads to a prolonged prometaphase arrest and the formation of monopolar spindles. This results from collapse of bipolar spindles, as previously described in cells deficient for the mitotic kinase PLK1. ATAT1-depleted mitotic cells have defective recruitment of PLK1 to centrosomes, defects in centrosome maturation and thus microtubule nucleation, as well as labile microtubule-kinetochore attachments. Spindle bipolarity could be restored, in the absence of ATAT1, by stabilizing microtubule plus-ends or by increasing PLK1 activity at centrosomes, demonstrating that the phenotype is not just a consequence of lack of K-fiber stability. We propose that microtubule acetylation of K-fibers is required for a recently evidenced cross talk between centrosomes and kinetochores.
Collapse
Affiliation(s)
- Sylvia Fenosoa Rasamizafy
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Claude Delsert
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
- Institut Français de Recherche pour l’Exploitation de la mer, L3AS, 34250 Palavas-les-Flots, France
| | - Gabriel Rabeharivelo
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Julien Cau
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- IGH, CNRS UMR 9002, 141, rue de la Cardonille, 34396 Montpellier, France
- Montpellier Rio Imaging, 34293 Montpellier, France
| | - Nathalie Morin
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Juliette van Dijk
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| |
Collapse
|
16
|
Kim J. In silico analysis of differentially expressed genesets in metastatic breast cancer identifies potential prognostic biomarkers. World J Surg Oncol 2021; 19:188. [PMID: 34172056 PMCID: PMC8235641 DOI: 10.1186/s12957-021-02301-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Identification of specific biological functions, pathways, and appropriate prognostic biomarkers is essential to accurately predict the clinical outcomes of and apply efficient treatment for breast cancer patients. METHODS To search for metastatic breast cancer-specific biological functions, pathways, and novel biomarkers in breast cancer, gene expression datasets of metastatic breast cancer were obtained from Oncomine, an online data mining platform. Over- and under-expressed genesets were collected and the differentially expressed genes were screened from four datasets with large sample sizes (N > 200). They were analyzed for gene ontology (GO), KEGG pathway, protein-protein interaction, and hub gene analyses using online bioinformatic tools (Enrichr, STRING, and Cytoscape) to find enriched functions and pathways in metastatic breast cancer. To identify novel prognostic biomarkers in breast cancer, differentially expressed genes were screened from the entire twelve datasets with any sample sizes and tested for expression correlation and survival analyses using online tools such as KM plotter and bc-GenExMiner. RESULTS Compared to non-metastatic breast cancer, 193 and 144 genes were differentially over- and under-expressed in metastatic breast cancer, respectively, and they were significantly enriched in regulating cell death, epidermal growth factor receptor signaling, and membrane and cytoskeletal structures according to the GO analyses. In addition, genes involved in progesterone- and estrogen-related signalings were enriched according to KEGG pathway analyses. Hub genes were identified via protein-protein interaction network analysis. Moreover, four differentially over-expressed (CCNA2, CENPN, DEPDC1, and TTK) and three differentially under-expressed genes (ABAT, LRIG1, and PGR) were further identified as novel biomarker candidate genes from the entire twelve datasets. Over- and under-expressed biomarker candidate genes were positively and negatively correlated with the aggressive and metastatic nature of breast cancer and were associated with poor and good prognosis of breast cancer patients, respectively. CONCLUSIONS Transcriptome datasets of metastatic breast cancer obtained from Oncomine allow the identification of metastatic breast cancer-specific biological functions, pathways, and novel biomarkers to predict clinical outcomes of breast cancer patients. Further functional studies are needed to warrant validation of their roles as functional tumor-promoting or tumor-suppressing genes.
Collapse
Affiliation(s)
- Jongchan Kim
- Department of Life Sciences, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
17
|
Hao X, Qiu Y, Cao L, Yang X, Zhou D, Liu J, Shi Z, Zhao S, Zhang J. Over-Expression of Centromere Protein U Participates in the Malignant Neoplastic Progression of Breast Cancer. Front Oncol 2021; 11:615427. [PMID: 33833984 PMCID: PMC8021899 DOI: 10.3389/fonc.2021.615427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/27/2021] [Indexed: 01/02/2023] Open
Abstract
The expression of Centromere Protein U (CENP-U) is closely related to tumor malignancy. Till now, the role of CENP-U in the malignant progression of breast cancer remains unclear. In this study, we found that CENP-U protein was highly expressed in the primary invasive breast cancer tissues compared to the paired adjacent histologically normal tissues and ductal carcinoma in situ (DCIS) tissues. After CENP-U was knocked down, the proliferation and colony-forming abilities of breast cancer cells were significantly suppressed, whereas the portion of apoptotic cells was increased. Meanwhile, the PI3K/AKT/NF-κB pathway was significantly inhibited. In vivo studies showed that, the inhibition of CENP-U repressed the tumor growth in orthotopic breast cancer models. Therefore, our study demonstrated that the CENP-U might act as an oncogene and promote breast cancer progression via activation of the PI3K/AKT/NF-κB pathway, which suggests a promising direction for targeting therapy in breast cancer.
Collapse
Affiliation(s)
- Xiaomeng Hao
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yufan Qiu
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Lixia Cao
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xiaonan Yang
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Dongdong Zhou
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jingjing Liu
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Zhendong Shi
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Shaorong Zhao
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jin Zhang
- Third Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
18
|
Singh P, Pesenti ME, Maffini S, Carmignani S, Hedtfeld M, Petrovic A, Srinivasamani A, Bange T, Musacchio A. BUB1 and CENP-U, Primed by CDK1, Are the Main PLK1 Kinetochore Receptors in Mitosis. Mol Cell 2021; 81:67-87.e9. [PMID: 33248027 PMCID: PMC7837267 DOI: 10.1016/j.molcel.2020.10.040] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 09/08/2020] [Accepted: 10/28/2020] [Indexed: 02/02/2023]
Abstract
Reflecting its pleiotropic functions, Polo-like kinase 1 (PLK1) localizes to various sub-cellular structures during mitosis. At kinetochores, PLK1 contributes to microtubule attachments and mitotic checkpoint signaling. Previous studies identified a wealth of potential PLK1 receptors at kinetochores, as well as requirements for various mitotic kinases, including BUB1, Aurora B, and PLK1 itself. Here, we combine ectopic localization, in vitro reconstitution, and kinetochore localization studies to demonstrate that most and likely all of the PLK1 is recruited through BUB1 in the outer kinetochore and centromeric protein U (CENP-U) in the inner kinetochore. BUB1 and CENP-U share a constellation of sequence motifs consisting of a putative PP2A-docking motif and two neighboring PLK1-docking sites, which, contingent on priming phosphorylation by cyclin-dependent kinase 1 and PLK1 itself, bind PLK1 and promote its dimerization. Our results rationalize previous observations and describe a unifying mechanism for recruitment of PLK1 to human kinetochores.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Marion E Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Stefano Maffini
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Sara Carmignani
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Marius Hedtfeld
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Arsen Petrovic
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Anupallavi Srinivasamani
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Tanja Bange
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany; Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Universitätsstrasse, 45141 Essen, Germany.
