51
|
Joglekar AP, Kukreja AA. How Kinetochore Architecture Shapes the Mechanisms of Its Function. Curr Biol 2018; 27:R816-R824. [PMID: 28829971 DOI: 10.1016/j.cub.2017.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The eukaryotic kinetochore is a sophisticated multi-protein machine that segregates chromosomes during cell division. To ensure accurate chromosome segregation, it performs three major functions using disparate molecular mechanisms. It operates a mechanosensitive signaling cascade known as the spindle assembly checkpoint (SAC) to detect and signal the lack of attachment to spindle microtubules, and delay anaphase onset in response. In addition, after attaching to spindle microtubules, the kinetochore generates the force necessary to move chromosomes. Finally, if the two sister kinetochores on a chromosome are both attached to microtubules emanating from the same spindle pole, they activate another mechanosensitive mechanism to correct the monopolar attachments. All three of these functions maintain genome stability during cell division. The outlines of the biochemical activities responsible for these functions are now available. How the kinetochore integrates the underlying molecular mechanisms is still being elucidated. In this Review, we discuss how the nanoscale protein organization in the kinetochore, which we refer to as kinetochore 'architecture', organizes its biochemical activities to facilitate the realization and integration of emergent mechanisms underlying its three major functions. For this discussion, we will use the relatively simple budding yeast kinetochore as a model, and extrapolate insights gained from this model to elucidate functional roles of the architecture of the much more complex human kinetochore.
Collapse
Affiliation(s)
- Ajit P Joglekar
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
52
|
Saurin AT. Kinase and Phosphatase Cross-Talk at the Kinetochore. Front Cell Dev Biol 2018; 6:62. [PMID: 29971233 PMCID: PMC6018199 DOI: 10.3389/fcell.2018.00062] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023] Open
Abstract
Multiple kinases and phosphatases act on the kinetochore to control chromosome segregation: Aurora B, Mps1, Bub1, Plk1, Cdk1, PP1, and PP2A-B56, have all been shown to regulate both kinetochore-microtubule attachments and the spindle assembly checkpoint. Given that so many kinases and phosphatases converge onto two key mitotic processes, it is perhaps not surprising to learn that they are, quite literally, entangled in cross-talk. Inhibition of any one of these enzymes produces secondary effects on all the others, which results in a complicated picture that is very difficult to interpret. This review aims to clarify this picture by first collating the direct effects of each enzyme into one overarching schematic of regulation at the Knl1/Mis12/Ndc80 (KMN) network (a major signaling hub at the outer kinetochore). This schematic will then be used to discuss the implications of the cross-talk that connects these enzymes; both in terms of why it may be needed to produce the right type of kinetochore signals and why it nevertheless complicates our interpretations about which enzymes control what processes. Finally, some general experimental approaches will be discussed that could help to characterize kinetochore signaling by dissociating the direct from indirect effect of kinase or phosphatase inhibition in vivo. Together, this review should provide a framework to help understand how a network of kinases and phosphatases cooperate to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Adrian T. Saurin
- Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
53
|
Chaurasia S, Lehner CF. Dynamics and control of sister kinetochore behavior during the meiotic divisions in Drosophila spermatocytes. PLoS Genet 2018; 14:e1007372. [PMID: 29734336 PMCID: PMC5957430 DOI: 10.1371/journal.pgen.1007372] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/17/2018] [Accepted: 04/19/2018] [Indexed: 11/19/2022] Open
Abstract
Sister kinetochores are connected to the same spindle pole during meiosis I and to opposite poles during meiosis II. The molecular mechanisms controlling the distinct behavior of sister kinetochores during the two meiotic divisions are poorly understood. To study kinetochore behavior during meiosis, we have optimized time lapse imaging with Drosophila spermatocytes, enabling kinetochore tracking with high temporal and spatial resolution through both meiotic divisions. The correct bipolar orientation of chromosomes within the spindle proceeds rapidly during both divisions. Stable bi-orientation of the last chromosome is achieved within ten minutes after the onset of kinetochore-microtubule interactions. Our analyses of mnm and tef mutants, where univalents instead of bivalents are present during meiosis I, indicate that the high efficiency of normal bi-orientation depends on pronounced stabilization of kinetochore attachments to spindle microtubules by the mechanical tension generated by spindle forces upon bi-orientation. Except for occasional brief separation episodes, sister kinetochores are so closely associated that they cannot be resolved individually by light microscopy during meiosis I, interkinesis and at the start of meiosis II. Permanent evident separation of sister kinetochores during M II depends on spindle forces resulting from bi-orientation. In mnm and tef mutants, sister kinetochore separation can be observed already during meiosis I in bi-oriented univalents. Interestingly, however, this sister kinetochore separation is delayed until the metaphase to anaphase transition and depends on the Fzy/Cdc20 activator of the anaphase-promoting complex/cyclosome. We propose that univalent bi-orientation in mnm and tef mutants exposes a release of sister kinetochore conjunction that occurs also during normal meiosis I in preparation for bi-orientation of dyads during meiosis II. For production of oocytes and sperm, cells have to complete meiosis which includes two successive divisions. These divisions convert diploid cells with a maternal and a paternal copy of each chromosome into haploid cells with only one copy of each chromosome. Chromosome copy reduction requires regulation of sister kinetochore behavior during the meiotic divisions. Kinetochores are protein networks assembled at the start of divisions within the centromeric region of chromosomes. They provide attachment sites for spindle microtubules which in turn exert poleward pulling forces. During pre-meiotic S phase, each chromosome is duplicated into two closely associated sister chromatids. At the start of the first meiotic division, both sister chromatids together assemble only one functional kinetochore, permitting subsequent separation of paired homologous chromosomes to opposite spindle poles. In contrast, at the onset of the second meiotic division, each sister chromatid organizes its own kinetochore followed by separation of sister chromatids to opposite spindle poles. To analyze when and how sister kinetochores are individualized, we have improved time lapse imaging with Drosophila spermatocytes. Our analyses in normal and genetically altered spermatocytes suggest that the release of sister kinetochore conjunction occurs during the first meiotic division after activation of the anaphase promoting complex/cyclosome.
Collapse
Affiliation(s)
- Soumya Chaurasia
- Institute of Molecular Life Sciences (IMLS), University of Zurich, Zurich, Switzerland
| | - Christian F. Lehner
- Institute of Molecular Life Sciences (IMLS), University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
54
|
Ruppert JG, Samejima K, Platani M, Molina O, Kimura H, Jeyaprakash AA, Ohta S, Earnshaw WC. HP1α targets the chromosomal passenger complex for activation at heterochromatin before mitotic entry. EMBO J 2018; 37:e97677. [PMID: 29467217 PMCID: PMC5852645 DOI: 10.15252/embj.201797677] [Citation(s) in RCA: 31] [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: 06/26/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 12/18/2022] Open
Abstract
The chromosomal passenger complex (CPC) is directed to centromeres during mitosis via binding to H3T3ph and Sgo1. Whether and how heterochromatin protein 1α (HP1α) influences CPC localisation and function during mitotic entry is less clear. Here, we alter HP1α dynamics by fusing it to a CENP-B DNA-binding domain. Tethered HP1 strongly recruits the CPC, destabilising kinetochore-microtubule interactions and activating the spindle assembly checkpoint. During mitotic exit, the tethered HP1 traps active CPC at centromeres. These HP1-CPC clusters remain catalytically active throughout the subsequent cell cycle. We also detect interactions between endogenous HP1 and the CPC during G2 HP1α and HP1γ cooperate to recruit the CPC to active foci in a CDK1-independent process. Live cell tracking with Fab fragments reveals that H3S10ph appears well before H3T3 is phosphorylated by Haspin kinase. Our results suggest that HP1 may concentrate and activate the CPC at centromeric heterochromatin in G2 before Aurora B-mediated phosphorylation of H3S10 releases HP1 from chromatin and allows pathways dependent on H3T3ph and Sgo1 to redirect the CPC to mitotic centromeres.
Collapse
Affiliation(s)
- Jan G Ruppert
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Kumiko Samejima
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Melpomeni Platani
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Oscar Molina
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Hiroshi Kimura
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | | | - Shinya Ohta
- Department of Biochemistry, Medical School, Kochi University, Nankoku, Kochi, Japan
| | - William C Earnshaw
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
55
|
Adamowicz M, Morgan CC, Haubner BJ, Noseda M, Collins MJ, Abreu Paiva M, Srivastava PK, Gellert P, Razzaghi B, O’Gara P, Raina P, Game L, Bottolo L, Schneider MD, Harding SE, Penninger J, Aitman TJ. Functionally Conserved Noncoding Regulators of Cardiomyocyte Proliferation and Regeneration in Mouse and Human. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018; 11:e001805. [DOI: 10.1161/circgen.117.001805] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
The adult mammalian heart has little regenerative capacity after myocardial infarction (MI), whereas neonatal mouse heart regenerates without scarring or dysfunction. However, the underlying pathways are poorly defined. We sought to derive insights into the pathways regulating neonatal development of the mouse heart and cardiac regeneration post-MI.
Methods and Results:
Total RNA-seq of mouse heart through the first 10 days of postnatal life (referred to as P3, P5, P10) revealed a previously unobserved transition in microRNA (miRNA) expression between P3 and P5 associated specifically with altered expression of protein-coding genes on the focal adhesion pathway and cessation of cardiomyocyte cell division. We found profound changes in the coding and noncoding transcriptome after neonatal MI, with evidence of essentially complete healing by P10. Over two-thirds of each of the messenger RNAs, long noncoding RNAs, and miRNAs that were differentially expressed in the post-MI heart were differentially expressed during normal postnatal development, suggesting a common regulatory pathway for normal cardiac development and post-MI cardiac regeneration. We selected exemplars of miRNAs implicated in our data set as regulators of cardiomyocyte proliferation. Several of these showed evidence of a functional influence on mouse cardiomyocyte cell division. In addition, a subset of these miRNAs, miR-144-3p, miR-195a-5p, miR-451a, and miR-6240 showed evidence of functional conservation in human cardiomyocytes.
Conclusions:
The sets of messenger RNAs, miRNAs, and long noncoding RNAs that we report here merit further investigation as gatekeepers of cell division in the postnatal heart and as targets for extension of the period of cardiac regeneration beyond the neonatal period.
