1
|
Yildiz A. Mechanism and regulation of kinesin motors. Nat Rev Mol Cell Biol 2025; 26:86-103. [PMID: 39394463 DOI: 10.1038/s41580-024-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
Kinesins are a diverse superfamily of microtubule-based motors that perform fundamental roles in intracellular transport, cytoskeletal dynamics and cell division. These motors share a characteristic motor domain that powers unidirectional motility and force generation along microtubules, and they possess unique tail domains that recruit accessory proteins and facilitate oligomerization, regulation and cargo recognition. The location, direction and timing of kinesin-driven processes are tightly regulated by various cofactors, adaptors, microtubule tracks and microtubule-associated proteins. This Review focuses on recent structural and functional studies that reveal how members of the kinesin superfamily use the energy of ATP hydrolysis to transport cargoes, depolymerize microtubules and regulate microtubule dynamics. I also survey how accessory proteins and post-translational modifications regulate the autoinhibition, cargo binding and motility of some of the best-studied kinesins. Despite much progress, the mechanism and regulation of kinesins are still emerging, and unresolved questions can now be tackled using newly developed approaches in biophysics and structural biology.
Collapse
Affiliation(s)
- Ahmet Yildiz
- Physics Department, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
2
|
Kreis NN, Moon HH, Wordeman L, Louwen F, Solbach C, Yuan J, Ritter A. KIF2C/MCAK a prognostic biomarker and its oncogenic potential in malignant progression, and prognosis of cancer patients: a systematic review and meta-analysis as biomarker. Crit Rev Clin Lab Sci 2024; 61:404-434. [PMID: 38344808 PMCID: PMC11815995 DOI: 10.1080/10408363.2024.2309933] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 03/24/2024]
Abstract
KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Ha Hyung Moon
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Frank Louwen
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
3
|
Gao JP, Liang W, Liu CW, Xie F, Murray JD. Unraveling the rhizobial infection thread. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:2235-2245. [PMID: 38262702 DOI: 10.1093/jxb/erae017] [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: 10/20/2023] [Accepted: 01/23/2024] [Indexed: 01/25/2024]
Abstract
Most legumes can form an endosymbiotic association with soil bacteria called rhizobia, which colonize specialized root structures called nodules where they fix nitrogen. To colonize nodule cells, rhizobia must first traverse the epidermis and outer cortical cell layers of the root. In most legumes, this involves formation of the infection thread, an intracellular structure that becomes colonized by rhizobia, guiding their passage through the outer cell layers of the root and into the newly formed nodule cells. In this brief review, we recount the early research milestones relating to the rhizobial infection thread and highlight two relatively recent advances in the symbiotic infection mechanism, the eukaryotically conserved 'MYB-AUR1-MAP' mitotic module, which links cytokinesis mechanisms to intracellular infection, and the discovery of the 'infectosome' complex, which guides infection thread growth. We also discuss the potential intertwining of the two modules and the hypothesis that cytokinesis served as a foundation for intracellular infection of symbiotic microbes.
Collapse
Affiliation(s)
- Jin-Peng Gao
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Wenjie Liang
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Cheng-Wu Liu
- School of Life Sciences, Division of Life Sciences and Medicine, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science and Technology of China, Hefei 230026, China
| | - Fang Xie
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jeremy D Murray
- National Key Laboratory of Plant Molecular Genetics, Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- John Innes Centre, CAS-JIC Centre of Excellence for Plant and Microbial Science (CEPAMS), Norwich Research Park, Norwich NR4 7UH, UK
| |
Collapse
|
4
|
Kalkan BM, Ozcan SC, Cicek E, Gonen M, Acilan C. Nek2A prevents centrosome clustering and induces cell death in cancer cells via KIF2C interaction. Cell Death Dis 2024; 15:222. [PMID: 38493150 PMCID: PMC10944510 DOI: 10.1038/s41419-024-06601-0] [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: 11/24/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Unlike normal cells, cancer cells frequently exhibit supernumerary centrosomes, leading to formation of multipolar spindles that can trigger cell death. Nevertheless, cancer cells with supernumerary centrosomes escape the deadly consequences of unequal segregation of genomic material by coalescing their centrosomes into two poles. This unique trait of cancer cells presents a promising target for cancer therapy, focusing on selectively attacking cells with supernumerary centrosomes. Nek2A is a kinase involved in mitotic regulation, including the centrosome cycle, where it phosphorylates linker proteins to separate centrosomes. In this study, we investigated if Nek2A also prevents clustering of supernumerary centrosomes, akin to its separation function. Reduction of Nek2A activity, achieved through knockout, silencing, or inhibition, promotes centrosome clustering, whereas its overexpression results in inhibition of clustering. Significantly, prevention of centrosome clustering induces cell death, but only in cancer cells with supernumerary centrosomes, both in vitro and in vivo. Notably, none of the known centrosomal (e.g., CNAP1, Rootletin, Gas2L1) or non-centrosomal (e.g., TRF1, HEC1) Nek2A targets were implicated in this machinery. Additionally, Nek2A operated via a pathway distinct from other proteins involved in centrosome clustering mechanisms, like HSET and NuMA. Through TurboID proximity labeling analysis, we identified novel proteins associated with the centrosome or microtubules, expanding the known interaction partners of Nek2A. KIF2C, in particular, emerged as a novel interactor, confirmed through coimmunoprecipitation and localization analysis. The silencing of KIF2C diminished the impact of Nek2A on centrosome clustering and rescued cell viability. Additionally, elevated Nek2A levels were indicative of better patient outcomes, specifically in those predicted to have excess centrosomes. Therefore, while Nek2A is a proposed target, its use must be specifically adapted to the broader cellular context, especially considering centrosome amplification. Discovering partners such as KIF2C offers fresh insights into cancer biology and new possibilities for targeted treatment.
Collapse
Affiliation(s)
- Batuhan Mert Kalkan
- Koç University, Graduate School of Health Sciences, Istanbul, Turkey
- Koç University, Research Center for Translational Medicine, Istanbul, Turkey
| | | | - Enes Cicek
- Koç University, Graduate School of Health Sciences, Istanbul, Turkey
- Koç University, Research Center for Translational Medicine, Istanbul, Turkey
| | - Mehmet Gonen
- Koç University, School of Medicine, Istanbul, Turkey
- Koç University, College of Engineering, Department of Industrial Engineering, Istanbul, Turkey
| | - Ceyda Acilan
- Koç University, Research Center for Translational Medicine, Istanbul, Turkey.
- Koç University, School of Medicine, Istanbul, Turkey.
| |
Collapse
|
5
|
Li RQ, Yang Y, Qiao L, Yang L, Shen DD, Zhao XJ. KIF2C: An important factor involved in signaling pathways, immune infiltration, and DNA damage repair in tumorigenesis. Biomed Pharmacother 2024; 171:116173. [PMID: 38237349 DOI: 10.1016/j.biopha.2024.116173] [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: 11/01/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUNDS Poorly regulated mitosis and chromosomal instability are common characteristics in malignant tumor cells. Kinesin family member 2 C (KIF2C), also known as mitotic centromere-associated kinesin (MCAK) is an essential component during mitotic regulation. In recent years, KIF2C was shown to be dysregulated in several tumors and was involved in many aspects of tumor self-regulation. Research on KIF2C may be a new direction and target for anti-tumor therapy. OBJECT The article aims at reviewing current literatures and summarizing the research status of KIF2C in malignant tumors as well as the oncogenic signaling pathways associated with KIF2C and its role in immune infiltration. RESULT In this review, we summarize the KIF2C mechanisms and signaling pathways in different malignant tumors, and briefly describe its involvement in pathways related to classical chemotherapeutic drug resistance, such as MEK/ERK, mTOR, Wnt/β-catenin, P53 and TGF-β1/Smad pathways. KIF2C upregulation was shown to promote tumor cell migration, invasion, chemotherapy resistance and inhibit DNA damage repair. It was also highly correlated with microRNAs, and CD4 +T cell and CD8 +T cell tumor immune infiltration. CONCLUSION This review shows that KIF2C may function as a new anticancer drug target with great potential for malignant tumor treatment and the mitigation of chemotherapy resistance.
Collapse
Affiliation(s)
- Rui-Qing Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lin Qiao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China.
| | - Dan-Dan Shen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Jing Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Zhang B, Liu P, Li Y, Hu Q, Li H, Pang X, Wu H. Multi-omics analysis of kinesin family member 2C in human tumors: novel prognostic biomarker and tumor microenvironment regulator. Am J Cancer Res 2022; 12:4954-4976. [PMID: 36504885 PMCID: PMC9729912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/26/2022] [Indexed: 12/15/2022] Open
Abstract
Kinesin family member 2C (KIF2C) is the best-characterized member of the kinesin-13 family and is involved in accurately fine-tuned dynamics of mitotic spindles. As KIF2C is involved in both spindle formation and regulation of DNA double-strand breaks, precise regulation of KIF2C is essential to prevent malignant transformation associated with gains and losses of DNA content. In the present study, we initially reviewed The Cancer Genome Atlas database and observed that KIF2C is abundantly expressed in most tumor types. We then analyzed the gene alteration profile, protein expression, prognosis, and immune reactivities of KIF2C in more than 10,000 samples from several well-established databases. In addition, we conducted a gene enrichment set analysis to investigate the potential mechanisms underlying the role of KIF2C in tumorigenesis. Multi-omics analysis of KIF2C demonstrated significant statistical correlations between KIF2C expression and clinical prognosis, oncogenic signature gene sets, myeloid-derived suppressor cell infiltration, ImmunoScore, immune checkpoints, microsatellite instability, and tumor mutational burden across multiple tumors. Single-cell data showed that KIF2C is abundantly expressed in malignant cells. The experimental validation demonstrated that KIF2C is highly expressed in gastric cancer cell lines, gastric adenocarcinoma, and hepatocelluar carcinoma. The findings of this study provide important insight for understanding the role and mechanisms of KIF2C in tumorigenesis and immunotherapy in a variety of cancers.