| |
Collapse
|
19
|
Movsisyan N, Pardo LA. Kv10.1 Regulates Microtubule Dynamics during Mitosis. Cancers (Basel) 2020; 12:cancers12092409. [PMID: 32854244 PMCID: PMC7564071 DOI: 10.3390/cancers12092409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022] Open
Abstract
Kv10.1 (potassium voltage-gated channel subfamily H member 1, known as EAG1 or Ether-à-go-go 1), is a voltage-gated potassium channel, prevailingly expressed in the central nervous system. The aberrant expression of Kv10.1 is detected in over 70% of all human tumor tissues and correlates with poorer prognosis. In peripheral tissues, Kv10.1 is expressed almost exclusively during the G2/M phase of the cell cycle and regulates its progression-downregulation of Kv10.1 extends the duration of the G2/M phase both in cancer and healthy cells. Here, using biochemical and imaging techniques, such as live-cell measurements of microtubule growth and of cytosolic calcium, we elucidate the mechanisms of Kv10.1-mediated regulation at the G2/M phase. We show that Kv10.1 has a dual effect on mitotic microtubule dynamics. Through the functional interaction with ORAI1 (calcium release-activated calcium channel protein 1), it modulates cytosolic calcium oscillations, thereby changing microtubule behavior. The inhibition of either Kv10.1 or ORAI1 stabilizes the microtubules. In contrast, the knockdown of Kv10.1 increases the dynamicity of mitotic microtubules, resulting in a stronger spindle assembly checkpoint, greater mitotic spindle angle, and a decrease in lagging chromosomes. Understanding of Kv10.1-mediated modulation of the microtubule architecture will help to comprehend how cancer tissue benefits from the presence of Kv10.1, and thereby increase the efficacy and safety of Kv10.1-directed therapeutic strategies.
Collapse
|
20
|
Yu R, Wu H, Ismail H, Du S, Cao J, Wang J, Ward T, Yang F, Gui P, Ali M, Chu L, Mo F, Wang Q, Chu Y, Zang J, Zhao Y, Ye M, Fang G, Chen PR, Dou Z, Gao X, Wang W, Liu X, Yao X. Methylation of PLK1 by SET7/9 ensures accurate kinetochore-microtubule dynamics. J Mol Cell Biol 2020; 12:462-476. [PMID: 31863092 PMCID: PMC7333475 DOI: 10.1093/jmcb/mjz107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023] Open
Abstract
Faithful segregation of mitotic chromosomes requires bi-orientation of sister chromatids, which relies on the sensing of correct attachments between spindle microtubules and kinetochores. Although the mechanisms underlying PLK1 activation have been extensively studied, the regulatory mechanisms that couple PLK1 activity to accurate chromosome segregation are not well understood. In particular, PLK1 is implicated in stabilizing kinetochore-microtubule attachments, but how kinetochore PLK1 activity is regulated to avoid hyperstabilized kinetochore-microtubules in mitosis remains elusive. Here, we show that kinetochore PLK1 kinase activity is modulated by SET7/9 via lysine methylation during early mitosis. The SET7/9-elicited dimethylation occurs at the Lys191 of PLK1, which tunes down its activity by limiting ATP utilization. Overexpression of the non-methylatable PLK1 mutant or chemical inhibition of SET7/9 methyltransferase activity resulted in mitotic arrest due to destabilized kinetochore-microtubule attachments. These data suggest that kinetochore PLK1 is essential for stable kinetochore-microtubule attachments and methylation by SET7/9 promotes dynamic kinetochore-microtubule attachments for accurate error correction. Our findings define a novel homeostatic regulation at the kinetochore that integrates protein phosphorylation and methylation with accurate chromosome segregation for maintenance of genomic stability.
Collapse
Affiliation(s)
- Ruoying Yu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Huihui Wu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shihao Du
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Jun Cao
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Jianyu Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Tarsha Ward
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Fengrui Yang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ping Gui
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Mahboob Ali
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- BUCM-MSM-USTC Joint Program on Global Health Equity, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lingluo Chu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Harvard Medical School, Boston, MA 02115, USA
| | - Fei Mo
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Harvard Medical School, Boston, MA 02115, USA
| | - Qi Wang
- Dalian Institute for Physical Chemistry, Dalian 116023, China
| | - Youjun Chu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Jianye Zang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Yun Zhao
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingliang Ye
- Dalian Institute for Physical Chemistry, Dalian 116023, China
| | - Guowei Fang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Peng R Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xinjiao Gao
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Wenwen Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
21
|
Li L, Huang K, Zhao H, Chen B, Ye Q, Yue J. CDK1-PLK1/SGOL2/ANLN pathway mediating abnormal cell division in cell cycle may be a critical process in hepatocellular carcinoma. Cell Cycle 2020; 19:1236-1252. [PMID: 32275843 PMCID: PMC7217380 DOI: 10.1080/15384101.2020.1749471] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/13/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022] Open
Abstract
This study aims to investigate the potential mechanisms and identify core biomarkers of Hepatocellular carcinoma (HCC). The profile GSE113850 was downloaded to analyze the differentially expressed genes. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction network analysis were used to reveal the main signal pathways of the differentially expressed genes (DEGs) and hub genes. The correlation between core gene expression and pathological stages, the disease-free survival analysis, the overall survival analysis were analyzed by Gene Expression Profiling Interactive Analysis. Furthermore, we reidentified the expression level of core genes of carcinoma tissues and para-carcinoma tissues from 14 HCC patients with real-time reverse transcription-polymerase chain reaction analysis (RT-PCR) and western blotting. After SK-Hep1 cell was treated with cyclin-dependent kinase 1 (CDK1) siRNA for 72 h, we detected the expression of the core genes and fluorescence-activated cell sorting analysis. A total of 378 DEGs were found. GO and KEGG analysis revealed that the DEGs were mainly enriched in the cell cycle. There were positive correlations among CDK1, polo-like kinase 1, shugoshin2 and anillin actin-binding protein. Moreover, the expression levels of four core genes were related to the HCC occurrence, pathological stages, and survivorship curve. The clinical HCC specimens verified the higher expression level of core genes by real-time RT-PCR. The transfection of siCDK1 in SK-Hep1 resulted in a disordered cell cycle. Furthermore, CDK1 knockdown suppressed the expression of PLK1, ANLN, and SGOL2. The CDK1-PLK1/SGOL2/ANLN pathway mediating abnormal cell division in the cell cycle might be a critical process in HCC.
