51
|
Lee KY, Green RA, Gutierrez E, Gomez-Cavazos JS, Kolotuev I, Wang S, Desai A, Groisman A, Oegema K. CYK-4 functions independently of its centralspindlin partner ZEN-4 to cellularize oocytes in germline syncytia. eLife 2018; 7:36919. [PMID: 29989548 PMCID: PMC6056237 DOI: 10.7554/elife.36919] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
Throughout metazoans, germ cells undergo incomplete cytokinesis to form syncytia connected by intercellular bridges. Gamete formation ultimately requires bridge closure, yet how bridges are reactivated to close is not known. The most conserved bridge component is centralspindlin, a complex of the Rho family GTPase-activating protein (GAP) CYK-4/MgcRacGAP and the microtubule motor ZEN-4/kinesin-6. Here, we show that oocyte production by the syncytial Caenorhabditis elegans germline requires CYK-4 but not ZEN-4, which contrasts with cytokinesis, where both are essential. Longitudinal imaging after conditional inactivation revealed that CYK-4 activity is important for oocyte cellularization, but not for the cytokinesis-like events that generate syncytial compartments. CYK-4’s lipid-binding C1 domain and the GTPase-binding interface of its GAP domain were both required to target CYK-4 to intercellular bridges and to cellularize oocytes. These results suggest that the conserved C1-GAP region of CYK-4 constitutes a targeting module required for closure of intercellular bridges in germline syncytia.
Collapse
Affiliation(s)
- Kian-Yong Lee
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Rebecca A Green
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, United States
| | - J Sebastian Gomez-Cavazos
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Irina Kolotuev
- Microscopy Rennes Imaging Center and Biosit, University of Rennes 1, Rennes, France
| | - Shaohe Wang
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Arshad Desai
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, United States
| | - Karen Oegema
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| |
Collapse
|
52
|
Dix CL, Matthews HK, Uroz M, McLaren S, Wolf L, Heatley N, Win Z, Almada P, Henriques R, Boutros M, Trepat X, Baum B. The Role of Mitotic Cell-Substrate Adhesion Re-modeling in Animal Cell Division. Dev Cell 2018; 45:132-145.e3. [PMID: 29634933 DOI: 10.1016/j.devcel.2018.03.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 01/17/2018] [Accepted: 03/13/2018] [Indexed: 12/24/2022]
Abstract
Animal cells undergo a dramatic series of shape changes as they divide, which depend on re-modeling of cell-substrate adhesions. Here, we show that while focal adhesion complexes are disassembled during mitotic rounding, integrins remain in place. These integrin-rich contacts connect mitotic cells to the underlying substrate throughout mitosis, guide polarized cell migration following mitotic exit, and are functionally important, since adherent cells undergo division failure when removed from the substrate. Further, the ability of cells to re-spread along pre-existing adhesive contacts is essential for division in cells compromised in their ability to construct a RhoGEF-dependent (Ect2) actomyosin ring. As a result, following Ect2 depletion, cells fail to divide on small adhesive islands but successfully divide on larger patterns, as the connection between daughter cells narrows and severs as they migrate away from one another. In this way, regulated re-modeling of cell-substrate adhesions during mitotic rounding aids division in animal cells.
Collapse
Affiliation(s)
- Christina L Dix
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Helen K Matthews
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Marina Uroz
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Susannah McLaren
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Lucie Wolf
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department for Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg 69120, Germany
| | - Nicholas Heatley
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Zaw Win
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Pedro Almada
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Ricardo Henriques
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department for Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg 69120, Germany
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona 08036, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona 08028, Spain
| | - Buzz Baum
- MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK.
| |
Collapse
|
53
|
Comparative evaluation of three proliferation markers, Ki-67, TOP2A, and RacGAP1, in bronchopulmonary neuroendocrine neoplasms: Issues and prospects. Oncotarget 2018; 7:41959-41973. [PMID: 27259241 PMCID: PMC5173108 DOI: 10.18632/oncotarget.9747] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 05/16/2016] [Indexed: 02/06/2023] Open
Abstract
The classification of bronchopulmonary neuroendocrine neoplasms (BP-NEN) into four tumor entities (typical carcinoids (TC), atypical carcinoids (AC), small cell lung cancers (SCLC), large cell neuroendocrine lung carcinomas (LCNEC)) is difficult to perform accurately, but important for prognostic statements and therapeutic management decisions. In this regard, we compared the expression of three proliferation markers, Ki-67, Topoisomerase II alpha (TOP2A), and RacGAP1, in a series of tumor samples from 104 BP-NEN patients (24 TC, 21 AC, 52 SCLC, 7 LCNEC) using different evaluation methods (immunohistochemistry (IHC): Average evaluation, Hotspot evaluation, digital image analysis; RT-qPCR). The results indicated that all three markers had increased protein and mRNA expression with poorer differentiation and correlated well with each other, as well as with grading, staging, and poor survival. Compared with Ki-67 and TOP2A, RacGAP1 allowed for a clearer prognostic statement. The cut-off limits obtained for Ki-67-Average (IHC) were TC-AC 1.5, AC-SCLC 19, and AC-LCNEC 23.5. The Hotspot evaluation generated equal to higher, the digital image analysis generally lower between-entity cut-off limits. All three markers enabled a clear-cut differentiation between the BP-NEN entities, and all methods evaluated were suitable for marker assessment. However, to define optimal cut-off limits, the Ki-67 evaluation methods should be standardized. RacGAP1 appeared to be a new marker with great potential.
Collapse
|
54
|
Li Y, Cai X, Chen B, Gu H, Liu C. Overexpression of epithelial cell transforming 2 protein in colorectal carcinoma predicts a poor prognosis. Exp Ther Med 2017; 14:4862-4868. [PMID: 29109759 PMCID: PMC5663027 DOI: 10.3892/etm.2017.5132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 02/17/2017] [Indexed: 02/07/2023] Open
Abstract
Epithelial cell transforming 2 (Ect2) protein is a member of the human diffuse B-cell lymphoma family of guanine nucleotide exchange factors, which activate the Ras homolog gene family of small GTPases; however, the clinical implications of Ect2 in colorectal carcinoma (CRC) are unclear. The present study aimed to determine the relationship between Ect2 expression and prognosis in patients with CRC. Western blot analysis and immunohistochemistry assays were used to determine the expression of Ect2 in CRC and paired non-cancerous tissues from 66 patients. The correlation between Ect2 expression and clinicopathological parameters was assessed using χ2 tests. Patient survival was determined using the Kaplan-Meier method and log-rank test. Cox regression was used for multivariate analysis of prognostic factors. Results demonstrated that Ect2 protein was highly expressed in human CRC samples [29/45 (64.45%)] and significantly correlated with a poor prognosis (P<0.05). Compared with normal tissues, CRC tissues demonstrated higher expression levels of Ect2 mRNA [44/66 (66.67%)]. In addition, highly-expressed Ect2 was significantly associated with recurrence (P=0.023) and invasion (P=0.008) of CRC. High Ect2 expression levels in patients were associated with poorer overall survival (OS) and disease-free survival (DFS) compared with lower expression levels of Ect2. Based on multivariate analysis, Ect2 overexpression was significantly correlated with OS and DFS (P=0.015 and 0.020, respectively). In conclusion, Ect2 overexpression is an independent and important prognostic factor for OS and DFS in patients with CRC.
Collapse
Affiliation(s)
- Yiming Li
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China.,Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Xiangjun Cai
- Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Bo Chen
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hanbo Gu
- Department of General Surgery, 202 Hospital of People's Liberation Army, Shenyang, Liaoning 110812, P.R. China
| | - Caigang Liu
- Division of Breast Surgery, Department of Surgical Oncology, General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
55
|
Abstract
Cytokinesis in metazoan cells is mediated by an actomyosin-based contractile ring that assembles in response to activation of the small GTPase RhoA. The guanine nucleotide exchange factor that activates RhoA during cytokinesis, ECT-2, is highly regulated. In most metazoan cells, with the notable exception of the early
Caenorhabditis elegans embryo, RhoA activation and furrow ingression require the centralspindlin complex. This exception is due to the existence of a parallel pathway for RhoA activation in
C. elegans. Centralspindlin contains CYK-4 which contains a predicted Rho family GTPase-activating protein (GAP) domain. The function of this domain has been the subject of considerable debate. Some publications suggest that the GAP domain promotes RhoA activation (for example, Zhang and Glotzer, 2015; Loria, Longhini and Glotzer, 2012), whereas others suggest that it functions to inactivate the GTPase Rac1 (for example, Zhuravlev
et al., 2017). Here, we review the mechanisms underlying RhoA activation during cytokinesis, primarily focusing on data in
C. elegans. We highlight the importance of considering the parallel pathway for RhoA activation and detailed analyses of
cyk-4 mutant phenotypes when evaluating the role of the GAP domain of CYK-4.
Collapse
Affiliation(s)
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
56
|
Abstract
SUMMARYCell division-cytokinesis-involves large-scale rearrangements of the entire cell. Primarily driven by cytoskeletal proteins, cytokinesis also depends on topological rearrangements of the plasma membrane, which are coordinated with nuclear division in both space and time. Despite the fundamental nature of the process, different types of eukaryotic cells show variations in both the structural mechanisms of cytokinesis and the regulatory controls. In animal cells and fungi, a contractile actomyosin-based structure plays a central, albeit flexible, role. Here, the underlying molecular mechanisms are summarized and integrated and common themes are highlighted.
