51
|
Asai Y, Matsumura R, Hasumi Y, Susumu H, Nagata K, Watanabe Y, Terada Y. SET/TAF1 forms a distance-dependent feedback loop with Aurora B and Bub1 as a tension sensor at centromeres. Sci Rep 2020; 10:15653. [PMID: 32973131 PMCID: PMC7518443 DOI: 10.1038/s41598-020-71955-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 06/26/2020] [Indexed: 12/28/2022] Open
Abstract
During mitosis, spatiotemporal regulation of phosphorylation at the kinetochore is essential for accurate chromosome alignment and proper chromosome segregation. Aurora B kinase phosphorylates kinetochore substrates to correct improper kinetochore-microtubule (KT-MT) attachments, whereas tension across the centromeres inactivates Aurora B kinase, and PP2A phosphatase dephosphorylates the kinetochore proteins to stabilize the attachments. However, the molecular entity of the tension sensing mechanism remains elusive. In a previous report, we showed that centromeric SET/TAF1 on Sgo2 up-regulates Aurora B kinase activity via PP2A inhibition in prometaphase. Here we show that Aurora B and Bub1 at the centromere/kinetochore regulate both kinase activities one another in an inter-kinetochore distance-dependent manner, indicating a positive feedback loop. We further show that the centromeric pool of SET on Sgo2 depends on Bub1 kinase activity, and the centromeric localization of SET decreases in a distance-dependent manner, thereby inactivating Aurora B in metaphase. Consistently, ectopic targeting of SET to the kinetochores during metaphase hyperactivates Aurora B via PP2A inhibition, and thereby rescues the feedback loop. Thus, we propose that SET, Aurora B and Bub1 form a distance-dependent positive feedback loop, which spatiotemporally may act as a tension sensor at centromeres.
Collapse
Affiliation(s)
- Yuichiro Asai
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Rieko Matsumura
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Yurina Hasumi
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Hiroaki Susumu
- Graduate Program in Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Yayoi, Tokyo, 113-0032, Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Yoshinori Watanabe
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, BN1 9RQ, Sussex, UK
| | - Yasuhiko Terada
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
| |
Collapse
|
52
|
Audett MR, Maresca TJ. The whole is greater than the sum of its parts: at the intersection of order, disorder, and kinetochore function. Essays Biochem 2020; 64:349-358. [PMID: 32756877 PMCID: PMC8011995 DOI: 10.1042/ebc20190069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 11/17/2022]
Abstract
The kinetochore (KT) field has matured tremendously since Earnshaw first identified CENP-A, CENP-B, and CENP-C [1,2]. In the past 35 years, the accumulation of knowledge has included: defining the parts list, identifying epistatic networks of interdependence within the parts list, understanding the spatial organization of subcomplexes into a massive structure - hundreds of megadaltons in size, and dissecting the functions of the KT in its entirety as well as of its individual parts. Like nearly all cell and molecular biology fields, the structure-function paradigm has been foundational to advances in the KT field. A point nicely highlighted by the fact that we are at the precipice of the in vitro reconstitution of a functional KT holo complex. Yet conventional notions of structure cannot provide a complete picture of the KT especially since it contains an abundance of unstructured or intrinsically disordered constituents. The combination of structured and disordered proteins within the KT results in an assembled system that is functionally greater than the sum of its parts.
Collapse
Affiliation(s)
- Margaux R Audett
- Biology Department, University of Massachusetts, Amherst, MA, U.S.A
| | - Thomas J Maresca
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, U.S.A
| |
Collapse
|
53
|
Kuijt TEF, Lambers MLA, Weterings S, Ponsioen B, Bolhaqueiro ACF, Staijen DHM, Kops GJPL. A Biosensor for the Mitotic Kinase MPS1 Reveals Spatiotemporal Activity Dynamics and Regulation. Curr Biol 2020; 30:3862-3870.e6. [PMID: 32888483 DOI: 10.1016/j.cub.2020.07.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 01/10/2023]
Abstract
Accurate chromosome segregation during cell division critically depends on error correction of chromosome-spindle interactions and the spindle assembly checkpoint (SAC) [1-3]. The kinase MPS1 is an essential regulator of both processes, ensuring full chromosome biorientation before anaphase onset [3, 4]. To understand when and where MPS1 activation occurs and how MPS1 signaling is modulated during mitosis, we developed MPS1sen, a sensitive and specific FRET-based biosensor for MPS1 activity. By placing MPS1sen at different subcellular locations, we show that MPS1 activity initiates in the nucleus ∼9-12 min prior to nuclear envelope breakdown (NEB) in a kinetochore-dependent manner and reaches the cytoplasm at the start of NEB. Soon after initiation, MPS1 activity increases with switch-like kinetics, peaking at completion of NEB. We further show that timing and extent of pre-NEB MPS1 activity is regulated by Aurora B and PP2A-B56. MPS1sen phosphorylation declines in prometaphase as a result of formation of kinetochore-microtubule attachments, reaching low but still detectable levels at metaphase. Finally, leveraging the sensitivity and dynamic range of MPS1sen, we show deregulated MPS1 signaling dynamics in colorectal cancer cell lines and tumor organoids with diverse genomic instability phenotypes.
Collapse
Affiliation(s)
- Timo E F Kuijt
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Maaike L A Lambers
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Sonja Weterings
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Bas Ponsioen
- Cancer Genomics Netherlands, UMC Utrecht, 3584CG Utrecht, the Netherlands; Molecular Cancer Research, Centre for Molecular Medicine, UMC Utrecht, 3584CG Utrecht, the Netherlands
| | - Ana C F Bolhaqueiro
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Debbie H M Staijen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584CT Utrecht, the Netherlands.
| |
Collapse
|
54
|
A stochastic model for error correction of kinetochore-microtubule attachments in budding yeast. PLoS One 2020; 15:e0236293. [PMID: 32760074 PMCID: PMC7410253 DOI: 10.1371/journal.pone.0236293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/01/2020] [Indexed: 12/05/2022] Open
Abstract
To divide replicated chromosomes equally between daughter cells, kinetochores must attach to microtubules emanating from opposite poles of the mitotic spindle (biorientation). An error correction mechanism facilitates this process by destabilizing erroneous kinetochore-microtubule attachments. Here we present a stochastic model of kinetochore-microtubule attachments, via an essential protein Ndc80 in budding yeast, Saccharomyces cerevisiae. Using the model, we calculate the stochastic dynamics of a pair of sister kinetochores as they transition among different attachment states. First of all, we determine the kinase-to-phosphatase balance point that maximizes the probability of biorientation, while starting from an erroneous attachment state. We find that the balance point is sensitive to the rates of microtubule-Ndc80 dissociation and derive an approximate analytical formula that defines the balance point. Secondly, we determine the probability of transition from low-tension amphitelic to monotelic attachment and find that, despite this probability being approximately 33%, biorientation can be achieved with high probability. Thirdly, we calculate the contribution of the geometrical orientation of sister kinetochores to the probability of biorientation and show that, in the absence of geometrical orientation, the biorientation error rate is much larger than that observed in experiments. Finally, we study the coupling of the error correction mechanism to the spindle assembly checkpoint by calculating the average binding of checkpoint-related proteins to the kinetochore during the error correction process.
Collapse
|
55
|
Benzi G, Piatti S. Killing two birds with one stone: how budding yeast Mps1 controls chromosome segregation and spindle assembly checkpoint through phosphorylation of a single kinetochore protein. Curr Genet 2020; 66:1037-1044. [PMID: 32632756 DOI: 10.1007/s00294-020-01091-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
During mitosis, the identical sister chromatids of each chromosome must attach through their kinetochores to microtubules emanating from opposite spindle poles. This process, referred to as chromosome biorientation, is essential for equal partitioning of the genetic information to the two daughter cells. Defects in chromosome biorientation can give rise to aneuploidy, a hallmark of cancer and genetic diseases. A conserved surveillance mechanism called spindle assembly checkpoint (SAC) prevents the onset of anaphase until biorientation is attained. Key to chromosome biorientation is an error correction mechanism that allows kinetochores to establish proper bipolar attachments by disengaging faulty kinetochore-microtubule connections. Error correction relies on the Aurora B and Mps1 kinases that also promote SAC signaling, raising the possibility that they are part of a single sensory device responding to improper attachments and concomitantly controlling both their disengagement and a temporary mitotic arrest. In budding yeast, Aurora B and Mps1 promote error correction independently from one another, but while the substrates of Aurora B in this process are at least partially known, the mechanism underlying the involvement of Mps1 in the error correction pathway is unknown. Through the characterization of a novel mps1 mutant and an unbiased genetic screen for extragenic suppressors, we recently gained evidence that a common mechanism based on Mps1-dependent phosphorylation of the Knl1/Spc105 kinetochore scaffold and subsequent recruitment of the Bub1 kinase is critical for the function of Mps1 in chromosome biorientation as well as for SAC activation (Benzi et al. EMBO Rep, 2020).