Collapse
Affiliation(s)
- Martyna Adamowicz
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Claire C. Morgan
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Bernhard J. Haubner
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Michela Noseda
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Melissa J. Collins
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Marta Abreu Paiva
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Prashant K. Srivastava
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Pascal Gellert
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Bonnie Razzaghi
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Peter O’Gara
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Priyanka Raina
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Laurence Game
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Leonardo Bottolo
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Michael D. Schneider
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Sian E. Harding
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Josef Penninger
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| | - Timothy J. Aitman
- From the Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, United Kingdom (T.J.A.); National Heart and Lung Institute (M.A., C.C.M., M.N., M.A.P., P.O., M.D.S., S.E.H.), Department of Medicine (C.C.M., M.J.C., P.K.S., B.R., P.R., T.J.A.), Department of Mathematics (L.B.), Imperial College London, United Kingdom; IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria (B.J.H., J.P.)
| |
Collapse
|
56
|
Kitajima TS. Mechanisms of kinetochore-microtubule attachment errors in mammalian oocytes. Dev Growth Differ 2018; 60:33-43. [PMID: 29318599 PMCID: PMC11520954 DOI: 10.1111/dgd.12410] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022]
Abstract
Proper kinetochore-microtubule attachment is essential for correct chromosome segregation. Therefore, cells normally possess multiple mechanisms for the prevention of errors in kinetochore-microtubule attachments and for selective stabilization of correct attachments. However, the oocyte, a cell that produces an egg through meiosis, exhibits a high frequency of errors in kinetochore-microtubule attachments. These attachment errors predispose oocytes to chromosome segregation errors, resulting in aneuploidy in eggs. This review aims to provide possible explanations for the error-prone nature of oocytes by examining key differences among other cell types in the mechanisms for the establishment of kinetochore-microtubule attachments.
Collapse
Affiliation(s)
- Tomoya S. Kitajima
- Laboratory for Chromosome SegregationRIKEN Center for Developmental BiologyKobe650‐0047Japan
| |
Collapse
|
57
|
Dumitru AMG, Rusin SF, Clark AEM, Kettenbach AN, Compton DA. Cyclin A/Cdk1 modulates Plk1 activity in prometaphase to regulate kinetochore-microtubule attachment stability. eLife 2017; 6:e29303. [PMID: 29154753 PMCID: PMC5706962 DOI: 10.7554/elife.29303] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/10/2017] [Indexed: 12/24/2022] Open
Abstract
The fidelity of chromosome segregation in mitosis is safeguarded by the precise regulation of kinetochore microtubule (k-MT) attachment stability. Previously, we demonstrated that Cyclin A/Cdk1 destabilizes k-MT attachments to promote faithful chromosome segregation. Here, we use quantitative phosphoproteomics to identify 156 Cyclin A/Cdk1 substrates in prometaphase. One Cyclin A/Cdk1 substrate is myosin phosphatase targeting subunit 1 (MYPT1), and we show that MYPT1 localization to kinetochores depends on Cyclin A/Cdk1 activity and that MYPT1 destabilizes k-MT attachments by negatively regulating Plk1 at kinetochores. Thus, Cyclin A/Cdk1 phosphorylation primes MYPT1 for Plk1 binding. Interestingly, priming of PBIP1 by Plk1 itself (self-priming) increased in MYPT1-depleted cells showing that MYPT1 provides a molecular link between the processes of Cdk1-dependent priming and self-priming of Plk1 substrates. These data demonstrate cross-regulation between Cyclin A/Cdk1-dependent and Plk1-dependent phosphorylation of substrates during mitosis to ensure efficient correction of k-MT attachment errors necessary for high mitotic fidelity.
Collapse
Affiliation(s)
- Ana Maria G Dumitru
- Department of Biochemistry and Cell BiologyGeisel School of Medicine at DartmouthHanoverUnited States
- Norris Cotton Cancer CenterLebanonUnited States
| | - Scott F Rusin
- Department of Biochemistry and Cell BiologyGeisel School of Medicine at DartmouthHanoverUnited States
- Norris Cotton Cancer CenterLebanonUnited States
| | - Amber E M Clark
- Department of Biochemistry and Cell BiologyGeisel School of Medicine at DartmouthHanoverUnited States
- Norris Cotton Cancer CenterLebanonUnited States
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell BiologyGeisel School of Medicine at DartmouthHanoverUnited States
- Norris Cotton Cancer CenterLebanonUnited States
| | - Duane A Compton
- Department of Biochemistry and Cell BiologyGeisel School of Medicine at DartmouthHanoverUnited States
- Norris Cotton Cancer CenterLebanonUnited States
| |
Collapse
|
58
|
Gu WW, Lin J, Hong XY. Cyclin A2 regulates homologous recombination DNA repair and sensitivity to DNA damaging agents and poly(ADP-ribose) polymerase (PARP) inhibitors in human breast cancer cells. Oncotarget 2017; 8:90842-90851. [PMID: 29207607 PMCID: PMC5710888 DOI: 10.18632/oncotarget.20412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/25/2017] [Indexed: 11/25/2022] Open
Abstract
Defects in homologous recombination (HR) repair are found in breast cancers. Intriguingly, breast cancers with defective HR show increased sensitivity to DNA crosslinking agents and poly(ADP-ribose) polymerase (PARP) inhibitors. As such, genes that can affect HR functions have been of high interest in studies aiming to develop biomarkers for predicting response to treatment with these agents. Cyclin A2 is a key component of the core cell cycle machinery. However, whether cyclin A2 dysfunctions could cause HR defect and mediate sensitivity to DNA damaging agents remain unclear. Here we show that loss of cyclin A2 causes high rates of double-strand breaks (DSB) in MCF-7 and MDA-MB-231 cells. The increased DSB was due to defective HR-mediated repair of the breaks, resulting from reduced MRE11 and RAD51 proteins. Cyclin A2 mediates MRE11 abundance through its MRE11 mRNA binding property and RAD51 abundance through inhibition of proteasome degradation of RAD51. Moreover, cyclin A2 depletion hypersensitized the cells to DNA damaging agents, such as cisplatin and melphalan. Our results demonstrate novel roles for cyclin A2 in regulating HR repair and determining sensitivity to DNA cross linkers and PARP inhibitors in breast cancer cells.
Collapse
Affiliation(s)
- Wei Wei Gu
- Department of Hepatopancreatobility Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jie Lin
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xing Yu Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
59
|
Tipton AR, Wren JD, Daum JR, Siefert JC, Gorbsky GJ. GTSE1 regulates spindle microtubule dynamics to control Aurora B kinase and Kif4A chromokinesin on chromosome arms. J Cell Biol 2017; 216:3117-3132. [PMID: 28821562 PMCID: PMC5626529 DOI: 10.1083/jcb.201610012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 04/20/2017] [Accepted: 07/12/2017] [Indexed: 12/24/2022] Open
Abstract
In mitosis, the dynamic assembly and disassembly of microtubules are critical for normal chromosome movement and segregation. Microtubule turnover varies among different mitotic spindle microtubules, dictated by their spatial distribution within the spindle. How turnover among the various classes of spindle microtubules is differentially regulated and the resulting significance of differential turnover for chromosome movement remains a mystery. As a new tactic, we used global microarray meta-analysis (GAMMA), a bioinformatic method, to identify novel regulators of mitosis, and in this study, we describe G2- and S phase-expressed protein 1 (GTSE1). GTSE1 is expressed exclusively in late G2 and M phase. From nuclear envelope breakdown until anaphase onset, GTSE1 binds preferentially to the most stable mitotic spindle microtubules and promotes their turnover. Cells depleted of GTSE1 show defects in chromosome alignment at the metaphase plate and in spindle pole integrity. These defects are coupled with an increase in the proportion of stable mitotic spindle microtubules. A consequence of this reduced microtubule turnover is diminished recruitment and activity of Aurora B kinase on chromosome arms. This decrease in Aurora B results in diminished binding of the chromokinesin Kif4A to chromosome arms.
Collapse
Affiliation(s)
- Aaron R Tipton
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - John R Daum
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Joseph C Siefert
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gary J Gorbsky
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
60
|
Zhang QH, Yuen WS, Adhikari D, Flegg JA, FitzHarris G, Conti M, Sicinski P, Nabti I, Marangos P, Carroll J. Cyclin A2 modulates kinetochore-microtubule attachment in meiosis II. J Cell Biol 2017; 216:3133-3143. [PMID: 28819014 PMCID: PMC5626527 DOI: 10.1083/jcb.201607111] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022] Open
Abstract
Cyclin A2 is a crucial mitotic Cdk regulatory partner that coordinates entry into mitosis and is then destroyed in prometaphase within minutes of nuclear envelope breakdown. The role of cyclin A2 in female meiosis and its dynamics during the transition from meiosis I (MI) to meiosis II (MII) remain unclear. We found that cyclin A2 decreases in prometaphase I but recovers after the first meiotic division and persists, uniquely for metaphase, in MII-arrested oocytes. Conditional deletion of cyclin A2 from mouse oocytes has no discernible effect on MI but leads to disrupted MII spindles and increased merotelic attachments. On stimulation of exit from MII, there is a dramatic increase in lagging chromosomes and an inhibition of cytokinesis. These defects are associated with an increase in microtubule stability in MII spindles, suggesting that cyclin A2 mediates the fidelity of MII by maintaining microtubule dynamics during the rapid formation of the MII spindle.
Collapse
Affiliation(s)
- Qing-Hua Zhang
- Development and Stem Cell Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia .,Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Wai Shan Yuen
- Development and Stem Cell Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Deepak Adhikari
- Development and Stem Cell Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Jennifer A Flegg
- Monash Academy for Cross and Interdisciplinary Mathematical Applications, Monash University, Melbourne, Victoria, Australia
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada.,Department of Obstetrics and Gynaecology, University of Montréal, Montréal, Québec, Canada
| | - Marco Conti
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA
| | - Piotr Sicinski
- Dana-Farber Cancer Institute, Boston, MA.,Department of Genetics, Harvard Medical School, Boston, MA
| | - Ibtissem Nabti
- Department of Cell and Developmental Biology, University College London, London, England, UK.,Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Petros Marangos
- Department of Cell and Developmental Biology, University College London, London, England, UK.,Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece.,Department of Biomedical Research, Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, Ioannina, Greece
| | - John Carroll
- Development and Stem Cell Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia .,Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia.,Department of Cell and Developmental Biology, University College London, London, England, UK
| |
Collapse
|
61
|
Vleugel M, Kok M, Dogterom M. Understanding force-generating microtubule systems through in vitro reconstitution. Cell Adh Migr 2017; 10:475-494. [PMID: 27715396 PMCID: PMC5079405 DOI: 10.1080/19336918.2016.1241923] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Microtubules switch between growing and shrinking states, a feature known as dynamic instability. The biochemical parameters underlying dynamic instability are modulated by a wide variety of microtubule-associated proteins that enable the strict control of microtubule dynamics in cells. The forces generated by controlled growth and shrinkage of microtubules drive a large range of processes, including organelle positioning, mitotic spindle assembly, and chromosome segregation. In the past decade, our understanding of microtubule dynamics and microtubule force generation has progressed significantly. Here, we review the microtubule-intrinsic process of dynamic instability, the effect of external factors on this process, and how the resulting forces act on various biological systems. Recently, reconstitution-based approaches have strongly benefited from extensive biochemical and biophysical characterization of individual components that are involved in regulating or transmitting microtubule-driven forces. We will focus on the current state of reconstituting increasingly complex biological systems and provide new directions for future developments.