Collapse
Affiliation(s)
- Ling Li
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, RP China
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei, RP China
| | - Kang Huang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, RP China
| | - Huijia Zhao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, RP China
| | - Binyao Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, RP China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, RP China
- The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, Hubei, RP China
| | - Jiang Yue
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei, RP China
| |
Collapse
|
22
|
Abstract
The conserved serine-threonine kinase, Cdc7, plays a crucial role in initiation of DNA replication by facilitating the assembly of an initiation complex. Cdc7 is expressed at a high level and exhibits significant kinase activity not only during S-phase but also during G2/M-phases. A conserved mitotic kinase, Aurora B, is activated during M-phase by association with INCENP, forming the chromosome passenger complex with Borealin and Survivin. We show that Cdc7 phosphorylates and stimulates Aurora B kinase activity in vitro. We identified threonine-236 as a critical phosphorylation site on Aurora B that could be a target of Cdc7 or could be an autophosphorylation site stimulated by Cdc7-mediated phosphorylation elsewhere. We found that threonines at both 232 (that has been identified as an autophosphorylation site) and 236 are essential for the kinase activity of Aurora B. Cdc7 down regulation or inhibition reduced Aurora B activity in vivo and led to retarded M-phase progression. SAC imposed by paclitaxel was dramatically reversed by Cdc7 inhibition, similar to the effect of Aurora B inhibition under the similar situation. Our data show that Cdc7 contributes to M-phase progression and to spindle assembly checkpoint most likely through Aurora B activation.
Collapse
|
23
|
Belsham HR, Friel CT. Identification of key residues that regulate the interaction of kinesins with microtubule ends. Cytoskeleton (Hoboken) 2019; 76:440-446. [PMID: 31574569 PMCID: PMC6899999 DOI: 10.1002/cm.21568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 11/16/2022]
Abstract
Kinesins are molecular motors that use energy derived from ATP turnover to walk along microtubules, or when at the microtubule end, regulate growth or shrinkage. All kinesins that regulate microtubule dynamics have long residence times at microtubule ends, whereas those that only walk have short end‐residence times. Here, we identify key amino acids involved in end binding by showing that when critical residues from Kinesin‐13, which depolymerises microtubules, are introduced into Kinesin‐1, a walking kinesin with no effect on microtubule dynamics, the end‐residence time is increased up to several‐fold. This indicates that the interface between the kinesin motor domain and the microtubule is malleable and can be tuned to favour either lattice or end binding.
Collapse
Affiliation(s)
- Hannah R Belsham
- School of Life Sciences, University of Nottingham, Medical School, QMC, Nottingham, NG7 2UH, United Kingdom
| | - Claire T Friel
- School of Life Sciences, University of Nottingham, Medical School, QMC, Nottingham, NG7 2UH, United Kingdom
| |
Collapse
|
24
|
Gan H, Lin L, Hu N, Yang Y, Gao Y, Pei Y, Chen K, Sun B. KIF2C exerts an oncogenic role in nonsmall cell lung cancer and is negatively regulated by miR‐325‐3p. Cell Biochem Funct 2019; 37:424-431. [PMID: 31328811 DOI: 10.1002/cbf.3420] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/11/2019] [Accepted: 06/05/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Huizhu Gan
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lin Lin
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nanjun Hu
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Yang
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yu Gao
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yu Pei
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kang Chen
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Butong Sun
- Department of Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Joukov V, De Nicolo A. The Centrosome and the Primary Cilium: The Yin and Yang of a Hybrid Organelle. Cells 2019; 8:E701. [PMID: 31295970 PMCID: PMC6678760 DOI: 10.3390/cells8070701] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 12/27/2022] Open
Abstract
Centrosomes and primary cilia are usually considered as distinct organelles, although both are assembled with the same evolutionary conserved, microtubule-based templates, the centrioles. Centrosomes serve as major microtubule- and actin cytoskeleton-organizing centers and are involved in a variety of intracellular processes, whereas primary cilia receive and transduce environmental signals to elicit cellular and organismal responses. Understanding the functional relationship between centrosomes and primary cilia is important because defects in both structures have been implicated in various diseases, including cancer. Here, we discuss evidence that the animal centrosome evolved, with the transition to complex multicellularity, as a hybrid organelle comprised of the two distinct, but intertwined, structural-functional modules: the centriole/primary cilium module and the pericentriolar material/centrosome module. The evolution of the former module may have been caused by the expanding cellular diversification and intercommunication, whereas that of the latter module may have been driven by the increasing complexity of mitosis and the requirement for maintaining cell polarity, individuation, and adhesion. Through its unique ability to serve both as a plasma membrane-associated primary cilium organizer and a juxtanuclear microtubule-organizing center, the animal centrosome has become an ideal integrator of extracellular and intracellular signals with the cytoskeleton and a switch between the non-cell autonomous and the cell-autonomous signaling modes. In light of this hypothesis, we discuss centrosome dynamics during cell proliferation, migration, and differentiation and propose a model of centrosome-driven microtubule assembly in mitotic and interphase cells. In addition, we outline the evolutionary benefits of the animal centrosome and highlight the hierarchy and modularity of the centrosome biogenesis networks.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, 197758 Saint-Petersburg, Russia.