Collapse
Affiliation(s)
- Giorgia Benzi
- CRBM, University of Montpellier, CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Simonetta Piatti
- CRBM, University of Montpellier, CNRS, 1919 Route de Mende, 34293, Montpellier, France.
| |
Collapse
|
56
|
Allan LA, Camacho Reis M, Ciossani G, Huis in ‘t Veld PJ, Wohlgemuth S, Kops GJPL, Musacchio A, Saurin AT. Cyclin B1 scaffolds MAD1 at the kinetochore corona to activate the mitotic checkpoint. EMBO J 2020; 39:e103180. [PMID: 32202322 PMCID: PMC7298293 DOI: 10.15252/embj.2019103180] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/20/2020] [Accepted: 03/02/2020] [Indexed: 11/28/2022] Open
Abstract
Cyclin B:CDK1 is the master kinase regulator of mitosis. We show here that, in addition to its kinase functions, mammalian Cyclin B also scaffolds a localised signalling pathway to help preserve genome stability. Cyclin B1 localises to an expanded region of the outer kinetochore, known as the corona, where it scaffolds the spindle assembly checkpoint (SAC) machinery by binding directly to MAD1. In vitro reconstitutions map the key binding interface to a few acidic residues in the N-terminal region of MAD1, and point mutations in this sequence abolish MAD1 corona localisation and weaken the SAC. Therefore, Cyclin B1 is the long-sought-after scaffold that links MAD1 to the corona, and this specific pool of MAD1 is needed to generate a robust SAC response. Robustness arises because Cyclin B1:MAD1 localisation loses dependence on MPS1 kinase after the corona has been established, ensuring that corona-localised MAD1 can still be phosphorylated when MPS1 activity is low. Therefore, this study explains how corona-MAD1 generates a robust SAC signal, and it reveals a scaffolding role for the key mitotic kinase, Cyclin B1:CDK1, which ultimately helps to inhibit its own degradation.
Collapse
Affiliation(s)
- Lindsey A Allan
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| | - Magda Camacho Reis
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| | - Giuseppe Ciossani
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Pim J Huis in ‘t Veld
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Geert JPL Kops
- Oncode InstituteHubrecht Institute—KNAW and University Medical Centre UtrechtUtrechtThe Netherlands
| | - Andrea Musacchio
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Adrian T Saurin
- Division of Cellular MedicineSchool of MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
57
|
Roy B, Han SJ, Fontan AN, Joglekar AP. The copy-number and varied strengths of MELT motifs in Spc105 balance the strength and responsiveness of the spindle assembly checkpoint. eLife 2020; 9:55096. [PMID: 32479259 PMCID: PMC7292645 DOI: 10.7554/elife.55096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
During mitosis, the Spindle Assembly Checkpoint (SAC) maintains genome stability while also ensuring timely anaphase onset. To maintain genome stability, the SAC must be strong to delay anaphase even if just one chromosome is unattached, but for timely anaphase onset, it must promptly respond to silencing mechanisms. How the SAC meets these potentially antagonistic requirements is unclear. Here we show that the balance between SAC strength and responsiveness is determined by the number of ‘MELT’ motifs in the kinetochore protein Spc105/KNL1 and their Bub3-Bub1 binding affinities. Many strong MELT motifs per Spc105/KNL1 minimize chromosome missegregation, but too many delay anaphase onset. We demonstrate this by constructing a Spc105 variant that trades SAC responsiveness for much more accurate chromosome segregation. We propose that the necessity of balancing SAC strength and responsiveness drives the dual evolutionary trend of the amplification of MELT motif number, but degeneration of their functionally optimal amino acid sequence.
Collapse
Affiliation(s)
- Babhrubahan Roy
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Simon Jy Han
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Adrienne Nicole Fontan
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Ajit P Joglekar
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
58
|
Roscioli E, Germanova TE, Smith CA, Embacher PA, Erent M, Thompson AI, Burroughs NJ, McAinsh AD. Ensemble-Level Organization of Human Kinetochores and Evidence for Distinct Tension and Attachment Sensors. Cell Rep 2020; 31:107535. [PMID: 32348762 PMCID: PMC7196887 DOI: 10.1016/j.celrep.2020.107535] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/10/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
Kinetochores are multi-protein machines that form dynamic attachments to microtubules and control chromosome segregation. High fidelity is ensured because kinetochores can monitor attachment status and tension, using this information to activate checkpoints and error-correction mechanisms. To explore how kinetochores achieve this, we used two- and three-color subpixel fluorescence localization to define how proteins from six major complexes (CCAN, MIS12, NDC80, KNL1, RZZ, and SKA) and the checkpoint proteins Bub1, Mad1, and Mad2 are organized in the human kinetochore. This reveals how the outer kinetochore has a high nematic order and is largely invariant to the loss of attachment or tension, except for two mechanical sensors. First, Knl1 unravels to relay tension, and second, NDC80 undergoes jackknifing and loss of nematic order under microtubule detachment, with only the latter wired up to the checkpoint signaling system. This provides insight into how kinetochores integrate mechanical signals to promote error-free chromosome segregation.
Collapse
Affiliation(s)
- Emanuele Roscioli
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Tsvetelina E Germanova
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Christopher A Smith
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Peter A Embacher
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Mathematics Institute, University of Warwick, Coventry, UK
| | - Muriel Erent
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Amelia I Thompson
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Nigel J Burroughs
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Mathematics Institute, University of Warwick, Coventry, UK.
| | - Andrew D McAinsh
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, UK; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.
| |
Collapse
|
59
|
Pachis ST, Hiruma Y, Tromer EC, Perrakis A, Kops GJPL. Interactions between N-terminal Modules in MPS1 Enable Spindle Checkpoint Silencing. Cell Rep 2020; 26:2101-2112.e6. [PMID: 30784592 DOI: 10.1016/j.celrep.2019.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/13/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022] Open
Abstract
Faithful chromosome segregation relies on the ability of the spindle assembly checkpoint (SAC) to delay anaphase onset until chromosomes are attached to the mitotic spindle via their kinetochores. MPS1 kinase is recruited to kinetochores to initiate SAC signaling and is removed from kinetochores once stable microtubule attachments have been formed to allow normal mitotic progression. Here, we show that a helical fragment within the kinetochore-targeting N-terminal extension (NTE) module of MPS1 is required for interactions with kinetochores and forms intramolecular interactions with its adjacent tetratricopeptide repeat (TPR) domain. Bypassing this NTE-TPR interaction results in high MPS1 levels at kinetochores due to loss of regulatory input into MPS1 localization, inefficient MPS1 delocalization upon microtubule attachment, and SAC silencing defects. These results show that SAC responsiveness to attachments relies on regulated intramolecular interactions in MPS1 and highlight the sensitivity of mitosis to perturbations in the dynamics of the MPS1-NDC80-C interactions.
Collapse
Affiliation(s)
- Spyridon T Pachis
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, 3584 CT, the Netherlands
| | - Yoshitaka Hiruma
- Department of Biochemistry, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands
| | - Eelco C Tromer
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Anastassis Perrakis
- Department of Biochemistry, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, 3584 CT, the Netherlands.
| |
Collapse
|
60
|
Curtis NL, Ruda GF, Brennan P, Bolanos-Garcia VM. Deregulation of Chromosome Segregation and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2020. [DOI: 10.1146/annurev-cancerbio-030419-033541] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The mitotic spindle assembly checkpoint (SAC) is an intricate cell signaling system that ensures the high fidelity and timely segregation of chromosomes during cell division. Mistakes in this process can lead to the loss, gain, or rearrangement of the genetic material. Gross chromosomal aberrations are usually lethal but can cause birth and development defects as well as cancer. Despite advances in the identification of SAC protein components, important details of the interactions underpinning chromosome segregation regulation remain to be established. This review discusses the current understanding of the function, structure, mode of regulation, and dynamics of the assembly and disassembly of SAC subcomplexes, which ultimately safeguard the accurate transmission of a stable genome to descendants. We also discuss how diverse oncoviruses take control of human cell division by exploiting the SAC and the potential of this signaling circuitry as a pool of drug targets to develop effective cancer therapies.