Collapse
Affiliation(s)
- Mathijs Vleugel
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| | - Maurits Kok
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| | - Marileen Dogterom
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| |
Collapse
|
62
|
Wei Y, Mo X, Zhang P, Li Y, Liao J, Li Y, Zhang J, Ning C, Wang S, Deng X, Jiang L. Directing Stem Cell Differentiation via Electrochemical Reversible Switching between Nanotubes and Nanotips of Polypyrrole Array. ACS NANO 2017; 11:5915-5924. [PMID: 28587445 DOI: 10.1021/acsnano.7b01661] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Control of stem cell behaviors at solid biointerfaces is critical for stem-cell-based regeneration and generally achieved by engineering chemical composition, topography, and stiffness. However, the influence of dynamic stimuli at the nanoscale from solid biointerfaces on stem cell fate remains unclear. Herein, we show that electrochemical switching of a polypyrrole (Ppy) array between nanotubes and nanotips can alter surface adhesion, which can strongly influence mechanotransduction activation and guide differentiation of mesenchymal stem cells (MSCs). The Ppy array, prepared via template-free electrochemical polymerization, can be reversibly switched between highly adhesive hydrophobic nanotubes and poorly adhesive hydrophilic nanotips through an electrochemical oxidation/reduction process, resulting in dynamic attachment and detachment to MSCs at the nanoscale. Multicyclic attachment/detachment of the Ppy array to MSCs can activate intracellular mechanotransduction and osteogenic differentiation independent of surface stiffness and chemical induction. This smart surface, permitting transduction of nanoscaled dynamic physical inputs into biological outputs, provides an alternative to classical cell culture substrates for regulating stem cell fate commitment. This study represents a general strategy to explore nanoscaled interactions between stem cells and stimuli-responsive surfaces.
Collapse
Affiliation(s)
- Yan Wei
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Xiaoju Mo
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Pengchao Zhang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Yingying Li
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Jingwen Liao
- School of Materials Science and Engineering, South China University of Technology , Guangzhou 510641, China
| | - Yongjun Li
- Department of Physics, Beijing Normal University , Beijing 100875, China
| | - Jinxing Zhang
- Department of Physics, Beijing Normal University , Beijing 100875, China
| | - Chengyun Ning
- School of Materials Science and Engineering, South China University of Technology , Guangzhou 510641, China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Lei Jiang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| |
Collapse
|
63
|
Cirillo L, Gotta M, Meraldi P. The Elephant in the Room: The Role of Microtubules in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:93-124. [DOI: 10.1007/978-3-319-57127-0_5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
64
|
Cheerambathur DK, Prevo B, Hattersley N, Lewellyn L, Corbett KD, Oegema K, Desai A. Dephosphorylation of the Ndc80 Tail Stabilizes Kinetochore-Microtubule Attachments via the Ska Complex. Dev Cell 2017; 41:424-437.e4. [PMID: 28535376 PMCID: PMC5572820 DOI: 10.1016/j.devcel.2017.04.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 03/16/2017] [Accepted: 04/19/2017] [Indexed: 12/24/2022]
Abstract
During cell division, genome inheritance is orchestrated by microtubule attachments formed at kinetochores of mitotic chromosomes. The primary microtubule coupler at the kinetochore, the Ndc80 complex, is regulated by Aurora kinase phosphorylation of its N-terminal tail. Dephosphorylation is proposed to stabilize kinetochore-microtubule attachments by strengthening electrostatic interactions of the tail with the microtubule lattice. Here, we show that removal of the Ndc80 tail, which compromises in vitro microtubule binding, has no effect on kinetochore-microtubule attachments in the Caenorhabditis elegans embryo. Despite this, preventing Aurora phosphorylation of the tail results in prematurely stable attachments that restrain spindle elongation. This premature stabilization requires the conserved microtubule-binding Ska complex, which enriches at attachment sites prior to anaphase onset to dampen chromosome motion. We propose that Ndc80-tail dephosphorylation promotes stabilization of kinetochore-microtubule attachments via the Ska complex and that this mechanism ensures accurate segregation by constraining chromosome motion following biorientation on the spindle.
Collapse
Affiliation(s)
- Dhanya K Cheerambathur
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | - Bram Prevo
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Neil Hattersley
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Lindsay Lewellyn
- Department of Biological Sciences, Butler University, 4600 Sunset Boulevard, Indianapolis, IN 46208, USA
| | - Kevin D Corbett
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Karen Oegema
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Arshad Desai
- Ludwig Institute for Cancer Research, CMM-E Room 3052, 9500 Gilman Drive, La Jolla, CA 92093-0653, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
65
|
Moura M, Osswald M, Leça N, Barbosa J, Pereira AJ, Maiato H, Sunkel CE, Conde C. Protein Phosphatase 1 inactivates Mps1 to ensure efficient Spindle Assembly Checkpoint silencing. eLife 2017; 6. [PMID: 28463114 PMCID: PMC5433843 DOI: 10.7554/elife.25366] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/29/2017] [Indexed: 12/13/2022] Open
Abstract
Faithfull genome partitioning during cell division relies on the Spindle Assembly Checkpoint (SAC), a conserved signaling pathway that delays anaphase onset until all chromosomes are attached to spindle microtubules. Mps1 kinase is an upstream SAC regulator that promotes the assembly of an anaphase inhibitor through a sequential multi-target phosphorylation cascade. Thus, the SAC is highly responsive to Mps1, whose activity peaks in early mitosis as a result of its T-loop autophosphorylation. However, the mechanism controlling Mps1 inactivation once kinetochores attach to microtubules and the SAC is satisfied remains unknown. Here we show in vitro and in Drosophila that Protein Phosphatase 1 (PP1) inactivates Mps1 by dephosphorylating its T-loop. PP1-mediated dephosphorylation of Mps1 occurs at kinetochores and in the cytosol, and inactivation of both pools of Mps1 during metaphase is essential to ensure prompt and efficient SAC silencing. Overall, our findings uncover a mechanism of SAC inactivation required for timely mitotic exit. DOI:http://dx.doi.org/10.7554/eLife.25366.001
Collapse
Affiliation(s)
- Margarida Moura
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Mariana Osswald
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Nelson Leça
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - João Barbosa
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - António J Pereira
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Helder Maiato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Biomedicina, Unidade de Biologia Experimental, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Claudio E Sunkel
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Biologia Molecular, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
66
|
Kanakkanthara A, Jeganathan KB, Limzerwala JF, Baker DJ, Hamada M, Nam HJ, van Deursen WH, Hamada N, Naylor RM, Becker NA, Davies BA, van Ree JH, Mer G, Shapiro VS, Maher LJ, Katzmann DJ, van Deursen JM. Cyclin A2 is an RNA binding protein that controls Mre11 mRNA translation. Science 2017; 353:1549-1552. [PMID: 27708105 DOI: 10.1126/science.aaf7463] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 08/29/2016] [Indexed: 12/23/2022]
Abstract
Cyclin A2 activates the cyclin-dependent kinases Cdk1 and Cdk2 and is expressed at elevated levels from S phase until early mitosis. We found that mutant mice that cannot elevate cyclin A2 are chromosomally unstable and tumor-prone. Underlying the chromosomal instability is a failure to up-regulate the meiotic recombination 11 (Mre11) nuclease in S phase, which leads to impaired resolution of stalled replication forks, insufficient repair of double-stranded DNA breaks, and improper segregation of sister chromosomes. Unexpectedly, cyclin A2 controlled Mre11 abundance through a C-terminal RNA binding domain that selectively and directly binds Mre11 transcripts to mediate polysome loading and translation. These data reveal cyclin A2 as a mechanistically diverse regulator of DNA replication combining multifaceted kinase-dependent functions with a kinase-independent, RNA binding-dependent role that ensures adequate repair of common replication errors.
Collapse
Affiliation(s)
- Arun Kanakkanthara
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jazeel F Limzerwala
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hyun-Ja Nam
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Naomi Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ryan M Naylor
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Nicole A Becker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Brian A Davies
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Janine H van Ree
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Georges Mer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | - L James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - David J Katzmann
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA. Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
67
|
Chen X, Wang M, Xu X, Liu J, Mei B, Fu P, Zhao D, Sun L. Panax ginseng total protein promotes proliferation and secretion of collagen in NIH/3T3 cells by activating extracellular signal-related kinase pathway. J Ginseng Res 2017; 41:411-418. [PMID: 28701885 PMCID: PMC5489768 DOI: 10.1016/j.jgr.2017.02.001] [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: 11/09/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recently, protein from ginseng was studied and used for the treatment of several kinds of diseases. However, the effect of ginseng total protein (GTP) on proliferation and wound healing in fibroblast cells remains unclear. METHODS In this study, cell viability was analyzed using the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. Cell cycle distribution was analyzed by flow cytometer. The levels of transforming growth factor β1, vascular endothelial growth factor, and collagens were analyzed by enzyme-linked immunosorbent assay and immunofluorescence staining. The expressions of cyclin A, phosphorylation of extracellular signal-related kinase (p-ERK1/2), and ERK1/2 were analyzed by Western blotting. RESULTS Our results showed that GTP promoted cell proliferation and increased the percentage of cells in S phase through the upregulation of cyclin A in NIH/3T3 cells. We also found that GTP induced the secretion of type I collagen, and promoted the expression of other factors that regulate the synthesis of collagen such as transforming growth factor β1 and vascular endothelial growth factor. In addition, the phosphorylation of ERK1/2 at Thr202/Tyr204 was also increased by GTP. CONCLUSION Our studies suggest that GTP promoted proliferation and secretion of collagen in NIH/3T3 cells by activating the ERK signal pathway, which shed light on a potential function of GTP in promoting wound healing.
Collapse
Affiliation(s)
- Xuenan Chen
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Manying Wang
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaohao Xu
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
| | - Jianzeng Liu
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bing Mei
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
- College of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Pingping Fu
- China–Japan Union Hospital and First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Daqing Zhao
- Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, Changchun, Jilin, China
- Corresponding author. Research and Development Center of Traditional Chinese Medicine and Biological Engineering, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, Jilin Province 130021, China.Research and Development Center of Traditional Chinese Medicine and Biological EngineeringChangchun University of Chinese Medicine1035 Boshuo RoadChangchunJilin Province130021China
| | - Liwei Sun
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, Jilin, China
- Corresponding author. Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Biology and Chemistry, Beihua University, 15 Jilin Street, Jilin, Jilin Province 132013, China.Jilin Technology Innovation Center for Chinese Medicine BiotechnologyCollege of Biology and ChemistryBeihua University15 Jilin StreetJilinJilin Province132013China
| |
Collapse
|
68
|
Bakhoum SF, Kabeche L, Compton DA, Powell SN, Bastians H. Mitotic DNA Damage Response: At the Crossroads of Structural and Numerical Cancer Chromosome Instabilities. Trends Cancer 2017; 3:225-234. [PMID: 28718433 DOI: 10.1016/j.trecan.2017.02.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/04/2017] [Accepted: 02/06/2017] [Indexed: 11/29/2022]
Abstract
DNA double-strand breaks (DSBs) prevent cells from entering mitosis allowing cells to repair their genomic damage. Little is known about the response to DSBs once cells have already committed to mitosis. Here, we review the genome-protective role of the mitotic DNA damage response (DDR) and evidence suggesting that its untimely activation induces chromosome segregation errors and paradoxically undermines genomic integrity. In contrast to normal cells, cancer cells coopt this pathway to propagate structural and numerical chromosomal instabilities. Cells derived from genomically unstable tumors exhibit evidence for a partially activated DDR during mitosis, which leads to ongoing chromosome segregation errors. Thus, a thorough understanding of the consequences of mitotic DNA damage is key to our ability to devise novel anticancer therapeutic strategies.