| | | |
Collapse
|
26
|
Vallardi G, Cordeiro MH, Saurin AT. A Kinase-Phosphatase Network that Regulates Kinetochore-Microtubule Attachments and the SAC. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:457-484. [PMID: 28840249 DOI: 10.1007/978-3-319-58592-5_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The KMN network (for KNL1, MIS12 and NDC80 complexes) is a hub for signalling at the outer kinetochore. It integrates the activities of two kinases (MPS1 and Aurora B) and two phosphatases (PP1 and PP2A-B56) to regulate kinetochore-microtubule attachments and the spindle assembly checkpoint (SAC). We will first discuss each of these enzymes separately, to describe how they are regulated at kinetochores and why this is important for their primary function in controlling either microtubule attachments or the SAC. We will then discuss why inhibiting any one of them individually produces secondary effects on all the others. This cross-talk may help to explain why all enzymes have been linked to both processes, even though the direct evidence suggests they each control only one. This chapter therefore describes how a network of kinases and phosphatases work together to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Marilia Henriques Cordeiro
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian Thomas Saurin
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
27
|
Bai Y, Xiong L, Zhu M, Yang Z, Zhao J, Tang H. Co-expression network analysis identified KIF2C in association with progression and prognosis in lung adenocarcinoma. Cancer Biomark 2019; 24:371-382. [DOI: 10.3233/cbm-181512] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
Moura M, Conde C. Phosphatases in Mitosis: Roles and Regulation. Biomolecules 2019; 9:E55. [PMID: 30736436 PMCID: PMC6406801 DOI: 10.3390/biom9020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Mitosis requires extensive rearrangement of cellular architecture and of subcellular structures so that replicated chromosomes can bind correctly to spindle microtubules and segregate towards opposite poles. This process originates two new daughter nuclei with equal genetic content and relies on highly-dynamic and tightly regulated phosphorylation of numerous cell cycle proteins. A burst in protein phosphorylation orchestrated by several conserved kinases occurs as cells go into and progress through mitosis. The opposing dephosphorylation events are catalyzed by a small set of protein phosphatases, whose importance for the accuracy of mitosis is becoming increasingly appreciated. This review will focus on the established and emerging roles of mitotic phosphatases, describe their structural and biochemical properties, and discuss recent advances in understanding the regulation of phosphatase activity and function.
Collapse
Affiliation(s)
- Margarida Moura
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| | - Carlos Conde
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
29
|
Oser MG, Fonseca R, Chakraborty AA, Brough R, Spektor A, Jennings RB, Flaifel A, Novak JS, Gulati A, Buss E, Younger ST, McBrayer SK, Cowley GS, Bonal DM, Nguyen QD, Brulle-Soumare L, Taylor P, Cairo S, Ryan CJ, Pease EJ, Maratea K, Travers J, Root DE, Signoretti S, Pellman D, Ashton S, Lord CJ, Barry ST, Kaelin WG. Cells Lacking the RB1 Tumor Suppressor Gene Are Hyperdependent on Aurora B Kinase for Survival. Cancer Discov 2019; 9:230-247. [PMID: 30373918 PMCID: PMC6368871 DOI: 10.1158/2159-8290.cd-18-0389] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/22/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022]
Abstract
Small cell lung cancer (SCLC) accounts for 15% of lung cancers and is almost always linked to inactivating RB1 and TP53 mutations. SCLC frequently responds, albeit briefly, to chemotherapy. The canonical function of the RB1 gene product RB1 is to repress the E2F transcription factor family. RB1 also plays both E2F-dependent and E2F-independent mitotic roles. We performed a synthetic lethal CRISPR/Cas9 screen in an RB1 -/- SCLC cell line that conditionally expresses RB1 to identify dependencies that are caused by RB1 loss and discovered that RB1 -/- SCLC cell lines are hyperdependent on multiple proteins linked to chromosomal segregation, including Aurora B kinase. Moreover, we show that an Aurora B kinase inhibitor is efficacious in multiple preclinical SCLC models at concentrations that are well tolerated in mice. These results suggest that RB1 loss is a predictive biomarker for sensitivity to Aurora B kinase inhibitors in SCLC and perhaps other RB1 -/- cancers. SIGNIFICANCE: SCLC is rarely associated with actionable protooncogene mutations. We did a CRISPR/Cas9-based screen that showed that RB1 -/- SCLC are hyperdependent on AURKB, likely because both genes control mitotic fidelity, and confirmed that Aurora B kinase inhibitors are efficacious against RB1 -/- SCLC tumors in mice at nontoxic doses.See related commentary by Dick and Li, p. 169.This article is highlighted in the In This Issue feature, p. 151.
Collapse
Affiliation(s)
- Matthew G Oser
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raquel Fonseca
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Abhishek A Chakraborty
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rachel Brough
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Alexander Spektor
- Howard Hughes Medical Institute, Chevy Chase, Maryland
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rebecca B Jennings
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Abdallah Flaifel
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jesse S Novak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aditi Gulati
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Elizabeth Buss
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Scott T Younger
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Samuel K McBrayer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Dennis M Bonal
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Paula Taylor
- IMED Oncology, AstraZeneca, Cheshire, United Kingdom
| | | | - Colm J Ryan
- Systems Biology Ireland, University College Dublin, Dublin, Republic of Ireland
| | | | - Kim Maratea
- IMED Drug Safety and Metabolism, AstraZeneca, Boston, Massachusetts
| | - Jon Travers
- IMED Oncology, AstraZeneca, Cheshire, United Kingdom
| | - David E Root
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Pellman
- Howard Hughes Medical Institute, Chevy Chase, Maryland
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Susan Ashton
- IMED Oncology, AstraZeneca, Cheshire, United Kingdom
| | - Christopher J Lord
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Simon T Barry
- IMED Oncology, AstraZeneca, Cambridge, United Kingdom
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
30
|
Parts list for a microtubule depolymerising kinesin. Biochem Soc Trans 2018; 46:1665-1672. [PMID: 30467119 PMCID: PMC6299235 DOI: 10.1042/bst20180350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
The Kinesin superfamily is a large group of molecular motors that use the turnover of ATP to regulate their interaction with the microtubule cytoskeleton. The coupled relationship between nucleotide turnover and microtubule binding is harnessed in various ways by these motors allowing them to carry out a variety of cellular functions. The Kinesin-13 family is a group of specialist microtubule depolymerising motors. Members of this family use their microtubule destabilising activity to regulate processes such as chromosome segregation, maintenance of cilia and neuronal development. Here, we describe the current understanding of the structure of this family of kinesins and the role different parts of these proteins play in their microtubule depolymerisation activity and in the wider function of this family of kinesins.