Collapse
Affiliation(s)
- Bixi Zhang
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Peng Liu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Yanchun Li
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Qing Hu
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Xiaoyang Pang
- Department of Orthopaedics, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Hao Wu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
- Center for Precision Medicine, University of Missouri School of MedicineColumbia, MO, USA
| |
Collapse
|
7
|
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
|
8
|
Merenich D, Nakos K, Pompan T, Donovan SJ, Gill A, Patel P, Spiliotis ET, Myers KA. Septins guide noncentrosomal microtubules to promote focal adhesion disassembly in migrating cells. Mol Biol Cell 2022; 33:ar40. [PMID: 35274967 PMCID: PMC9282018 DOI: 10.1091/mbc.e21-06-0334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/07/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cell migration is critical for vascular angiogenesis and is compromised to facilitate tumor metastasis. The migratory process requires the coordinated assembly and disassembly of focal adhesions (FA), actin, and microtubules (MT). MT dynamics at FAs deliver vesicular cargoes and enhance actomyosin contractility to promote FA turnover and facilitate cell advance. Noncentrosomal (NC) MTs regulate FA dynamics and are sufficient to drive cell polarity, but how NC MTs target FAs to control FA turnover is not understood. Here, we show that Rac1 induces the assembly of FA-proximal septin filaments that promote NC MT growth into FAs and inhibit mitotic centromere-associated kinesin (MCAK)-associated MT disassembly, thereby maintaining intact MT plus ends proximal to FAs. Septin-associated MT rescue is coupled with accumulation of Aurora-A kinase and cytoplasmic linker-associated protein (CLASP) localization to the MT between septin and FAs. In this way, NC MTs are strategically positioned to undergo MCAK- and CLASP-regulated bouts of assembly and disassembly into FAs, thereby regulating FA turnover and cell migration.
Collapse
Affiliation(s)
- Daniel Merenich
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | | | - Taylor Pompan
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Samantha J. Donovan
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Amrik Gill
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Pranav Patel
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | | | - Kenneth A. Myers
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
9
|
Jaunky DB, Larocque K, Husser MC, Liu JT, Forgione P, Piekny A. Characterization of a recently synthesized microtubule-targeting compound that disrupts mitotic spindle poles in human cells. Sci Rep 2021; 11:23665. [PMID: 34880347 PMCID: PMC8655040 DOI: 10.1038/s41598-021-03076-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022] Open
Abstract
We reveal the effects of a new microtubule-destabilizing compound in human cells. C75 has a core thienoisoquinoline scaffold with several functional groups amenable to modification. Previously we found that sub micromolar concentrations of C75 caused cytotoxicity. We also found that C75 inhibited microtubule polymerization and competed with colchicine for tubulin-binding in vitro. However, here we found that the two compounds synergized suggesting differences in their mechanism of action. Indeed, live imaging revealed that C75 causes different spindle phenotypes compared to colchicine. Spindles remained bipolar and collapsed after colchicine treatment, while C75 caused bipolar spindles to become multipolar. Importantly, microtubules rapidly disappeared after C75-treatment, but then grew back unevenly and from multiple poles. The C75 spindle phenotype is reminiscent of phenotypes caused by depletion of ch-TOG, a microtubule polymerase, suggesting that C75 blocks microtubule polymerization in metaphase cells. C75 also caused an increase in the number of spindle poles in paclitaxel-treated cells, and combining low amounts of C75 and paclitaxel caused greater regression of multicellular tumour spheroids compared to each compound on their own. These findings warrant further exploration of C75’s anti-cancer potential.
Collapse
Affiliation(s)
| | - Kevin Larocque
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Mathieu C Husser
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Jiang Tian Liu
- Department of Chemistry and Biochemistry, Concordia University, Montreal, QC, Canada
| | - Pat Forgione
- Department of Chemistry and Biochemistry, Concordia University, Montreal, QC, Canada
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Gräf R, Grafe M, Meyer I, Mitic K, Pitzen V. The Dictyostelium Centrosome. Cells 2021; 10:cells10102657. [PMID: 34685637 PMCID: PMC8534566 DOI: 10.3390/cells10102657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
The centrosome of Dictyostelium amoebae contains no centrioles and consists of a cylindrical layered core structure surrounded by a corona harboring microtubule-nucleating γ-tubulin complexes. It is the major centrosomal model beyond animals and yeasts. Proteomics, protein interaction studies by BioID and superresolution microscopy methods led to considerable progress in our understanding of the composition, structure and function of this centrosome type. We discuss all currently known components of the Dictyostelium centrosome in comparison to other centrosomes of animals and yeasts.
Collapse
|
11
|
Campos Medina MA, Iemura K, Kimura A, Tanaka K. A mathematical model of kinetochore-microtubule attachment regulated by Aurora A activity gradient describes chromosome oscillation and correction of erroneous attachments. Biomed Res 2021; 42:203-219. [PMID: 34544996 DOI: 10.2220/biomedres.42.203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chromosome oscillation during metaphase is attenuated in cancer cell lines, concomitant with the reduction of Aurora A activity on kinetochores, which results in reduced mitotic fidelity. To verify the correlation between Aurora A activity, chromosome oscillation, and error correction efficiency, we developed a mathematical model of kinetochore-microtubule dynamics, based on stochastic attachment/detachment events regulated by Aurora A activity gradient centered at spindle poles. The model accurately reproduced the oscillatory movements of chromosomes, which were suppressed not only when Aurora A activity was inhibited, but also when it was upregulated, mimicking the situation in cancer cells. Our simulation also predicted efficient correction of erroneous attachments through chromosome oscillation, which was hampered by both inhibition and upregulation of Aurora A activity. Our model provides a framework to understand the physiological role of chromosome oscillation in the correction of erroneous attachments that is intrinsically related to Aurora A activity.
Collapse
Affiliation(s)
- Manuel Alejandro Campos Medina
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University.,Department of Systemic Cell Biology, Max Planck Institute for Molecular Physiology
| | - Kenji Iemura
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Akatsuki Kimura
- Cell Architecture Laboratory, Department of Chromosome Science, National Institute of Genetics; Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI)
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University
| |
Collapse
|
12
|
Yang L, Baumann C, De La Fuente R, Viveiros MM. Mechanisms underlying disruption of oocyte spindle stability by bisphenol compounds. Reproduction 2021; 159:383-396. [PMID: 31990668 DOI: 10.1530/rep-19-0494] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/17/2022]
Abstract
Accurate chromosome segregation relies on correct chromosome-microtubule interactions within a stable bipolar spindle apparatus. Thus, exposure to spindle disrupting compounds can impair meiotic division and genomic stability in oocytes. The endocrine disrupting activity of bisphenols such as bisphenol A (BPA) is well recognized, yet their damaging effects on spindle microtubules (MTs) is poorly understood. Here, we tested the effect(s) of acute exposure to BPA and bisphenol F (BPF) on assembled spindle stability in ovulated oocytes. Brief (4 h) exposure to increasing concentrations (5, 25, and 50 µg/mL) of BPA or BPF disrupted spindle organization in a dose-dependent manner, resulting in significantly shorter spindles with highly unfocused poles and fragmented pericentrin. The chromosomes remained congressed in an abnormally elongated metaphase-like configuration, yet normal end-on chromosome-MT attachments were reduced in BPF-treated oocytes. Live-cell imaging revealed a rapid onset of bisphenol-mediated spindle MT disruption that was reversed upon compound removal. Moreover, MT stability and regrowth were impaired in BPA-exposed oocytes, with few cold-stable MTs and formation of multipolar spindles upon MT regrowth. MT-associated kinesin-14 motor protein (HSET/KIFC1) labeling along the spindle was also lower in BPA-treated oocytes. Conversely, cold stable MTs and HSET labeling persisted after BPF exposure. Notably, inhibition of Aurora Kinase A limited bisphenol-mediated spindle pole widening, revealing a potential interaction. These results demonstrate rapid MT disrupting activity by bisphenols, which is highly detrimental to meiotic spindle stability and organization. Moreover, we identify an important link between these defects and altered distribution of key spindle associated factors as well as Aurora Kinase A activity.
Collapse
Affiliation(s)
- Luhan Yang
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Claudia Baumann
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Rabindranth De La Fuente
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Regenerative Biosciences Center (RBC), University of Georgia, Athens, Georgia, USA
| | - Maria M Viveiros
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Regenerative Biosciences Center (RBC), University of Georgia, Athens, Georgia, USA
| |
Collapse
|
13
|
Zong H, Hazelbaker M, Moe C, Ems-McClung SC, Hu K, Walczak CE. Spatial regulation of MCAK promotes cell polarization and focal adhesion turnover to drive robust cell migration. Mol Biol Cell 2021; 32:590-604. [PMID: 33566676 PMCID: PMC8101467 DOI: 10.1091/mbc.e20-05-0301] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The asymmetric distribution of microtubule (MT) dynamics in migrating cells is important for cell polarization, yet the underlying regulatory mechanisms remain underexplored. Here, we addressed this question by studying the role of the MT depolymerase, MCAK (mitotic centromere-associated kinesin), in the highly persistent migration of RPE-1 cells. MCAK knockdown leads to slowed migration and poor directional movement. Fixed and live cell imaging revealed that MCAK knockdown results in excessive membrane ruffling as well as defects in cell polarization and the maintenance of a major protrusive front. Additionally, loss of MCAK increases the lifetime of focal adhesions by decreasing their disassembly rate. These functions correlate with a spatial distribution of MCAK activity, wherein activity is higher in the trailing edge of cells compared with the leading edge. Overexpression of Rac1 has a dominant effect over MCAK activity, placing it downstream of or in a parallel pathway to MCAK function in migration. Together, our data support a model in which the polarized distribution of MCAK activity and subsequent differential regulation of MT dynamics contribute to cell polarity, centrosome positioning, and focal adhesion dynamics, which all help facilitate robust directional migration.