Collapse
Affiliation(s)
- Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
57
|
Wu CG, Chen H, Guo F, Yadav VK, Mcilwain SJ, Rowse M, Choudhary A, Lin Z, Li Y, Gu T, Zheng A, Xu Q, Lee W, Resch E, Johnson B, Day J, Ge Y, Ong IM, Burkard ME, Ivarsson Y, Xing Y. PP2A-B' holoenzyme substrate recognition, regulation and role in cytokinesis. Cell Discov 2017; 3:17027. [PMID: 28884018 PMCID: PMC5586252 DOI: 10.1038/celldisc.2017.27] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a major Ser/Thr phosphatase; it forms diverse heterotrimeric holoenzymes that counteract kinase actions. Using a peptidome that tiles the disordered regions of the human proteome, we identified proteins containing [LMFI]xx[ILV]xEx motifs that serve as interaction sites for B′-family PP2A regulatory subunits and holoenzymes. The B′-binding motifs have important roles in substrate recognition and in competitive inhibition of substrate binding. With more than 100 novel ligands identified, we confirmed that the recently identified LxxIxEx B′α-binding motifs serve as common binding sites for B′ subunits with minor variations, and that S/T phosphorylation or D/E residues at positions 2, 7, 8 and 9 of the motifs reinforce interactions. Hundreds of proteins in the human proteome harbor intrinsic or phosphorylation-responsive B′-interaction motifs, and localize at distinct cellular organelles, such as midbody, predicting kinase-facilitated recruitment of PP2A-B′ holoenzymes for tight spatiotemporal control of phosphorylation at mitosis and cytokinesis. Moroever, Polo-like kinase 1-mediated phosphorylation of Cyk4/RACGAP1, a centralspindlin component at the midbody, facilitates binding of both RhoA guanine nucleotide exchange factor (epithelial cell transforming sequence 2 (Ect2)) and PP2A-B′ that in turn dephosphorylates Cyk4 and disrupts Ect2 binding. This feedback signaling loop precisely controls RhoA activation and specifies a restricted region for cleavage furrow ingression. Our results provide a framework for further investigation of diverse signaling circuits formed by PP2A-B′ holoenzymes in various cellular processes.
Collapse
Affiliation(s)
- Cheng-Guo Wu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA.,Biophysics Program, University of Wisconsin at Madison, Madison, WI, USA
| | - Hui Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Feng Guo
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Vikash K Yadav
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Sean J Mcilwain
- Biostatistics and Medical Informatics, Wisconsin Institutes of Medical Research, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Michael Rowse
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Alka Choudhary
- Department of Medicine, Hematology/Oncology, UW Carbone Cancer Center, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Yitong Li
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Tingjia Gu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Aiping Zheng
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Qingge Xu
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Woojong Lee
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Eduard Resch
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Frankfurt am Main, Germany
| | - Benjamin Johnson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Jenny Day
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, Human Proteomic Program, School of Medicine and Public Health, Madison, WI, USA
| | - Irene M Ong
- Biostatistics and Medical Informatics, Wisconsin Institutes of Medical Research, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Mark E Burkard
- Department of Medicine, Hematology/Oncology, UW Carbone Cancer Center, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Ylva Ivarsson
- Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Yongna Xing
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA.,Biophysics Program, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
58
|
de Cárcer G, Wachowicz P, Martínez-Martínez S, Oller J, Méndez-Barbero N, Escobar B, González-Loyola A, Takaki T, El Bakkali A, Cámara JA, Jiménez-Borreguero LJ, Bustelo XR, Cañamero M, Mulero F, de Los Ángeles Sevilla M, Montero MJ, Redondo JM, Malumbres M. Plk1 regulates contraction of postmitotic smooth muscle cells and is required for vascular homeostasis. Nat Med 2017; 23:964-974. [PMID: 28692064 DOI: 10.1038/nm.4364] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 06/13/2017] [Indexed: 12/19/2022]
Abstract
Polo-like kinase 1 (PLK1), an essential regulator of cell division, is currently undergoing clinical evaluation as a target for cancer therapy. We report an unexpected function of Plk1 in sustaining cardiovascular homeostasis. Plk1 haploinsufficiency in mice did not induce obvious cell proliferation defects but did result in arterial structural alterations, which frequently led to aortic rupture and death. Specific ablation of Plk1 in vascular smooth muscle cells (VSMCs) led to reduced arterial elasticity, hypotension, and an impaired arterial response to angiotensin II in vivo. Mechanistically, we found that Plk1 regulated angiotensin II-dependent activation of RhoA and actomyosin dynamics in VSMCs in a mitosis-independent manner. This regulation depended on Plk1 kinase activity, and the administration of small-molecule Plk1 inhibitors to angiotensin II-treated mice led to reduced arterial fitness and an elevated risk of aneurysm and aortic rupture. We thus conclude that a partial reduction of Plk1 activity that does not block cell division can nevertheless impair aortic homeostasis. Our findings have potentially important implications for current approaches aimed at PLK1 inhibition for cancer therapy.
Collapse
Affiliation(s)
- Guillermo de Cárcer
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Paulina Wachowicz
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sara Martínez-Martínez
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Jorge Oller
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Nerea Méndez-Barbero
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
| | - Beatriz Escobar
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Tohru Takaki
- Clare Hall Laboratories, London Research Institute, London, UK
| | - Aicha El Bakkali
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Juan A Cámara
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Luis J Jiménez-Borreguero
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
- Advanced Imaging Unit, Spanish National Cardiovascular Centre (CNIC), and Cardiac Imaging Department, Hospital de la Princesa, Madrid, Spain
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer de Salamanca, University of Salamanca-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marta Cañamero
- Comparative Pathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María de Los Ángeles Sevilla
- Department of Physiology and Pharmacology and Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - María Jose Montero
- Department of Physiology and Pharmacology and Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Spanish National Cardiovascular Centre (CNIC), Madrid, Spain
- Centro de Investigaciones Biomédicas en RED (CIBERCV), Madrid, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
59
|
Kumar S, Sharma G, Chakraborty C, Sharma AR, Kim J. Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget 2017; 8:37942-37962. [PMID: 28415805 PMCID: PMC5514964 DOI: 10.18632/oncotarget.16290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/03/2017] [Indexed: 12/04/2022] Open
Abstract
Polo-like kinase 1 (PLK-1) is a well-known (Ser/Thr) mitotic protein kinase and is considered as a proto-oncogene. As hyper-activation of PLK-1 is broadly associated with poor prognosis and cancer progression, it is one of the most extensively studied mitotic kinases. During mitosis, PLK-1 regulates various cell cycle events, such as spindle pole maturation, chromosome segregation and cytokinesis. However, studies have demonstrated that the role of PLK-1 is not only restricted to mitosis, but PLK-1 can also regulate other vital events beyond mitosis, including transcription, translation, ciliogenesis, checkpoint adaptation and recovery, apoptosis, chromosomes dynamics etc. Recent reviews have tried to define the regulatory role of PLK-1 during mitosis progression and tumorigenesis, but its' functional role beyond mitosis is still largely unexplored. PLK-1 can regulate the activity of many proteins that work outside of its conventional territory. The dysregulation of these proteins can cause diseases such as Alzheimer's disease, tumorigenesis etc. and may also lead to drug resistance. Thus, in this review, we discussed the versatile role of PLK-1 and tried to collect data to validate its' functional role in cell cycle regulation apart from mitosis.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Garima Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Ashish Ranjan Sharma
- Institute For Skeletal Aging & Orthopedic Surgery, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, Hallym University, College of Medicine, Chucheonsi, Gangwondo, Republic of Korea
| |
Collapse
|
60
|
Luo R, Reed CE, Sload JA, Wordeman L, Randazzo PA, Chen PW. Arf GAPs and molecular motors. Small GTPases 2017; 10:196-209. [PMID: 28430047 DOI: 10.1080/21541248.2017.1308850] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Arf GTPase-activating proteins (Arf GAPs) were first identified as regulators of the small GTP-binding proteins ADP-ribosylation factors (Arfs). The Arf GAPs are a large family of proteins in metazoans, outnumbering the Arfs that they regulate. The members of the Arf GAP family have complex domain structures and some have been implicated in particular cellular functions, such as cell migration, or with particular pathologies, such as tumor invasion and metastasis. The specific effects of Arfs sometimes depend on the Arf GAP involved in their regulation. These observations have led to speculation that the Arf GAPs themselves may affect cellular activities in capacities beyond the regulation of Arfs. Recently, 2 Arf GAPs, ASAP1 and AGAP1, have been found to bind directly to and influence the activity of myosins and kinesins, motor proteins associated with filamentous actin and microtubules, respectively. The Arf GAP-motor protein interaction is critical for cellular behaviors involving the actin cytoskeleton and microtubules, such as cell migration and other cell movements. Arfs, then, may function with molecular motors through Arf GAPs to regulate microtubule and actin remodeling.
Collapse
Affiliation(s)
- Ruibai Luo
- a Laboratory of Cellular and Molecular Biology , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Christine E Reed
- c Department of Biology , Williams College , Williamstown , MA , USA
| | - Jeffrey A Sload
- c Department of Biology , Williams College , Williamstown , MA , USA
| | - Linda Wordeman
- b Department of Physiology and Biophysics , University of Washington School of Medicine , Seattle , WA , USA
| | - Paul A Randazzo
- a Laboratory of Cellular and Molecular Biology , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Pei-Wen Chen
- c Department of Biology , Williams College , Williamstown , MA , USA
| |
Collapse
|
61
|
Zhuravlev Y, Hirsch SM, Jordan SN, Dumont J, Shirasu-Hiza M, Canman JC. CYK-4 regulates Rac, but not Rho, during cytokinesis. Mol Biol Cell 2017; 28:1258-1270. [PMID: 28298491 PMCID: PMC5415020 DOI: 10.1091/mbc.e17-01-0020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 12/18/2022] Open
Abstract
The roles of the Rho-family GAP CYK-4 and small GTPase Rac during cytokinesis are examined in Caenorhabditis elegans embryos. CYK-4 opposes Rac (and potentially Cdc42) activity during cytokinesis. There is no evidence that CYK-4 is upstream of Rho activity or that Rac disruption is a general suppressor of cytokinesis failure. Cytokinesis is driven by constriction of an actomyosin contractile ring that is controlled by Rho-family small GTPases. Rho, activated by the guanine-nucleotide exchange factor ECT-2, is upstream of both myosin-II activation and diaphanous formin-mediated filamentous actin (f-actin) assembly, which drive ring constriction. The role for Rac and its regulators is more controversial, but, based on the finding that Rac inactivation can rescue cytokinesis failure when the GTPase-activating protein (GAP) CYK-4 is disrupted, Rac activity was proposed to be inhibitory to contractile ring constriction and thus specifically inactivated by CYK-4 at the division plane. An alternative model proposes that Rac inactivation generally rescues cytokinesis failure by reducing cortical tension, thus making it easier for the cell to divide when ring constriction is compromised. In this alternative model, CYK-4 was instead proposed to activate Rho by binding ECT-2. Using a combination of time-lapse in vivo single-cell analysis and Caenorhabditis elegans genetics, our evidence does not support this alternative model. First, we found that Rac disruption does not generally rescue cytokinesis failure: inhibition of Rac specifically rescues cytokinesis failure due to disruption of CYK-4 or ECT-2 but does not rescue cytokinesis failure due to disruption of two other contractile ring components, the Rho effectors diaphanous formin and myosin-II. Second, if CYK-4 regulates cytokinesis through Rho rather than Rac, then CYK-4 inhibition should decrease levels of downstream targets of Rho. Inconsistent with this, we found no change in the levels of f-actin or myosin-II at the division plane when CYK-4 GAP activity was reduced, suggesting that CYK-4 is not upstream of ECT-2/Rho activation. Instead, we found that the rescue of cytokinesis in CYK-4 mutants by Rac inactivation was Cdc42 dependent. Together our data suggest that CYK-4 GAP activity opposes Rac (and perhaps Cdc42) during cytokinesis.