Collapse
Affiliation(s)
- Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Lilian Kabeche
- Massachusetts General Hospital Cancer Center, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Duane A Compton
- Department of Biochemistry and the Norris-Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Holger Bastians
- Institute of Molecular Oncology, Section for Cellular Oncology, Goettingen Center for Molecular Biosciences (GZMB) and University Medical Center, University of Göttingen, D-37077 Goettingen, Germany
| |
Collapse
|
69
|
Meadows JC, Lancaster TC, Buttrick GJ, Sochaj AM, Messin LJ, Del Mar Mora-Santos M, Hardwick KG, Millar JBA. Identification of a Sgo2-Dependent but Mad2-Independent Pathway Controlling Anaphase Onset in Fission Yeast. Cell Rep 2017; 18:1422-1433. [PMID: 28178520 PMCID: PMC5316559 DOI: 10.1016/j.celrep.2017.01.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/08/2016] [Accepted: 01/15/2017] [Indexed: 10/28/2022] Open
Abstract
The onset of anaphase is triggered by activation of the anaphase-promoting complex/cyclosome (APC/C) following silencing of the spindle assembly checkpoint (SAC). APC/C triggers ubiquitination of Securin and Cyclin B, which leads to loss of sister chromatid cohesion and inactivation of Cyclin B/Cdk1, respectively. This promotes relocalization of Aurora B kinase and other components of the chromosome passenger complex (CPC) from centromeres to the spindle midzone. In fission yeast, this is mediated by Clp1 phosphatase-dependent interaction of CPC with Klp9/MKLP2 (kinesin-6). When this interaction is disrupted, kinetochores bi-orient normally, but APC/C activation is delayed via a mechanism that requires Sgo2 and some (Bub1, Mph1/Mps1, and Mad3), but not all (Mad1 and Mad2), components of the SAC and the first, but not second, lysine, glutamic acid, glutamine (KEN) box in Mad3. These data indicate that interaction of CPC with Klp9 terminates a Sgo2-dependent, but Mad2-independent, APC/C-inhibitory pathway that is distinct from the canonical SAC.
Collapse
Affiliation(s)
- John C Meadows
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; Institute of Advanced Study, University of Warwick, Coventry CV4 7AL, UK
| | - Theresa C Lancaster
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Graham J Buttrick
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Alicja M Sochaj
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| | - Liam J Messin
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Maria Del Mar Mora-Santos
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Kevin G Hardwick
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| | - Jonathan B A Millar
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
70
|
Sansregret L, Patterson JO, Dewhurst S, López-García C, Koch A, McGranahan N, Chao WCH, Barry DJ, Rowan A, Instrell R, Horswell S, Way M, Howell M, Singleton MR, Medema RH, Nurse P, Petronczki M, Swanton C. APC/C Dysfunction Limits Excessive Cancer Chromosomal Instability. Cancer Discov 2017; 7:218-233. [PMID: 28069571 PMCID: PMC5300100 DOI: 10.1158/2159-8290.cd-16-0645] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 01/25/2023]
Abstract
Intercellular heterogeneity, exacerbated by chromosomal instability (CIN), fosters tumor heterogeneity and drug resistance. However, extreme CIN correlates with improved cancer outcome, suggesting that karyotypic diversity required to adapt to selection pressures might be balanced in tumors against the risk of excessive instability. Here, we used a functional genomics screen, genome editing, and pharmacologic approaches to identify CIN-survival factors in diploid cells. We find partial anaphase-promoting complex/cyclosome (APC/C) dysfunction lengthens mitosis, suppresses pharmacologically induced chromosome segregation errors, and reduces naturally occurring lagging chromosomes in cancer cell lines or following tetraploidization. APC/C impairment caused adaptation to MPS1 inhibitors, revealing a likely resistance mechanism to therapies targeting the spindle assembly checkpoint. Finally, CRISPR-mediated introduction of cancer somatic mutations in the APC/C subunit cancer driver gene CDC27 reduces chromosome segregation errors, whereas reversal of an APC/C subunit nonsense mutation increases CIN. Subtle variations in mitotic duration, determined by APC/C activity, influence the extent of CIN, allowing cancer cells to dynamically optimize fitness during tumor evolution. SIGNIFICANCE We report a mechanism whereby cancers balance the evolutionary advantages associated with CIN against the fitness costs caused by excessive genome instability, providing insight into the consequence of CDC27 APC/C subunit driver mutations in cancer. Lengthening of mitosis through APC/C modulation may be a common mechanism of resistance to cancer therapeutics that increase chromosome segregation errors. Cancer Discov; 7(2); 218-33. ©2017 AACR.See related commentary by Burkard and Weaver, p. 134This article is highlighted in the In This Issue feature, p. 115.
Collapse
Affiliation(s)
| | | | | | | | - André Koch
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nicholas McGranahan
- The Francis Crick Institute, London, United Kingdom
- CRUK UCL/Manchester Lung Cancer Centre of Excellence
| | | | | | - Andrew Rowan
- The Francis Crick Institute, London, United Kingdom
| | | | | | - Michael Way
- The Francis Crick Institute, London, United Kingdom
| | | | | | - René H. Medema
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul Nurse
- The Francis Crick Institute, London, United Kingdom
| | - Mark Petronczki
- The Francis Crick Institute, London, United Kingdom
- Boehringer Ingelheim, Vienna, Austria
| | - Charles Swanton
- The Francis Crick Institute, London, United Kingdom
- CRUK UCL/Manchester Lung Cancer Centre of Excellence
| |
Collapse
|
71
|
Lampson MA, Grishchuk EL. Mechanisms to Avoid and Correct Erroneous Kinetochore-Microtubule Attachments. BIOLOGY 2017; 6:E1. [PMID: 28067761 PMCID: PMC5371994 DOI: 10.3390/biology6010001] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/24/2016] [Accepted: 12/28/2016] [Indexed: 12/19/2022]
Abstract
In dividing vertebrate cells multiple microtubules must connect to mitotic kinetochores in a highly stereotypical manner, with each sister kinetochore forming microtubule attachments to only one spindle pole. The exact sequence of events by which this goal is achieved varies considerably from cell to cell because of the variable locations of kinetochores and spindle poles, and randomness of initial microtubule attachments. These chance encounters with the kinetochores nonetheless ultimately lead to the desired outcome with high fidelity and in a limited time frame, providing one of the most startling examples of biological self-organization. This chapter discusses mechanisms that contribute to accurate chromosome segregation by helping dividing cells to avoid and resolve improper microtubule attachments.
Collapse
Affiliation(s)
- Michael A Lampson
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
72
|
Abstract
Chromosomal aberrations during cell division represent one of the first recognized features of human cancer cells, and modern detection methods have revealed the pervasiveness of aneuploidy in cancer. The ongoing karyotypic changes brought about by chromosomal instability (CIN) contribute to tumor heterogeneity, drug resistance, and treatment failure. Whole-chromosome and segmental aneuploidies resulting from CIN have been proposed to allow "macroevolutionary" leaps that may contribute to profound phenotypic change. In this review, we will outline evidence indicating that aneuploidy and CIN contribute to cancer evolution.
Collapse
Affiliation(s)
- Laurent Sansregret
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratories, London WC2A 3LY, United Kingdom
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratories, London WC2A 3LY, United Kingdom
- CRUK Lung Cancer Centre of Excellence/UCL Cancer Institute, London WC1E 6BT, United Kingdom
| |
Collapse
|
73
|
Radford SJ, Nguyen AL, Schindler K, McKim KS. The chromosomal basis of meiotic acentrosomal spindle assembly and function in oocytes. Chromosoma 2016; 126:351-364. [PMID: 27837282 DOI: 10.1007/s00412-016-0618-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022]
Abstract
Several aspects of meiosis are impacted by the absence of centrosomes in oocytes. Here, we review four aspects of meiosis I that are significantly affected by the absence of centrosomes in oocyte spindles. One, microtubules tend to assemble around the chromosomes. Two, the organization of these microtubules into a bipolar spindle is directed by the chromosomes. Three, chromosome bi-orientation and attachment to microtubules from the correct pole require modification of the mechanisms used in mitotic cells. Four, chromosome movement to the poles at anaphase cannot rely on polar anchoring of spindle microtubules by centrosomes. Overall, the chromosomes are more active participants during acentrosomal spindle assembly in oocytes, compared to mitotic and male meiotic divisions where centrosomes are present. The chromosomes are endowed with information that can direct the meiotic divisions and dictate their own behavior in oocytes. Processes beyond those known from mitosis appear to be required for their bi-orientation at meiosis I. As mitosis occurs without centrosomes in many systems other than oocytes, including all plants, the concepts discussed here may not be limited to oocytes. The study of meiosis in oocytes has revealed mechanisms that are operating in mitosis and will probably continue to do so.
Collapse
Affiliation(s)
- Sarah J Radford
- Waksman Institute, 190 Frelinghuysen Rd, Piscataway, NJ, 08854, USA
| | | | - Karen Schindler
- Department of Genetics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Kim S McKim
- Waksman Institute, 190 Frelinghuysen Rd, Piscataway, NJ, 08854, USA.
- Department of Genetics, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
74
|
Distinct and Overlapping Requirements for Cyclins A, B, and B3 in Drosophila Female Meiosis. G3-GENES GENOMES GENETICS 2016; 6:3711-3724. [PMID: 27652889 PMCID: PMC5100870 DOI: 10.1534/g3.116.033050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Meiosis, like mitosis, depends on the activity of the cyclin dependent kinase Cdk1 and its cyclin partners. Here, we examine the specific requirements for the three mitotic cyclins, A, B, and B3 in meiosis of Drosophila melanogaster. We find that all three cyclins contribute redundantly to nuclear envelope breakdown, though cyclin A appears to make the most important individual contribution. Cyclin A is also required for biorientation of homologs in meiosis I. Cyclin B3, as previously reported, is required for anaphase progression in meiosis I and in meiosis II. We find that it also plays a redundant role, with cyclin A, in preventing DNA replication during meiosis. Cyclin B is required for maintenance of the metaphase I arrest in mature oocytes, for spindle organization, and for timely progression through the second meiotic division. It is also essential for polar body formation at the completion of meiosis. With the exception of its redundant role in meiotic maturation, cyclin B appears to function independently of cyclins A and B3 through most of meiosis. We conclude that the three mitotic cyclin-Cdk complexes have distinct and overlapping functions in Drosophila female meiosis.