Collapse
Affiliation(s)
- Natalie L. Curtis
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, United Kingdom
| | - Gian Filippo Ruda
- Target Discovery Institute and Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Paul Brennan
- Target Discovery Institute and Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Victor M. Bolanos-Garcia
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, United Kingdom
| |
Collapse
|
61
|
Tsai YM, Wu KL, Chang YY, Hung JY, Chang WA, Chang CY, Jian SF, Tsai PH, Huang YC, Chong IW, Hsu YL. Upregulation of Thr/ Tyr kinase Increases the Cancer Progression by Neurotensin and Dihydropyrimidinase-Like 3 in Lung Cancer. Int J Mol Sci 2020; 21:ijms21051640. [PMID: 32121246 PMCID: PMC7084211 DOI: 10.3390/ijms21051640] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death globally, thus elucidation of its molecular pathology is highly highlighted. Aberrant alterations of the spindle assembly checkpoint (SAC) are implicated in the development of cancer due to abnormal cell division. TTK (Thr/Tyr kinase), a dual serine/threonine kinase, is considered to act as a cancer promoter by controlling SAC. However, the mechanistic details of how TTK-mediated signaling network supports cancer development is still a mystery. Here, we found that TTK was upregulated in the tumor tissue of patients with lung cancer, and enhanced tumor growth and metastasis in vitro and in vivo. Mechanistically, TTK exerted a significant enhancement in cancer growth by neurotensin (NTS) upregulation, and subsequently increased the expression of cyclin A and cdk2, which was resulting in the increase of DNA synthesis. In contrast, TTK increased cell migration and epithelial-to-mesenchymal transition (EMT) by enhancing the expression of dihydropyrimidinase-like 3 (DPYSL3) followed by the increase of snail-regulated EMT, thus reinforce metastatic potential and ultimately tumor metastasis. TTK and DPYSL3 upregulation was positively correlated with a poor clinical outcome in patients with lung cancer. Together, our findings revealed a novel mechanism underlying the oncogenic potential effect of TTK and clarified its downstream factors NTS and DPYSL3 might represent a novel, promising candidate oncogenes with potential therapeutic vulnerabilities in lung cancer.
Collapse
Affiliation(s)
- Ying-Ming Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kuan-Li Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Yun Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Jen-Yu Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wei-An Chang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chao-Yuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shu-Fang Jian
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
| | - Pei-Hsun Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
| | - Inn-Wen Chong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-M.T.); (K.-L.W.); (Y.-Y.C.); (J.-Y.H.); (C.-Y.C.); (S.-F.J.); (P.-H.T.); (Y.-C.H.); (I.-W.C.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan
- Correspondence:
| |
Collapse
|
62
|
Keating L, Touati SA, Wassmann K. A PP2A-B56-Centered View on Metaphase-to-Anaphase Transition in Mouse Oocyte Meiosis I. Cells 2020; 9:E390. [PMID: 32046180 PMCID: PMC7072534 DOI: 10.3390/cells9020390] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Meiosis is required to reduce to haploid the diploid genome content of a cell, generating gametes-oocytes and sperm-with the correct number of chromosomes. To achieve this goal, two specialized cell divisions without intermediate S-phase are executed in a time-controlled manner. In mammalian female meiosis, these divisions are error-prone. Human oocytes have an exceptionally high error rate that further increases with age, with significant consequences for human fertility. To understand why errors in chromosome segregation occur at such high rates in oocytes, it is essential to understand the molecular players at work controlling these divisions. In this review, we look at the interplay of kinase and phosphatase activities at the transition from metaphase-to-anaphase for correct segregation of chromosomes. We focus on the activity of PP2A-B56, a key phosphatase for anaphase onset in both mitosis and meiosis. We start by introducing multiple roles PP2A-B56 occupies for progression through mitosis, before laying out whether or not the same principles may apply to the first meiotic division in oocytes, and describing the known meiosis-specific roles of PP2A-B56 and discrepancies with mitotic cell cycle regulation.
Collapse
Affiliation(s)
- Leonor Keating
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Sandra A. Touati
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Katja Wassmann
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
63
|
Lok TM, Wang Y, Xu WK, Xie S, Ma HT, Poon RYC. Mitotic slippage is determined by p31 comet and the weakening of the spindle-assembly checkpoint. Oncogene 2020; 39:2819-2834. [PMID: 32029899 PMCID: PMC7098889 DOI: 10.1038/s41388-020-1187-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/14/2019] [Accepted: 01/23/2020] [Indexed: 11/09/2022]
Abstract
Mitotic slippage involves cells exiting mitosis without proper chromosome segregation. Although degradation of cyclin B1 during prolonged mitotic arrest is believed to trigger mitotic slippage, its upstream regulation remains obscure. Whether mitotic slippage is caused by APC/CCDC20 activity that is able to escape spindle-assembly checkpoint (SAC)-mediated inhibition, or is actively promoted by a change in SAC activity remains an outstanding issue. We found that a major culprit for mitotic slippage involves reduction of MAD2 at the kinetochores, resulting in a progressive weakening of SAC during mitotic arrest. A further level of control of the timing of mitotic slippage is through p31comet-mediated suppression of MAD2 activation. The loss of kinetochore MAD2 was dependent on APC/CCDC20, indicating a feedback control of APC/C to SAC during prolonged mitotic arrest. The gradual weakening of SAC during mitotic arrest enables APC/CCDC20 to degrade cyclin B1, cumulating in the cell exiting mitosis by mitotic slippage.
Collapse
Affiliation(s)
- Tsun Ming Lok
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Yang Wang
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wendy Kaichun Xu
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.,Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Siwei Xie
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| |
Collapse
|
64
|
Abstract
The goal of mitosis is to form two daughter cells each containing one copy of each mother cell chromosome, replicated in the previous S phase. To achieve this, sister chromatids held together back-to-back at their primary constriction, the centromere, have to interact with microtubules of the mitotic spindle so that each chromatid takes connections with microtubules emanating from opposite spindle poles (we will refer to this condition as bipolar attachment). Only once all replicated chromosomes have reached bipolar attachments can sister chromatids lose cohesion with each other, at the onset of anaphase, and move toward opposite spindle poles, being segregated into what will soon become the daughter cell nucleus. Prevention of errors in chromosome segregation is granted by a safeguard mechanism called Spindle Assembly Checkpoint (SAC). Until all chromosomes are bipolarly oriented at the equator of the mitotic spindle, the SAC prevents loss of sister chromatid cohesion, thus anaphase onset, and maintains the mitotic state by inhibiting inactivation of the major M phase promoting kinase, the cyclin B-cdk1 complex (Cdk1). Here, we review recent mechanistic insights about the circuitry that links Cdk1 to the SAC to ensure correct achievement of the goal of mitosis.
Collapse
Affiliation(s)
- Angela Flavia Serpico
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,DMMBM, University of Naples "Federico II", Naples, 80131, Italy
| | - Domenico Grieco
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,Department of Pharmacy, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
65
|
Wild T, Budzowska M, Hellmuth S, Eibes S, Karemore G, Barisic M, Stemmann O, Choudhary C. Deletion of APC7 or APC16 Allows Proliferation of Human Cells without the Spindle Assembly Checkpoint. Cell Rep 2019; 25:2317-2328.e5. [PMID: 30485802 PMCID: PMC6289045 DOI: 10.1016/j.celrep.2018.10.104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 09/07/2018] [Accepted: 10/29/2018] [Indexed: 11/17/2022] Open
Abstract
The multisubunit ubiquitin ligase APC/C (anaphase-promoting complex/cyclosome) is essential for mitosis by promoting timely degradation of cyclin B1. APC/C is tightly regulated by the spindle assembly checkpoint (SAC), which involves MPS1 and MAD2-dependent temporal inhibition of APC/C. We analyzed the contribution of the APC/C subunits APC7 and APC16 to APC/C composition and function in human cells. APC16 is required for APC7 assembly into APC/C, whereas APC16 assembles independently of APC7. APC7 and APC16 knockout cells display no major defects in mitotic progression, cyclin B1 degradation, or SAC response, but APC/C lacking these two subunits shows reduced ubiquitylation activity in vitro. Strikingly, deletion of APC7 or APC16 is sufficient to provide synthetic viability to MAD2 deletion. ΔAPC7ΔMAD2 cells display accelerated mitosis and require SAC-independent MPS1 function for genome stability. These findings reveal that the composition of APC/C critically influences the importance of the SAC in humans. APC16 is required for in vivo assembly of APC7 into APC/C APC7 or APC16 deletion has no major effect on mitosis Deletion of APC7 or APC16 provides synthetic viability to MAD2 deletion
Collapse
Affiliation(s)
- Thomas Wild
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Magda Budzowska
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Center for Chromosome Stability (CCS), Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Susanne Hellmuth
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Susana Eibes
- Danish Cancer Society Research Center, Cell Division Laboratory, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Gopal Karemore
- Protein Imaging Platform, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Marin Barisic
- Danish Cancer Society Research Center, Cell Division Laboratory, Strandboulevarden 49, 2100 Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Olaf Stemmann
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Chunaram Choudhary
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
66
|
Conti D, Gul P, Islam A, Martín-Durán JM, Pickersgill RW, Draviam VM. Kinetochores attached to microtubule-ends are stabilised by Astrin bound PP1 to ensure proper chromosome segregation. eLife 2019; 8:49325. [PMID: 31808746 PMCID: PMC6930079 DOI: 10.7554/elife.49325] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/01/2019] [Indexed: 12/12/2022] Open
Abstract
Microtubules segregate chromosomes by attaching to macromolecular kinetochores. Only microtubule-end attached kinetochores can be pulled apart; how these end-on attachments are selectively recognised and stabilised is not known. Using the kinetochore and microtubule-associated protein, Astrin, as a molecular probe, we show that end-on attachments are rapidly stabilised by spatially-restricted delivery of PP1 near the C-terminus of Ndc80, a core kinetochore-microtubule linker. PP1 is delivered by the evolutionarily conserved tail of Astrin and this promotes Astrin’s own enrichment creating a highly-responsive positive feedback, independent of biorientation. Abrogating Astrin:PP1-delivery disrupts attachment stability, which is not rescued by inhibiting Aurora-B, an attachment destabiliser, but is reversed by artificially tethering PP1 near the C-terminus of Ndc80. Constitutive Astrin:PP1-delivery disrupts chromosome congression and segregation, revealing a dynamic mechanism for stabilising attachments. Thus, Astrin-PP1 mediates a dynamic ‘lock’ that selectively and rapidly stabilises end-on attachments, independent of biorientation, and ensures proper chromosome segregation.