Collapse
|
31
|
Su CC, Chen NC, Chyau CC, Tseng HC, Chou FP. Induction of Mitotic Catastrophe via Inhibition of Aurora B by Ionizing Radiation With Additive of Mulberry Water Extract in Human Bladder Cancer Cells. Integr Cancer Ther 2018; 18:1534735418808586. [PMID: 30428726 PMCID: PMC7290068 DOI: 10.1177/1534735418808586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mulberry fruit water extract (MWE) has been reported to synergistically enhance the cytotoxic effect of paclitaxel by promoting mitotic catastrophe to induce apoptosis in bladder cancer cells in our previous work. The aim of this study was to evaluate and to mechanistically explore the effects of MWE on bladder cancer responses to ionizing radiation (IR) by treating TSGH 8301 bladder carcinoma cells with MWE after exposing to IR. The results of MTT assay showed a synergistic cytotoxicity of IR with the co-treatment of MWE (IR/MWE) by inducing G2/M phase arrest as demonstrated by flow cytometry analysis in TSGH 8301, HT1367 and HT1197 bladder carcinoma cells lines. The IR/MWE-treated cells expressed increased levels of the G2/M phase arrest-related proteins cdc2/cyclin B1 and displayed giant multinucleated morphology, a typical characteristic of mitotic catastrophe. Immunofluorescent confocal microscopy revealed that the combined strategy inhibited Aurora B phosphorylation through Ras/Raf/MEK/ERK signaling cascade as demonstrated by Western blotting analysis. IR/MWE also caused an inhibitory effect on Plk1 and the subsequent downstream regulator RhoA repression and Cep55 induction, which would influence cell cycle progression in the early steps of cytokinesis. A profound tumor growth suppression and inactivation of Aurora B activity in the tumor tissues by IR/MWE treatment were confirmed in the TSGH 8301 xenograft model in vivo. These data demonstrated that MWE could be an effective auxiliary to synergize with radiation on the anticancer efficacy by promoting mitotic catastrophe through inhibition of Aurora B, providing a novel and effective therapeutic option for bladder cancer management.
Collapse
Affiliation(s)
- Chia-Cheng Su
- 1 Chi Mei Medical Center, Tainan City, Taiwan.,2 Chia Nan University of Pharmacy & Science, Tainan City, Taiwan
| | | | | | - Hsien-Chun Tseng
- 3 Chung Shan Medical University, Taichung City, Taiwan.,5 Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Fen-Pi Chou
- 3 Chung Shan Medical University, Taichung City, Taiwan.,5 Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
32
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
33
|
Kumar A, Srivastava P, Sirisena P, Dubey SK, Kumar R, Shrinet J, Sunil S. Mosquito Innate Immunity. INSECTS 2018; 9:insects9030095. [PMID: 30096752 PMCID: PMC6165528 DOI: 10.3390/insects9030095] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
Mosquitoes live under the endless threat of infections from different kinds of pathogens such as bacteria, parasites, and viruses. The mosquito defends itself by employing both physical and physiological barriers that resist the entry of the pathogen and the subsequent establishment of the pathogen within the mosquito. However, if the pathogen does gain entry into the insect, the insect mounts a vigorous innate cellular and humoral immune response against the pathogen, thereby limiting the pathogen's propagation to nonpathogenic levels. This happens through three major mechanisms: phagocytosis, melanization, and lysis. During these processes, various signaling pathways that engage intense mosquito⁻pathogen interactions are activated. A critical overview of the mosquito immune system and latest information about the interaction between mosquitoes and pathogens are provided in this review. The conserved, innate immune pathways and specific anti-pathogenic strategies in mosquito midgut, hemolymph, salivary gland, and neural tissues for the control of pathogen propagation are discussed in detail.
Collapse
Affiliation(s)
- Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Priyanshu Srivastava
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Pdnn Sirisena
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sunil Kumar Dubey
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Ramesh Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Jatin Shrinet
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| |
Collapse
|
34
|
Saurin AT. Kinase and Phosphatase Cross-Talk at the Kinetochore. Front Cell Dev Biol 2018; 6:62. [PMID: 29971233 PMCID: PMC6018199 DOI: 10.3389/fcell.2018.00062] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023] Open
Abstract
Multiple kinases and phosphatases act on the kinetochore to control chromosome segregation: Aurora B, Mps1, Bub1, Plk1, Cdk1, PP1, and PP2A-B56, have all been shown to regulate both kinetochore-microtubule attachments and the spindle assembly checkpoint. Given that so many kinases and phosphatases converge onto two key mitotic processes, it is perhaps not surprising to learn that they are, quite literally, entangled in cross-talk. Inhibition of any one of these enzymes produces secondary effects on all the others, which results in a complicated picture that is very difficult to interpret. This review aims to clarify this picture by first collating the direct effects of each enzyme into one overarching schematic of regulation at the Knl1/Mis12/Ndc80 (KMN) network (a major signaling hub at the outer kinetochore). This schematic will then be used to discuss the implications of the cross-talk that connects these enzymes; both in terms of why it may be needed to produce the right type of kinetochore signals and why it nevertheless complicates our interpretations about which enzymes control what processes. Finally, some general experimental approaches will be discussed that could help to characterize kinetochore signaling by dissociating the direct from indirect effect of kinase or phosphatase inhibition in vivo. Together, this review should provide a framework to help understand how a network of kinases and phosphatases cooperate to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Adrian T. Saurin
- Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
35
|
Mchedlishvili N, Matthews HK, Corrigan A, Baum B. Two-step interphase microtubule disassembly aids spindle morphogenesis. BMC Biol 2018; 16:14. [PMID: 29361957 PMCID: PMC5778756 DOI: 10.1186/s12915-017-0478-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Entry into mitosis triggers profound changes in cell shape and cytoskeletal organisation. Here, by studying microtubule remodelling in human flat mitotic cells, we identify a two-step process of interphase microtubule disassembly. RESULTS First, a microtubule-stabilising protein, Ensconsin/MAP7, is inactivated in prophase as a consequence of its phosphorylation downstream of Cdk1/cyclin B. This leads to a reduction in interphase microtubule stability that may help to fuel the growth of centrosomally nucleated microtubules. The peripheral interphase microtubules that remain are then rapidly lost as the concentration of tubulin heterodimers falls following dissolution of the nuclear compartment boundary. Finally, we show that a failure to destabilise microtubules in prophase leads to the formation of microtubule clumps, which interfere with spindle assembly. CONCLUSIONS This analysis highlights the importance of the step-wise remodelling of the microtubule cytoskeleton and the significance of permeabilisation of the nuclear envelope in coordinating the changes in cellular organisation and biochemistry that accompany mitotic entry.