Collapse
|
75
|
CDK-1 Inhibition in G2 Stabilizes Kinetochore-Microtubules in the following Mitosis. PLoS One 2016; 11:e0157491. [PMID: 27281342 PMCID: PMC4900577 DOI: 10.1371/journal.pone.0157491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/30/2016] [Indexed: 11/18/2022] Open
Abstract
Cell proliferation is driven by cyclical activation of cyclin-dependent kinases (CDKs), which produce distinct biochemical cell cycle phases. Mitosis (M phase) is orchestrated by CDK-1, complexed with mitotic cyclins. During M phase, chromosomes are segregated by a bipolar array of microtubules called the mitotic spindle. The essential bipolarity of the mitotic spindle is established by the kinesin-5 Eg5, but factors influencing the maintenance of spindle bipolarity are not fully understood. Here, we describe an unexpected link between inhibiting CDK-1 before mitosis and bipolar spindle maintenance. Spindles in human RPE-1 cells normally collapse to monopolar structures when Eg5 is inhibited at metaphase. However, we found that inhibition of CDK-1 in the G2 phase of the cell cycle improved the ability of RPE-1 cells to maintain spindle bipolarity without Eg5 activity in the mitosis immediately after release from CDK-1 inhibition. This improved bipolarity maintenance correlated with an increase in the stability of kinetochore-microtubules, the subset of microtubules that link chromosomes to the spindle. The improvement in bipolarity maintenance after CDK-1 inhibition in G2 required both the kinesin-12 Kif15 and increased stability of kinetochore-microtubules. Consistent with increased kinetochore-microtubule stability, we find that inhibition of CDK-1 in G2 impairs mitotic fidelity by increasing the incidence of lagging chromosomes in anaphase. These results suggest that inhibition of CDK-1 in G2 causes unpredicted effects in mitosis, even after CDK-1 inhibition is relieved.
Collapse
|
76
|
Carniel FC, Gerdol M, Montagner A, Banchi E, De Moro G, Manfrin C, Muggia L, Pallavicini A, Tretiach M. New features of desiccation tolerance in the lichen photobiont Trebouxia gelatinosa are revealed by a transcriptomic approach. PLANT MOLECULAR BIOLOGY 2016; 91:319-339. [PMID: 26992400 DOI: 10.1007/s11103-016-0468-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/04/2016] [Indexed: 06/05/2023]
Abstract
Trebouxia is the most common lichen-forming genus of aero-terrestrial green algae and all its species are desiccation tolerant (DT). The molecular bases of this remarkable adaptation are, however, still largely unknown. We applied a transcriptomic approach to a common member of the genus, T. gelatinosa, to investigate the alteration of gene expression occurring after dehydration and subsequent rehydration in comparison to cells kept constantly hydrated. We sequenced, de novo assembled and annotated the transcriptome of axenically cultured T. gelatinosa by using Illumina sequencing technology. We tracked the expression profiles of over 13,000 protein-coding transcripts. During the dehydration/rehydration cycle c. 92 % of the total protein-coding transcripts displayed a stable expression, suggesting that the desiccation tolerance of T. gelatinosa mostly relies on constitutive mechanisms. Dehydration and rehydration affected mainly the gene expression for components of the photosynthetic apparatus, the ROS-scavenging system, Heat Shock Proteins, aquaporins, expansins, and desiccation related proteins (DRPs), which are highly diversified in T. gelatinosa, whereas Late Embryogenesis Abundant Proteins were not affected. Only some of these phenomena were previously observed in other DT green algae, bryophytes and resurrection plants, other traits being distinctive of T. gelatinosa, and perhaps related to its symbiotic lifestyle. Finally, the phylogenetic inference extended to DRPs of other chlorophytes, embryophytes and bacteria clearly pointed out that DRPs of chlorophytes are not orthologous to those of embryophytes: some of them were likely acquired through horizontal gene transfer from extremophile bacteria which live in symbiosis within the lichen thallus.
Collapse
Affiliation(s)
- Fabio Candotto Carniel
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
- Institute of Botany, University of Innsbruck, Sternwartestraße, 15, 6020, Innsbruck, Austria
| | - Marco Gerdol
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy.
| | - Alice Montagner
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Elisa Banchi
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Gianluca De Moro
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Chiara Manfrin
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Lucia Muggia
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Alberto Pallavicini
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| | - Mauro Tretiach
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via L. Giorgieri, 10, 34127, Trieste, Italy
| |
Collapse
|
77
|
Li C, Xue C, Yang Q, Low BC, Liou YC. NuSAP governs chromosome oscillation by facilitating the Kid-generated polar ejection force. Nat Commun 2016; 7:10597. [PMID: 26839278 PMCID: PMC4742958 DOI: 10.1038/ncomms10597] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022] Open
Abstract
In vertebrate cells, chromosomes oscillate to align precisely during metaphase. NuSAP, a microtubule-associated protein, plays a critical role in stabilizing spindle microtubules. In this study, we utilize 3D time-lapse live-cell imaging to monitor the role of NuSAP in chromosome oscillation and identify NuSAP as a novel regulator of the chromokinesin, Kid. Depletion of NuSAP significantly suppresses the amplitude and velocity of chromosome oscillation. We analyse the effects of NuSAP and Kid depletion in monopolar and bipolar cells with or without kinetochore microtubule depletion. Twelve postulated conditions are deciphered to reveal the contribution of NuSAP to the polar force generated at kinetochore microtubules and to the regulation of the polar ejection force generated by Kid, thus revealing a pivotal role of NuSAP in chromosome oscillation. During metaphase, alignment of chromosomes is facilitated by oscillations driven by the chromokinesin Kid. Here Li et al. show that the microtubule-associated protein NuSAP is a novel regulator of Kid, regulating the amplitude and velocity of chromosome oscillation.
Collapse
Affiliation(s)
- Chenyu Li
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore
| | - Chenyi Xue
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Qiaoyun Yang
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore
| | - Boon Chuan Low
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore.,Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore.,Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117573, Republic of Singapore
| |
Collapse
|
78
|
Acetylation of Aurora B by TIP60 ensures accurate chromosomal segregation. Nat Chem Biol 2016; 12:226-32. [PMID: 26829474 PMCID: PMC4798883 DOI: 10.1038/nchembio.2017] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/11/2015] [Indexed: 01/19/2023]
Abstract
Faithful segregation of chromosomes in mammalian cells requires bi-orientation of sister chromatids, which relies on the sensing of correct attachments between spindle microtubules and kinetochores. Although the mechanisms underlying cyclin-dependent kinase 1 (CDK1) activation, which triggers mitotic entry, have been extensively studied, the regulatory mechanisms that couple CDK1-cyclin B activity to chromosome stability are not well understood. Here, we identified a signaling axis in which Aurora B activity is modulated by CDK1-cyclin B via the acetyltransferase TIP60 in human cell division. CDK1-cyclin B phosphorylates Ser90 of TIP60, which elicits TIP60-dependent acetylation of Aurora B and promotes accurate chromosome segregation in mitosis. Mechanistically, TIP60 acetylation of Aurora B at Lys215 protects Aurora B's activation loop from dephosphorylation by the phosphatase PP2A to ensure a robust, error-free metaphase-anaphase transition. These findings delineate a conserved signaling cascade that integrates protein phosphorylation and acetylation with cell cycle progression for maintenance of genomic stability.
Collapse
|
79
|
DeLuca KF, Herman JA, DeLuca JG. Measuring Kinetochore-Microtubule Attachment Stability in Cultured Cells. Methods Mol Biol 2016; 1413:147-168. [PMID: 27193848 DOI: 10.1007/978-1-4939-3542-0_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Duplicated sister chromatids connect to the mitotic spindle through kinetochores, large proteinaceous structures built at sites of centromeric heterochromatin. Kinetochores are responsible for harnessing the forces generated by microtubule polymerization and depolymerization to power chromosome movements. The fidelity of chromosome segregation relies on proper kinetochore function, as precise regulation of the attachment between kinetochores and microtubules is essential to prevent mitotic errors, which are linked to the initiation and progression of cancer and the formation of birth defects (Godek et al., Nat Rev Mol Cell Biol 16(1):57-64, 2014; Ricke and van Deursen, Semin Cell Dev Biol 22(6):559-565, 2011; Holland and Cleveland, EMBO Rep 13(6):501-514, 2012). Here we describe assays to quantitatively measure kinetochore-microtubule attachment stability in cultured cells.
Collapse
Affiliation(s)
- Keith F DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, MRB 237, Fort Collins, CO, 80523, USA
| | - Jacob A Herman
- Department of Biochemistry and Molecular Biology, Colorado State University, MRB 237, Fort Collins, CO, 80523, USA
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, MRB 237, Fort Collins, CO, 80523, USA.
| |
Collapse
|
80
|
Drpic D, Pereira AJ, Barisic M, Maresca TJ, Maiato H. Polar Ejection Forces Promote the Conversion from Lateral to End-on Kinetochore-Microtubule Attachments on Mono-oriented Chromosomes. Cell Rep 2015; 13:460-468. [PMID: 26456825 PMCID: PMC4623360 DOI: 10.1016/j.celrep.2015.08.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/08/2015] [Accepted: 08/03/2015] [Indexed: 11/17/2022] Open
Abstract
Chromosome bi-orientation occurs after conversion of initial lateral attachments between kinetochores and spindle microtubules into stable end-on attachments near the cell equator. After bi-orientation, chromosomes experience tension from spindle forces, which plays a key role in the stabilization of correct kinetochore-microtubule attachments. However, how end-on kinetochore-microtubule attachments are first stabilized in the absence of tension remains a key unanswered question. To address this, we generated Drosophila S2 cells undergoing mitosis with unreplicated genomes (SMUGs). SMUGs retained single condensed chromatids that attached laterally to spindle microtubules. Over time, laterally attached kinetochores converted into end-on attachments and experienced intra-kinetochore stretch/structural deformation, and SMUGs eventually exited a delayed mitosis with mono-oriented chromosomes after satisfying the spindle-assembly checkpoint (SAC). Polar ejection forces (PEFs) generated by Chromokinesins promoted the conversion from lateral to end-on kinetochore-microtubule attachments that satisfied the SAC in SMUGs. Thus, PEFs convert lateral to stable end-on kinetochore-microtubule attachments, independently of chromosome bi-orientation. Spindle assembly checkpoint (SAC) can be satisfied after a delay in cells with mono-oriented chromosomes Mono-oriented chromosomes experience intra-kinetochore stretch Polar ejection forces promote SAC satisfaction independently of bi-orientation Polar ejection forces promote the conversion from lateral to end-on attachments
Collapse
Affiliation(s)
- Danica Drpic
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4099-002 Porto, Portugal; Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciencias Biomedicas Abel Salazar da Universidade do Porto, Rua de Jorge Viterbo Ferreira no. 228, 4050-313 Porto, Portugal
| | - António J Pereira
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4099-002 Porto, Portugal
| | - Marin Barisic
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4099-002 Porto, Portugal
| | - Thomas J Maresca
- Biology Department, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Helder Maiato
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4099-002 Porto, Portugal; Cell Division Unit, Department of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
81
|
Defective sister chromatid cohesion is synthetically lethal with impaired APC/C function. Nat Commun 2015; 6:8399. [PMID: 26423134 PMCID: PMC4600715 DOI: 10.1038/ncomms9399] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 08/19/2015] [Indexed: 01/05/2023] Open
Abstract
Warsaw breakage syndrome (WABS) is caused by defective DDX11, a DNA helicase that is essential for chromatid cohesion. Here, a paired genome-wide siRNA screen in patient-derived cell lines reveals that WABS cells do not tolerate partial depletion of individual APC/C subunits or the spindle checkpoint inhibitor p31comet. A combination of reduced cohesion and impaired APC/C function also leads to fatal mitotic arrest in diploid RPE1 cells. Moreover, WABS cell lines, and several cancer cell lines with cohesion defects, display a highly increased response to a new cell-permeable APC/C inhibitor, apcin, but not to the spindle poison paclitaxel. Synthetic lethality of APC/C inhibition and cohesion defects strictly depends on a functional mitotic spindle checkpoint as well as on intact microtubule pulling forces. This indicates that the underlying mechanism involves cohesion fatigue in response to mitotic delay, leading to spindle checkpoint re-activation and lethal mitotic arrest. Our results point to APC/C inhibitors as promising therapeutic agents targeting cohesion-defective cancers. Cohesion is associated with many forms of cancer. De Lange et al. show that such cohesion defects can sensitise cells to apoptosis in response to a new APC/C ubiquitin ligase inhibitor, by prolonging mitotic arrest and checkpoint activation due to cohesion fatigue.