Collapse
Affiliation(s)
- Hailing Zong
- Department of Biology, Indiana University, Bloomington, IN 47405
| | - Mark Hazelbaker
- Medical Sciences, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405
| | - Christina Moe
- Department of Biology, Indiana University, Bloomington, IN 47405
| | | | - Ke Hu
- Department of Biology, Indiana University, Bloomington, IN 47405
| | - Claire E Walczak
- Medical Sciences, Indiana University School of Medicine-Bloomington, Bloomington, IN 47405
| |
Collapse
|
14
|
Chen A, Wen S, Liu F, Zhang Z, Liu M, Wu Y, He B, Yan M, Kang T, Lam EWF, Wang Z, Liu Q. CRISPR/Cas9 screening identifies a kinetochore-microtubule dependent mechanism for Aurora-A inhibitor resistance in breast cancer. Cancer Commun (Lond) 2021; 41:121-139. [PMID: 33471959 PMCID: PMC7896750 DOI: 10.1002/cac2.12125] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/31/2022] Open
Abstract
Background Overexpression of Aurora‐A (AURKA) is a feature of breast cancer and associates with adverse prognosis. The selective Aurora‐A inhibitor alisertib (MLN8237) has recently demonstrated promising antitumor responses as a single agent in various cancer types but its phase III clinical trial was reported as a failure since MLN8237 did not show an apparent effect in prolonging the survival of patients. Thus, identification of potential targets that could enhance the activity of MLN8237 would provide a rationale for drug combination to achieve better therapeutic outcome. Methods Here, we conducted a systematic synthetic lethality CRISPR/Cas9 screening of 507 kinases using MLN8237 in breast cancer cells and identified a number of targetable kinases that displayed synthetic lethality interactions with MLN8237. Then, we performed competitive growth assays, colony formation assays, cell viability assays, apoptosis assays, and xenograft murine model to evaluate the synergistic therapeutic effects of Haspin (GSG2) depletion or inhibition with MLN8237. For mechanistic studies, immunofluorescence was used to detect the state of microtubules and the localization of Aurora‐B and mitotic centromere‐associated kinesin (MCAK). Results Among the hits, we observed that Haspin depletion or inhibition marginally inhibited breast cancer cell growth but could substantially enhance the killing effects of MLN8237. Mechanistic studies showed that co‐treatment with Aurora‐A and Haspin inhibitors abolished the recruitment of Aurora‐B and mitotic centromere‐associated kinesin (MCAK) to centromeres which were associated with excessive microtubule depolymerization, kinetochore‐microtubule (KT‐MT) attachment failure, and severe mitotic catastrophe. We further showed that the combination of MLN8237 and the Haspin inhibitor CHR‐6494 synergistically reduced breast cancer cell viability and significantly inhibited both in vitro and in vivo tumor growth. Conclusions These findings establish Haspin as a synthetic lethal target and demonstrate CHR‐6494 as a potential combinational drug for promoting the therapeutic effects of MLN8237 on breast cancer.
Collapse
Affiliation(s)
- Ailin Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Shijun Wen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Fang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Zijian Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Meiling Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Yuanzhong Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Bin He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Min Yan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Eric W-F Lam
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.,Department of Surgery and Cancer, Imperial College London, W12 0NN, London, UK
| | - Zifeng Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Quentin Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.,Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, 116044, P. R. China
| |
Collapse
|
15
|
Kapoor S, Kotak S. Centrosome Aurora A regulates RhoGEF ECT-2 localisation and ensures a single PAR-2 polarity axis in C. elegans embryos. Development 2019; 146:dev174565. [PMID: 31636075 PMCID: PMC7115938 DOI: 10.1242/dev.174565] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
Abstract
Proper establishment of cell polarity is essential for development. In the one-cell C. elegans embryo, a centrosome-localised signal provides spatial information for polarity establishment. It is hypothesised that this signal causes local inhibition of the cortical actomyosin network, and breaks symmetry to direct partitioning of the PAR proteins. However, the molecular nature of the centrosomal signal that triggers cortical anisotropy in the actomyosin network to promote polarity establishment remains elusive. Here, we discover that depletion of Aurora A kinase (AIR-1 in C. elegans) causes pronounced cortical contractions on the embryo surface, and this creates more than one PAR-2 polarity axis. This function of AIR-1 appears to be independent of its role in microtubule nucleation. Importantly, upon AIR-1 depletion, centrosome positioning becomes dispensable in dictating the PAR-2 axis. Moreover, we uncovered that a Rho GEF, ECT-2, acts downstream of AIR-1 in regulating contractility and PAR-2 localisation, and, notably, AIR-1 depletion influences ECT-2 cortical localisation. Overall, this study provides a novel insight into how an evolutionarily conserved centrosome Aurora A kinase inhibits promiscuous PAR-2 domain formation to ensure singularity in the polarity establishment axis.
Collapse
Affiliation(s)
- Sukriti Kapoor
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science, Bangalore 560012, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
16
|
Bisht JS, Tomschik M, Gatlin JC. Induction of a Spindle-Assembly-Competent M Phase in Xenopus Egg Extracts. Curr Biol 2019; 29:1273-1285.e5. [PMID: 30930041 DOI: 10.1016/j.cub.2019.02.061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/12/2019] [Accepted: 02/28/2019] [Indexed: 11/25/2022]
Abstract
Normal mitotic spindle assembly is a prerequisite for faithful chromosome segregation and unperturbed cell-cycle progression. Precise functioning of the spindle machinery relies on conserved architectural features, such as focused poles, chromosome alignment at the metaphase plate, and proper spindle length. These morphological requirements can be achieved only within a compositionally distinct cytoplasm that results from cell-cycle-dependent regulation of specific protein levels and specific post-translational modifications. Here, we used cell-free extracts derived from Xenopus laevis eggs to recapitulate different phases of the cell cycle in vitro and to determine which components are required to render interphase cytoplasm spindle-assembly competent in the absence of protein translation. We found that addition of a nondegradable form of the master cell-cycle regulator cyclin B1 can indeed induce some biochemical and phenomenological characteristics of mitosis, but cyclin B1 alone is insufficient and actually deleterious at high levels for normal spindle assembly. In contrast, addition of a phosphomimetic form of the Greatwall-kinase effector Arpp19 with a specific concentration of nondegradable cyclin B1 rescued spindle bipolarity but resulted in larger-than-normal bipolar spindles with a misalignment of chromosomes. Both were corrected by the addition of exogenous Xkid (Xenopus homolog of human Kid/KIF22), indicating a role for this chromokinesin in regulating spindle length. These observations suggest that, of the many components degraded at mitotic exit and then replenished during the subsequent interphase, only a few are required to induce a cell-cycle transition that produces a spindle-assembly-competent cytoplasm.
Collapse
Affiliation(s)
- Jitender S Bisht
- Department of Molecular Biology, University of Wyoming, 1000 E. University Ave., Laramie, WY 82071, USA; Marine Biological Laboratory, Cell Division and Organization Group, 7 MBL Street, Woods Hole, MA 02543, USA
| | - Miroslav Tomschik
- Department of Molecular Biology, University of Wyoming, 1000 E. University Ave., Laramie, WY 82071, USA
| | - Jesse C Gatlin
- Department of Molecular Biology, University of Wyoming, 1000 E. University Ave., Laramie, WY 82071, USA; Marine Biological Laboratory, Cell Division and Organization Group, 7 MBL Street, Woods Hole, MA 02543, USA.
| |
Collapse
|
17
|
Poser E, Caous R, Gruneberg U, Barr FA. Aurora A promotes chromosome congression by activating the condensin-dependent pool of KIF4A. J Cell Biol 2019; 219:e201905194. [PMID: 31881080 PMCID: PMC7041678 DOI: 10.1083/jcb.201905194] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/08/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Aurora kinases create phosphorylation gradients within the spindle during prometaphase and anaphase, thereby locally regulating factors that promote spindle organization, chromosome condensation and movement, and cytokinesis. We show that one such factor is the kinesin KIF4A, which is present along the chromosome axes throughout mitosis and the central spindle in anaphase. These two pools of KIF4A depend on condensin I and PRC1, respectively. Previous work has shown KIF4A is activated by Aurora B at the anaphase central spindle. However, whether or not chromosome-associated KIF4A bound to condensin I is regulated by Aurora kinases remain unclear. To determine the roles of the two different pools of KIF4A, we generated specific point mutants that are unable to interact with either condensin I or PRC1 or are deficient for Aurora kinase regulation. By analyzing these mutants, we show that Aurora A phosphorylates the condensin I-dependent pool of KIF4A and thus actively promotes chromosome congression from the spindle poles to the metaphase plate.
Collapse
Affiliation(s)
- Elena Poser
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Renaud Caous
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Deretic J, Kerr A, Welburn JPI. A rapid computational approach identifies SPICE1 as an Aurora kinase substrate. Mol Biol Cell 2019; 30:312-323. [PMID: 30485161 PMCID: PMC6589576 DOI: 10.1091/mbc.e18-08-0495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/08/2018] [Accepted: 11/20/2018] [Indexed: 11/11/2022] Open
Abstract
Aurora kinases play a major role in mitosis by regulating diverse substrates. Defining their critical downstream targets is important in understanding Aurora kinase function. Here we have developed an unbiased computational approach to identify new Aurora kinase substrates based on phosphorylation site clustering, protein localization, protein structure, and species conservation. We validate the microtubule-associated proteins Clasp2, Elys, tubulin tyrosine ligase-like polyglutamylase residues 330-624 and spindle and centriole associated protein 1, residues 549-855 (SPICE1), as Aurora A and B kinases substrates in vitro. We also demonstrate that SPICE1 localization is regulated by Aurora kinases during mitosis. In the absence of Aurora kinase activity, SPICE1 remains at centrioles but does not target to the spindle. Similarly, a nonphosphorylatable SPICE1 mutant no longer localizes to the spindle. Finally, we show that misregulating SPICE1 phosphorylation results in abnormal centriole number, spindle multipolarity, and chromosome alignment defects. Overall, our work indicates that temporal and spatial Aurora kinase-mediated regulation of SPICE1 is important for correct chromosome segregation. In addition, our work provides a database-search tool that enables rapid identification of Aurora kinase substrates.
Collapse
Affiliation(s)
- Jovana Deretic
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Alastair Kerr
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| | - Julie P. I. Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, Scotland, UK
| |
Collapse
|
19
|
Potential involvement of RITA in the activation of Aurora A at spindle poles during mitosis. Oncogene 2019; 38:4199-4214. [PMID: 30705408 DOI: 10.1038/s41388-019-0716-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/27/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
The mitotic kinase Aurora A is crucial for various mitotic events. Its activation has been intensively investigated and is not yet completely understood. RITA, the RBP-J interacting and tubulin-associated protein, has been shown to modulate microtubule dynamics in mitosis. We asked if RITA could be related to the activation of Aurora A. We show here that RITA is colocalized with Aurora A and its activator TPX2 at spindle poles during mitosis. FLAG-RITA is precipitated with the complex of Aurora A, TPX2 and tubulin. Depletion of RITA increases exclusively active Aurora A and TPX2 at spindle poles in diverse cancer cell lines and in RITA knockout mouse embryonic fibroblasts. The enhanced active Aurora A, its substrate p-TACC3 and TPX2 are restored by adding back of RITA but not its Δtub mutant with an impaired tubulin-binding capability, indicating that RITA's role as Aurora A's modulator is mediated through its interaction with tubulin. Also, the mitotic failures in cells depleted of RITA are rescued by the inhibition of Aurora A. RITA itself does not directly interfere with the catalytic activity of Aurora A, instead, affects the microtubule binding of its activator TPX2. Moreover, Aurora A's activation correlates with microtubule stabilization induced by the microtubule stabilizer paclitaxel, implicating that stabilized microtubules caused by RITA depletion could also account for increased active Aurora A. Our data suggest a potential role for RITA in the activation of Aurora A at spindle poles by modulating the microtubule binding of TPX2 and the microtubule stability during mitosis.