Collapse
Affiliation(s)
- Duccio Conti
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom.,Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Parveen Gul
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Asifa Islam
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - José M Martín-Durán
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Richard W Pickersgill
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Viji M Draviam
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom.,Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
67
|
Roy B, Verma V, Sim J, Fontan A, Joglekar AP. Delineating the contribution of Spc105-bound PP1 to spindle checkpoint silencing and kinetochore microtubule attachment regulation. J Cell Biol 2019; 218:3926-3942. [PMID: 31649151 PMCID: PMC6891095 DOI: 10.1083/jcb.201810172] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 06/19/2019] [Accepted: 09/18/2019] [Indexed: 02/01/2023] Open
Abstract
Roy et al. highlight a harmful cross-talk that can arise between spindle assembly checkpoint silencing and chromosome biorientation due to the involvement of protein phosphatase 1 in both the processes. Accurate chromosome segregation during cell division requires the spindle assembly checkpoint (SAC), which detects unattached kinetochores, and an error correction mechanism that destabilizes incorrect kinetochore–microtubule attachments. While the SAC and error correction are both regulated by protein phosphatase 1 (PP1), which silences the SAC and stabilizes kinetochore–microtubule attachments, how these distinct PP1 functions are coordinated remains unclear. Here, we investigate the contribution of PP1, docked on its conserved kinetochore receptor Spc105/Knl1, to SAC silencing and attachment regulation. We find that Spc105-bound PP1 is critical for SAC silencing but dispensable for error correction; in fact, reduced PP1 docking on Spc105 improved chromosome segregation and viability of mutant/stressed states. We additionally show that artificially recruiting PP1 to Spc105/Knl1 before, but not after, chromosome biorientation interfered with error correction. These observations lead us to propose that recruitment of PP1 to Spc105/Knl1 is carefully regulated to ensure that chromosome biorientation precedes SAC silencing, thereby ensuring accurate chromosome segregation.
Collapse
Affiliation(s)
- Babhrubahan Roy
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Vikash Verma
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Janice Sim
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Adrienne Fontan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Ajit P Joglekar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI .,Department of Biophysics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
68
|
Anderhub SJ, Mak GWY, Gurden MD, Faisal A, Drosopoulos K, Walsh K, Woodward HL, Innocenti P, Westwood IM, Naud S, Hayes A, Theofani E, Filosto S, Saville H, Burke R, van Montfort RLM, Raynaud FI, Blagg J, Hoelder S, Eccles SA, Linardopoulos S. High Proliferation Rate and a Compromised Spindle Assembly Checkpoint Confers Sensitivity to the MPS1 Inhibitor BOS172722 in Triple-Negative Breast Cancers. Mol Cancer Ther 2019; 18:1696-1707. [PMID: 31575759 DOI: 10.1158/1535-7163.mct-18-1203] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/21/2019] [Accepted: 07/01/2019] [Indexed: 11/16/2022]
Abstract
BOS172722 (CCT289346) is a highly potent, selective, and orally bioavailable inhibitor of spindle assembly checkpoint kinase MPS1. BOS172722 treatment alone induces significant sensitization to death, particularly in highly proliferative triple-negative breast cancer (TNBC) cell lines with compromised spindle assembly checkpoint activity. BOS172722 synergizes with paclitaxel to induce gross chromosomal segregation defects caused by MPS1 inhibitor-mediated abrogation of the mitotic delay induced by paclitaxel treatment. In in vivo pharmacodynamic experiments, BOS172722 potently inhibits the spindle assembly checkpoint induced by paclitaxel in human tumor xenograft models of TNBC, as measured by inhibition of the phosphorylation of histone H3 and the phosphorylation of the MPS1 substrate, KNL1. This mechanistic synergy results in significant in vivo efficacy, with robust tumor regressions observed for the combination of BOS172722 and paclitaxel versus either agent alone in long-term efficacy studies in multiple human tumor xenograft TNBC models, including a patient-derived xenograft and a systemic metastasis model. The current target indication for BOS172722 is TNBC, based on their high sensitivity to MPS1 inhibition, the well-defined clinical patient population with high unmet need, and the synergy observed with paclitaxel.
Collapse
Affiliation(s)
- Simon J Anderhub
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Grace Wing-Yan Mak
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Mark D Gurden
- The Breast Cancer Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Amir Faisal
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Konstantinos Drosopoulos
- The Breast Cancer Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Katie Walsh
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Hannah L Woodward
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Paolo Innocenti
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Isaac M Westwood
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sébastien Naud
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Angela Hayes
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Efthymia Theofani
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Simone Filosto
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Harry Saville
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Rob L M van Montfort
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Florence I Raynaud
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Swen Hoelder
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Suzanne A Eccles
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Spiros Linardopoulos
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.
- The Breast Cancer Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
69
|
Bonner MK, Haase J, Swinderman J, Halas H, Miller Jenkins LM, Kelly AE. Enrichment of Aurora B kinase at the inner kinetochore controls outer kinetochore assembly. J Cell Biol 2019; 218:3237-3257. [PMID: 31527147 PMCID: PMC6781445 DOI: 10.1083/jcb.201901004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/19/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
Outer kinetochore assembly enables chromosome attachment to microtubules and spindle assembly checkpoint (SAC) signaling in mitosis. Aurora B kinase controls kinetochore assembly by phosphorylating the Mis12 complex (Mis12C) subunit Dsn1. Current models propose Dsn1 phosphorylation relieves autoinhibition, allowing Mis12C binding to inner kinetochore component CENP-C. Using Xenopus laevis egg extracts and biochemical reconstitution, we found that autoinhibition of the Mis12C by Dsn1 impedes its phosphorylation by Aurora B. Our data indicate that the INCENP central region increases Dsn1 phosphorylation by enriching Aurora B at inner kinetochores, close to CENP-C. Furthermore, centromere-bound CENP-C does not exchange in mitosis, and CENP-C binding to the Mis12C dramatically increases Dsn1 phosphorylation by Aurora B. We propose that the coincidence of Aurora B and CENP-C at inner kinetochores ensures the fidelity of kinetochore assembly. We also found that the central region is required for the SAC beyond its role in kinetochore assembly, suggesting that kinetochore enrichment of Aurora B promotes the phosphorylation of other kinetochore substrates.