Collapse
|
82
|
Zaytsev AV, Grishchuk EL. Basic mechanism for biorientation of mitotic chromosomes is provided by the kinetochore geometry and indiscriminate turnover of kinetochore microtubules. Mol Biol Cell 2015; 26:3985-98. [PMID: 26424798 PMCID: PMC4710231 DOI: 10.1091/mbc.e15-06-0384] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/22/2015] [Indexed: 12/22/2022] Open
Abstract
Accuracy of chromosome segregation relies on the ill-understood ability of mitotic kinetochores to biorient, whereupon each sister kinetochore forms microtubule (MT) attachments to only one spindle pole. Because initial MT attachments result from chance encounters with the kinetochores, biorientation must rely on specific mechanisms to avoid and resolve improper attachments. Here we use mathematical modeling to critically analyze the error-correction potential of a simplified biorientation mechanism, which involves the back-to-back arrangement of sister kinetochores and the marked instability of kinetochore-MT attachments. We show that a typical mammalian kinetochore operates in a near-optimal regime, in which the back-to-back kinetochore geometry and the indiscriminate kinetochore-MT turnover provide strong error-correction activity. In human cells, this mechanism alone can potentially enable normal segregation of 45 out of 46 chromosomes during one mitotic division, corresponding to a mis-segregation rate in the range of 10(-1)-10(-2) per chromosome. This theoretical upper limit for chromosome segregation accuracy predicted with the basic mechanism is close to the mis-segregation rate in some cancer cells; however, it cannot explain the relatively low chromosome loss in diploid human cells, consistent with their reliance on additional mechanisms.
Collapse
Affiliation(s)
- Anatoly V Zaytsev
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
83
|
Uniparental disomy of the entire X chromosome in Turner syndrome patient-specific induced pluripotent stem cells. Cell Discov 2015; 1:15022. [PMID: 27462421 PMCID: PMC4860828 DOI: 10.1038/celldisc.2015.22] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/30/2015] [Indexed: 12/21/2022] Open
Abstract
The human induced pluripotent stem cell (iPSC) technique promises to provide an unlimited, reliable source of genetically matched pluripotent cells for personalized therapy and disease modeling. Recently, it is observed that cells with ring chromosomes 13 or 17 autonomously correct the defects via compensatory uniparental disomy during cellular reprogramming to iPSCs. This breakthrough finding suggests a potential therapeutic approach to repair large-scale chromosomal aberrations. However, due to the scarceness of ring chromosome samples, the reproducibility of this approach in different individuals is not carefully evaluated yet. Moreover, the underlying mechanism and the applicability to other types of chromosomal aberrations remain unknown. Here we generated iPSCs from four 45,X chorionic villous fibroblast lines and found that only one reprogrammed line acquired 46,XX karyotype via uniparental disomy of the entire X chromosome. The karyotype correction was reproducible in the same cell line by either retroviral or episomal reprogramming. The karyotype-corrected iPSCs were subject to X chromosome inactivation and obtained better colony morphology and higher proliferation rate than other uncorrected ones. Further transcriptomic comparison among the fibroblast lines identified a distinct expression pattern of cell cycle regulators in the uncorrectable ones. These findings demonstrate that the iPSC technique holds the potential to correct X monosomy, but the correction rate is very low, probably due to differential regulation of cell cycle genes between individuals. Our data strongly suggest that more systematic investigations are needed before defining the iPSC technique as a novel means of chromosome therapy.
Collapse
|
84
|
Touati SA, Wassmann K. How oocytes try to get it right: spindle checkpoint control in meiosis. Chromosoma 2015; 125:321-35. [PMID: 26255654 DOI: 10.1007/s00412-015-0536-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/09/2015] [Accepted: 07/20/2015] [Indexed: 11/27/2022]
Abstract
The generation of a viable, diploid organism depends on the formation of haploid gametes, oocytes, and spermatocytes, with the correct number of chromosomes. Halving the genome requires the execution of two consecutive specialized cell divisions named meiosis I and II. Unfortunately, and in contrast to male meiosis, chromosome segregation in oocytes is error prone, with human oocytes being extraordinarily "meiotically challenged". Aneuploid oocytes, that are with the wrong number of chromosomes, give rise to aneuploid embryos when fertilized. In humans, most aneuploidies are lethal and result in spontaneous abortions. However, some trisomies survive to birth or even adulthood, such as the well-known trisomy 21, which gives rise to Down syndrome (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012). A staggering 20-25 % of oocytes ready to be fertilized are aneuploid in humans. If this were not bad enough, there is an additional increase in meiotic missegregations as women get closer to menopause. A woman above 40 has a risk of more than 30 % of getting pregnant with a trisomic child. Worse still, in industrialized western societies, child birth is delayed, with women getting their first child later in life than ever. This trend has led to an increase of trisomic pregnancies by 70 % in the last 30 years (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012; Schmidt et al. in Hum Reprod Update 18:29-43, 2012). To understand why errors occur so frequently during the meiotic divisions in oocytes, we review here the molecular mechanisms at works to control chromosome segregation during meiosis. An important mitotic control mechanism, namely the spindle assembly checkpoint or SAC, has been adapted to the special requirements of the meiotic divisions, and this review will focus on our current knowledge of SAC control in mammalian oocytes. Knowledge on how chromosome segregation is controlled in mammalian oocytes may help to identify risk factors important for questions related to human reproductive health.
Collapse
Affiliation(s)
- Sandra A Touati
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.,Chromosome Segregation Laboratory, Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London, UK
| | - Katja Wassmann
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France. .,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.
| |
Collapse
|
85
|
Papini D, Langemeyer L, Abad MA, Kerr A, Samejima I, Eyers PA, Jeyaprakash AA, Higgins JMG, Barr FA, Earnshaw WC. TD-60 links RalA GTPase function to the CPC in mitosis. Nat Commun 2015; 6:7678. [PMID: 26158537 PMCID: PMC4510650 DOI: 10.1038/ncomms8678] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 05/29/2015] [Indexed: 12/26/2022] Open
Abstract
TD-60 (also known as RCC2) is a highly conserved protein that structurally resembles the Ran guanine exchange factor (GEF) RCC1, but has not previously been shown to have GEF activity. TD-60 has a typical chromosomal passenger complex (CPC) distribution in mitotic cells, but associates with integrin complexes and is involved in cell motility during interphase. Here we show that TD-60 exhibits GEF activity, in vitro and in cells, for the small GTPase RalA. TD-60 or RalA depletion causes spindle abnormalities in prometaphase associated with abnormal centromeric accumulation of CPC components. TD-60 and RalA apparently work together to contribute to the regulation of kinetochore-microtubule interactions in early mitosis. Importantly, several mitotic phenotypes caused by TD-60 depletion are reverted by the expression of a GTP-locked mutant, RalA (Q72L). The demonstration that a small GTPase participates in the regulation of the CPC reveals a level of mitotic regulation not suspected in previous studies.
Collapse
Affiliation(s)
- Diana Papini
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Lars Langemeyer
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Maria A. Abad
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Alastair Kerr
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Itaru Samejima
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Patrick A. Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
| | - A. Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Jonathan M. G. Higgins
- Institute for Cell and Molecular Biosciences (ICaMB), Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - William C. Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| |
Collapse
|
86
|
Yoshida S, Kaido M, Kitajima TS. Inherent Instability of Correct Kinetochore-Microtubule Attachments during Meiosis I in Oocytes. Dev Cell 2015; 33:589-602. [PMID: 26028219 DOI: 10.1016/j.devcel.2015.04.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/03/2015] [Accepted: 04/24/2015] [Indexed: 01/08/2023]
Abstract
A model for mitosis suggests that correct kinetochore-microtubule (KT-MT) attachments are stabilized by spatial separation of the attachment sites from Aurora B kinase through sister KT stretching. However, the spatiotemporal regulation of attachment stability during meiosis I (MI) in oocytes remains unclear. Here, we found that in mouse oocytes, Aurora B and C (B/C) are located in close proximity to KT-MT attachment sites after bivalent stretching due to an intrinsic property of the MI chromosomes. The Aurora B/C activity destabilizes correct attachments while allowing a considerable amount of incorrect attachments to form. KT-MT attachments are eventually stabilized through KT dephosphorylation by PP2A-B56 phosphatase, which is progressively recruited to KTs depending on the BubR1 phosphorylation resulting from the timer Cdk1 and independent of bivalent stretching. Thus, oocytes lack a mechanism for coordinating bivalent stretching and KT phosphoregulation during MI, which may explain the high frequency of KT-MT attachment errors.
Collapse
Affiliation(s)
- Shuhei Yoshida
- Laboratory for Chromosome Segregation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Masako Kaido
- Laboratory for Chromosome Segregation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Tomoya S Kitajima
- Laboratory for Chromosome Segregation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| |
Collapse
|
87
|
Darzynkiewicz Z, Zhao H, Zhang S, Marietta YL, Ernest YL, Zhang Z. Initiation and termination of DNA replication during S phase in relation to cyclins D1, E and A, p21WAF1, Cdt1 and the p12 subunit of DNA polymerase δ revealed in individual cells by cytometry. Oncotarget 2015; 6:11735-50. [PMID: 26059433 PMCID: PMC4494901 DOI: 10.18632/oncotarget.4149] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 05/03/2015] [Indexed: 12/18/2022] Open
Abstract
During our recent studies on mechanism of the regulation of human DNA polymerase δ in preparation for DNA replication or repair, multiparameter imaging cytometry as exemplified by laser scanning cytometry (LSC) has been used to assess changes in expression of the following nuclear proteins associated with initiation of DNA replication: cyclin A, PCNA, Ki-67, p21(WAF1), DNA replication factor Cdt1 and the smallest subunit of DNA polymerase δ, p12. In the present review, rather than focusing on Pol δ, we emphasize the application of LSC in these studies and outline possibilities offered by the concurrent differential analysis of DNA replication in conjunction with expression of the nuclear proteins. A more extensive analysis of the data on a correlation between rates of EdU incorporation, likely reporting DNA replication, and expression of these proteins, is presently provided. New data, specifically on the expression of cyclin D1 and cyclin E with respect to EdU incorporation as well as on a relationship between expression of cyclin A vs. p21(WAF1) and Ki-67 vs. Cdt1, are also reported. Of particular interest is the observation that this approach makes it possible to assess the temporal sequence of degradation of cyclin D1, p21(WAF1), Cdt1 and p12, each with respect to initiation of DNA replication and with respect to each other. Also the sequence or reappearance of these proteins in G2 after termination of DNA replication is assessed. The reviewed data provide a more comprehensive presentation of potential markers, whose presence or absence marks the DNA replicating cells. Discussed is also usefulness of these markers as indicators of proliferative activity in cancer tissues that may bear information on tumor progression and have a prognostic value.