Collapse
|
20
|
Magnaghi-Jaulin L, Eot-Houllier G, Gallaud E, Giet R. Aurora A Protein Kinase: To the Centrosome and Beyond. Biomolecules 2019; 9:biom9010028. [PMID: 30650622 PMCID: PMC6359016 DOI: 10.3390/biom9010028] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Accurate chromosome segregation requires the perfect spatiotemporal rearrangement of the cellular cytoskeleton. Isolated more than two decades ago from Drosophila, Aurora A is a widespread protein kinase that plays key roles during cell division. Numerous studies have described the localisation of Aurora A at centrosomes, the mitotic spindle, and, more recently, at mitotic centromeres. In this review, we will summarise the cytoskeletal rearrangements regulated by Aurora A during cell division. We will also discuss the recent discoveries showing that Aurora A also controls not only the dynamics of the cortical proteins but also regulates the centromeric proteins, revealing new roles for this kinase during cell division.
Collapse
Affiliation(s)
- Laura Magnaghi-Jaulin
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Grégory Eot-Houllier
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Emmanuel Gallaud
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Régis Giet
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| |
Collapse
|
21
|
McHugh T, Zou J, Volkov VA, Bertin A, Talapatra SK, Rappsilber J, Dogterom M, Welburn JPI. The depolymerase activity of MCAK shows a graded response to Aurora B kinase phosphorylation through allosteric regulation. J Cell Sci 2019; 132:jcs.228353. [PMID: 30578316 PMCID: PMC6398471 DOI: 10.1242/jcs.228353] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/02/2023] Open
Abstract
Kinesin-13 motors regulate precise microtubule dynamics and limit microtubule length throughout metazoans by depolymerizing microtubule ends. Recently, the kinesin-13 motor family member MCAK (also known Kif2C) has been proposed to undergo large conformational changes during its catalytic cycle, as it switches from being in solution to being bound to microtubules. Here, we reveal that MCAK has a compact conformation in solution through crosslinking and electron microscopy experiments. When MCAK is bound to the microtubule ends, it adopts an extended conformation with the N-terminus and neck region of MCAK interacting with the microtubule. Interestingly, the region of MCAK that interacts with the microtubule is the region phosphorylated by Aurora B and contains an end binding (EB) protein-binding motif. The level of phosphorylation of the N-terminus results in a graded microtubule depolymerase activity. Here, we show that the N-terminus of MCAK forms a platform to integrate Aurora B kinase downstream signals and in response fine-tunes its depolymerase activity during mitosis. We propose that this allosteric control mechanism allows decoupling of the N-terminus from the motor domain of MCAK to allow MCAK depolymerase activity at kinetochores. Summary: The kinesin-13 MCAK has a compact conformation in solution but is extended when bound to microtubules. Aurora B phosphorylation of MCAK inhibits depolymerase activity by disrupting its extended conformation.
Collapse
Affiliation(s)
- Toni McHugh
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Juan Zou
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Vladimir A Volkov
- Department of Bionanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft 2629, The Netherlands
| | - Aurélie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France.,Sorbonne Universités, UPMC University Paris 06, 75005 Paris, France
| | - Sandeep K Talapatra
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Juri Rappsilber
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK.,Chair of Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin 10623, Germany
| | - Marileen Dogterom
- Department of Bionanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft 2629, The Netherlands
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| |
Collapse
|
22
|
Willems E, Dedobbeleer M, Digregorio M, Lombard A, Lumapat PN, Rogister B. The functional diversity of Aurora kinases: a comprehensive review. Cell Div 2018; 13:7. [PMID: 30250494 PMCID: PMC6146527 DOI: 10.1186/s13008-018-0040-6] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Aurora kinases are serine/threonine kinases essential for the onset and progression of mitosis. Aurora members share a similar protein structure and kinase activity, but exhibit distinct cellular and subcellular localization. AurA favors the G2/M transition by promoting centrosome maturation and mitotic spindle assembly. AurB and AurC are chromosome-passenger complex proteins, crucial for chromosome binding to kinetochores and segregation of chromosomes. Cellular distribution of AurB is ubiquitous, while AurC expression is mainly restricted to meiotically-active germ cells. In human tumors, all Aurora kinase members play oncogenic roles related to their mitotic activity and promote cancer cell survival and proliferation. Furthermore, AurA plays tumor-promoting roles unrelated to mitosis, including tumor stemness, epithelial-to-mesenchymal transition and invasion. In this review, we aim to understand the functional interplay of Aurora kinases in various types of human cells, including tumor cells. The understanding of the functional diversity of Aurora kinases could help to evaluate their relevance as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
- Estelle Willems
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Matthias Dedobbeleer
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Marina Digregorio
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium
| | - Arnaud Lombard
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,2Department of Neurosurgery, CHU of Liège, Liège, Belgium
| | - Paul Noel Lumapat
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,3Department of Neurology, CHU of Liège, Liège, Belgium
| | - Bernard Rogister
- 1Laboratory of Nervous System Diseases and Therapy, GIGA-Neuroscience, University of Liège, Avenue Hippocrate, 15, 4000 Liège, Belgium.,3Department of Neurology, CHU of Liège, Liège, Belgium
| |
Collapse
|
23
|
Rosas-Salvans M, Cavazza T, Espadas G, Sabido E, Vernos I. Proteomic Profiling of Microtubule Self-organization in M-phase. Mol Cell Proteomics 2018; 17:1991-2004. [PMID: 29970457 DOI: 10.1074/mcp.ra118.000745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/11/2018] [Indexed: 01/08/2023] Open
Abstract
Microtubules (MTs) and associated proteins can self-organize into complex structures such as the bipolar spindle, a process in which RanGTP plays a major role. Addition of RanGTP to M-phase Xenopus egg extracts promotes the nucleation and self-organization of MTs into asters and bipolar-like structures in the absence of centrosomes or chromosomes. We show here that the complex proteome of these RanGTP-induced MT assemblies is similar to that of mitotic spindles. Using proteomic profiling we show that MT self-organization in the M-phase cytoplasm involves the non-linear and non-stoichiometric recruitment of proteins from specific functional groups. Our study provides for the first time a temporal understanding of the protein dynamics driving MT self-organization in M-phase.
Collapse
Affiliation(s)
- Miquel Rosas-Salvans
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Tommaso Cavazza
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Guadalupe Espadas
- **Proteomics Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Eduard Sabido
- **Proteomics Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Isabelle Vernos
- From the ‡Cell and Developmental Biology Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; .,§Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain.,‡‡Institució Catalana de Recerca I Estudis Avançats (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
24
|
Courtheoux T, Diallo A, Damodaran AP, Reboutier D, Watrin E, Prigent C. Aurora A kinase activity is required to maintain an active spindle assembly checkpoint during prometaphase. J Cell Sci 2018; 131:jcs.191353. [PMID: 29555820 DOI: 10.1242/jcs.191353] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
During the prometaphase stage of mitosis, the cell builds a bipolar spindle of microtubules that mechanically segregates sister chromatids between two daughter cells in anaphase. The spindle assembly checkpoint (SAC) is a quality control mechanism that monitors proper attachment of microtubules to chromosome kinetochores during prometaphase. Segregation occurs only when each chromosome is bi-oriented with each kinetochore pair attached to microtubules emanating from opposite spindle poles. Overexpression of the protein kinase Aurora A is a feature of various cancers and is thought to enable tumour cells to bypass the SAC, leading to aneuploidy. Here, we took advantage of a chemical and chemical-genetic approach to specifically inhibit Aurora A kinase activity in late prometaphase. We observed that a loss of Aurora A activity directly affects SAC function, that Aurora A is essential for maintaining the checkpoint protein Mad2 on unattached kinetochores and that inhibition of Aurora A leads to loss of the SAC, even in the presence of nocodazole or Taxol. This is a new finding that should affect the way Aurora A inhibitors are used in cancer treatments.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Thibault Courtheoux
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Alghassimou Diallo
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Arun Prasath Damodaran
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - David Reboutier
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Erwan Watrin
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| | - Claude Prigent
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, Équipe labellisée Ligue contre le Cancer 2014-2016, F-35000 Rennes, France
| |
Collapse
|
25
|
Jun DY, Lee JY, Park HS, Lee YH, Kim YH. Tumor suppressor protein p53-mediated repression of human mitotic centromere-associated kinesin gene expression is exerted via down-regulation of Sp1 level. PLoS One 2017; 12:e0189698. [PMID: 29244835 PMCID: PMC5731752 DOI: 10.1371/journal.pone.0189698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
The repressive role of p53 on the human mitotic centromere-associated kinesin (MCAK) core promoter from ‒266 to +54, relative to the transcription start site, has been determined. The MCAK mRNA and protein levels were 2.1- and 3.0-fold higher, respectively, in HCT116 (p53‒/‒) than in HCT116 (p53+/+) cells. Enforced down-regulation of p53 levels either in HCT116 (p53+/+) cells by p53 RNAi treatment or in MCF-7 cells using shRNA for p53 (shp53) resulted in a remarkable increase in the MCAK protein level. Site-directed mutagenesis and ChIP analyses showed that p53-mediated repression of the MCAK core promoter activity was not directly exerted by p53-binding to putative p53-response elements (p53-RE1 at −173/−166 and p53-RE2 at −245/−238), but indirectly by attenuating Sp1 binding to GC-motifs (GC1 at −93/−84 and GC2 at −119/−110). Treatment of HEK-293 cells bearing the MCAK core promoter-reporter (pGL2-320-Luc) with mithramycin A, which down-regulates Sp1 gene expression, reduced the promoter activity as well as endogenous MCAK levels. Exposure of HCT116 (p53+/+) cells to nutlin-3a, a validated activator of p53, caused a simultaneous reduction in Sp1 and MCAK protein levels, but not in HCT116 (p53−/−) cells. In contrast to wild-type (wt)-p53, tumor-derived p53 mutants (p53V143A, p53R248W, and p53R273H) failed to repress the Sp1-dependent activation of the MCAK promoter and to down-regulate endogenous levels of Sp1 and MCAK proteins. Collectively, these findings demonstrate that p53 can repress MCAK promoter activity indirectly via down-regulation of Sp1 expression level, and suggest that MCAK elevation in human tumor cells might be due to p53 mutation.