Collapse
Affiliation(s)
- Nunu Mchedlishvili
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Helen K Matthews
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Adam Corrigan
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Buzz Baum
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
36
|
Lee M, Kim IS, Park KC, Kim JS, Baek SH, Kim KI. Mitosis-specific phosphorylation of Mis18α by Aurora B kinase enhances kinetochore recruitment of polo-like kinase 1. Oncotarget 2017; 9:1563-1576. [PMID: 29416714 PMCID: PMC5788582 DOI: 10.18632/oncotarget.22707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/28/2017] [Indexed: 01/11/2023] Open
Abstract
Mis18α, a component of Mis18 complex comprising of Mis18α, Mis18β, and M18BP1, is known to localize at the centromere from late telophase to early G1 phase and plays a priming role in CENP-A deposition. Although its role in CENP-A deposition is well established, the other function of Mis18α remains unknown. Here, we elucidate a new function of Mis18α that is critical for the proper progression of cell cycle independent of its role in CENP-A deposition. We find that Aurora B kinase phosphorylates Mis18α during mitosis not affecting neither centromere localization of Mis18 complex nor centromere loading of CENP-A. However, the replacement of endogenous Mis18α by phosphorylation-defective mutant causes mitotic defects including micronuclei formation, chromosome misalignment, and chromosomal bridges. Together, our data demonstrate that Aurora B kinase-mediated mitotic phosphorylation of Mis18α is a crucial event for faithful cell cycle progression through the enhanced recruitment of polo-like kinase 1 to the kinetochore.
Collapse
Affiliation(s)
- Minkyoung Lee
- Creative Research Initiatives Center for Chromatin Dynamics, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Ik Soo Kim
- Creative Research Initiatives Center for Chromatin Dynamics, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Koog Chan Park
- Department of Biological Sciences, Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jong-Seo Kim
- Center for RNA Research, Institute for Basic Science, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Chromatin Dynamics, Department of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Keun Il Kim
- Department of Biological Sciences, Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul 04310, South Korea
| |
Collapse
|
37
|
Kinetochore-microtubule interactions in chromosome segregation: lessons from yeast and mammalian cells. Biochem J 2017; 474:3559-3577. [PMID: 29046344 DOI: 10.1042/bcj20170518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023]
Abstract
Chromosome congression and segregation require robust yet dynamic attachment of the kinetochore with the spindle microtubules. Force generated at the kinetochore-microtubule interface plays a vital role to drive the attachment, as it is required to move chromosomes and to provide signal to sense correct attachments. To understand the mechanisms underlying these processes, it is critical to describe how the force is generated and how the molecules at the kinetochore-microtubule interface are organized and assembled to withstand the force and respond to it. Research in the past few years or so has revealed interesting insights into the structural organization and architecture of kinetochore proteins that couple kinetochore attachment to the spindle microtubules. Interestingly, despite diversities in the molecular players and their modes of action, there appears to be architectural similarity of the kinetochore-coupling machines in lower to higher eukaryotes. The present review focuses on the most recent advances in understanding of the molecular and structural aspects of kinetochore-microtubule interaction based on the studies in yeast and vertebrate cells.
Collapse
|
38
|
Gutteridge REA, Singh CK, Ndiaye MA, Ahmad N. Targeted knockdown of polo-like kinase 1 alters metabolic regulation in melanoma. Cancer Lett 2017; 394:13-21. [PMID: 28235541 PMCID: PMC5415376 DOI: 10.1016/j.canlet.2017.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 01/05/2023]
Abstract
A limited number of studies have indicated an association of the mitotic kinase polo-like kinase 1 (PLK1) and cellular metabolism. Here, employing an inducible RNA interference approach in A375 melanoma cells coupled with a PCR array and multiple validation approaches, we demonstrated that PLK1 alters a number of genes associated with cellular metabolism. PLK1 knockdown resulted in a significant downregulation of IDH1, PDP2 and PCK1 and upregulation of FBP1. Ingenuity Pathway Analysis (IPA) identified that 1) glycolysis and the pentose phosphate pathway are major canonical pathways associated with PLK1, and 2) PLK1 inhibition-modulated genes were largely associated with cellular proliferation, with FBP1 being the key modulator. Further, BI 6727-mediated inhibition of PLK1 caused a decrease in PCK1 and increase in FBP1 in A375 melanoma cell implanted xenografts in vivo. Furthermore, an inverse correlation between PLK1 and FBP1 was found in melanoma cells, with FBP1 expression significantly downregulated in a panel of melanoma cells. In addition, BI 6727 treatment resulted in an upregulation in FBP1 in A375, Hs294T and G361 melanoma cells. Overall, our study suggests that PLK1 may be an important regulator of metabolism maintenance in melanoma cells.