Collapse
Affiliation(s)
- Zbigniew Darzynkiewicz
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY
| | - Hong Zhao
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY
| | - Sufang Zhang
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY
| | - Y.W.T. Lee Marietta
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY
| | - Y.C. Lee Ernest
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY
| | - Zhongtao Zhang
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY
| |
Collapse
|
88
|
Voets E, Wolthuis R. MASTL promotes cyclin B1 destruction by enforcing Cdc20-independent binding of cyclin B1 to the APC/C. Biol Open 2015; 4:484-95. [PMID: 25750436 PMCID: PMC4400591 DOI: 10.1242/bio.201410793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
When cells enter mitosis, the anaphase-promoting complex/cyclosome (APC/C) is activated by phosphorylation and binding of Cdc20. The RXXL destruction box (D-box) of cyclin B1 only binds Cdc20 after release of the spindle checkpoint in metaphase, initiating cyclin B1 ubiquitination upon chromosome bi-orientation. However, we found that cyclin B1, through Cdk1 and Cks, is targeted to the phosphorylated APC/CCdc20 at the start of prometaphase, when the spindle checkpoint is still active. Here, we show that MASTL is essential for cyclin B1 recruitment to the mitotic APC/C and that this occurs entirely independently of Cdc20. Importantly, MASTL-directed binding of cyclin B1 to spindle checkpoint-inhibited APC/CCdc20 critically supports efficient cyclin B1 destruction after checkpoint release. A high incidence of anaphase bridges observed in response to MASTL RNAi may result from cyclin B1 remaining after securin destruction, which is insufficient to keep MASTL-depleted cells in mitosis but delays the activation of separase.
Collapse
Affiliation(s)
- Erik Voets
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Institute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Rob Wolthuis
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Institute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands Section of Oncogenetics, Department of Clinical Genetics and CCA/V-ICI Research Program Oncogenesis, VUmc Medical Faculty, van de Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|
89
|
Di Fiore B, Davey NE, Hagting A, Izawa D, Mansfeld J, Gibson TJ, Pines J. The ABBA motif binds APC/C activators and is shared by APC/C substrates and regulators. Dev Cell 2015; 32:358-372. [PMID: 25669885 PMCID: PMC4713905 DOI: 10.1016/j.devcel.2015.01.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 09/03/2014] [Accepted: 01/05/2015] [Indexed: 11/30/2022]
Abstract
The anaphase-promoting complex or cyclosome (APC/C) is the ubiquitin ligase that regulates mitosis by targeting specific proteins for degradation at specific times under the control of the spindle assembly checkpoint (SAC). How the APC/C recognizes its different substrates is a key problem in the control of cell division. Here, we have identified the ABBA motif in cyclin A, BUBR1, BUB1, and Acm1, and we show that it binds to the APC/C coactivator CDC20. The ABBA motif in cyclin A is required for its proper degradation in prometaphase through competing with BUBR1 for the same site on CDC20. Moreover, the ABBA motifs in BUBR1 and BUB1 are necessary for the SAC to work at full strength and to recruit CDC20 to kinetochores. Thus, we have identified a conserved motif integral to the proper control of mitosis that connects APC/C substrate recognition with the SAC.
Collapse
Affiliation(s)
- Barbara Di Fiore
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Norman E. Davey
- Department of Physiology and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Baden-Württemberg 69117, Germany
| | - Anja Hagting
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Daisuke Izawa
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Jörg Mansfeld
- Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Toby J. Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Baden-Württemberg 69117, Germany
| | - Jonathon Pines
- The Gurdon Institute and Department of Zoology, University of Cambridge, Cambridge, CB2 1QN, UK
| |
Collapse
|
90
|
Abstract
In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.
Collapse
Affiliation(s)
- Samuel Wieser
- The Gurdon Institute, Cambridge CB2 1QN, United Kingdom
| | | |
Collapse
|
91
|
Abstract
The majority of human cancer cells are highly aneuploid harboring chromosome numbers deviating from the modal number of 46. In cancer, aneuploidy is a consequence of an increased rate of whole chromosome missegregation during mitosis, a process known as chromosomal instability (CIN). In fact, CIN is a hallmark of human cancer and is thought to contribute to tumorigenesis, tumor progression, and the development of therapy resistance by providing a high genetic variability that might foster rapid adaptation processes. However, the molecular mechanisms that cause chromosome missegregation in cancer cells are still poorly understood. So far, several mechanisms underlying CIN have been proposed and some of them are indeed detectable in human cancer cells exhibiting CIN. Examples include, for instance, weakened spindle checkpoint signaling, supernumerary centrosomes, defects in chromatid cohesion, abnormal kinetochore-microtubule attachments and increased spindle microtubule dynamics. Here, the mechanisms leading to CIN in human cancer cells are summarized.
Collapse
Affiliation(s)
- Holger Bastians
- Goettingen Center for Molecular Biosciences (GZMB), University Medical Center, Institute of Molecular Oncology, Section for Cellular Oncology, Georg-August University Goettingen, Grisebachstrasse 8, 37077, Goettingen, Germany.
| |
Collapse
|
92
|
Boekhout M, Wolthuis R. Nek2A destruction marks APC/C activation at the prophase-to-prometaphase transition by spindle-checkpoint restricted Cdc20. J Cell Sci 2015; 128:1639-53. [DOI: 10.1242/jcs.163279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/06/2015] [Indexed: 12/31/2022] Open
Abstract
Nek2A is a presumed APC/CCdc20 substrate, which, like cyclin A, is degraded in mitosis while the spindle checkpoint is active. Cyclin A prevents spindle checkpoint proteins from binding to Cdc20 and is recruited to the APC/C in prometaphase. We found that Nek2A and cyclin A avoid stabilization by the spindle checkpoint in different ways. First, enhancing mitotic checkpoint complex (MCC) formation by nocodazole treatment inhibited the degradation of geminin and cyclin A while Nek2A disappeared at normal rate. Secondly, depleting Cdc20 effectively stabilized cyclin A but not Nek2A. Nevertheless, Nek2A destruction critically depended on Cdc20 binding to the APC/C. Thirdly, in contrast to cyclin A, Nek2A was recruited to the APC/C before the start of mitosis. Interestingly, the spindle checkpoint very effectively stabilized an APC/C-binding mutant of Nek2A, which required the Nek2A KEN box. Apparently, in cells, the spindle checkpoint primarily prevents Cdc20 from binding destruction motifs. Nek2A disappearance marks the prophase-to-prometaphase transition, when Cdc20, regardless of the spindle checkpoint, activates the APC/C. However, Mad2 depletion accelerated Nek2A destruction, showing that spindle checkpoint release further increases APC/CCdc20 catalytic activity.
Collapse
|
93
|
Dong C, Wang XL, Ma BL. Expression of Spindle and Kinetochore-Associated Protein 1 Is Associated with Poor Prognosis in Papillary Thyroid Carcinoma. DISEASE MARKERS 2015; 2015:616541. [PMID: 26063960 PMCID: PMC4434216 DOI: 10.1155/2015/616541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/16/2015] [Indexed: 11/17/2022]
Abstract
AIM Spindle and kinetochore-associated protein 1 (SKA1) is one subtype of SKA, whose protein can make spindle microtubules attach steadily to the kinetochore in the middle of mitosis. At present, there are fewer researches on the relationship between SKA1 expression and tumor development. METHODS In this study, immunohistochemical analysis was used to determine the expression of SKA1 in papillary thyroid carcinoma (PTC) and adjacent tissues. We used quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis to further verify the results. RESULTS We found that SKA1 expression was significantly higher in PTC tissues than normal adjacent tissues (P < 0.05). There existed a significant correlation among a higher SKA1 expression, including lymphoid node (P = 0.005), clinical stage (P = 0.015), and extrathyroid invasion (P = 0.004). Survival analysis showed high SKA1 expression in PTC patients more likely to relapse after surgery. CONCLUSION High SKA1 expression is predictive of poor prognosis of PTC, implying that SKA1 may be a promising new target for targeted therapies for PTC.
Collapse
Affiliation(s)
- Chao Dong
- Department of Head and Neck Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Xiao-li Wang
- Department of Head and Neck Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Bin-lin Ma
- Department of Head and Neck Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, China
- *Bin-lin Ma:
| |
Collapse
|
94
|
The architecture of CCAN proteins creates a structural integrity to resist spindle forces and achieve proper Intrakinetochore stretch. Dev Cell 2014; 30:717-30. [PMID: 25268173 DOI: 10.1016/j.devcel.2014.08.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/26/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
Constitutive centromere-associated network (CCAN) proteins, particularly CENP-C, CENP-T, and the CENP-H/-I complex, mechanically link CENP-A-containing centromeric chromatin within the inner kinetochore to outer kinetochore proteins, such as the Ndc80 complex, that bind kinetochore microtubules. Accuracy of chromosome segregation depends critically upon Aurora B phosphorylation of Ndc80/Hec1. To determine how CCAN protein architecture mechanically constrains intrakinetochore stretch between CENP-A and Ndc80/Hec1 for proper Ndc80/Hec1 phosphorylation, we used super-resolution fluorescence microscopy and selective protein depletion. We found that at bi-oriented chromosomes in late prometaphase cells, CENP-T is stretched ∼16 nm to the inner end of Ndc80/Hec1, much less than expected for full-length CENP-T. Depletion of various CCAN linker proteins induced hyper-intrakinetochore stretch (an additional 20-60 nm) with corresponding significant decreases in Aurora B phosphorylation of Ndc80/Hec1. Thus, proper intrakinetochore stretch is required for normal kinetochore function and depends critically on all the CCAN mechanical linkers to the Ndc80 complex.
Collapse
|
95
|
Regulation of kinetochore-microtubule attachments through homeostatic control during mitosis. Nat Rev Mol Cell Biol 2014; 16:57-64. [PMID: 25466864 DOI: 10.1038/nrm3916] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Faithful chromosome segregation during mitosis is essential for genome integrity and is mediated by the bi-oriented attachment of replicated chromosomes to spindle microtubules through kinetochores. Errors in kinetochore-microtubule (k-MT) attachment that could cause chromosome mis-segregation are frequent and are corrected by the dynamic turnover of k-MT attachments. Thus, regulating the rate of spindle microtubule attachment and detachment to kinetochores is crucial for mitotic fidelity and is frequently disrupted in cancer cells displaying chromosomal instability. A model based on homeostatic principles involving receptors, a core control network, effectors and feedback control may explain the precise regulation of k-MT attachment stability during mitotic progression to ensure error-free mitosis.