Collapse
Affiliation(s)
- Do Youn Jun
- Laboratory of Immunobiology, School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Ji Young Lee
- Laboratory of Immunobiology, School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Hae Sun Park
- Laboratory of Immunobiology, School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Yun Han Lee
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Young Ho Kim
- Laboratory of Immunobiology, School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- * E-mail:
| |
Collapse
|
26
|
Belsham HR, Friel CT. A Cdk1 phosphomimic mutant of MCAK impairs microtubule end recognition. PeerJ 2017; 5:e4034. [PMID: 29230353 PMCID: PMC5723132 DOI: 10.7717/peerj.4034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/24/2017] [Indexed: 12/04/2022] Open
Abstract
The microtubule depolymerising kinesin-13, MCAK, is phosphorylated at residue T537 by Cdk1. This is the only known phosphorylation site within MCAK’s motor domain. To understand the impact of phosphorylation by Cdk1 on microtubule depolymerisation activity, we have investigated the molecular mechanism of the phosphomimic mutant T537E. This mutant significantly impairs microtubule depolymerisation activity and when transfected into cells causes metaphase arrest and misaligned chromosomes. We show that the molecular mechanism underlying the reduced depolymerisation activity of this phosphomimic mutant is an inability to recognise the microtubule end. The microtubule-end residence time is reduced relative to wild-type MCAK, whereas the lattice residence time is unchanged by the phosphomimic mutation. Further, the microtubule-end specific stimulation of ADP dissociation, characteristic of MCAK, is abolished by this mutation. Our data shows that T537E is unable to distinguish between the microtubule end and the microtubule lattice.
Collapse
Affiliation(s)
- Hannah R Belsham
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Claire T Friel
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
27
|
Huang Y, Li T, Ems-McClung SC, Walczak CE, Prigent C, Zhu X, Zhang X, Zheng Y. Aurora A activation in mitosis promoted by BuGZ. J Cell Biol 2017; 217:107-116. [PMID: 29074706 PMCID: PMC5748987 DOI: 10.1083/jcb.201706103] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 09/22/2017] [Accepted: 09/27/2017] [Indexed: 12/25/2022] Open
Abstract
Mitotic spindle component BuGZ is known to undergo phase separation. Huang et al. show that BuGZ promotes Aurora A phosphorylation and activation and that this is inhibited when BuGZ phase separation is disrupted. Protein phase separation or coacervation has emerged as a potential mechanism to regulate biological functions. We have shown that coacervation of a mostly unstructured protein, BuGZ, promotes assembly of spindle and its matrix. BuGZ in the spindle matrix binds and concentrates tubulin to promote microtubule (MT) assembly. It remains unclear, however, whether BuGZ could regulate additional proteins to promote spindle assembly. In this study, we report that BuGZ promotes Aurora A (AurA) activation in vitro. Depletion of BuGZ in cells reduces the amount of phosphorylated AurA on spindle MTs. BuGZ also enhances MCAK phosphorylation. The two zinc fingers in BuGZ directly bind to the kinase domain of AurA, which allows AurA to incorporate into the coacervates formed by BuGZ in vitro. Importantly, mutant BuGZ that disrupts the coacervation activity in vitro fails to promote AurA phosphorylation in Xenopus laevis egg extracts. These results suggest that BuGZ coacervation promotes AurA activation in mitosis.
Collapse
Affiliation(s)
- Yuejia Huang
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD
| | - Teng Li
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD.,Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | | | | | - Claude Prigent
- Institut de Génétique et Développement de Rennes, Equipe laboratoryélisée Ligue Nationale Contre la Cancer 2014-2017, Centre National de la Recherche Scientifique, Université Rennes 1, Rennes, France
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuemin Zhang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD
| |
Collapse
|
28
|
Goldstein A, Siegler N, Goldman D, Judah H, Valk E, Kõivomägi M, Loog M, Gheber L. Three Cdk1 sites in the kinesin-5 Cin8 catalytic domain coordinate motor localization and activity during anaphase. Cell Mol Life Sci 2017; 74:3395-3412. [PMID: 28455557 PMCID: PMC11107736 DOI: 10.1007/s00018-017-2523-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
The bipolar kinesin-5 motors perform essential functions in mitotic spindle dynamics. We previously demonstrated that phosphorylation of at least one of the Cdk1 sites in the catalytic domain of the Saccharomyces cerevisiae kinesin-5 Cin8 (S277, T285, S493) regulates its localization to the anaphase spindle. The contribution of these three sites to phospho-regulation of Cin8, as well as the timing of such contributions, remains unknown. Here, we examined the function and spindle localization of phospho-deficient (serine/threonine to alanine) and phospho-mimic (serine/threonine to aspartic acid) Cin8 mutants. In vitro, the three Cdk1 sites undergo phosphorylation by Clb2-Cdk1. In cells, phosphorylation of Cin8 affects two aspects of its localization to the anaphase spindle, translocation from the spindle-pole bodies (SPBs) region to spindle microtubules (MTs) and the midzone, and detachment from the mitotic spindle. We found that phosphorylation of S277 is essential for the translocation of Cin8 from SPBs to spindle MTs and the subsequent detachment from the spindle. Phosphorylation of T285 mainly affects the detachment of Cin8 from spindle MTs during anaphase, while phosphorylation at S493 affects both the translocation of Cin8 from SPBs to the spindle and detachment from the spindle. Only S493 phosphorylation affected the anaphase spindle elongation rate. We conclude that each phosphorylation site plays a unique role in regulating Cin8 functions and postulate a model in which the timing and extent of phosphorylation of the three sites orchestrates the anaphase function of Cin8.
Collapse
Affiliation(s)
- Alina Goldstein
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, PO Box 653, 84105, Beer-Sheva, Israel
| | - Nurit Siegler
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, PO Box 653, 84105, Beer-Sheva, Israel
| | - Darya Goldman
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, PO Box 653, 84105, Beer-Sheva, Israel
| | - Haim Judah
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, PO Box 653, 84105, Beer-Sheva, Israel
| | - Ervin Valk
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mardo Kõivomägi
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mart Loog
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Larisa Gheber
- Department of Chemistry and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, PO Box 653, 84105, Beer-Sheva, Israel.
| |
Collapse
|
29
|
Cirillo L, Gotta M, Meraldi P. The Elephant in the Room: The Role of Microtubules in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:93-124. [DOI: 10.1007/978-3-319-57127-0_5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
D'Angelo L, Myer NM, Myers KA. MCAK-mediated regulation of endothelial cell microtubule dynamics is mechanosensitive to myosin-II contractility. Mol Biol Cell 2017; 28:1223-1237. [PMID: 28298485 PMCID: PMC5415018 DOI: 10.1091/mbc.e16-05-0306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 01/16/2023] Open
Abstract
This study indicates that MCAK contributes to the mechanosensing-mediated regulation of MT dynamics through a myosin-II–dependent mechanism that becomes uncoupled in response to 3D ECM engagement specifically within EC branches. Compliance and dimensionality mechanosensing, the processes by which cells sense the physical attributes of the extracellular matrix (ECM), are known to drive cell branching and shape change largely through a myosin-II–mediated reorganization of the actin and microtubule (MT) cytoskeletons. Subcellular regulation of MT dynamics is spatially controlled through a Rac1–Aurora-A kinase pathway that locally inhibits the MT depolymerizing activity of mitotic centromere–associated kinesin (MCAK), thereby promoting leading-edge MT growth and cell polarization. These results suggest that the regulation of MT growth dynamics is intimately linked to physical engagement of the cell with the ECM. Here, we tested the hypothesis that MCAK contributes to compliance and dimensionality mechanosensing-mediated regulation of MT growth dynamics through a myosin-II–dependent signaling pathway. We cultured endothelial cells (ECs) on collagen-coupled stiff or compliant polyacrylamide ECMs to examine the effects of MCAK expression on MT growth dynamics and EC branching morphology. Our results identify that MCAK promotes fast MT growth speeds in ECs cultured on compliant 2D ECMs but promotes slow MT growth speeds in ECs cultured on compliant 3D ECMs, and these effects are myosin-II dependent. Furthermore, we find that 3D ECM engagement uncouples MCAK-mediated regulation of MT growth persistence from myosin-II–mediated regulation of growth persistence specifically within EC branched protrusions.
Collapse
Affiliation(s)
- Lauren D'Angelo
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Nicole M Myer
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Kenneth A Myers
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
31
|
Kumar R, Sanawar R, Li X, Li F. Structure, biochemistry, and biology of PAK kinases. Gene 2016; 605:20-31. [PMID: 28007610 DOI: 10.1016/j.gene.2016.12.014] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
PAKs, p21-activated kinases, play central roles and act as converging junctions for discrete signals elicited on the cell surface and for a number of intracellular signaling cascades. PAKs phosphorylate a vast number of substrates and act by remodeling cytoskeleton, employing scaffolding, and relocating to distinct subcellular compartments. PAKs affect wide range of processes that are crucial to the cell from regulation of cell motility, survival, redox, metabolism, cell cycle, proliferation, transformation, stress, inflammation, to gene expression. Understandably, their dysregulation disrupts cellular homeostasis and severely impacts key cell functions, and many of those are implicated in a number of human diseases including cancers, neurological disorders, and cardiac disorders. Here we provide an overview of the members of the PAK family and their current status. We give special emphasis to PAK1 and PAK4, the prototypes of groups I and II, for their profound roles in cancer, the nervous system, and the heart. We also highlight other family members. We provide our perspective on the current advancements, their growing importance as strategic therapeutic targets, and our vision on the future of PAKs.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA; Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India.
| | - Rahul Sanawar
- Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India
| | - Xiaodong Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China.
| |
Collapse
|
32
|
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]
|
33
|
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
|
34
|
Kumar R, Li DQ. PAKs in Human Cancer Progression: From Inception to Cancer Therapeutic to Future Oncobiology. Adv Cancer Res 2016; 130:137-209. [PMID: 27037753 DOI: 10.1016/bs.acr.2016.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the initial recognition of a mechanistic role of p21-activated kinase 1 (PAK1) in breast cancer invasion, PAK1 has emerged as one of the widely overexpressed or hyperactivated kinases in human cancer at-large, allowing the PAK family to make in-roads in cancer biology, tumorigenesis, and cancer therapeutics. Much of our current understanding of the PAK family in cancer progression relates to a central role of the PAK family in the integration of cancer-promoting signals from cell membrane receptors as well as function as a key nexus-modifier of complex, cytoplasmic signaling network. Another core aspect of PAK signaling that highlights its importance in cancer progression is through PAK's central role in the cross talk with signaling and interacting proteins, as well as PAK's position as a key player in the phosphorylation of effector substrates to engage downstream components that ultimately leads to the development cancerous phenotypes. Here we provide a comprehensive review of the recent advances in PAK cancer research and its downstream substrates in the context of invasion, nuclear signaling and localization, gene expression, and DNA damage response. We discuss how a deeper understanding of PAK1's pathobiology over the years has widened research interest to the PAK family and human cancer, and positioning the PAK family as a promising cancer therapeutic target either alone or in combination with other therapies. With many landmark findings and leaps in the progress of PAK cancer research since the infancy of this field nearly 20 years ago, we also discuss postulated advances in the coming decade as the PAK family continues to shape the future of oncobiology.