Collapse
Affiliation(s)
| | - Chandra K Singh
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; William S. Middleton VA Medical Center, 2500 Overlook Terrace, Madison, WI 53705, USA.
| |
Collapse
|
39
|
Awad MM, Chu QSC, Gandhi L, Stephenson JJ, Govindan R, Bradford DS, Bonomi PD, Ellison DM, Eaton KD, Fritsch H, Munzert G, Johnson BE, Socinski MA. An open-label, phase II study of the polo-like kinase-1 (Plk-1) inhibitor, BI 2536, in patients with relapsed small cell lung cancer (SCLC). Lung Cancer 2017; 104:126-130. [PMID: 28212994 DOI: 10.1016/j.lungcan.2016.12.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/20/2016] [Accepted: 12/26/2016] [Indexed: 12/01/2022]
Abstract
OBJECTIVES This phase II, open-label study was designed to evaluate the response rate to the polo-like kinase 1 (Plk-1) inhibitor BI 2536 in patients with sensitive-relapsed small cell lung cancer (SCLC). Secondary endpoints included progression-free survival (PFS), overall survival (OS), duration of response, and safety. MATERIALS AND METHODS Patients were treated with the recommended phase II dose of 200mg of BI 2536 intravenously every 21days. This was a two-stage design with an early stopping rule in place if responses were not seen in at least 2 of the first 18 enrolled patients. RESULTS AND CONCLUSION Twenty-three patients were enrolled in the study and 21 patients were evaluable for response. No responses were observed and all 23 patients have progressed. The median PFS was 1.4 months. Treatment was generally well tolerated and the most frequent adverse events were neutropenia, fatigue, nausea, vomiting, and constipation. BI 2536 is not effective in the treatment of sensitive relapsed SCLC. The criteria for expanding the trial to the second stage were not achieved, and the study was terminated for a lack of efficacy.
Collapse
Affiliation(s)
- Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Quincy S-C Chu
- Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Leena Gandhi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - David M Ellison
- Charleston Hematology Oncology Associates, Charleston, SC, USA
| | | | | | | | - Bruce E Johnson
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
40
|
Qin B, Cao D, Wu H, Mo F, Shao H, Chu J, Powell M, Aikhionbare F, Wang D, Fu C, He P, Pan W, Wang W, Liu X, Yao X. Phosphorylation of SKAP by GSK3β ensures chromosome segregation by a temporal inhibition of Kif2b activity. Sci Rep 2016; 6:38791. [PMID: 27982129 PMCID: PMC5159797 DOI: 10.1038/srep38791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/11/2016] [Indexed: 12/28/2022] Open
Abstract
Chromosome segregation in mitosis is orchestrated by the dynamic interactions between the kinetochore and spindle microtubules. Our recent study shows SKAP is an EB1-dependent, microtubule plus-end tracking protein essential for kinetochore oscillations during mitosis. Here we show that phosphorylation of SKAP by GSK3β regulates Kif2b depolymerase activity by competing Kif2b for microtubule plus-end binding. SKAP is a bona fide substrate of GSK3β in vitro and the phosphorylation is essential for an accurate kinetochore-microtubule attachment in cells. The GSK3β-elicited phosphorylation sites were mapped by mass spectrometry and the phosphomimetic mutant of SKAP can rescue the phenotype of chromosome missegregation in SKAP-suppressed cells. Importantly, GSK3β-elicited phosphorylation promotes SKAP binding to Kif2b to regulate its depolymerase activity at the microtubule plus-ends. Based on those findings, we reason that GSK3β-SKAP-Kif2b signaling axis constitutes a dynamic link between spindle microtubule plus-ends and mitotic chromosomes to achieve faithful cell division.
Collapse
Affiliation(s)
- Bo Qin
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China
| | - Dan Cao
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China
| | - Huihui Wu
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China
| | - Fei Mo
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China
| | - Hengyi Shao
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China
| | - Jane Chu
- Molecular Imaging Center, Atlanta Clinical &Translational Science Institute, Atlanta, GA 30310
| | - Michael Powell
- Molecular Imaging Center, Atlanta Clinical &Translational Science Institute, Atlanta, GA 30310
| | - Felix Aikhionbare
- Molecular Imaging Center, Atlanta Clinical &Translational Science Institute, Atlanta, GA 30310
| | - Dongmei Wang
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China.,Center of Excellence on Molecular Cell Sciences, Chinese Academy of Sciences, Hefei 230026, China
| | - Chuanhai Fu
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China.,Center of Excellence on Molecular Cell Sciences, Chinese Academy of Sciences, Hefei 230026, China
| | - Ping He
- Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Weijun Pan
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenwen Wang
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China.,Center of Excellence on Molecular Cell Sciences, Chinese Academy of Sciences, Hefei 230026, China
| | - Xing Liu
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China.,Center of Excellence on Molecular Cell Sciences, Chinese Academy of Sciences, Hefei 230026, China
| | - Xuebiao Yao
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science &Technology of China, Hefei 230027, China.,Center of Excellence on Molecular Cell Sciences, Chinese Academy of Sciences, Hefei 230026, China
| |
Collapse
|
41
|
Duan H, Wang C, Wang M, Gao X, Yan M, Akram S, Peng W, Zou H, Wang D, Zhou J, Chu Y, Dou Z, Barrett G, Green HN, Wang F, Tian R, He P, Wang W, Liu X, Yao X. Phosphorylation of PP1 Regulator Sds22 by PLK1 Ensures Accurate Chromosome Segregation. J Biol Chem 2016; 291:21123-21136. [PMID: 27557660 PMCID: PMC5076521 DOI: 10.1074/jbc.m116.745372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/22/2016] [Indexed: 11/06/2022] Open
Abstract
During cell division, accurate chromosome segregation is tightly regulated by Polo-like kinase 1 (PLK1) and opposing activities of Aurora B kinase and protein phosphatase 1 (PP1). However, the regulatory mechanisms underlying the aforementioned hierarchical signaling cascade during mitotic chromosome segregation have remained elusive. Sds22 is a conserved regulator of PP1 activity, but how it regulates PP1 activity in space and time during mitosis remains elusive. Here we show that Sds22 is a novel and cognate substrate of PLK1 in mitosis, and the phosphorylation of Sds22 by PLK1 elicited an inhibition of PP1-mediated dephosphorylation of Aurora B at threonine 232 (Thr232) in a dose-dependent manner. Overexpression of a phosphomimetic mutant of Sds22 causes a dramatic increase in mitotic delay, whereas overexpression of a non-phosphorylatable mutant of Sds22 results in mitotic arrest. Mechanistically, the phosphorylation of Sds22 by PLK1 strengthens the binding of Sds22 to PP1 and inhibits the dephosphorylation of Thr232 of Aurora B to ensure a robust, error-free metaphase-anaphase transition. These findings delineate a conserved signaling hierarchy that orchestrates dynamic protein phosphorylation and dephosphorylation of critical mitotic regulators during chromosome segregation to guard chromosome stability.