Collapse
|
96
|
Lu D, Hsiao JY, Davey NE, Van Voorhis VA, Foster SA, Tang C, Morgan DO. Multiple mechanisms determine the order of APC/C substrate degradation in mitosis. ACTA ACUST UNITED AC 2014; 207:23-39. [PMID: 25287299 PMCID: PMC4195823 DOI: 10.1083/jcb.201402041] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To ensure proper mitotic progression, robust ordering of the destruction of APC/CCdc20 substrates is driven by the integration of molecular mechanisms ranging from phosphorylation-dependent interaction with substrates to sensing of the status of the spindle assembly checkpoint. The ubiquitin protein ligase anaphase-promoting complex or cyclosome (APC/C) controls mitosis by promoting ordered degradation of securin, cyclins, and other proteins. The mechanisms underlying the timing of APC/C substrate degradation are poorly understood. We explored these mechanisms using quantitative fluorescence microscopy of GFP-tagged APC/CCdc20 substrates in living budding yeast cells. Degradation of the S cyclin, Clb5, begins early in mitosis, followed 6 min later by the degradation of securin and Dbf4. Anaphase begins when less than half of securin is degraded. The spindle assembly checkpoint delays the onset of Clb5 degradation but does not influence securin degradation. Early Clb5 degradation depends on its interaction with the Cdk1–Cks1 complex and the presence of a Cdc20-binding “ABBA motif” in its N-terminal region. The degradation of securin and Dbf4 is delayed by Cdk1-dependent phosphorylation near their Cdc20-binding sites. Thus, a remarkably diverse array of mechanisms generates robust ordering of APC/CCdc20 substrate destruction.
Collapse
Affiliation(s)
- Dan Lu
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Jennifer Y Hsiao
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Norman E Davey
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Vanessa A Van Voorhis
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Scott A Foster
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| | - Chao Tang
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - David O Morgan
- Department of Physiology and Department of Biochemistry and Biophysics and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| |
Collapse
|
97
|
Orticello M, Fiore M, Totta P, Desideri M, Barisic M, Passeri D, Lenzi J, Rosa A, Orlandi A, Maiato H, Del Bufalo D, Degrassi F. N-terminus-modified Hec1 suppresses tumour growth by interfering with kinetochore-microtubule dynamics. Oncogene 2014; 34:3325-35. [PMID: 25132262 DOI: 10.1038/onc.2014.265] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 06/11/2014] [Accepted: 07/03/2014] [Indexed: 01/12/2023]
Abstract
Mitotic proteins are attractive targets to develop molecular cancer therapeutics due to the intimate interdependence between cell proliferation and mitosis. In this work, we have explored the therapeutic potential of the kinetochore (KT) protein Hec1 (Highly Expressed in Cancer protein 1) as a molecular target to produce massive chromosome missegregation and cell death in cancer cells. Hec1 is a constituent of the Ndc80 complex, which mediates KT-microtubule (MT) attachments at mitosis and is upregulated in various cancer types. We expressed Hec1 fused with enhanced green fluorescent protein (EGFP) at its N-terminus MT-interaction domain in HeLa cells and showed that expression of this modified Hec1, which localized at KTs, blocked cell proliferation and promoted apoptosis in tumour cells. EGFP-Hec1 was extremely potent in tumour cell killing and more efficient than siRNA-induced Hec1 depletion. In striking contrast, normal cells showed no apparent cell proliferation defects or cell death following EGFP-Hec1 expression. Live-cell imaging demonstrated that cancer cell death was associated with massive chromosome missegregation within multipolar spindles after a prolonged mitotic arrest. Moreover, EGFP-Hec1 expression was found to increase KT-MT attachment stability, providing a molecular explanation for the abnormal spindle architecture and the cytotoxic activity of this modified protein. Consistent with cell culture data, EGFP-Hec1 expression was found to strongly inhibit tumour growth in a mouse xenograft model by disrupting mitosis and inducing multipolar spindles. Taken together, these findings demonstrate that stimulation of massive chromosome segregation defects can be used as an anti-cancer strategy through the activation of mitotic catastrophe after a multipolar mitosis. Importantly, this study represents a clear proof of concept that targeting KT proteins required for proper KT-MT attachment dynamics constitutes a powerful approach in cancer therapy.
Collapse
Affiliation(s)
- M Orticello
- Institute of Biology, Molecular Medicine and Nanobiotechnology, CNR National Research Council, Rome, Italy
| | - M Fiore
- Institute of Biology, Molecular Medicine and Nanobiotechnology, CNR National Research Council, Rome, Italy
| | - P Totta
- Institute of Biology, Molecular Medicine and Nanobiotechnology, CNR National Research Council, Rome, Italy
| | - M Desideri
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - M Barisic
- Chromosome Instability and Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - D Passeri
- Anatomic Pathology Institute, Tor Vergata University, Rome, Italy
| | - J Lenzi
- Department of Biology and Biotechnology 'Charles Darwin', Sapienza University, Rome, Italy
| | - A Rosa
- Department of Biology and Biotechnology 'Charles Darwin', Sapienza University, Rome, Italy
| | - A Orlandi
- Anatomic Pathology Institute, Tor Vergata University, Rome, Italy
| | - H Maiato
- 1] Chromosome Instability and Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal [2] Cell Division Unit, Department of Experimental Biology, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - D Del Bufalo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - F Degrassi
- Institute of Biology, Molecular Medicine and Nanobiotechnology, CNR National Research Council, Rome, Italy
| |
Collapse
|
98
|
Kleyman M, Kabeche L, Compton DA. STAG2 promotes error correction in mitosis by regulating kinetochore-microtubule attachments. J Cell Sci 2014; 127:4225-33. [PMID: 25074805 DOI: 10.1242/jcs.151613] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mutations in the STAG2 gene are present in ∼20% of tumors from different tissues of origin. STAG2 encodes a subunit of the cohesin complex, and tumors with loss-of-function mutations are usually aneuploid and display elevated frequencies of lagging chromosomes during anaphase. Lagging chromosomes are a hallmark of chromosomal instability (CIN) arising from persistent errors in kinetochore-microtubule (kMT) attachment. To determine whether the loss of STAG2 increases the rate of formation of kMT attachment errors or decreases the rate of their correction, we examined mitosis in STAG2-deficient cells. STAG2 depletion does not impair bipolar spindle formation or delay mitotic progression. Instead, loss of STAG2 permits excessive centromere stretch along with hyperstabilization of kMT attachments. STAG2-deficient cells display mislocalization of Bub1 kinase, Bub3 and the chromosome passenger complex. Importantly, strategically destabilizing kMT attachments in tumor cells harboring STAG2 mutations by overexpression of the microtubule-destabilizing enzymes MCAK (also known as KIF2C) and Kif2B decreased the rate of lagging chromosomes and reduced the rate of chromosome missegregation. These data demonstrate that STAG2 promotes the correction of kMT attachment errors to ensure faithful chromosome segregation during mitosis.
Collapse
Affiliation(s)
- Marianna Kleyman
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA Norris Cotton Cancer Center, Lebanon, NH 03766, USA
| | - Lilian Kabeche
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA Norris Cotton Cancer Center, Lebanon, NH 03766, USA
| | - Duane A Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA Norris Cotton Cancer Center, Lebanon, NH 03766, USA
| |
Collapse
|
99
|
Häfner J, Mayr MI, Möckel MM, Mayer TU. Pre-anaphase chromosome oscillations are regulated by the antagonistic activities of Cdk1 and PP1 on Kif18A. Nat Commun 2014; 5:4397. [PMID: 25048371 DOI: 10.1038/ncomms5397] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 06/13/2014] [Indexed: 12/16/2022] Open
Abstract
Upon congression at the spindle equator, vertebrate chromosomes display oscillatory movements which typically decline as cells progress towards anaphase. Kinesin-8 Kif18A has been identified as a suppressor of chromosome movements, but how its activity is temporally regulated to dampen chromosome oscillations before anaphase onset remained mysterious. Here, we identify a regulatory network composed of cyclin-dependent kinase-1 (Cdk1) and protein phosphatase-1 (PP1) that antagonistically regulate Kif18A. Cdk1-mediated inhibitory phosphorylation of Kif18A promotes chromosome oscillations in early metaphase. PP1 induces metaphase plate thinning by directly dephosphorylating Kif18A. Chromosome attachment induces Cdk1 inactivation and kinetochore recruitment of PP1α/γ. Thus, we propose that chromosome biorientation mediates the alignment of chromosomes at the metaphase plate by tipping the balance in favour of dephosphorylated Kif18A capable of suppressing the oscillatory movements of chromosomes. Notably, interfering with chromosome oscillations severely impairs the fidelity of sister chromatid segregation demonstrating the importance of timely controlled chromosome dynamics for the maintenance of genome integrity.
Collapse
Affiliation(s)
- Julia Häfner
- Department of Molecular Genetics, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany
| | - Monika I Mayr
- Department of Biology, Institute of Biochemistry, ETH Zurich, Schafmattstrasse 18, 8093 Zurich, Switzerland
| | - Martin M Möckel
- Department of Molecular Genetics, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany
| | - Thomas U Mayer
- Department of Molecular Genetics, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany
| |
Collapse
|
100
|
Abstract
The importance of Eg5 at metaphase is linked to kinetochore-microtubule stability: short-lived kinetochore-microtubules impose a need for Eg5 in bipolar spindle maintenance, but longer-lived kinetochore-microtubules do not. Kinetochore-microtubule dynamics defines their contribution to force balance in the metaphase spindle. The mitotic spindle is a bipolar, microtubule (MT)-based cellular machine that segregates the duplicated genome into two daughter cells. The kinesin-5 Eg5 establishes the bipolar geometry of the mitotic spindle, but previous work in mammalian cells suggested that this motor is unimportant for the maintenance of spindle bipolarity. Although it is known that Kif15, a second mitotic kinesin, enforces spindle bipolarity in the absence of Eg5, how Kif15 functions in this capacity and/or whether other biochemical or physical properties of the spindle promote its bipolarity have been poorly studied. Here we report that not all human cell lines can efficiently maintain bipolarity without Eg5, despite their expressing Kif15. We show that the stability of chromosome-attached kinetochore-MTs (K-MTs) is important for bipolar spindle maintenance without Eg5. Cells that efficiently maintain bipolar spindles without Eg5 have more stable K-MTs than those that collapse without Eg5. Consistent with this observation, artificial destabilization of K-MTs promotes spindle collapse without Eg5, whereas stabilizing K-MTs improves bipolar spindle maintenance without Eg5. Our findings suggest that either rapid K-MT turnover pulls poles inward or slow K-MT turnover allows for greater resistance to inward-directed forces.
Collapse
Affiliation(s)
- A Sophia Gayek
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|