Collapse
Affiliation(s)
- Yelena Zhuravlev
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Sophia M Hirsch
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Shawn N Jordan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Julien Dumont
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
62
|
Thieleke-Matos C, Osório DS, Carvalho AX, Morais-de-Sá E. Emerging Mechanisms and Roles for Asymmetric Cytokinesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:297-345. [PMID: 28526136 DOI: 10.1016/bs.ircmb.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytokinesis completes cell division by physically separating the contents of the mother cell between the two daughter cells. This event requires the highly coordinated reorganization of the cytoskeleton within a precise window of time to ensure faithful genomic segregation. In addition, recent progress in the field highlighted the importance of cytokinesis in providing particularly important cues in the context of multicellular tissues. The organization of the cytokinetic machinery and the asymmetric localization or inheritance of the midbody remnants is critical to define the spatial distribution of mechanical and biochemical signals. After a brief overview of the conserved steps of animal cytokinesis, we review the mechanisms controlling polarized cytokinesis focusing on the challenges of epithelial cytokinesis. Finally, we discuss the significance of these asymmetries in defining embryonic body axes, determining cell fate, and ensuring the correct propagation of epithelial organization during proliferation.
Collapse
Affiliation(s)
- C Thieleke-Matos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - D S Osório
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A X Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - E Morais-de-Sá
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
63
|
Mishima M. Preparation of centralspindlin as an active heterotetramer of kinesin and GTPase activating protein subunits for in vitro structural and functional assays. Methods Cell Biol 2017; 137:371-385. [DOI: 10.1016/bs.mcb.2016.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
64
|
Kotýnková K, Su KC, West SC, Petronczki M. Plasma Membrane Association but Not Midzone Recruitment of RhoGEF ECT2 Is Essential for Cytokinesis. Cell Rep 2016; 17:2672-2686. [PMID: 27926870 PMCID: PMC5177604 DOI: 10.1016/j.celrep.2016.11.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/29/2016] [Accepted: 11/07/2016] [Indexed: 11/27/2022] Open
Abstract
Cytokinesis, the final step of cell division, begins with the formation of a cleavage furrow. How the mitotic spindle specifies the furrow at the equator in animal cells remains unknown. Current models propose that the concentration of the RhoGEF ECT2 at the spindle midzone and the equatorial plasma membrane directs furrow formation. Using chemical genetic and optogenetic tools, we demonstrate that the association of ECT2 with the plasma membrane during anaphase is required and sufficient for cytokinesis. Local membrane targeting of ECT2 leads to unilateral furrowing, highlighting the importance of local ECT2 activity. ECT2 mutations that prevent centralspindlin binding compromise concentration of ECT2 at the midzone and equatorial membrane but sustain cytokinesis. While the association of ECT2 with the plasma membrane is essential for cytokinesis, our data suggest that ECT2 recruitment to the spindle midzone is insufficient to account for equatorial furrowing and may act redundantly with yet-uncharacterized signals.
Collapse
Affiliation(s)
- Kristýna Kotýnková
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; DNA Recombination and Repair Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kuan-Chung Su
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; Whitehead Institute and Department of Biology, MIT, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Stephen C West
- DNA Recombination and Repair Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mark Petronczki
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Herts EN6 3LD, UK; Boehringer Ingelheim RCV, Dr.-Boehringer-Gasse 5-11, 1121 Vienna, Austria.
| |
Collapse
|
65
|
Warga RM, Wicklund A, Webster SE, Kane DA. Progressive loss of RacGAP1/ ogre activity has sequential effects on cytokinesis and zebrafish development. Dev Biol 2016; 418:307-22. [PMID: 27339293 DOI: 10.1016/j.ydbio.2016.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022]
|
66
|
Hertz EPT, Kruse T, Davey NE, López-Méndez B, Sigurðsson JO, Montoya G, Olsen JV, Nilsson J. A Conserved Motif Provides Binding Specificity to the PP2A-B56 Phosphatase. Mol Cell 2016; 63:686-695. [PMID: 27453045 DOI: 10.1016/j.molcel.2016.06.024] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/05/2016] [Accepted: 06/15/2016] [Indexed: 01/17/2023]
Abstract
Dynamic protein phosphorylation is a fundamental mechanism regulating biological processes in all organisms. Protein phosphatase 2A (PP2A) is the main source of phosphatase activity in the cell, but the molecular details of substrate recognition are unknown. Here, we report that a conserved surface-exposed pocket on PP2A regulatory B56 subunits binds to a consensus sequence on interacting proteins, which we term the LxxIxE motif. The composition of the motif modulates the affinity for B56, which in turn determines the phosphorylation status of associated substrates. Phosphorylation of amino acid residues within the motif increases B56 binding, allowing integration of kinase and phosphatase activity. We identify conserved LxxIxE motifs in essential proteins throughout the eukaryotic domain of life and in human viruses, suggesting that the motifs are required for basic cellular function. Our study provides a molecular description of PP2A binding specificity with broad implications for understanding signaling in eukaryotes.
Collapse
Affiliation(s)
- Emil Peter Thrane Hertz
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Thomas Kruse
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Norman E Davey
- Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Dublin 4, Ireland
| | - Blanca López-Méndez
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jón Otti Sigurðsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Guillermo Montoya
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
67
|
An Amino-Terminal Polo Kinase Interaction Motif Acts in the Regulation of Centrosome Formation and Reveals a Novel Function for centrosomin (cnn) in Drosophila. Genetics 2016; 201:685-706. [PMID: 26447129 DOI: 10.1534/genetics.115.181842] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The formation of the pericentriolar matrix (PCM) and a fully functional centrosome in syncytial Drosophila melanogaster embryos requires the rapid transport of Cnn during initiation of the centrosome replication cycle. We show a Cnn and Polo kinase interaction is apparently required during embryogenesis and involves the exon 1A-initiating coding exon, suggesting a subset of Cnn splice variants is regulated by Polo kinase. During PCM formation exon 1A Cnn-Long Form proteins likely bind Polo kinase before phosphorylation by Polo for Cnn transport to the centrosome. Loss of either of these interactions in a portion of the total Cnn protein pool is sufficient to remove native Cnn from the pool, thereby altering the normal localization dynamics of Cnn to the PCM. Additionally, Cnn-Short Form proteins are required for polar body formation, a process known to require Polo kinase after the completion of meiosis. Exon 1A Cnn-LF and Cnn-SF proteins, in conjunction with Polo kinase, are required at the completion of meiosis and for the formation of functional centrosomes during early embryogenesis.
Collapse
|
68
|
Wachowicz P, Fernández-Miranda G, Marugán C, Escobar B, de Cárcer G. Genetic depletion of Polo-like kinase 1 leads to embryonic lethality due to mitotic aberrancies. Bioessays 2016; 38 Suppl 1:S96-S106. [PMID: 27417127 DOI: 10.1002/bies.201670908] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/17/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022]
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase that plays multiple and essential roles during the cell division cycle. Its inhibition in cultured cells leads to severe mitotic aberrancies and cell death. Whereas previous reports suggested that Plk1 depletion in mice leads to a non-mitotic arrest in early embryos, we show here that the bi-allelic Plk1 depletion in mice certainly results in embryonic lethality due to extensive mitotic aberrations at the morula stage, including multi- and mono-polar spindles, impaired chromosome segregation and cytokinesis failure. In addition, the conditional depletion of Plk1 during mid-gestation leads also to severe mitotic aberrancies. Our data also confirms that Plk1 is completely dispensable for mitotic entry in vivo. On the other hand, Plk1 haploinsufficient mice are viable, and Plk1-heterozygous fibroblasts do not harbor any cell cycle alterations. Plk1 is overexpressed in many human tumors, suggesting a therapeutic benefit of inhibiting Plk1, and specific small-molecule inhibitors for this kinase are now being evaluated in clinical trials. Therefore, the different Plk1 mouse models here presented are a valuable tool to reexamine the relevance of the mitotic kinase Plk1 during mammalian development and animal physiology.
Collapse
Affiliation(s)
- Paulina Wachowicz
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Gonzalo Fernández-Miranda
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
| | - Carlos Marugán
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Beatriz Escobar
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Spanish National Cardiovascular Research Centre (CNIC), Madrid, Spain
| | | |
Collapse
|
69
|
Wagner E, Glotzer M. Local RhoA activation induces cytokinetic furrows independent of spindle position and cell cycle stage. J Cell Biol 2016; 213:641-9. [PMID: 27298323 PMCID: PMC4915195 DOI: 10.1083/jcb.201603025] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022] Open
Abstract
The GTPase RhoA promotes contractile ring assembly and furrow ingression during cytokinesis. Although many factors that regulate RhoA during cytokinesis have been characterized, the spatiotemporal regulatory logic remains undefined. We have developed an optogenetic probe to gain tight spatial and temporal control of RhoA activity in mammalian cells and demonstrate that cytokinetic furrowing is primarily regulated at the level of RhoA activation. Light-mediated recruitment of a RhoGEF domain to the plasma membrane leads to rapid induction of RhoA activity, leading to assembly of cytokinetic furrows that partially ingress. Furthermore, furrow formation in response to RhoA activation is not temporally or spatially restricted. RhoA activation is sufficient to generate furrows at both the cell equator and cell poles, in both metaphase and anaphase. Remarkably, furrow formation can be initiated in rounded interphase cells, but not adherent cells. These results indicate that RhoA activation is sufficient to induce assembly of functional contractile rings and that cell rounding facilitates furrow formation.