Collapse
Affiliation(s)
- Mary Kate Bonner
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Julian Haase
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jason Swinderman
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hyunmi Halas
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alexander E Kelly
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
70
|
Hayward D, Alfonso-Pérez T, Gruneberg U. Orchestration of the spindle assembly checkpoint by CDK1-cyclin B1. FEBS Lett 2019; 593:2889-2907. [PMID: 31469407 DOI: 10.1002/1873-3468.13591] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
In mitosis, the spindle assembly checkpoint (SAC) monitors the formation of microtubule-kinetochore attachments during capture of chromosomes by the mitotic spindle. Spindle assembly is complete once there are no longer any unattached kinetochores. Here, we will discuss the mechanism and key components of spindle checkpoint signalling. Unattached kinetochores bind the principal spindle checkpoint kinase monopolar spindle 1 (MPS1). MPS1 triggers the recruitment of other spindle checkpoint proteins and the formation of a soluble inhibitor of anaphase, thus preventing exit from mitosis. On microtubule attachment, kinetochores become checkpoint silent due to the actions of PP2A-B56 and PP1. This SAC responsive period has to be coordinated with mitotic spindle formation to ensure timely mitotic exit and accurate chromosome segregation. We focus on the molecular mechanisms by which the SAC permissive state is created, describing a central role for CDK1-cyclin B1 and its counteracting phosphatase PP2A-B55. Furthermore, we discuss how CDK1-cyclin B1, through its interaction with MAD1, acts as an integral component of the SAC, and actively orchestrates checkpoint signalling and thus contributes to the faithful execution of mitosis.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, UK
| | | | | |
Collapse
|
71
|
Bao X, Liu H, Liu X, Ruan K, Zhang Y, Zhang Z, Hu Q, Liu Y, Akram S, Zhang J, Gong Q, Wang W, Yuan X, Li J, Zhao L, Dou Z, Tian R, Yao X, Wu J, Shi Y. Mitosis-specific acetylation tunes Ran effector binding for chromosome segregation. J Mol Cell Biol 2019; 10:18-32. [PMID: 29040603 PMCID: PMC6041754 DOI: 10.1093/jmcb/mjx045] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/03/2017] [Indexed: 11/12/2022] Open
Abstract
Stable transmission of genetic information during cell division requires faithful mitotic spindle assembly and chromosome segregation. The Ran GTPase plays a key role in mitotic spindle assembly. However, how the generation of a chemical gradient of Ran-GTP at the spindle is coupled to mitotic post-translational modifications has never been characterized. Here, we solved the complex structure of Ran with the nucleotide release factor Mog1 and delineated a novel mitosis-specific acetylation-regulated Ran-Mog1 interaction during chromosome segregation. Our structure-guided functional analyses revealed that Mog1 competes with RCC1 for Ran binding in a GTP/GDP-dependent manner. Biochemical characterization demonstrated that Mog1-bound Ran prevents RCC1 binding and subsequent GTP loading. Surprisingly, Ran is a bona fide substrate of TIP60, and the acetylation of Lys134 by TIP60 liberates Mog1 from Ran binding during mitosis. Importantly, this acetylation-elicited switch of Ran binding to RCC1 promotes high level of Ran-GTP, which is essential for chromosome alignment. These results establish a previously uncharacterized regulatory mechanism in which TIP60 provides a homeostatic control of Ran-GTP level by tuning Ran effector binding for chromosome segregation in mitosis.
Collapse
Affiliation(s)
- Xiaoling Bao
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Heng Liu
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xing Liu
- Center of Excellence in Molecular Cell Sciences, Chinese Academy of Sciences & Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Ke Ruan
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yonghui Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Zhiyong Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Qi Hu
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Ying Liu
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Saima Akram
- Center of Excellence in Molecular Cell Sciences, Chinese Academy of Sciences & Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Jiahai Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Qingguo Gong
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Wenwen Wang
- Center of Excellence in Molecular Cell Sciences, Chinese Academy of Sciences & Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xiao Yuan
- Southern University of Science & Technology, Shenzhen 518055, China
| | - Jian Li
- Keck Center for Molecular Imaging, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Lingli Zhao
- Keck Center for Molecular Imaging, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Zhen Dou
- Center of Excellence in Molecular Cell Sciences, Chinese Academy of Sciences & Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Ruijun Tian
- Southern University of Science & Technology, Shenzhen 518055, China
| | - Xuebiao Yao
- Center of Excellence in Molecular Cell Sciences, Chinese Academy of Sciences & Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Jihui Wu
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yunyu Shi
- Hefei National Laboratory for Physical Sciences at Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
72
|
Kuhn J, Dumont S. Mammalian kinetochores count attached microtubules in a sensitive and switch-like manner. J Cell Biol 2019; 218:3583-3596. [PMID: 31492713 PMCID: PMC6829666 DOI: 10.1083/jcb.201902105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 07/10/2019] [Accepted: 08/08/2019] [Indexed: 01/09/2023] Open
Abstract
Kinetochores monitor their attachment to spindle microtubules to control spindle assembly checkpoint (SAC) signaling and cell cycle progression. Kuhn and Dumont show that individual mammalian kinetochores monitor the number of attached microtubules as a single unit in a sensitive and switch-like manner. The spindle assembly checkpoint (SAC) prevents anaphase until all kinetochores attach to the spindle. Each mammalian kinetochore binds many microtubules, but how many attached microtubules are required to turn off the checkpoint, and how the kinetochore monitors microtubule numbers, are not known and are central to understanding SAC mechanisms and function. To address these questions, here we systematically tune and fix the fraction of Hec1 molecules capable of microtubule binding. We show that Hec1 molecules independently bind microtubules within single kinetochores, but that the kinetochore does not independently process attachment information from different molecules. Few attached microtubules (20% occupancy) can trigger complete Mad1 loss, and Mad1 loss is slower in this case. Finally, we show using laser ablation that individual kinetochores detect changes in microtubule binding, not in spindle forces that accompany attachment. Thus, the mammalian kinetochore responds specifically to the binding of each microtubule and counts microtubules as a single unit in a sensitive and switch-like manner. This may allow kinetochores to rapidly react to early attachments and maintain a robust SAC response despite dynamic microtubule numbers.
Collapse
Affiliation(s)
- Jonathan Kuhn
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA .,Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Sophie Dumont
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA .,Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA.,Department of Cell and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
73
|
Nellikka RK, Sreeja JS, Dharmapal D, John R, Monteiro A, Macedo JC, Conde C, Logarinho E, Sunkel CE, Sengupta S. α-Fodrin is required for the organization of functional microtubules during mitosis. Cell Cycle 2019; 18:2713-2726. [PMID: 31455186 DOI: 10.1080/15384101.2019.1656476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The cytoskeleton protein α-fodrin plays a major role in maintaining structural stability of membranes. It was also identified as part of the brain γ-tubulin ring complex, the major microtubule nucleator. Here, we investigated the requirement of α-fodrin for microtubule spindle assembly during mitotic progression. We found that α-fodrin depletion results in abnormal mitosis with uncongressed chromosomes, leading to prolonged activation of the spindle assembly checkpoint and a severe mitotic delay. Further, α-fodrin repression led to the formation of shortened spindles with unstable kinetochore-microtubule attachments. We also found that the mitotic kinesin CENP-E had reduced levels at kinetochores to likely account for the chromosome misalignment defects in α-fodrin-depleted cells. Importantly, we showed these cells to exhibit reduced levels of detyrosinated α-tubulin, which primarily drives CENP-E localization. Since proper microtubule dynamics and chromosome alignment are required for completion of normal mitosis, this study reveals an unforeseen role of α-fodrin in regulating mitotic progression. Future studies on these lines of observations should reveal important mechanistic insight for fodrin's involvement in cancer.
Collapse
Affiliation(s)
- Rohith Kumar Nellikka
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Jamuna S Sreeja
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Dhrishya Dharmapal
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | - Rince John
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| | | | | | - Carlos Conde
- i3S-IBMC, Universidade do Porto , Porto , Portugal
| | | | - Claudio E Sunkel
- i3S-IBMC, Universidade do Porto , Porto , Portugal.,ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto , Porto , Portugal
| | - Suparna Sengupta
- Cancer Research Program-III, Rajiv Gandhi Centre for Biotechnology, University of Kerala , Thiruvananthapuram , India
| |
Collapse
|
74
|
Autophosphorylation is sufficient to release Mps1 kinase from native kinetochores. Proc Natl Acad Sci U S A 2019; 116:17355-17360. [PMID: 31405987 DOI: 10.1073/pnas.1901653116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Accurate mitosis depends on a surveillance system called the spindle assembly checkpoint. This checkpoint acts at kinetochores, which attach chromosomes to the dynamic tips of spindle microtubules. When a kinetochore is unattached or improperly attached, the protein kinase Mps1 phosphorylates kinetochore components, catalyzing the generation of a diffusible "wait" signal that delays anaphase and gives the cell time to correct the error. When a kinetochore becomes properly attached, its checkpoint signal is silenced to allow progression into anaphase. Recently, microtubules were found to compete directly against recombinant human Mps1 fragments for binding to the major microtubule-binding kinetochore element Ndc80c, suggesting a direct competition model for silencing the checkpoint signal at properly attached kinetochores. Here, by developing single-particle fluorescence-based assays, we tested whether such direct competition occurs in the context of native kinetochores isolated from yeast. Mps1 levels were not reduced on kinetochore particles bound laterally to the sides of microtubules or on particles tracking processively with disassembling tips. Instead, we found that Mps1 kinase activity was sufficient to promote its release from the isolated kinetochores. Mps1 autophosphorylation, rather than phosphorylation of other kinetochore components, was responsible for this dissociation. Our findings suggest that checkpoint silencing in yeast does not arise from a direct competition between Mps1 and microtubules, and that phosphoregulation of Mps1 may be a critical aspect of the silencing mechanism.