Collapse
Affiliation(s)
- R Kumar
- School of Medicine and Health Sciences, George Washington University, Washington, DC, United States; Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram, India.
| | - D-Q Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Epigenetics in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Connolly AA, Sugioka K, Chuang CH, Lowry JB, Bowerman B. KLP-7 acts through the Ndc80 complex to limit pole number in C. elegans oocyte meiotic spindle assembly. J Cell Biol 2015; 210:917-32. [PMID: 26370499 PMCID: PMC4576866 DOI: 10.1083/jcb.201412010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
KLP-7/MCAK regulates kinetochore–microtubule attachment and spindle tension to promote the coalescence of early spindle pole foci, which produces a bipolar structure during the acentrosomal process of oocyte meiotic spindle assembly in C. elegans. During oocyte meiotic cell division in many animals, bipolar spindles assemble in the absence of centrosomes, but the mechanisms that restrict pole assembly to a bipolar state are unknown. We show that KLP-7, the single mitotic centromere–associated kinesin (MCAK)/kinesin-13 in Caenorhabditis elegans, is required for bipolar oocyte meiotic spindle assembly. In klp-7(−) mutants, extra microtubules accumulated, extra functional spindle poles assembled, and chromosomes frequently segregated as three distinct masses during meiosis I anaphase. Moreover, reducing KLP-7 function in monopolar klp-18(−) mutants often restored spindle bipolarity and chromosome segregation. MCAKs act at kinetochores to correct improper kinetochore–microtubule (k–MT) attachments, and depletion of the Ndc-80 kinetochore complex, which binds microtubules to mediate kinetochore attachment, restored bipolarity in klp-7(−) mutant oocytes. We propose a model in which KLP-7/MCAK regulates k–MT attachment and spindle tension to promote the coalescence of early spindle pole foci that produces a bipolar structure during the acentrosomal process of oocyte meiotic spindle assembly.
Collapse
Affiliation(s)
- Amy A Connolly
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Kenji Sugioka
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Chien-Hui Chuang
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Joshua B Lowry
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Bruce Bowerman
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| |
Collapse
|
36
|
Reboutier D, Benaud C, Prigent C. Aurora A's Functions During Mitotic Exit: The Guess Who Game. Front Oncol 2015; 5:290. [PMID: 26734572 PMCID: PMC4685928 DOI: 10.3389/fonc.2015.00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/07/2015] [Indexed: 11/24/2022] Open
Abstract
Until recently, the knowledge of Aurora A kinase functions during mitosis was limited to pre-metaphase events, particularly centrosome maturation, G2/M transition, and mitotic spindle assembly. However, an involvement of Aurora A in post-metaphase events was also suspected, but not clearly demonstrated due to the technical difficulty to perform the appropriate experiments. Recent developments of both an analog-specific version of Aurora A and small molecule inhibitors have led to the first demonstration that Aurora A is required for the early steps of cytokinesis. As in pre-metaphase, Aurora A plays diverse functions during anaphase, essentially participating in astral microtubules dynamics and central spindle assembly and functioning. The present review describes the experimental systems used to decipher new functions of Aurora A during late mitosis and situate these functions into the context of cytokinesis mechanisms.
Collapse
Affiliation(s)
- David Reboutier
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Christelle Benaud
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| | - Claude Prigent
- Unité Mixte de Recherche 6290, Équipe labellisée Ligue, Centre National de la Recherche Scientifique, Rennes, France; Institut de Génétique et Développement de Rennes, Université Rennes 1, Rennes, France
| |
Collapse
|
37
|
Carvalhal S, Ribeiro SA, Arocena M, Kasciukovic T, Temme A, Koehler K, Huebner A, Griffis ER. The nucleoporin ALADIN regulates Aurora A localization to ensure robust mitotic spindle formation. Mol Biol Cell 2015; 26:3424-38. [PMID: 26246606 PMCID: PMC4591688 DOI: 10.1091/mbc.e15-02-0113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022] Open
Abstract
The nucleoporin ALADIN, which is mutated in patients with triple A syndrome, is necessary for proper spindle formation. Without ALADIN, active Aurora A moves away from centrosomes. The relocalization of active Aurora A leads to a redistribution of specific spindle assembly factors that make spindles less stable and slows their formation. The formation of the mitotic spindle is a complex process that requires massive cellular reorganization. Regulation by mitotic kinases controls this entire process. One of these mitotic controllers is Aurora A kinase, which is itself highly regulated. In this study, we show that the nuclear pore protein ALADIN is a novel spatial regulator of Aurora A. Without ALADIN, Aurora A spreads from centrosomes onto spindle microtubules, which affects the distribution of a subset of microtubule regulators and slows spindle assembly and chromosome alignment. ALADIN interacts with inactive Aurora A and is recruited to the spindle pole after Aurora A inhibition. Of interest, mutations in ALADIN cause triple A syndrome. We find that some of the mitotic phenotypes that we observe after ALADIN depletion also occur in cells from triple A syndrome patients, which raises the possibility that mitotic errors may underlie part of the etiology of this syndrome.
Collapse
Affiliation(s)
- Sara Carvalhal
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Susana Abreu Ribeiro
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543 Wellcome Trust Centre for Cell Biology, Institute of Cell and Molecular Biology, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Miguel Arocena
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Taciana Kasciukovic
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom
| | - Achim Temme
- Department of Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Katrin Koehler
- Department of Paediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Angela Huebner
- Department of Paediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Eric R Griffis
- Centre for Gene Regulation and Expression, University of Dundee, College of Life Sciences, Dundee DD1 5EH, United Kingdom Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543
| |
Collapse
|
38
|
Shao H, Huang Y, Zhang L, Yuan K, Chu Y, Dou Z, Jin C, Garcia-Barrio M, Liu X, Yao X. Spatiotemporal dynamics of Aurora B-PLK1-MCAK signaling axis orchestrates kinetochore bi-orientation and faithful chromosome segregation. Sci Rep 2015; 5:12204. [PMID: 26206521 PMCID: PMC4513279 DOI: 10.1038/srep12204] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/13/2015] [Indexed: 12/01/2022] Open
Abstract
Chromosome segregation in mitosis is orchestrated by the dynamic interactions between the kinetochore and spindle microtubules. The microtubule depolymerase mitotic centromere-associated kinesin (MCAK) is a key regulator for an accurate kinetochore-microtubule attachment. However, the regulatory mechanism underlying precise MCAK depolymerase activity control during mitosis remains elusive. Here, we describe a novel pathway involving an Aurora B-PLK1 axis for regulation of MCAK activity in mitosis. Aurora B phosphorylates PLK1 on Thr210 to activate its kinase activity at the kinetochores during mitosis. Aurora B-orchestrated PLK1 kinase activity was examined in real-time mitosis using a fluorescence resonance energy transfer-based reporter and quantitative analysis of native PLK1 substrate phosphorylation. Active PLK1, in turn, phosphorylates MCAK at Ser715 which promotes its microtubule depolymerase activity essential for faithful chromosome segregation. Importantly, inhibition of PLK1 kinase activity or expression of a non-phosphorylatable MCAK mutant prevents correct kinetochore-microtubule attachment, resulting in abnormal anaphase with chromosome bridges. We reason that the Aurora B-PLK1 signaling at the kinetochore orchestrates MCAK activity, which is essential for timely correction of aberrant kinetochore attachment to ensure accurate chromosome segregation during mitosis.
Collapse
Affiliation(s)
- Hengyi Shao
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Yuejia Huang
- Anhui-MSM Joint Research Group for Cellular Dynamics, Hefei National Laboratory for Physical Sciences at Nanoscale, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Liangyu Zhang
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Kai Yuan
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | - Youjun Chu
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Zhen Dou
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Anhui-MSM Joint Research Group for Cellular Dynamics, Hefei National Laboratory for Physical Sciences at Nanoscale, Hefei 230027, China
| | - Changjiang Jin
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| | | | - Xing Liu
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
- Anhui-MSM Joint Research Group for Cellular Dynamics, Hefei National Laboratory for Physical Sciences at Nanoscale, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xuebiao Yao
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science & Technology of China, Hefei 230027, China
| |
Collapse
|
39
|
The KLP-7 Residue S546 Is a Putative Aurora Kinase Site Required for Microtubule Regulation at the Centrosome in C. elegans. PLoS One 2015; 10:e0132593. [PMID: 26168236 PMCID: PMC4500558 DOI: 10.1371/journal.pone.0132593] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 06/16/2015] [Indexed: 12/20/2022] Open
Abstract
Regulation of microtubule dynamics is essential for many cellular processes, including proper assembly and function of the mitotic spindle. The kinesin-13 microtubule-depolymerizing enzymes provide one mechanism to regulate microtubule behaviour temporally and spatially. Vertebrate MCAK locates to chromatin, kinetochores, spindle poles, microtubule tips, and the cytoplasm, implying that the regulation of kinesin-13 activity and subcellular targeting is complex. Phosphorylation of kinesin-13 by Aurora kinase inhibits microtubule depolymerization activity and some Aurora phosphorylation sites on kinesin-13 are required for subcellular localization. Herein, we determine that a C. elegans deletion mutant klp-7(tm2143) causes meiotic and mitotic defects that are consistent with an increase in the amount of microtubules in the cytoplasmic and spindle regions of meiotic embryos, and an increase in microtubules emanating from centrosomes. We show that KLP-7 is phosphorylated by Aurora A and Aurora B kinases in vitro, and that the phosphorylation by Aurora A is stimulated by TPXL-1. Using a structure-function approach, we establish that one putative Aurora kinase site, S546, within the C-terminal part of the core domain is required for the function, but not subcellular localization, of KLP-7 in vivo. Furthermore, FRAP analysis reveals microtubule-dependent differences in the turnover of KLP-7(S546A) and KLP-7(S546E) mutant proteins at the centrosome, suggesting a possible mechanism for the regulation of KLP-7 by Aurora kinase.