Collapse
Affiliation(s)
- Elizabeth Wagner
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
70
|
Mishima M. Centralspindlin in Rappaport’s cleavage signaling. Semin Cell Dev Biol 2016; 53:45-56. [DOI: 10.1016/j.semcdb.2016.03.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 03/02/2016] [Indexed: 02/07/2023]
|
71
|
Uehara R, Kamasaki T, Hiruma S, Poser I, Yoda K, Yajima J, Gerlich DW, Goshima G. Augmin shapes the anaphase spindle for efficient cytokinetic furrow ingression and abscission. Mol Biol Cell 2016; 27:812-27. [PMID: 26764096 PMCID: PMC4803307 DOI: 10.1091/mbc.e15-02-0101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/11/2022] Open
Abstract
During anaphase, distinct populations of microtubules (MTs) form by either centrosome-dependent or augmin-dependent nucleation. It remains largely unknown whether these different MT populations contribute distinct functions to cytokinesis. Here we show that augmin-dependent MTs are required for the progression of both furrow ingression and abscission. Augmin depletion reduced the accumulation of anillin, a contractile ring regulator at the cell equator, yet centrosomal MTs were sufficient to mediate RhoA activation at the furrow. This defect in contractile ring organization, combined with incomplete spindle pole separation during anaphase, led to impaired furrow ingression. During the late stages of cytokinesis, astral MTs formed bundles in the intercellular bridge, but these failed to assemble a focused midbody structure and did not establish tight linkage to the plasma membrane, resulting in furrow regression. Thus augmin-dependent acentrosomal MTs and centrosomal MTs contribute to nonredundant targeting mechanisms of different cytokinesis factors, which are required for the formation of a functional contractile ring and midbody.
Collapse
Affiliation(s)
- Ryota Uehara
- Creative Research Institution, Hokkaido University, Sapporo 001-0021, Japan Department of Life Sciences, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Tomoko Kamasaki
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Shota Hiruma
- Creative Research Institution, Hokkaido University, Sapporo 001-0021, Japan
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Kinya Yoda
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Junichiro Yajima
- Department of Life Sciences, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter Campus, 1030 Vienna, Austria
| | - Gohta Goshima
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| |
Collapse
|
72
|
Lasek AL, McPherson BM, Trueman NG, Burkard ME. The Functional Significance of Posttranslational Modifications on Polo-Like Kinase 1 Revealed by Chemical Genetic Complementation. PLoS One 2016; 11:e0150225. [PMID: 26919439 PMCID: PMC4769148 DOI: 10.1371/journal.pone.0150225] [Citation(s) in RCA: 11] [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: 09/29/2015] [Accepted: 02/10/2016] [Indexed: 01/19/2023] Open
Abstract
Mitosis is coordinated by carefully controlled phosphorylation and ubiquitin-mediated proteolysis. Polo-like kinase 1 (Plk1) plays a central role in regulating mitosis and cytokinesis by phosphorylating target proteins. Yet, Plk1 is itself a target for posttranslational modification by phosphorylation and ubiquitination. We developed a chemical-genetic complementation assay to evaluate the functional significance of 34 posttranslational modifications (PTMs) on human Plk1. To do this, we used human cells that solely express a modified analog-sensitive Plk1 (Plk1AS) and complemented with wildtype Plk1. The wildtype Plk1 provides cells with a functional Plk1 allele in the presence of 3-MB-PP1, a bulky ATP-analog inhibitor that specifically inhibits Plk1AS. Using this approach, we evaluated the ability of 34 singly non-modifiable Plk1 mutants to complement Plk1AS in the presence of 3-MB-PP1. Mutation of the T-loop activating residue T210 and adjacent T214 are lethal, but surprisingly individual mutation of the remaining 32 posttranslational modification sites did not disrupt the essential functions of Plk1. To evaluate redundancy, we simultaneously mutated all phosphorylation sites in the kinase domain except for T210 and T214 or all sites in the C-terminal polo-box domain (PBD). We discovered that redundant phosphorylation events within the kinase domain are required for accurate chromosome segregation in anaphase but those in the PBD are dispensable. We conclude that PTMs within the T-loop of Plk1 are essential and nonredundant, additional modifications in the kinase domain provide redundant control of Plk1 function, and those in the PBD are dispensable for essential mitotic functions of Plk1. This comprehensive evaluation of Plk1 modifications demonstrates that although phosphorylation and ubiquitination are important for mitotic progression, many individual PTMs detected in human tissue may have redundant, subtle, or dispensable roles in gene function.
Collapse
Affiliation(s)
- Amber L. Lasek
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- Department of Medicine, Hematology/Oncology Division, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| | - Brittany M. McPherson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- Department of Medicine, Hematology/Oncology Division, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| | - Natalie G. Trueman
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- Department of Medicine, Hematology/Oncology Division, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| | - Mark E. Burkard
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- Department of Medicine, Hematology/Oncology Division, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| |
Collapse
|
73
|
D'Avino PP, Capalbo L. Regulation of midbody formation and function by mitotic kinases. Semin Cell Dev Biol 2016; 53:57-63. [PMID: 26802517 DOI: 10.1016/j.semcdb.2016.01.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/14/2016] [Indexed: 12/22/2022]
Abstract
Cytokinesis is the final phase of cell division and safeguards the correct distribution of genomic and cytoplasmic materials between the two nascent daughter cells. The final separation, or abscission, of the daughter cells depends on the proper assembly of an organelle at the intercellular bridge, the midbody, which acts as a platform for the recruitment and organisation of various proteins involved in both the control and execution of the abscission process. Recent studies have led to the identification of the mechanisms, signalling pathways and molecules that control the two tightly linked processes of midbody formation and abscission. Here we review our current knowledge of the role that mitotic kinases play in these processes and offer our perspectives on the potential future challenges that await researchers in the field.
Collapse
Affiliation(s)
- Pier Paolo D'Avino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Luisa Capalbo
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
74
|
Fischer M, Quaas M, Nickel A, Engeland K. Indirect p53-dependent transcriptional repression of Survivin, CDC25C, and PLK1 genes requires the cyclin-dependent kinase inhibitor p21/CDKN1A and CDE/CHR promoter sites binding the DREAM complex. Oncotarget 2015; 6:41402-17. [PMID: 26595675 PMCID: PMC4747163 DOI: 10.18632/oncotarget.6356] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022] Open
Abstract
The transcription factor p53 is central to cell cycle control by downregulation of cell cycle-promoting genes upon cell stress such as DNA damage. Survivin (BIRC5), CDC25C, and PLK1 encode important cell cycle regulators that are repressed following p53 activation. Here, we provide evidence that p53-dependent repression of these genes requires activation of p21 (CDKN1A, WAF1, CIP1). Chromatin immunoprecipitation (ChIP) data indicate that promoter binding of B-MYB switches to binding of E2F4 and p130 resulting in a replacement of the MMB (Myb-MuvB) by the DREAM complex. We demonstrate that this replacement depends on p21. Furthermore, transcriptional repression by p53 requires intact DREAM binding sites in the target promoters. The CDE and CHR cell cycle promoter elements are the sites for DREAM binding. These elements as well as the p53 response of Survivin, CDC25C, and PLK1 are evolutionarily conserved. No binding of p53 to these genes is detected by ChIP and mutation of proposed p53 binding sites does not alter the p53 response. Thus, a mechanism for direct p53-dependent transcriptional repression is not supported by the data. In contrast, repression by DREAM is consistent with most previous findings and unifies models based on p21-, E2F4-, p130-, and CDE/CHR-dependent repression by p53. In conclusion, the presented data suggest that the p53-p21-DREAM-CDE/CHR pathway regulates p53-dependent repression of Survivin, CDC25C, and PLK1.
Collapse
Affiliation(s)
- Martin Fischer
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
- Department of Medical Oncology, Dana–Farber Cancer Institute, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marianne Quaas
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Annina Nickel
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Kurt Engeland
- Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| |
Collapse
|
75
|
Spindle Assembly and Chromosome Segregation Requires Central Spindle Proteins in Drosophila Oocytes. Genetics 2015; 202:61-75. [PMID: 26564158 DOI: 10.1534/genetics.115.181081] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/06/2015] [Indexed: 11/18/2022] Open
Abstract
Oocytes segregate chromosomes in the absence of centrosomes. In this situation, the chromosomes direct spindle assembly. It is still unclear in this system which factors are required for homologous chromosome bi-orientation and spindle assembly. The Drosophila kinesin-6 protein Subito, although nonessential for mitotic spindle assembly, is required to organize a bipolar meiotic spindle and chromosome bi-orientation in oocytes. Along with the chromosomal passenger complex (CPC), Subito is an important part of the metaphase I central spindle. In this study we have conducted genetic screens to identify genes that interact with subito or the CPC component Incenp. In addition, the meiotic mutant phenotype for some of the genes identified in these screens were characterized. We show, in part through the use of a heat-shock-inducible system, that the Centralspindlin component RacGAP50C and downstream regulators of cytokinesis Rho1, Sticky, and RhoGEF2 are required for homologous chromosome bi-orientation in metaphase I oocytes. This suggests a novel function for proteins normally involved in mitotic cell division in the regulation of microtubule-chromosome interactions. We also show that the kinetochore protein, Polo kinase, is required for maintaining chromosome alignment and spindle organization in metaphase I oocytes. In combination our results support a model where the meiotic central spindle and associated proteins are essential for acentrosomal chromosome segregation.