Collapse
|
75
|
Interplay between Phosphatases and the Anaphase-Promoting Complex/Cyclosome in Mitosis. Cells 2019; 8:cells8080814. [PMID: 31382469 PMCID: PMC6721574 DOI: 10.3390/cells8080814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Accurate division of cells into two daughters is a process that is vital to propagation of life. Protein phosphorylation and selective degradation have emerged as two important mechanisms safeguarding the delicate choreography of mitosis. Protein phosphatases catalyze dephosphorylation of thousands of sites on proteins, steering the cells through establishment of the mitotic phase and exit from it. A large E3 ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C) becomes active during latter stages of mitosis through G1 and marks hundreds of proteins for destruction. Recent studies have revealed the complex interregulation between these two classes of enzymes. In this review, we highlight the direct and indirect mechanisms by which phosphatases and the APC/C mutually influence each other to ensure accurate spatiotemporal and orderly progression through mitosis, with a particular focus on recent insights and conceptual advances.
Collapse
|
76
|
Human Papillomavirus 16 oncoprotein E7 retards mitotic progression by blocking Mps1-MAP4 signaling cascade. Oncogene 2019; 38:5959-5970. [PMID: 31253867 DOI: 10.1038/s41388-019-0851-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/04/2019] [Indexed: 11/09/2022]
Abstract
Human epithelial cells can be infected by more than 200 types of human papilloma viruses (HPVs), and persistent HPV infections lead to cervical cancer or other deadly cancers. It has been established that mitotic progression is critical for HPV16 infection, but the underlying mechanism remains unknown. Here, we report that oncoprotein E7 of HPV16 but not HPV18 retards mitotic progression in host cell by direct binding to the C terminus of Microtubule-Associated Protein 4 (MAP4). MAP4 is a novel bona fide target of HPV16E7 protein which binds and recruits the latter to spindle microtubule in mitosis. HPV16E7 protein promotes MAP4 stability by inhibiting MAP4 phosphorylation- mediated degradation to increase the stability of microtubule polymerization and cause an extend mitotic progression. We further uncovered that Mps1 is a kinase of MAP4, and E7-MAP4 binding blocks Mps1 phosphorylation of MAP4, thereby interrupting phosphorylation-dependent MAP4 degradation. Mutations of MAP4 at T927ES928E partially abolished E7-binding capacity and rescued mitotic progression in host cells. In conclusion, our study reveals a molecular mechanism by which HPV16E7 perturbs host mitotic progression by interfering Mps1-MAP4 signaling cascade, which results in an extended infection window and may facilitate the persistent HPV16 infection.
Collapse
|
77
|
Abstract
Mistakes in the process of cell division can lead to the loss, gain or rearrangement of chromosomes. Significant chromosomal abnormalities are usually lethal to the cells and cause spontaneous miscarriages. However, in some cases, defects in the spindle assembly checkpoint lead to severe diseases, such as cancer and birth and development defects, including Down's syndrome. The timely and accurate control of chromosome segregation in mitosis relies on the spindle assembly checkpoint (SAC), an evolutionary conserved, self-regulated signalling system present in higher organisms. The spindle assembly checkpoint is orchestrated by dynamic interactions between spindle microtubules and the kinetochore , a multiprotein complex that constitutes the site for attachment of chromosomes to microtubule polymers to pull sister chromatids apart during cell division. This chapter discusses the current molecular understanding of the essential, highly dynamic molecular interactions underpinning spindle assembly checkpoint signalling and how the complex choreography of interactions can be coordinated in time and space to finely regulate the process. The potential of targeting this signalling pathway to interfere with the abnormal segregation of chromosomes, which occurs in diverse malignancies and the new opportunities that recent technological developments are opening up for a deeper understanding of the spindle assembly checkpoint are also discussed.
Collapse
Affiliation(s)
- Victor M Bolanos-Garcia
- Faculty of Health and Life Sciences, Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
| |
Collapse
|
78
|
Etemad B, Vertesy A, Kuijt TEF, Sacristan C, van Oudenaarden A, Kops GJPL. Spindle checkpoint silencing at kinetochores with submaximal microtubule occupancy. J Cell Sci 2019; 132:jcs.231589. [PMID: 31138679 DOI: 10.1242/jcs.231589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/17/2019] [Indexed: 11/20/2022] Open
Abstract
The spindle assembly checkpoint (SAC) ensures proper chromosome segregation by monitoring kinetochore-microtubule interactions. SAC proteins are shed from kinetochores once stable attachments are achieved. Human kinetochores consist of hundreds of SAC protein recruitment modules and bind up to 20 microtubules, raising the question of how the SAC responds to intermediate attachment states. We show that one protein module ('RZZS-MAD1-MAD2') of the SAC is removed from kinetochores at low microtubule occupancy and remains absent at higher occupancies, while another module ('BUB1-BUBR1') is retained at substantial levels irrespective of attachment states. These behaviours reflect different silencing mechanisms: while BUB1 displacement is almost fully dependent on MPS1 inactivation, MAD1 (also known as MAD1L1) displacement is not. Artificially tuning the affinity of kinetochores for microtubules further shows that ∼50% occupancy is sufficient to shed MAD2 and silence the SAC. Kinetochores thus respond as a single unit to shut down SAC signalling at submaximal occupancy states, but retain one SAC module. This may ensure continued SAC silencing on kinetochores with fluctuating occupancy states while maintaining the ability for fast SAC re-activation.
Collapse
Affiliation(s)
- Banafsheh Etemad
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Abel Vertesy
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Timo E F Kuijt
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Carlos Sacristan
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Alexander van Oudenaarden
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, 3584 CT, The Netherlands
| |
Collapse
|
79
|
The mammalian kinetochore-microtubule interface: robust mechanics and computation with many microtubules. Curr Opin Cell Biol 2019; 60:60-67. [PMID: 31132675 DOI: 10.1016/j.ceb.2019.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022]
Abstract
The kinetochore drives chromosome segregation at cell division. It acts as a physical link between chromosomes and dynamic microtubules, and as a signaling hub detecting and processing microtubule attachments to control anaphase onset. The mammalian kinetochore is a large macromolecular machine that forms a dynamic interface with the many microtubules that it binds. While we know most of the kinetochore's component parts, how they work together to give rise to its robust functions remains poorly understood. Here we highlight recent findings that shed light on this question, driven by an expanding physical and molecular toolkit. We present emerging principles that underlie the kinetochore's robust microtubule grip, such as redundancy, specialization, and dynamicity, and present signal processing principles that connect this microtubule grip to robust computation. Throughout, we identify open questions, and define simple engineering concepts that provide insight into kinetochore function.
Collapse
|
80
|
Vallardi G, Cordeiro MH, Saurin AT. A Kinase-Phosphatase Network that Regulates Kinetochore-Microtubule Attachments and the SAC. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:457-484. [PMID: 28840249 DOI: 10.1007/978-3-319-58592-5_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The KMN network (for KNL1, MIS12 and NDC80 complexes) is a hub for signalling at the outer kinetochore. It integrates the activities of two kinases (MPS1 and Aurora B) and two phosphatases (PP1 and PP2A-B56) to regulate kinetochore-microtubule attachments and the spindle assembly checkpoint (SAC). We will first discuss each of these enzymes separately, to describe how they are regulated at kinetochores and why this is important for their primary function in controlling either microtubule attachments or the SAC. We will then discuss why inhibiting any one of them individually produces secondary effects on all the others. This cross-talk may help to explain why all enzymes have been linked to both processes, even though the direct evidence suggests they each control only one. This chapter therefore describes how a network of kinases and phosphatases work together to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Marilia Henriques Cordeiro
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian Thomas Saurin
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
81
|
French BT, Straight AF. The Power of Xenopus Egg Extract for Reconstitution of Centromere and Kinetochore Function. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:59-84. [PMID: 28840233 DOI: 10.1007/978-3-319-58592-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Faithful transmission of genetic information during cell division requires attachment of chromosomes to the mitotic spindle via the kinetochore. In vitro reconstitution studies are beginning to uncover how the kinetochore is assembled upon the underlying centromere, how the kinetochore couples chromosome movement to microtubule dynamics, and how cells ensure the site of kinetochore assembly is maintained from one generation to the next. Here we give special emphasis to advances made in Xenopus egg extract, which provides a unique, biochemically tractable in vitro system that affords the complexity of cytoplasm and nucleoplasm to permit reconstitution of the dynamic, cell cycle-regulated functions of the centromere and kinetochore.