Collapse
|
40
|
Ritter A, Sanhaji M, Friemel A, Roth S, Rolle U, Louwen F, Yuan J. Functional analysis of phosphorylation of the mitotic centromere-associated kinesin by Aurora B kinase in human tumor cells. Cell Cycle 2015; 14:3755-3767. [PMID: 26148251 PMCID: PMC4825789 DOI: 10.1080/15384101.2015.1068481] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/27/2015] [Indexed: 12/20/2022] Open
Abstract
Mitotic centromere-associated kinesin (MCAK) is the best characterized member of the kinesin-13 family and plays important roles in microtubule dynamics during mitosis. Its activity and subcellular localization is tightly regulated by an orchestra of mitotic kinases, such as Aurora B. It is well known that serine 196 of MCAK is the major phosphorylation site of Aurora B in Xenopus leavis extracts and that this phosphorylation regulates its catalytic activity and subcellular localization. In the current study, we have addressed the conserved phosphorylation site serine 192 in human MCAK to characterize its function in more depth in human cancer cells. Our data confirm that S192 is the major phosphorylation site of Aurora B in human MCAK and that this phosphorylation has crucial roles in regulating its catalytic activity and localization at the kinetochore/centromere region in mitosis. Interfering with this phosphorylation leads to a delayed progression through prometa- and metaphase associated with mitotic defects in chromosome alignment and segregation. We show further that MCAK is involved in directional migration and invasion of tumor cells, and interestingly, interference with the S192 phosphorylation affects this capability of MCAK. These data provide the first molecular explanation for clinical observation, where an overexpression of MCAK was associated with lymphatic invasion and lymph node metastasis in gastric and colorectal cancer patients.
Collapse
Affiliation(s)
- Andreas Ritter
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| | - Mourad Sanhaji
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| | - Alexandra Friemel
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| | - Susanne Roth
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology; School of Medicine; JW Goethe-University; Frankfurt, Germany
| | - Frank Louwen
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| | - Juping Yuan
- Department of Gynecology and Obstetrics; JW Goethe-University; Frankfurt, Germany
| |
Collapse
|
41
|
Platani M, Trinkle-Mulcahy L, Porter M, Jeyaprakash AA, Earnshaw WC. Mio depletion links mTOR regulation to Aurora A and Plk1 activation at mitotic centrosomes. J Cell Biol 2015; 210:45-62. [PMID: 26124292 PMCID: PMC4494011 DOI: 10.1083/jcb.201410001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Coordination of cell growth and proliferation in response to nutrient supply is mediated by mammalian target of rapamycin (mTOR) signaling. In this study, we report that Mio, a highly conserved member of the SEACAT/GATOR2 complex necessary for the activation of mTORC1 kinase, plays a critical role in mitotic spindle formation and subsequent chromosome segregation by regulating the proper concentration of active key mitotic kinases Plk1 and Aurora A at centrosomes and spindle poles. Mio-depleted cells showed reduced activation of Plk1 and Aurora A kinase at spindle poles and an impaired localization of MCAK and HURP, two key regulators of mitotic spindle formation and known substrates of Aurora A kinase, resulting in spindle assembly and cytokinesis defects. Our results indicate that a major function of Mio in mitosis is to regulate the activation/deactivation of Plk1 and Aurora A, possibly by linking them to mTOR signaling in a pathway to promote faithful mitotic progression.
Collapse
Affiliation(s)
- Melpomeni Platani
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H8M5, Canada Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H8M5, Canada
| | - Michael Porter
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - A Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - William C Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| |
Collapse
|
42
|
Talapatra SK, Harker B, Welburn JPI. The C-terminal region of the motor protein MCAK controls its structure and activity through a conformational switch. eLife 2015; 4. [PMID: 25915621 PMCID: PMC4443670 DOI: 10.7554/elife.06421] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/24/2015] [Indexed: 11/29/2022] Open
Abstract
The precise regulation of microtubule dynamics is essential during cell division. The
kinesin-13 motor protein MCAK is a potent microtubule depolymerase. The divergent
non-motor regions flanking the ATPase domain are critical in regulating its targeting
and activity. However, the molecular basis for the function of the non-motor regions
within the context of full-length MCAK is unknown. Here, we determine the structure
of MCAK motor domain bound to its regulatory C-terminus. Our analysis reveals that
the MCAK C-terminus binds to two motor domains in solution and is displaced
allosterically upon microtubule binding, which allows its robust accumulation at
microtubule ends. These results demonstrate that MCAK undergoes long-range
conformational changes involving its C-terminus during the soluble to
microtubule-bound transition and that the C-terminus-motor interaction represents a
structural intermediate in the MCAK catalytic cycle. Together, our work reveals
intrinsic molecular mechanisms underlying the regulation of kinesin-13 activity. DOI:http://dx.doi.org/10.7554/eLife.06421.001 Within a cell, there is a scaffold-like structure called the cytoskeleton that
provides shape and structural support, and acts as a transport network for the
movement of molecules around the cell. This scaffold contains highly dynamic polymers
called microtubules that are made from a protein called tubulin. The constant growth
and shrinking of the ends of the microtubules is essential to rebuild and adapt the
cytoskeleton according to the needs of the cell. A protein called MCAK belongs to a family of motor proteins that can move along
microtubules. It generally binds to the ends of the microtubules to shorten them.
Previous studies have found that a single MCAK protein binds to another MCAK protein
to form a larger molecule known as a dimer. Part of the MCAK protein forms a
so-called motor domain, which enables this protein to bind to the microtubules. One
end of the protein, known as the C-terminus, controls the activity of this motor
domain. However, it is not clear how this works. Talapatra et al. have now revealed the three-dimensional structure of MCAK's
motor domain with the C-terminus using a technique called X-ray crystallography. The
experiments show that the C-terminus binds to the motor domain, which promotes the
formation of the dimers. A short stretch of amino acids—the building blocks of
proteins—in the C-terminus interacts with two motor molecules. This
‘motif’ is also found in other similar proteins from a variety of
animals. However, once MCAK binds to a microtubule, the microtubule triggers the
release of the C-terminus from the motor domain. This allows MCAK to bind more
strongly to the microtubule. The experiments also show that the binding of the C-terminus to the motor domain
alters the ability of MCAK to associate with microtubules, which encourages the
protein to reach the ends of the polymers. Future work is required to see whether
other motor proteins work in a similar way. DOI:http://dx.doi.org/10.7554/eLife.06421.002
Collapse
Affiliation(s)
- Sandeep K Talapatra
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bethany Harker
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Ritter A, Sanhaji M, Steinhäuser K, Roth S, Louwen F, Yuan J. The activity regulation of the mitotic centromere-associated kinesin by Polo-like kinase 1. Oncotarget 2015; 6:6641-55. [PMID: 25504441 PMCID: PMC4466640 DOI: 10.18632/oncotarget.2843] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/01/2014] [Indexed: 01/10/2023] Open
Abstract
The mitotic centromere-associated kinesin (MCAK), a potent microtubule depolymerase, is involved in regulating microtubule dynamics. The activity and subcellular localization of MCAK are tightly regulated by key mitotic kinases, such as Polo-like kinase 1 (Plk1) by phosphorylating multiple residues in MCAK. Since Plk1 phosphorylates very often different residues of substrates at different stages, we have dissected individual phosphorylation of MCAK by Plk1 and characterized its function in more depth. We have recently shown that S621 in MCAK is the major phosphorylation site of Plk1, which is responsible for regulating MCAK's degradation by promoting the association of MCAK with APC/CCdc20. In the present study, we have addressed another two residues phosphorylated by Plk1, namely S632/S633 in the C-terminus of MCAK. Our data suggest that Plk1 phosphorylates S632/S633 and regulates its catalytic activity in mitosis. This phosphorylation is required for proper spindle assembly during early phases of mitosis. The subsequent dephosphorylation of S632/S633 might be necessary to timely align the chromosomes onto the metaphase plate. Therefore, our studies suggest new mechanisms by which Plk1 regulates MCAK: the degradation of MCAK is controlled by Plk1 phosphorylation on S621, whereas its activity is modulated by Plk1 phosphorylation on S632/S633 in mitosis.
Collapse
Affiliation(s)
- Andreas Ritter
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Mourad Sanhaji
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- Present address: University Hospital Jena, Institute for Diagnostic and Interventional Radiology, Experimental Radiology, Erlanger Allee 101, 07747 Jena, Germany
| | - Kerstin Steinhäuser
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Susanne Roth
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Frank Louwen
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Juping Yuan
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
44
|
Gottardo M, Callaini G, Riparbelli MG. Aurora A inhibition by MNL8054 promotes centriole elongation during Drosophila male meiosis. Cell Cycle 2015; 14:2844-52. [PMID: 25785740 DOI: 10.1080/15384101.2015.1026488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Aurora A kinase plays an important role in several aspects of cell division, including centrosome maturation and separation, a crucial step for the correct organization of the bipolar spindle. Although it has long been showed that this kinase accumulates at the centrosome throughout mitosis its precise contribution to centriole biogenesis and structure has until now not been reported. It is not surprising that so little is known, due to the small size of somatic centrioles, where only dramatic structural changes may be identified by careful electron microscopy analysis. Conversely, centrioles of Drosophila primary spermatocytes increase tenfold in length during the first prophase, thus making any change easily detectable. Therefore, we examined the consequence of the pharmacological inhibition of Aurora A by MLN8054 on centriole biogenesis during early Drosophila gametogenesis. Here, we show that depletion of this kinase results in longer centrioles, mainly during transition from prophase to prometaphase of the first meiosis. We also found abnormal ciliogenesis characterized by irregularly growing axonemal doublets. Our results represent the first documentation of a potential requirement of Aurora A in centriole integrity and elongation.