Collapse
|
76
|
van Adrichem AJ, Fagerholm A, Turunen L, Lehto A, Saarela J, Koskinen A, Repasky GA, Wennerberg K. Discovery of MINC1, a GTPase-activating protein small molecule inhibitor, targeting MgcRacGAP. Comb Chem High Throughput Screen 2015; 18:3-17. [PMID: 25479424 PMCID: PMC4435052 DOI: 10.2174/1386207318666141205112730] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/12/2014] [Accepted: 12/03/2014] [Indexed: 12/02/2022]
Abstract
The Rho family of Ras superfamily small GTPases regulates a broad range of biological processes such as migration, differentiation, cell growth and cell survival. Therefore, the availability of small molecule modulators as tool compounds could greatly enhance research on these proteins and their biological function. To this end, we designed a biochemical, high throughput screening assay with complementary follow-up assays to identify small molecule compounds inhibiting MgcRacGAP, a Rho family GTPase activating protein involved in cytokinesis and transcriptionally upregulated in many cancers. We first performed an in-house screen of 20,480 compounds, and later we tested the assay against 342,046 compounds from the NIH Molecular Libraries Small Molecule Repository. Primary screening hit rates were about 1% with the majority of those affecting the primary readout, an enzyme-coupled GDP detection assay. After orthogonal and counter screens, we identified two hits with high selectivity towards MgcRacGAP, compared with other RhoGAPs, and potencies in the low micromolar range. The most promising hit, termed MINC1, was then examined with cell-based testing where it was observed to induce an increased rate of cytokinetic failure and multinucleation in addition to other cell division defects, suggesting that it may act as an MgcRacGAP inhibitor also in cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Krister Wennerberg
- Institute for Molecular Medicine Finland FIMM, Nordic EMBL Partnership for Molecular Medicine, Biomedicum Helsinki 2U, P.O. Box 20 (Tukholmankatu 8), FI- 00014, University of Helsinki, Finland.
| |
Collapse
|
77
|
Archambault V, Lépine G, Kachaner D. Understanding the Polo Kinase machine. Oncogene 2015; 34:4799-807. [PMID: 25619835 DOI: 10.1038/onc.2014.451] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/27/2014] [Accepted: 11/28/2014] [Indexed: 12/26/2022]
Abstract
The Polo Kinase is a central regulator of cell division required for several events of mitosis and cytokinesis. In addition to a kinase domain (KD), Polo-like kinases (Plks) comprise a Polo-Box domain (PBD), which mediates protein interactions with targets and regulators of Plks. In all organisms that contain Plks, one Plk family member fulfills several essential functions in the regulation of cell division, and here we refer to this conserved protein as Polo Kinase (Plk1 in humans). The PBD and the KD are capable of both cooperation and mutual inhibition in their functions. Crystal structures of the PBD, the KD and, recently, a PBD-KD complex have helped understanding the inner workings of the Polo Kinase. In parallel, an impressive array of molecular mechanisms has been found to mediate the regulation of the protein. Moreover, the targeting of Polo Kinase in the development of anti-cancer drugs has yielded several molecules with which to chemically modulate Polo Kinase to study its biological functions. Here we review our current understanding of the protein function and regulation of Polo Kinase as a fascinating molecular device in control of cell division.
Collapse
Affiliation(s)
- V Archambault
- Institut de recherche en immunologie et en cancérologie, Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - G Lépine
- Institut de recherche en immunologie et en cancérologie, Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - D Kachaner
- Institut de recherche en immunologie et en cancérologie, Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
78
|
Zhang D, Glotzer M. The RhoGAP activity of CYK-4/MgcRacGAP functions non-canonically by promoting RhoA activation during cytokinesis. eLife 2015; 4. [PMID: 26252513 PMCID: PMC4552957 DOI: 10.7554/elife.08898] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/06/2015] [Indexed: 12/29/2022] Open
Abstract
Cytokinesis requires activation of the GTPase RhoA. ECT-2, the exchange factor responsible for RhoA activation, is regulated to ensure spatiotemporal control of contractile ring assembly. Centralspindlin, composed of the Rho family GTPase-activating protein (RhoGAP) MgcRacGAP/CYK-4 and the kinesin MKLP1/ZEN-4, is known to activate ECT-2, but the underlying mechanism is not understood. We report that ECT-2-mediated RhoA activation depends on the ability of CYK-4 to localize to the plasma membrane, bind RhoA, and promote GTP hydrolysis by RhoA. Defects resulting from loss of CYK-4 RhoGAP activity can be rescued by activating mutations in ECT-2 or depletion of RGA-3/4, which functions as a conventional RhoGAP for RhoA. Consistent with CYK-4 RhoGAP activity contributing to GEF activation, the catalytic domains of CYK-4 and ECT-2 directly interact. Thus, counterintuitively, CYK-4 RhoGAP activity promotes RhoA activation. We propose that the most active form of the cytokinetic RhoGEF involves complex formation between ECT-2, centralspindlin and RhoA. DOI:http://dx.doi.org/10.7554/eLife.08898.001 Cell division is a process in which a cell splits to form two daughter cells. In most cases, the cell first duplicates its genetic material and then the two copies are pulled to opposite ends of the cell. A ring of protein filaments—called the contractile ring—then assembles to form a band around the cell at the site of the division. This ring contracts and the force generated separates the cells in a step known as cytokinesis. A protein belonging to the Rho family, called RhoA, is essential for cytokinesis because it controls the formation of the contractile ring. Rho proteins are switched on by the activities of other proteins called guanine nucleotide exchange factors. Another group of proteins known as ‘GTPase activating proteins’ (or GAPs for short) generally act to promote the ability of Rho proteins to turn themselves off. In animals and other multicellular organisms, a GAP called CYK-4 largely concentrates on the spindle midzone, but some of the protein also moves to part of the cell membrane near the future site of cell division. It binds to a guanine nucleotide exchange factor called ECT-2 to switch RhoA on, which in turn promotes the formation of the contractile ring. However, it is not clear why a protein that activates RhoA is also able to trigger its inactivation. In this study, Zhang and Glotzer studied cell division in a roundworm called Caenorhabditis elegans. The experiments show that cells that lacked the GAP activity of CYK-4 were unable to complete cytokinesis because RhoA was not fully switched on. This requirement could be bypassed in cells with mutant forms of ECT-2 that were overactive. Therefore, an activity that was thought to inactivate RhoA actually promotes its activation. Further experiments show that the section (or ‘domain’) of CYK-4 that has GAP activity interacts directly with the guanine nucleotide exchange domain of ECT2. Zhang and Glotzer suggest that this interaction stimulates ECT2 and thereby promotes the activation of RhoA. Further experiments will reveal how CYK-4 stimulates ECT-2. In addition, it will be important to determine whether other proteins with GAP domains also work in this unconventional way. DOI:http://dx.doi.org/10.7554/eLife.08898.002
Collapse
Affiliation(s)
- Donglei Zhang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, United States
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, United States
| |
Collapse
|
79
|
Basant A, Lekomtsev S, Tse YC, Zhang D, Longhini KM, Petronczki M, Glotzer M. Aurora B kinase promotes cytokinesis by inducing centralspindlin oligomers that associate with the plasma membrane. Dev Cell 2015; 33:204-15. [PMID: 25898168 DOI: 10.1016/j.devcel.2015.03.015] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/06/2015] [Accepted: 02/06/2015] [Indexed: 02/03/2023]
Abstract
In metazoans, cytokinesis is triggered by activation of the GTPase RhoA at the equatorial plasma membrane. ECT-2, the guanine nucleotide exchange factor (GEF) required for RhoA activation, is activated by the centralspindlin complex that concentrates on spindle midzone microtubules. However, these microtubules and the plasma membrane are not generally in apposition, and thus the mechanism by which RhoA is activated at the cell equator remains unknown. Here we report that a regulated pool of membrane-bound, oligomeric centralspindlin stimulates RhoA activation. The membrane-binding C1 domain of CYK-4, a centralspindlin component, promotes furrow initiation in C. elegans embryos and human cells. Membrane localization of centralspindlin oligomers is globally inhibited by PAR-5/14-3-3. This activity is antagonized by the chromosome passenger complex (CPC), resulting in RhoA activation at the nascent cleavage site. Therefore, CPC-directed centralspindlin oligomerization during anaphase induces contractile ring assembly at the membrane.
Collapse
Affiliation(s)
- Angika Basant
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Sergey Lekomtsev
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Hertfordshire EN6 3LD, UK
| | - Yu Chung Tse
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Donglei Zhang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Katrina M Longhini
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Mark Petronczki
- Cell Division and Aneuploidy Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Blanche Lane, South Mimms, Hertfordshire EN6 3LD, UK
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
80
|
Carmena M, Lombardia MO, Ogawa H, Earnshaw WC. Polo kinase regulates the localization and activity of the chromosomal passenger complex in meiosis and mitosis in Drosophila melanogaster. Open Biol 2015; 4:140162. [PMID: 25376909 PMCID: PMC4248065 DOI: 10.1098/rsob.140162] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cell cycle progression is regulated by members of the cyclin-dependent kinase (CDK), Polo and Aurora families of protein kinases. The levels of expression and localization of the key regulatory kinases are themselves subject to very tight control. There is increasing evidence that crosstalk between the mitotic kinases provides for an additional level of regulation. We have previously shown that Aurora B activates Polo kinase at the centromere in mitosis, and that the interaction between Polo and the chromosomal passenger complex (CPC) component INCENP is essential in this activation. In this report, we show that Polo kinase is required for the correct localization and activity of the CPC in meiosis and mitosis. Study of the phenotype of different polo allele combinations compared to the effect of chemical inhibition revealed significant differences in the localization and activity of the CPC in diploid tissues. Our results shed new light on the mechanisms that control the activity of Aurora B in meiosis and mitosis.