Collapse
Affiliation(s)
- Bradley T French
- Department of Biochemistry, Stanford University, 279 Campus Drive, Beckman 409, Stanford, CA, 94305, USA
| | - Aaron F Straight
- Department of Biochemistry, Stanford University, 279 Campus Drive, Beckman 409, Stanford, CA, 94305, USA.
| |
Collapse
|
82
|
Riggs JR, Elsner J, Cashion D, Robinson D, Tehrani L, Nagy M, Fultz KE, Krishna Narla R, Peng X, Tran T, Kulkarni A, Bahmanyar S, Condroski K, Pagarigan B, Fenalti G, LeBrun L, Leftheris K, Zhu D, Boylan JF. Design and Optimization Leading to an Orally Active TTK Protein Kinase Inhibitor with Robust Single Agent Efficacy. J Med Chem 2019; 62:4401-4410. [PMID: 30998356 DOI: 10.1021/acs.jmedchem.8b01869] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Triple negative breast cancer (TNBC) is an aggressive disease with high relapse rates and few treatment options. Outlined in previous publications, we identified a series of potent, dual TTK/CLK2 inhibitors with strong efficacy in TNBC xenograft models. Pharmacokinetic properties and kinome selectivity were optimized, resulting in the identification of a new series of potent, selective, and orally bioavailable TTK inhibitors. We describe here the structure-activity relationship of the 2,4-disubstituted-7 H-pyrrolo[2,3- d]pyrimidine series, leading to significant single agent efficacy in a TNBC xenograft model without body weight loss. The design effort evolving an iv-dosed TTK/CLK2 inhibitor to an orally bioavailable TTK inhibitor is described.
Collapse
Affiliation(s)
- Jennifer R Riggs
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Jan Elsner
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Dan Cashion
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Dale Robinson
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Lida Tehrani
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Mark Nagy
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Kimberly E Fultz
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Rama Krishna Narla
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Xiaohui Peng
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Tam Tran
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Ashutosh Kulkarni
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Sogole Bahmanyar
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Kevin Condroski
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Barbra Pagarigan
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Gustavo Fenalti
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Laurie LeBrun
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Katerina Leftheris
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - Dan Zhu
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| | - John F Boylan
- Celgene Corporation , 10300 Campus Point Drive, Suite 100 , San Diego , California 92121 , United States
| |
Collapse
|
83
|
Hayward D, Alfonso-Pérez T, Cundell MJ, Hopkins M, Holder J, Bancroft J, Hutter LH, Novak B, Barr FA, Gruneberg U. CDK1-CCNB1 creates a spindle checkpoint-permissive state by enabling MPS1 kinetochore localization. J Cell Biol 2019; 218:1182-1199. [PMID: 30674582 PMCID: PMC6446832 DOI: 10.1083/jcb.201808014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/19/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
Spindle checkpoint signaling is initiated by recruitment of the kinase MPS1 to unattached kinetochores during mitosis. We show that CDK1-CCNB1 and a counteracting phosphatase PP2A-B55 regulate the engagement of human MPS1 with unattached kinetochores by controlling the phosphorylation status of S281 in the kinetochore-binding domain. This regulation is essential for checkpoint signaling, since MPS1S281A is not recruited to unattached kinetochores and fails to support the recruitment of other checkpoint proteins. Directly tethering MPS1S281A to the kinetochore protein Mis12 bypasses this regulation and hence the requirement for S281 phosphorylation in checkpoint signaling. At the metaphase-anaphase transition, MPS1 S281 dephosphorylation is delayed because PP2A-B55 is negatively regulated by CDK1-CCNB1 and only becomes fully active once CCNB1 concentration falls below a characteristic threshold. This mechanism prolongs the checkpoint-responsive period when MPS1 can localize to kinetochores and enables a response to late-stage spindle defects. By acting together, CDK1-CCNB1 and PP2A-B55 thus create a spindle checkpoint-permissive state and ensure the fidelity of mitosis.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| | - Tatiana Alfonso-Pérez
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Michael J Cundell
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Michael Hopkins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - James Bancroft
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| | - Lukas H Hutter
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Bela Novak
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| |
Collapse
|
84
|
Recent Progress on the Localization of the Spindle Assembly Checkpoint Machinery to Kinetochores. Cells 2019; 8:cells8030278. [PMID: 30909555 PMCID: PMC6468716 DOI: 10.3390/cells8030278] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/14/2022] Open
Abstract
Faithful chromosome segregation during mitosis is crucial for maintaining genome stability. The spindle assembly checkpoint (SAC) is a surveillance mechanism that ensures accurate mitotic progression. Defective SAC signaling leads to premature sister chromatid separation and aneuploid daughter cells. Mechanistically, the SAC couples the kinetochore microtubule attachment status to the cell cycle progression machinery. In the presence of abnormal kinetochore microtubule attachments, the SAC prevents the metaphase-to-anaphase transition through a complex kinase-phosphatase signaling cascade which results in the correct balance of SAC components recruited to the kinetochore. The correct kinetochore localization of SAC proteins is a prerequisite for robust SAC signaling and, hence, accurate chromosome segregation. Here, we review recent progresses on the kinetochore recruitment of core SAC factors.
Collapse
|
85
|
Pachis ST, Kops GJPL. Leader of the SAC: molecular mechanisms of Mps1/TTK regulation in mitosis. Open Biol 2019; 8:rsob.180109. [PMID: 30111590 PMCID: PMC6119859 DOI: 10.1098/rsob.180109] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Discovered in 1991 in a screen for genes involved in spindle pole body duplication, the monopolar spindle 1 (Mps1) kinase has since claimed a central role in processes that ensure error-free chromosome segregation. As a result, Mps1 kinase activity has become an attractive candidate for pharmaceutical companies in the search for compounds that target essential cellular processes to eliminate, for example, tumour cells or pathogens. Research in recent decades has offered many insights into the molecular function of Mps1 and its regulation. In this review, we integrate the latest knowledge regarding the regulation of Mps1 activity and its spatio-temporal distribution, highlight gaps in our understanding of these processes and propose future research avenues to address them.
Collapse
Affiliation(s)
- Spyridon T Pachis
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute - KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
86
|
Zhang Y, Dong J, Shi R, Feng L, Li Y, Cheng C, Zhang L, Song B, Bi Y, Huang H, Kong P, Guo J, Liu J. Mps1 is associated with the BRAF V600E mutation and predicts poor outcome in patients with colorectal cancer. Oncol Lett 2019; 17:2809-2817. [PMID: 30854056 PMCID: PMC6365956 DOI: 10.3892/ol.2019.9924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) with the V600E mutation of B-Raf proto-oncogene serine/threonine kinase (BRAFV600E) mutation is insensitive to chemotherapy and is indicative of a poor patient prognosis. Although BRAF inhibitors have a marked effect on malignant melanoma harboring the BRAFV600E mutation, they have a limited effect on patients with CRC with the same BRAF mutation. A previous study identified a novel gene, monopolar spindle protein kinase 1 (Mps1), a downstream target of BRAFV600E only, rather than of wild-type BRAF as well, which contributes to tumorigenesis in melanoma. In the present study, the incidence of BRAFV600E in patients with CRC was identified and the correlation of Mps1, phospho-extracellular-signal-regulated kinase (p-ERK) and BRAFV600E was investigated. The results indicated that the mutation rate of BRAFV600E was 5.2% in CRC. Poorly differentiated tumors and mucinous tumors have a significantly higher incidence of BRAFV600E compared with well-differentiated tumors and non-mucinous tumors (P<0.05). Kaplan-Meier survival analysis indicated that the survival rate was markedly lower in patients with BRAFV600E compared with in patients with wild-type BRAF (BRAFWT). The expression of p-ERK and Mps1 in CRC with BRAFV600E was significantly higher compared with in CRC with BRAFWT (P<0.05), and their expression is associated with cancer classification, degree of differentiation and lymph node transfusion (P<0.05). In addition p-ERK expression was positively correlated with Mps1 expression, with a contingency coefficient of 0.679 (P=0.002). In conclusion, the results of the present study indicated that Mps1 was significantly associated with BRAFV600E/p-ERK and may serve a crucial function in the development of CRC. The results of the present study raise the possibility that targeting the oncogenic BRAF and Mps1, particularly when in conjunction, could provide promising therapeutic opportunities for the treatment of CRC.