Collapse
Affiliation(s)
- Marco Gottardo
- a Department of Life Sciences ; University of Siena ; Siena , Italy
| | | | | |
Collapse
|
45
|
Yount AL, Zong H, Walczak CE. Regulatory mechanisms that control mitotic kinesins. Exp Cell Res 2015; 334:70-7. [PMID: 25576382 DOI: 10.1016/j.yexcr.2014.12.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 12/26/2014] [Indexed: 11/18/2022]
Abstract
During mitosis, the mitotic spindle is assembled to align chromosomes at the spindle equator in metaphase, and to separate the genetic material equally to daughter cells in anaphase. The spindle itself is a macromolecular machine composed of an array of dynamic microtubules and associated proteins that coordinate the diverse events of mitosis. Among the microtubule associated proteins are a plethora of molecular motor proteins that couple the energy of ATP hydrolysis to force production. These motors, including members of the kinesin superfamily, must function at the right time and in the right place to insure the fidelity of mitosis. Misregulation of mitotic motors in disease states, such as cancer, underlies their potential utility as targets for antitumor drug development and highlights the importance of understanding the molecular mechanisms for regulating their function. Here, we focus on recent progress about regulatory mechanisms that control the proper function of mitotic kinesins and highlight new findings that lay the path for future studies.
Collapse
Affiliation(s)
- Amber L Yount
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, United States
| | - Hailing Zong
- Department of Biology, Indiana University, Bloomington, IN 47405, United States
| | - Claire E Walczak
- Medical Sciences, Indiana University, Myers Hall 262, 915 East 3rd Street, Bloomington, IN 47405, United States.
| |
Collapse
|
46
|
Tanenbaum ME, Medema RH, Akhmanova A. Regulation of localization and activity of the microtubule depolymerase MCAK. BIOARCHITECTURE 2014; 1:80-87. [PMID: 21866268 DOI: 10.4161/bioa.1.2.15807] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 04/09/2011] [Indexed: 12/29/2022]
Abstract
Mitotic Centromere Associated Kinesin (MCAK) is a potent microtubule depolymerizing and catastrophe-inducing factor, which uses the energy of ATP hydrolysis to destabilize microtubule ends. MCAK is localized to inner centromeres, kinetochores and spindle poles of mitotic cells, and is also present in the cytoplasm. Both in interphase and in mitosis, MCAK can specifically accumulate at the growing microtubule ends. Here we discuss the mechanisms, which modulate subcellular localization and activity of MCAK through the interaction with the End Binding (EB) proteins and phosphorylation.
Collapse
Affiliation(s)
- Marvin E Tanenbaum
- Department of Medical Oncology and Cancer Genomics Center; University Medical Center; Utrecht, The Netherlands
| | | | | |
Collapse
|
47
|
Ferreira JG, Pereira AL, Maiato H. Microtubule plus-end tracking proteins and their roles in cell division. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 309:59-140. [PMID: 24529722 DOI: 10.1016/b978-0-12-800255-1.00002-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microtubules are cellular components that are required for a variety of essential processes such as cell motility, mitosis, and intracellular transport. This is possible because of the inherent dynamic properties of microtubules. Many of these properties are tightly regulated by a number of microtubule plus-end-binding proteins or +TIPs. These proteins recognize the distal end of microtubules and are thus in the right context to control microtubule dynamics. In this review, we address how microtubule dynamics are regulated by different +TIP families, focusing on how functionally diverse +TIPs spatially and temporally regulate microtubule dynamics during animal cell division.
Collapse
Affiliation(s)
- Jorge G Ferreira
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal; Cell Division Unit, Department of Experimental Biology, University of Porto, Porto, Portugal
| | - Ana L Pereira
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Helder Maiato
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal; Cell Division Unit, Department of Experimental Biology, University of Porto, Porto, Portugal.
| |
Collapse
|
48
|
Braun A, Dang K, Buslig F, Baird MA, Davidson MW, Waterman CM, Myers KA. Rac1 and Aurora A regulate MCAK to polarize microtubule growth in migrating endothelial cells. ACTA ACUST UNITED AC 2014; 206:97-112. [PMID: 25002679 PMCID: PMC4085700 DOI: 10.1083/jcb.201401063] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A Rac1–Aurora A–MCAK signaling pathway mediates endothelial cell polarization and directional migration by promoting regional differences in microtubule dynamics in the leading and trailing cell edges. Endothelial cells (ECs) migrate directionally during angiogenesis and wound healing by polarizing to extracellular cues to guide directional movement. EC polarization is controlled by microtubule (MT) growth dynamics, which are regulated by MT-associated proteins (MAPs) that alter MT stability. Mitotic centromere-associated kinesin (MCAK) is a MAP that promotes MT disassembly within the mitotic spindle, yet its function in regulating MT dynamics to promote EC polarity and migration has not been investigated. We used high-resolution fluorescence microscopy coupled with computational image analysis to elucidate the role of MCAK in regulating MT growth dynamics, morphology, and directional migration of ECs. Our results show that MCAK-mediated depolymerization of MTs is specifically targeted to the trailing edge of polarized wound-edge ECs. Regulation of MCAK function is dependent on Aurora A kinase, which is regionally enhanced by signaling from the small guanosine triphosphatase, Rac1. Thus, a Rac1–Aurora A–MCAK signaling pathway mediates EC polarization and directional migration by promoting regional differences in MT dynamics in the leading and trailing cell edges.
Collapse
Affiliation(s)
- Alexander Braun
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104
| | - Kyvan Dang
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104
| | - Felinah Buslig
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104
| | - Michelle A Baird
- National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, FL 32310 National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, FL 32310
| | - Michael W Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, FL 32310 National High Magnetic Field Laboratory and Department of Biological Science, The Florida State University, Tallahassee, FL 32310
| | - Clare M Waterman
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kenneth A Myers
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104 Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
49
|
Xia P, Zhou J, Song X, Wu B, Liu X, Li D, Zhang S, Wang Z, Yu H, Ward T, Zhang J, Li Y, Wang X, Chen Y, Guo Z, Yao X. Aurora A orchestrates entosis by regulating a dynamic MCAK-TIP150 interaction. J Mol Cell Biol 2014; 6:240-254. [PMID: 24847103 PMCID: PMC4034728 DOI: 10.1093/jmcb/mju016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 01/23/2014] [Accepted: 01/23/2014] [Indexed: 01/04/2023] Open
Abstract
Entosis, a cell-in-cell process, has been implicated in the formation of aneuploidy associated with an aberrant cell division control. Microtubule plus-end-tracking protein TIP150 facilitates the loading of MCAK onto the microtubule plus ends and orchestrates microtubule plus-end dynamics during cell division. Here we show that TIP150 cooperates with MCAK to govern entosis via a regulatory circuitry that involves Aurora A-mediated phosphorylation of MCAK. Our biochemical analyses show that MCAK forms an intra-molecular association, which is essential for TIP150 binding. Interestingly, Aurora A-mediated phosphorylation of MCAK modulates its intra-molecular association, which perturbs the MCAK-TIP150 interaction in vitro and inhibits entosis in vivo. To probe if MCAK-TIP150 interaction regulates microtubule plasticity to affect the mechanical properties of cells during entosis, we used an optical trap to measure the mechanical rigidity of live MCF7 cells. We find that the MCAK cooperates with TIP150 to promote microtubule dynamics and modulate the mechanical rigidity of the cells during entosis. Our results show that a dynamic interaction of MCAK-TIP150 orchestrated by Aurora A-mediated phosphorylation governs entosis via regulating microtubule plus-end dynamics and cell rigidity. These data reveal a previously unknown mechanism of Aurora A regulation in the control of microtubule plasticity during cell-in-cell processes.
Collapse
Affiliation(s)
- Peng Xia
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Jinhua Zhou
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Xiaoyu Song
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Bing Wu
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Xing Liu
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Di Li
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Shuyuan Zhang
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Zhikai Wang
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Huijuan Yu
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | - Tarsha Ward
- Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA Harvard Medical School, Boston, MA 02115, USA
| | - Jiancun Zhang
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China Guangzhou Institutes of Biomedicine and Health, Guangzhou 510513, China
| | - Yinmei Li
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| | | | - Yong Chen
- Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Zhen Guo
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xuebiao Yao
- Anhui Key Laboratory of Cellular Dynamics & Chemical Biology, Department of Optics and Optical Engineering, and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
50
|
Sanhaji M, Ritter A, Belsham HR, Friel CT, Roth S, Louwen F, Yuan J. Polo-like kinase 1 regulates the stability of the mitotic centromere-associated kinesin in mitosis. Oncotarget 2014; 5:3130-44. [PMID: 24931513 PMCID: PMC4102797 DOI: 10.18632/oncotarget.1861] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/24/2014] [Indexed: 12/13/2022] Open
Abstract
Proper bi-orientation of chromosomes is critical for the accurate segregation of chromosomes in mitosis. A key regulator of this process is MCAK, the mitotic centromere-associated kinesin. During mitosis the activity and localization of MCAK are regulated by mitotic key kinases including Plk1 and Aurora B. We show here that S621 in the MCAK's C-terminal domain is the major phosphorylation site for Plk1. This phosphorylation regulates MCAK's stability and facilitates its recognition by the ubiquitin/proteasome dependent APC/C(Cdc20) pathway leading to its D-box dependent degradation in mitosis. While phosphorylation of S621 does not directly affect its microtubule depolymerising activity, loss of Plk1 phosphorylation on S621 indirectly enhances its depolymerization activity in vivo by stabilizing MCAK, leading to an increased level of protein. Interfering with phosphorylation at S621 causes spindle formation defects and chromosome misalignments. Therefore, this study suggests a new mechanism by which Plk1 regulates MCAK: by regulating its degradation and hence controlling its turnover in mitosis.
Collapse
Affiliation(s)
- Mourad Sanhaji
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | - Andreas Ritter
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | - Hannah R. Belsham
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Claire T. Friel
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Susanne Roth
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | - Frank Louwen
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| | - Juping Yuan
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany
| |
Collapse
|