Collapse
Affiliation(s)
- Mar Carmena
- The Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| | - Miguel Ortiz Lombardia
- Centre National de la Recherche Scientifique, Aix-Marseille Université, CNRS UMR 7257, AFMB, 163 Avenue de Luminy, 13288 Marseille, France
| | - Hiromi Ogawa
- The Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| | - William C Earnshaw
- The Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| |
Collapse
|
81
|
Asano E, Hasegawa H, Hyodo T, Ito S, Maeda M, Chen D, Takahashi M, Hamaguchi M, Senga T. SHCBP1 is required for midbody organization and cytokinesis completion. Cell Cycle 2015; 13:2744-51. [PMID: 25486361 DOI: 10.4161/15384101.2015.945840] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The centralspindlin complex, which is composed of MKLP1 and MgcRacGAP, is one of the crucial factors involved in cytokinesis initiation. Centralspindlin is localized at the middle of the central spindle during anaphase and then concentrates at the midbody to control abscission. A number of proteins that associate with centralspindlin have been identified. These associating factors regulate furrowing and abscission in coordination with centralspindlin. A recent study identified a novel centralspindlin partner, called Nessun Dorma, which is essential for germ cell cytokinesis in Drosophila melanogaster. SHCBP1 is a human ortholog of Nessun Dorma that associates with human centralspindlin. In this report, we analyzed the interaction of SHCBP1 with centralspindlin in detail and determined the regions that are required for the interaction. In addition, we demonstrate that the central region is necessary for the SHCBP1 dimerization. Both MgcRacGAP and MKLP1 are degraded once cells exit mitosis. Similarly, endogenous and exogenous SHCBP1 were degraded with mitosis progression. Interestingly, SHCBP1 expression was significantly reduced in the absence of centralspindlin, whereas centralspindlin expression was not affected by SHCBP1 knockdown. Finally, we demonstrate that SHCBP1 depletion promotes midbody structure disruption and inhibits abscission, a final stage of cytokinesis. Our study gives novel insight into the role of SHCBP in cytokinesis completion.
Collapse
Affiliation(s)
- Eri Asano
- a Division of Cancer Biology ; Nagoya University Graduate School of Medicine ; Nagoya , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Kumar M, Pushpa K, Mylavarapu SVS. Splitting the cell, building the organism: Mechanisms of cell division in metazoan embryos. IUBMB Life 2015; 67:575-87. [PMID: 26173082 PMCID: PMC5937677 DOI: 10.1002/iub.1404] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 11/12/2022]
Abstract
The unicellular metazoan zygote undergoes a series of cell divisions that are central to its development into an embryo. Differentiation of embryonic cells leads eventually to the development of a functional adult. Fate specification of pluripotent embryonic cells occurs during the early embryonic cleavage divisions in several animals. Early development is characterized by well-known stages of embryogenesis documented across animals--morulation, blastulation, and morphogenetic processes such as gastrulation, all of which contribute to differentiation and tissue specification. Despite this broad conservation, there exist clearly discernible morphological and functional differences across early embryonic stages in metazoans. Variations in the mitotic mechanisms of early embryonic cell divisions play key roles in governing these gross differences that eventually encode developmental patterns. In this review, we discuss molecular mechanisms of both karyokinesis (nuclear division) and cytokinesis (cytoplasmic separation) during early embryonic divisions. We outline the broadly conserved molecular pathways that operate in these two stages in early embryonic mitoses. In addition, we highlight mechanistic variations in these two stages across different organisms. We finally discuss outstanding questions of interest, answers to which would illuminate the role of divergent mitotic mechanisms in shaping early animal embryogenesis.
Collapse
Affiliation(s)
- Megha Kumar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Kumari Pushpa
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Sivaram V. S. Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
83
|
Liu X. Targeting Polo-Like Kinases: A Promising Therapeutic Approach for Cancer Treatment. Transl Oncol 2015; 8:185-95. [PMID: 26055176 PMCID: PMC4486469 DOI: 10.1016/j.tranon.2015.03.010] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/19/2015] [Accepted: 03/24/2015] [Indexed: 12/29/2022] Open
Abstract
Polo-like kinases (Plks) are a family of serine-threonine kinases that regulate multiple intracellular processes including DNA replication, mitosis, and stress response. Plk1, the most well understood family member, regulates numerous stages of mitosis and is overexpressed in many cancers. Plk inhibitors are currently under clinical investigation, including phase III trials of volasertib, a Plk inhibitor, in acute myeloid leukemia and rigosertib, a dual inhibitor of Plk1/phosphoinositide 3-kinase signaling pathways, in myelodysplastic syndrome. Other Plk inhibitors, including the Plk1 inhibitors GSK461364A, TKM-080301, GW843682, purpurogallin, and poloxin and the Plk4 inhibitor CFI-400945 fumarate, are in earlier clinical development. This review discusses the biologic roles of Plks in cell cycle progression and cancer, and the mechanisms of action of Plk inhibitors currently in development as cancer therapies.
Collapse
Affiliation(s)
- Xiaoqi Liu
- Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
84
|
van Buul JD, Geerts D, Huveneers S. Rho GAPs and GEFs: controling switches in endothelial cell adhesion. Cell Adh Migr 2015; 8:108-24. [PMID: 24622613 PMCID: PMC4049857 DOI: 10.4161/cam.27599] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Within blood vessels, endothelial cell–cell and cell–matrix adhesions are crucial to preserve barrier function, and these adhesions are tightly controlled during vascular development, angiogenesis, and transendothelial migration of inflammatory cells. Endothelial cellular signaling that occurs via the family of Rho GTPases coordinates these cell adhesion structures through cytoskeletal remodelling. In turn, Rho GTPases are regulated by GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). To understand how endothelial cells initiate changes in the activity of Rho GTPases, and thereby regulate cell adhesion, we will discuss the role of Rho GAPs and GEFs in vascular biology. Many potentially important Rho regulators have not been studied in detail in endothelial cells. We therefore will first overview which GAPs and GEFs are highly expressed in endothelium, based on comparative gene expression analysis of human endothelial cells compared with other tissue cell types. Subsequently, we discuss the relevance of Rho GAPs and GEFs for endothelial cell adhesion in vascular homeostasis and disease.
Collapse
Affiliation(s)
- Jaap D van Buul
- Department of Molecular Cell Biology; Sanquin Research and Swammerdam Institute for Life Sciences; University of Amsterdam; The Netherlands
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology; Erasmus University Medical Center; Rotterdam, The Netherlands
| | - Stephan Huveneers
- Department of Molecular Cell Biology; Sanquin Research and Swammerdam Institute for Life Sciences; University of Amsterdam; The Netherlands
| |
Collapse
|
85
|
Gera N, Yang A, Holtzman TS, Lee SX, Wong ET, Swanson KD. Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PLoS One 2015; 10:e0125269. [PMID: 26010837 PMCID: PMC4444126 DOI: 10.1371/journal.pone.0125269] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/23/2015] [Indexed: 01/04/2023] Open
Abstract
The anti-tumor effects of chemotherapy and radiation are thought to be mediated by triggering G1/S or G2/M cell cycle checkpoints, while spindle poisons, such as paclitaxel, block metaphase exit by initiating the spindle assembly checkpoint. In contrast, we have found that 150 kilohertz (kHz) alternating electric fields, also known as Tumor Treating Fields (TTFields), perturbed cells at the transition from metaphase to anaphase. Cells exposed to the TTFields during mitosis showed normal progression to this point, but exhibited uncontrolled membrane blebbing that coincided with metaphase exit. The ability of such alternating electric fields to affect cellular physiology is likely to be dependent on their interactions with proteins possessing high dipole moments. The mitotic Septin complex consisting of Septin 2, 6 and 7, possesses a high calculated dipole moment of 2711 Debyes (D) and plays a central role in positioning the cytokinetic cleavage furrow, and governing its contraction during ingression. We showed that during anaphase, TTFields inhibited Septin localization to the anaphase spindle midline and cytokinetic furrow, as well as its association with microtubules during cell attachment and spreading on fibronectin. After aberrant metaphase exit as a consequence of TTFields exposure, cells exhibited aberrant nuclear architecture and signs of cellular stress including an overall decrease in cellular proliferation, followed by apoptosis that was strongly influenced by the p53 mutational status. Thus, TTFields are able to diminish cell proliferation by specifically perturbing key proteins involved in cell division, leading to mitotic catastrophe and subsequent cell death.
Collapse
Affiliation(s)
- Nidhi Gera
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aaron Yang
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Talia S. Holtzman
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Sze Xian Lee
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Eric T. Wong
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Kenneth D. Swanson
- Department of Neurology, Division of Neuro-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
86
|
Breznau EB, Semack AC, Higashi T, Miller AL. MgcRacGAP restricts active RhoA at the cytokinetic furrow and both RhoA and Rac1 at cell-cell junctions in epithelial cells. Mol Biol Cell 2015; 26:2439-55. [PMID: 25947135 PMCID: PMC4571299 DOI: 10.1091/mbc.e14-11-1553] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/30/2015] [Indexed: 12/17/2022] Open
Abstract
MgcRacGAP's role in regulating the spatiotemporal dynamics of active RhoA and Rac1 in epithelial cells is investigated. MgcRacGAP's GAP activity down-regulates RhoA at the furrow and both RhoA and Rac1 at cell–cell junctions in dividing epithelial cells and is required for successful cytokinesis and cell–cell junction structure. MgcRacGAP's ability to regulate adherens junctions is dependent on GAP activity and signaling via the RhoA pathway. Localized activation of Rho GTPases is essential for multiple cellular functions, including cytokinesis and formation and maintenance of cell–cell junctions. Although MgcRacGAP (Mgc) is required for spatially confined RhoA-GTP at the equatorial cortex of dividing cells, both the target specificity of Mgc's GAP activity and the involvement of phosphorylation of Mgc at Ser-386 are controversial. In addition, Mgc's function at cell–cell junctions remains unclear. Here, using gastrula-stage Xenopus laevis embryos as a model system, we examine Mgc's role in regulating localized RhoA-GTP and Rac1-GTP in the intact vertebrate epithelium. We show that Mgc's GAP activity spatially restricts accumulation of both RhoA-GTP and Rac1-GTP in epithelial cells—RhoA at the cleavage furrow and RhoA and Rac1 at cell–cell junctions. Phosphorylation at Ser-386 does not switch the specificity of Mgc's GAP activity and is not required for successful cytokinesis. Furthermore, Mgc regulates adherens junction but not tight junction structure, and the ability to regulate adherens junctions is dependent on GAP activity and signaling via the RhoA pathway. Together these results indicate that Mgc's GAP activity down-regulates the active populations of RhoA and Rac1 at localized regions of epithelial cells and is necessary for successful cytokinesis and cell–cell junction structure.