Collapse
Affiliation(s)
- Hequan Duan
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China, the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310
| | - Chunli Wang
- the National Chromatographic Research and Analysis Center, Chinesse Academy of Sciences, Dalian 116023, China
| | - Ming Wang
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Xinjiao Gao
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Maomao Yan
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Saima Akram
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Wei Peng
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Hanfa Zou
- the National Chromatographic Research and Analysis Center, Chinesse Academy of Sciences, Dalian 116023, China
| | - Dong Wang
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Jiajia Zhou
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Youjun Chu
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China, the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310
| | - Zhen Dou
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China
| | - Gregory Barrett
- the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310
| | - Hadiyah-Nicole Green
- the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310
| | - Fangjun Wang
- the National Chromatographic Research and Analysis Center, Chinesse Academy of Sciences, Dalian 116023, China
| | - Ruijun Tian
- the Guangzhou Women and Children's Medical Center, Guangzhou 510623, China, and the Center of Molecular Proteomics, South University of Science & Technology of China, Shenzhen 518055, China
| | - Ping He
- the Guangzhou Women and Children's Medical Center, Guangzhou 510623, China, and the Center of Molecular Proteomics, South University of Science & Technology of China, Shenzhen 518055, China
| | - Wenwen Wang
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China, the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310,
| | - Xing Liu
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China, the Morehouse School of Medicine and Atlanta Clinical & Translational Science Institute, Atlanta, Georgia 30310,
| | - Xuebiao Yao
- From the Anhui Key Laboratory for Cellular Dynamics & Chemical Biology, MOE Collaborative Innovation Center of Chemistry for Life Sciences, University of Science & Technology of China, Hefei 230027, China,
| |
Collapse
|
42
|
Gutteridge REA, Ndiaye MA, Liu X, Ahmad N. Plk1 Inhibitors in Cancer Therapy: From Laboratory to Clinics. Mol Cancer Ther 2016; 15:1427-35. [PMID: 27330107 PMCID: PMC4936921 DOI: 10.1158/1535-7163.mct-15-0897] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/06/2016] [Indexed: 01/06/2023]
Abstract
Polo-like kinase 1 (Plk1) overexpression has been shown to occur in a wide range of tumors, prompting research and development of Plk1 inhibitors as a means of cancer treatment. This review discusses recent advances in the development of Plk1 inhibitors for cancer management. Plk1 inhibition has been shown to cause mitotic block and apoptosis of cells with higher mitotic index and therefore higher Plk1 expression. The potential of Plk1 inhibitors as cancer therapeutics has been widely investigated. However, a complete understanding of Plk1 biology/mechanism is yet to be fully achieved. Resistance to certain chemotherapeutic drugs has been linked to Plk1 overexpression, and Plk1-mediated mitotic events such as microtubule rearrangement have been found to reduce the efficacy of chemotherapeutic agents. The Plk1 inhibitor volasertib has shown considerable promise in clinical studies, having reached phase III trials. However, preclinical success with Plk1 inhibitors has not translated well into clinical success. In our view, combined therapies targeting other relevant pathways together with Plk1 may be vital to combat issues observed with monotherapy, especially resistance. In addition, research should also be directed toward understanding the mechanisms of Plk1 and designing additional next generations of specific, potent Plk1 inhibitors to target cancer. Mol Cancer Ther; 15(7); 1427-35. ©2016 AACR.
Collapse
Affiliation(s)
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin. William S. Middleton Memorial VA Hospital, Madison, Wisconsin.
| |
Collapse
|
43
|
Ritter A, Kreis NN, Louwen F, Wordeman L, Yuan J. Molecular insight into the regulation and function of MCAK. Crit Rev Biochem Mol Biol 2016; 51:228-45. [DOI: 10.1080/10409238.2016.1178705] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
44
|
Zong H, Carnes SK, Moe C, Walczak CE, Ems-McClung SC. The far C-terminus of MCAK regulates its conformation and spindle pole focusing. Mol Biol Cell 2016; 27:1451-64. [PMID: 26941326 PMCID: PMC4850033 DOI: 10.1091/mbc.e15-10-0699] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/25/2016] [Indexed: 12/17/2022] Open
Abstract
Spatial regulation of microtubule dynamics is critical for proper spindle assembly. The far C-terminus of the microtubule-depolymerizing kinesin-13 MCAK regulates MCAK localization at spindle poles, which is needed for proper pole focusing. To ensure proper spindle assembly, microtubule (MT) dynamics needs to be spatially regulated within the cell. The kinesin-13 MCAK is a potent MT depolymerase with a complex subcellular localization, yet how MCAK spatial regulation contributes to spindle assembly is not understood. Here we show that the far C-terminus of MCAK plays a critical role in regulating MCAK conformation, subspindle localization, and spindle assembly in Xenopus egg extracts. Alteration of MCAK conformation by the point mutation E715A/E716A in the far C-terminus increased MCAK targeting to the poles and reduced MT lifetimes, which induced spindles with unfocused poles. These effects were phenocopied by the Aurora A phosphomimetic mutation, S719E. Furthermore, addition of the kinesin-14 XCTK2 to spindle assembly reactions rescued the unfocused-pole phenotype. Collectively our work shows how the regional targeting of MCAK regulates MT dynamics, highlighting the idea that multiple phosphorylation pathways of MCAK cooperate to spatially control MT dynamics to maintain spindle architecture.
Collapse
Affiliation(s)
- Hailing Zong
- Department of Biology, Indiana University, Bloomington, IN 47405
| | | | - Christina Moe
- Department of Biology, Indiana University, Bloomington, IN 47405
| | - Claire E Walczak
- Medical Sciences Program, Indiana University, Bloomington, IN 47405
| | | |
Collapse
|
45
|
Eichenlaub-Ritter U. Microtubule dynamics and tumor invasion involving MCAK. Cell Cycle 2015; 14:3353. [PMID: 26375511 PMCID: PMC4825562 DOI: 10.1080/15384101.2015.1093813] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/10/2015] [Indexed: 01/09/2023] Open
|