Collapse
Affiliation(s)
- Elaina B Breznau
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ansley C Semack
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Tomohito Higashi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L Miller
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109 Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
87
|
Abstract
Cytokinesis mediates the physical separation of dividing cells after chromosome segregation. In animal cell cytokinesis, a contractile ring, mainly composed of actin and myosin filaments, ingresses a cleavage furrow midway between the two spindle poles. A distinct machinery, involving the endosomal sorting complex required for transport III (ESCRT-III), subsequently splits the plasma membrane of nascent daughter cells in a process termed abscission. Here, we provide a brief overview of early cytokinesis events in animal cells and then cover in depth recently emerging models for the assembly and function of the abscission machinery and its temporal coordination with chromosome segregation.
Collapse
Affiliation(s)
- Beata Mierzwa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria.
| |
Collapse
|
88
|
Abstract
Cell division ends with the physical separation of the two daughter cells, a process known as cytokinesis. This final event ensures that nuclear and cytoplasmic contents are accurately partitioned between the two nascent cells. Cytokinesis is one of the most dramatic changes in cell shape and requires an extensive reorganization of the cell's cytoskeleton. Here, we describe the cytoskeletal structures, factors, and signaling pathways that orchestrate this robust and yet highly dynamic process in animal cells. Finally, we discuss possible future directions in this growing area of cell division research and its implications in human diseases, including cancer.
Collapse
Affiliation(s)
- Pier Paolo D'Avino
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari c/o Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, 00185 Roma, Italy
| | - Mark Petronczki
- Cell Division and Aneuploidy Laboratory, Cancer Research UK-London Research Institute, Clare Hall Laboratories, South Mimms, Hertfordshire EN6 3LD, United Kingdom
| |
Collapse
|
89
|
Cdk1-dependent mitotic enrichment of cortical myosin II promotes cell rounding against confinement. Nat Cell Biol 2015; 17:148-59. [DOI: 10.1038/ncb3098] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/17/2014] [Indexed: 12/16/2022]
|
90
|
|
91
|
Bastos RN, Cundell MJ, Barr FA. KIF4A and PP2A-B56 form a spatially restricted feedback loop opposing Aurora B at the anaphase central spindle. ACTA ACUST UNITED AC 2014; 207:683-93. [PMID: 25512391 PMCID: PMC4274259 DOI: 10.1083/jcb.201409129] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The mitotic kinase Aurora B is concentrated at the anaphase central spindle by the kinesin MKlp2 during mitotic exit and cytokinesis. This pool of Aurora B phosphorylates substrates including the kinesin KIF4A to regulate central spindle length. In this paper, we identify a counteracting system in which PP2A-B56γ and -ε, but not PP2A-B56α, -β, and -δ, are maintained at the central spindle by KIF4A. Biochemical assays show that PP2A-B56γ can dephosphorylate the T799 Aurora B site on KIF4A and thereby counteract the Aurora B- and microtubule-stimulated ATPase activity of KIF4A. In agreement with these observations, combined silencing of PP2A-B56γ and -ε resulted in increased phosphorylation of KIF4A T799 and decreased central spindle growth in anaphase B. Furthermore, reduced turnover of regulatory phosphorylation on another Aurora B substrate MKlp1 was observed, suggesting that PP2A-B56γ and -ε play a general role opposing Aurora B at the central spindle. KIF4A and PP2A-B56γ and -ε therefore create a spatially restricted negative feedback loop counteracting Aurora B in anaphase.
Collapse
Affiliation(s)
| | - Michael J Cundell
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| |
Collapse
|
92
|
Cannet A, Schmidt S, Delaval B, Debant A. Identification of a mitotic Rac-GEF, Trio, that counteracts MgcRacGAP function during cytokinesis. Mol Biol Cell 2014; 25:4063-71. [PMID: 25355950 PMCID: PMC4263449 DOI: 10.1091/mbc.e14-06-1153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inactivation of Rac1 by MgcRacGAP at the cleavage plane is essential to ensure cytokinesis. Trio activates Rac1 in dividing cells, and its depletion rescues the cytokinesis failure induced by MgcRacGAP. This work identifies for the first time a GEF-activating Rac1 in dividing cells that counteracts MgcRacGAP function in cytokinesis. The Rho GTPases RhoA and Rac1 function as master regulators of cytokinesis by controlling the actomyosin cytoskeleton. RhoA and Rac1 have to be respectively activated and inactivated at the division plane for cytokinesis to occur properly. The inactivation of Rac1 at the cleavage furrow is controlled by MgcRacGAP. However, the guanine-nucleotide exchange factor (GEF) that activates Rac1 during cell division remains unknown. Here, using a siRNA screening approach in HeLa cells, we identify Trio as a mitotic GEF of Rac1. We demonstrate that Trio controls Rac1 activation and subsequent F-actin remodeling in dividing cells. Moreover, Trio depletion specifically rescues the cytokinesis failure induced by MgcRacGAP depletion. Of importance, we demonstrate that this rescue is mediated by the Trio-Rac1 pathway, using GEF-dead mutants of Trio and a specific inhibitor of Rac1 activation by Trio. Overall this work identifies for the first time a GEF controlling Rac1 activation in dividing cells that counteracts MgcRacGAP function in cytokinesis.
Collapse
Affiliation(s)
- Aude Cannet
- Signaling and Cytoskeleton Dynamics Group, University of Montpellier, 34293 Montpellier, France
| | - Susanne Schmidt
- Signaling and Cytoskeleton Dynamics Group, University of Montpellier, 34293 Montpellier, France
| | - Bénédicte Delaval
- Centrosome, Cilia and Pathology Group, CRBM-CNRS, University of Montpellier, 34293 Montpellier, France
| | - Anne Debant
- Signaling and Cytoskeleton Dynamics Group, University of Montpellier, 34293 Montpellier, France
| |
Collapse
|
93
|
Wu D, Jiao M, Zu S, Sollecito CC, Jimenez-Cowell K, Mold AJ, Kennedy RM, Wei Q. Intramolecular interactions between the Dbl homology (DH) domain and the carboxyl-terminal region of myosin II-interacting guanine nucleotide exchange factor (MyoGEF) act as an autoinhibitory mechanism for the regulation of MyoGEF functions. J Biol Chem 2014; 289:34033-48. [PMID: 25336641 DOI: 10.1074/jbc.m114.607267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have reported previously that nonmuscle myosin II-interacting guanine nucleotide exchange factor (MyoGEF) plays an important role in the regulation of cell migration and cytokinesis. Like many other guanine nucleotide exchange factors (GEFs), MyoGEF contains a Dbl homology (DH) domain and a pleckstrin homology domain. In this study, we provide evidence demonstrating that intramolecular interactions between the DH domain (residues 162-351) and the carboxyl-terminal region (501-790) of MyoGEF can inhibit MyoGEF functions. In vitro and in vivo pulldown assays showed that the carboxyl-terminal region (residues 501-790) of MyoGEF could interact with the DH domain but not with the pleckstrin homology domain. Expression of a MyoGEF carboxyl-terminal fragment (residues 501-790) decreased RhoA activation and suppressed actin filament formation in MDA-MB-231 breast cancer cells. Additionally, Matrigel invasion assays showed that exogenous expression of the MyoGEF carboxyl-terminal region decreased the invasion activity of MDA-MB-231 cells. Moreover, coimmunoprecipitation assays showed that phosphorylation of the MyoGEF carboxyl-terminal region by aurora B kinase interfered with the intramolecular interactions of MyoGEF. Furthermore, expression of the MyoGEF carboxyl-terminal region interfered with RhoA localization during cytokinesis and led to an increase in multinucleation. Together, our findings suggest that binding of the carboxyl-terminal region of MyoGEF to its DH domain acts as an autoinhibitory mechanism for the regulation of MyoGEF activation.
Collapse
Affiliation(s)
- Di Wu
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Meng Jiao
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Shicheng Zu
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | | | - Kevin Jimenez-Cowell
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Alexander J Mold
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Ryan M Kennedy
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Qize Wei
- From the Department of Biological Sciences, Fordham University, Bronx, New York 10458
| |
Collapse
|
94
|
Kachaner D, Pinson X, El Kadhi KB, Normandin K, Talje L, Lavoie H, Lépine G, Carréno S, Kwok BH, Hickson GR, Archambault V. Interdomain allosteric regulation of Polo kinase by Aurora B and Map205 is required for cytokinesis. ACTA ACUST UNITED AC 2014; 207:201-11. [PMID: 25332165 PMCID: PMC4210448 DOI: 10.1083/jcb.201408081] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aurora B phosphorylation of the Polo kinase activation loop disrupts its binding to Map205 and central spindle microtubules, allowing it to be recruited to the site of cytokinesis. Drosophila melanogaster Polo and its human orthologue Polo-like kinase 1 fulfill essential roles during cell division. Members of the Polo-like kinase (Plk) family contain an N-terminal kinase domain (KD) and a C-terminal Polo-Box domain (PBD), which mediates protein interactions. How Plks are regulated in cytokinesis is poorly understood. Here we show that phosphorylation of Polo by Aurora B is required for cytokinesis. This phosphorylation in the activation loop of the KD promotes the dissociation of Polo from the PBD-bound microtubule-associated protein Map205, which acts as an allosteric inhibitor of Polo kinase activity. This mechanism allows the release of active Polo from microtubules of the central spindle and its recruitment to the site of cytokinesis. Failure in Polo phosphorylation results in both early and late cytokinesis defects. Importantly, the antagonistic regulation of Polo by Aurora B and Map205 in cytokinesis reveals that interdomain allosteric mechanisms can play important roles in controlling the cellular functions of Plks.
Collapse
Affiliation(s)
- David Kachaner
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Xavier Pinson
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Khaled Ben El Kadhi
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Karine Normandin
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Lama Talje
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Hugo Lavoie
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Guillaume Lépine
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Sébastien Carréno
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Benjamin H Kwok
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Gilles R Hickson
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Vincent Archambault
- Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada Institut de Recherche en Immunologie et en Cancérologie, Départment de Biochimie et Médecine Moléculaire, Centre Hospitalier Universitaire Sainte-Justine, Département de Pathologie et de Biologie Cellulaire, Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
95
|
Giansanti MG, Sechi S, Frappaolo A, Belloni G, Piergentili R. Cytokinesis in Drosophila male meiosis. SPERMATOGENESIS 2014; 2:185-196. [PMID: 23094234 PMCID: PMC3469441 DOI: 10.4161/spmg.21711] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytokinesis separates the cytoplasm and the duplicated genome into two daughter cells at the end of cell division. This process must be finely regulated to maintain ploidy and prevent tumor formation. Drosophila male meiosis provides an excellent cell system for investigating cytokinesis. Mutants affecting this process can be easily identified and spermatocytes are large cells particularly suitable for cytological analysis of cytokinetic structures. Over the past decade, the powerful tools of Drosophila genetics and the unique characteristics of this cell system have led researchers to identify molecular players of the cell cleavage machinery and to address important open questions. Although spermatocyte cytokinesis is incomplete, resulting in formation of stable intercellular bridges, the molecular mechanisms are largely conserved in somatic cells. Thus, studies of Drosophila male meiosis will shed new light on the complex cell circuits regulating furrow ingression and substantially further our knowledge of cancer and other human diseases.
Collapse
Affiliation(s)
- Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR; Dipartimento di Biologia e Biotecnologie Università Sapienza di Roma; Rome, Italy
| | | | | | | | | |
Collapse
|
96
|
Zuo Y, Oh W, Frost JA. Controlling the switches: Rho GTPase regulation during animal cell mitosis. Cell Signal 2014; 26:2998-3006. [PMID: 25286227 DOI: 10.1016/j.cellsig.2014.09.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 09/23/2014] [Indexed: 11/29/2022]
Abstract
Animal cell division is a fundamental process that requires complex changes in cytoskeletal organization and function. Aberrant cell division often has disastrous consequences for the cell and can lead to cell senescence, neoplastic transformation or death. As important regulators of the actin cytoskeleton, Rho GTPases play major roles in regulating many aspects of mitosis and cytokinesis. These include centrosome duplication and separation, generation of cortical rigidity, microtubule-kinetochore stabilization, cleavage furrow formation, contractile ring formation and constriction, and abscission. The ability of Rho proteins to function as regulators of cell division depends on their ability to cycle between their active, GTP-bound and inactive, GDP-bound states. However, Rho proteins are inherently inefficient at fulfilling this cycle and require the actions of regulatory proteins that enhance GTP binding (RhoGEFs), stimulate GTPase activity (RhoGAPs), and sequester inactive Rho proteins in the cytosol (RhoGDIs). The roles of these regulatory proteins in controlling cell division are an area of active investigation. In this review we will delineate the current state of knowledge of how specific RhoGEFs, RhoGAPs and RhoGDIs control mitosis and cytokinesis, and highlight the mechanisms by which their functions are controlled.
Collapse
Affiliation(s)
- Yan Zuo
- University of Texas Health Science Center at Houston, Department of Integrative Biology and Pharmacology, 6431 Fannin St., Houston, TX 77030, United States
| | - Wonkyung Oh
- University of Texas Health Science Center at Houston, Department of Integrative Biology and Pharmacology, 6431 Fannin St., Houston, TX 77030, United States
| | - Jeffrey A Frost
- University of Texas Health Science Center at Houston, Department of Integrative Biology and Pharmacology, 6431 Fannin St., Houston, TX 77030, United States.
| |
Collapse
|
97
|
Zou Y, Shao Z, Peng J, Li F, Gong D, Wang C, Zuo X, Zhang Z, Wu J, Shi Y, Gong Q. Crystal structure of triple-BRCT-domain of ECT2 and insights into the binding characteristics to CYK-4. FEBS Lett 2014; 588:2911-20. [PMID: 25068414 DOI: 10.1016/j.febslet.2014.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 06/29/2014] [Accepted: 07/14/2014] [Indexed: 11/18/2022]
Abstract
Homo sapiens ECT2 is a cell cycle regulator that plays critical roles in cytokinesis. ECT2 activity is restrained during interphase via intra-molecular interactions that involve its N-terminal triple-BRCT-domain and its C-terminal DH-PH domain. At anaphase, this self-inhibitory mechanism is relieved by Plk1-phosphorylated CYK-4, which directly engages the ECT2 BRCT domain. To provide a structural perspective for this auto-inhibitory property, we solved the crystal structure of the ECT2 triple-BRCT-domain. In addition, we systematically analyzed the interaction between the ECT2 BRCT domains with phospho-peptides derived from its binding partner CYK-4, and have identified Ser164 as the major phospho-residue that links CYK-4 to the second ECT2 BRCT domain.
Collapse
Affiliation(s)
- Yang Zou
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhenhua Shao
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Junhui Peng
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fudong Li
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Deshun Gong
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chongyuan Wang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaobing Zuo
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60349, USA
| | - Zhiyong Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jihui Wu
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yunyu Shi
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qingguo Gong
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
98
|
McKinley KL, Cheeseman IM. Polo-like kinase 1 licenses CENP-A deposition at centromeres. Cell 2014; 158:397-411. [PMID: 25036634 PMCID: PMC4192726 DOI: 10.1016/j.cell.2014.06.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/28/2014] [Accepted: 06/02/2014] [Indexed: 01/22/2023]
Abstract
To ensure the stable transmission of the genome during vertebrate cell division, the mitotic spindle must attach to a single locus on each chromosome, termed the centromere. The fundamental requirement for faithful centromere inheritance is the controlled deposition of the centromere-specifying histone, CENP-A. However, the regulatory mechanisms that ensure the precise control of CENP-A deposition have proven elusive. Here, we identify polo-like kinase 1 (Plk1) as a centromere-localized regulator required to initiate CENP-A deposition in human cells. We demonstrate that faithful CENP-A deposition requires integrated signals from Plk1 and cyclin-dependent kinase (CDK), with Plk1 promoting the localization of the key CENP-A deposition factor, the Mis18 complex, and CDK inhibiting Mis18 complex assembly. By bypassing these regulated steps, we uncoupled CENP-A deposition from cell-cycle progression, resulting in mitotic defects. Thus, CENP-A deposition is controlled by a two-step regulatory paradigm comprised of Plk1 and CDK that is crucial for genomic integrity.
Collapse
Affiliation(s)
- Kara L McKinley
- Whitehead Institute and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Iain M Cheeseman
- Whitehead Institute and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA.
| |
Collapse
|
99
|
Chircop M. Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells. Small GTPases 2014; 5:29770. [PMID: 24988197 DOI: 10.4161/sgtp.29770] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Rho GTPases regulate a diverse range of cellular functions primarily through their ability to modulate microtubule dynamics and the actin-myosin cytoskeleton. Both of these cytoskeletal structures are crucial for a mitotic cell division. Specifically, their assembly and disassembly is tightly regulated in a temporal manner to ensure that each mitotic stage occurs in the correct sequential order and not prematurely until the previous stage is completed. Thus, it is not surprising that the Rho GTPases, RhoA, and Cdc42, have reported roles in several stages of mitosis: cell cortex stiffening during cell rounding, mitotic spindle formation, and bi-orient attachment of the spindle microtubules to the kinetochore and during cytokinesis play multiple roles in establishing the division plane, assembly, and activation of the contractile ring, membrane ingression, and abscission. Here, I review the molecular mechanisms regulating the spatial and temporal activation of RhoA and Cdc42 during mitosis, and how this is critical for mitotic progression and completion.
Collapse
Affiliation(s)
- Megan Chircop
- Children's Medical Research Institute; The University of Sydney; Westmead, Australia
| |
Collapse
|
100
|
Kitazawa D, Matsuo T, Kaizuka K, Miyauchi C, Hayashi D, Inoue YH. Orbit/CLASP is required for myosin accumulation at the cleavage furrow in Drosophila male meiosis. PLoS One 2014; 9:e93669. [PMID: 24850412 PMCID: PMC4029619 DOI: 10.1371/journal.pone.0093669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 03/07/2014] [Indexed: 01/24/2023] Open
Abstract
Peripheral microtubules (MTs) near the cell cortex are essential for the positioning and continuous constriction of the contractile ring (CR) in cytokinesis. Time-lapse observations of Drosophila male meiosis showed that myosin II was first recruited along the cell cortex independent of MTs. Then, shortly after peripheral MTs made contact with the equatorial cortex, myosin II was concentrated there in a narrow band. After MT contact, anillin and F-actin abruptly appeared on the equatorial cortex, simultaneously with myosin accumulation. We found that the accumulation of myosin did not require centralspindlin, but was instead dependent on Orbit, a Drosophila ortholog of the MT plus-end tracking protein CLASP. This protein is required for stabilization of central spindle MTs, which are essential for cytokinesis. Orbit was also localized in a mid-zone of peripheral MTs, and was concentrated in a ring at the equatorial cortex during late anaphase. Fluorescence resonance energy transfer experiments indicated that Orbit is closely associated with F-actin in the CR. We also showed that the myosin heavy chain was in close proximity with Orbit in the cleavage furrow region. Centralspindlin was dispensable in Orbit ring formation. Instead, the Polo-KLP3A/Feo complex was required for the Orbit accumulation independently of the Orbit MT-binding domain. However, orbit mutations of consensus sites for the phosphorylation of Cdk1 or Polo did not influence the Orbit accumulation, suggesting an indirect regulatory role of these protein kinases in Orbit localization. Orbit was also necessary for the maintenance of the CR. Our data suggest that Orbit plays an essential role as a connector between MTs and the CR in Drosophila male meiosis.
Collapse
Affiliation(s)
- Daishi Kitazawa
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Tatsuru Matsuo
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Kana Kaizuka
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Chie Miyauchi
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Daisuke Hayashi
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
| | - Yoshihiro H. Inoue
- Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Kyoto, Japan
- * E-mail:
| |
Collapse
|