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jinyao Dong
- Endoscopy Center, Shanxi Cancer Hospital, Taiyuan, Shanxi 30013, P.R. China
| | - Ruyi Shi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Cell Biology and Genetics, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Liguo Feng
- Department of General Surgery, Taiyuan Municipal No. 2 People's Hospital, Taiyuan, Shanxi 030002, P.R. China
| | - Yike Li
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Caixia Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Pathology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ling Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bin Song
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yanghui Bi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - He Huang
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Pengzhou Kong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jiansheng Guo
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jing Liu
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
87
|
Yu KW, Zhong N, Xiao Y, She ZY. Mechanisms of kinesin-7 CENP-E in kinetochore-microtubule capture and chromosome alignment during cell division. Biol Cell 2019; 111:143-160. [PMID: 30784092 DOI: 10.1111/boc.201800082] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
Abstract
Chromosome congression is essential for faithful chromosome segregation and genomic stability in cell division. Centromere-associated protein E (CENP-E), a plus-end-directed kinesin motor, is required for congression of pole-proximal chromosomes in metaphase. CENP-E accumulates at the outer plate of kinetochores and mediates the kinetochore-microtubule capture. CENP-E also transports the chromosomes along spindle microtubules towards the equatorial plate. CENP-E interacts with Bub1-related kinase, Aurora B and core kinetochore components during kinetochore-microtubule attachment. In this review, we introduce the structures and mechanochemistry of kinesin-7 CENP-E. We highlight the complicated interactions between CENP-E and partner proteins during chromosome congression. We summarise the detailed roles and mechanisms of CENP-E in mitosis and meiosis, including the kinetochore-microtubule capture, chromosome congression/alignment in metaphase and the regulation of spindle assembly checkpoint. We also shed a light on the roles of CENP-E in tumourigenesis and CENP-E's specific inhibitors.
Collapse
Affiliation(s)
- Kai-Wei Yu
- Department of Cell Biology and Genetics/Center for Cell and Developmental Biology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Ning Zhong
- Department of Cell Biology and Genetics/Center for Cell and Developmental Biology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Yu Xiao
- Department of Cell Biology and Genetics/Center for Cell and Developmental Biology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics/Center for Cell and Developmental Biology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350108, China
| |
Collapse
|
88
|
Yang H, Zhang F, Huang CJ, Liao J, Han Y, Hao P, Chu Y, Lu X, Li W, Yu H, Kang J. Mps1 regulates spindle morphology through MCRS1 to promote chromosome alignment. Mol Biol Cell 2019; 30:1060-1068. [PMID: 30785839 PMCID: PMC6724509 DOI: 10.1091/mbc.e18-09-0546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Accurate partitioning of chromosomes during mitosis is essential for genetic stability and requires the assembly of the dynamic mitotic spindle and proper kinetochore–microtubule attachment. The spindle assembly checkpoint (SAC) monitors the incompleteness and errors in kinetochore–microtubule attachment and delays anaphase. The SAC kinase Mps1 regulates the recruitment of downstream effectors to unattached kinetochores. Mps1 also actively promotes chromosome alignment during metaphase, but the underlying mechanism is not completely understood. Here, we show that Mps1 regulates chromosome alignment through MCRS1, a spindle assembly factor that controls the dynamics of the minus end of kinetochore microtubules. Mps1 binds and phosphorylates MCRS1. This mechanism enables KIF2A localization to the minus end of spindle microtubules. Thus, our study reveals a novel role of Mps1 in regulating the dynamics of the minus end of microtubules and expands the functions of Mps1 in genome maintenance.
Collapse
Affiliation(s)
- Hongdan Yang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Fengxia Zhang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Ching-Jung Huang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Jun Liao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Ying Han
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Piliang Hao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Youjun Chu
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Xiaoai Lu
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Wenshu Li
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Hongtao Yu
- Howard Hughes Medical Institute, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jungseog Kang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China.,NYU-ECNU Center for Computational Chemistry, New York University at Shanghai, Shanghai 200062, China
| |
Collapse
|
89
|
Osswald M, Santos AF, Morais-de-Sá E. Light-Induced Protein Clustering for Optogenetic Interference and Protein Interaction Analysis in Drosophila S2 Cells. Biomolecules 2019; 9:biom9020061. [PMID: 30759894 PMCID: PMC6406598 DOI: 10.3390/biom9020061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022] Open
Abstract
Drosophila Schneider 2 (S2) cells are a simple and powerful system commonly used in cell biology because they are well suited for high resolution microscopy and RNAi-mediated depletion. However, understanding dynamic processes, such as cell division, also requires methodology to interfere with protein function with high spatiotemporal control. In this research study, we report the adaptation of an optogenetic tool to Drosophila S2 cells. Light-activated reversible inhibition by assembled trap (LARIAT) relies on the rapid light-dependent heterodimerization between cryptochrome 2 (CRY2) and cryptochrome-interacting bHLH 1 (CIB1) to form large protein clusters. An anti-green fluorescent protein (GFP) nanobody fused with CRY2 allows this method to quickly trap any GFP-tagged protein in these light-induced protein clusters. We evaluated clustering kinetics in response to light for different LARIAT modules, and showed the ability of GFP-LARIAT to inactivate the mitotic protein Mps1 and to disrupt the membrane localization of the polarity regulator Lethal Giant Larvae (Lgl). Moreover, we validated light-induced co-clustering assays to assess protein-protein interactions in S2 cells. In conclusion, GFP-based LARIAT is a versatile tool to answer different biological questions, since it enables probing of dynamic processes and protein-protein interactions with high spatiotemporal resolution in Drosophila S2 cells.
Collapse
Affiliation(s)
- Mariana Osswald
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - A Filipa Santos
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Eurico Morais-de-Sá
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
90
|
The binding of Borealin to microtubules underlies a tension independent kinetochore-microtubule error correction pathway. Nat Commun 2019; 10:682. [PMID: 30737408 PMCID: PMC6368601 DOI: 10.1038/s41467-019-08418-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
Proper chromosome segregation depends upon kinetochore phosphorylation by the Chromosome Passenger Complex (CPC). Current models suggest the activity of the CPC decreases in response to the inter-kinetochore stretch that accompanies the formation of bi-oriented microtubule attachments, however little is known about tension-independent CPC phosphoregulation. Microtubule bundles initially lie in close proximity to inner centromeres and become depleted by metaphase. Here we find these microtubules control kinetochore phosphorylation by the CPC in a tension independent manner via a microtubule-binding site on the Borealin subunit. Disruption of Borealin-microtubule interactions generates reduced phosphorylation of prometaphase kinetochores, improper kinetochore-microtubule attachments and weakened spindle checkpoint signals. Experimental and modeling evidence suggests that kinetochore phosphorylation is greatly stimulated when the CPC binds microtubules that lie near the inner centromere, even if kinetochores have high inter-kinetochore stretch. We propose the CPC senses its local environment through microtubule structures to control phosphorylation of kinetochores. How the chromosome passenger complex (CPC) phosphorylates the kinetochores that can be a micron away to control mitotic events is unknown. Here the authors find that the CPC directly binds microtubules near inner centromeres, which controls its ability to phosphorylate kinetochores independently of tension generated by kinetochore microtubule attachments.
Collapse
|
91
|
Moura M, Conde C. Phosphatases in Mitosis: Roles and Regulation. Biomolecules 2019; 9:E55. [PMID: 30736436 PMCID: PMC6406801 DOI: 10.3390/biom9020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Mitosis requires extensive rearrangement of cellular architecture and of subcellular structures so that replicated chromosomes can bind correctly to spindle microtubules and segregate towards opposite poles. This process originates two new daughter nuclei with equal genetic content and relies on highly-dynamic and tightly regulated phosphorylation of numerous cell cycle proteins. A burst in protein phosphorylation orchestrated by several conserved kinases occurs as cells go into and progress through mitosis. The opposing dephosphorylation events are catalyzed by a small set of protein phosphatases, whose importance for the accuracy of mitosis is becoming increasingly appreciated. This review will focus on the established and emerging roles of mitotic phosphatases, describe their structural and biochemical properties, and discuss recent advances in understanding the regulation of phosphatase activity and function.
Collapse
Affiliation(s)
- Margarida Moura
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| | - Carlos Conde
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
92
|
Han Y, Wu Y, Xu Y, Guo W, Zhang N, Wang X. Molecular mechanism of point mutation-induced Monopolar spindle 1 (Mps1/TTK) inhibitor resistance revealed by a comprehensive molecular modeling study. PeerJ 2019; 7:e6299. [PMID: 30693152 PMCID: PMC6345219 DOI: 10.7717/peerj.6299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022] Open
Abstract
Background Monopolar spindle 1 (Mps1/TTK) is an apical dual-specificity protein kinase in the spindle assembly checkpoint (SAC) that guarantees accurate segregation of chromosomes during mitosis. High levels of Mps1 are found in various types of human malignancies, such as glioblastoma, osteosarcoma, hepatocellular carcinoma, and breast cancer. Several potent inhibitors of Mps1 exist, and exhibit promising activity in many cell cultures and xenograft models. However, resistance due to point mutations in the kinase domain of Mps1 limits the therapeutic effects of these inhibitors. Understanding the detailed resistance mechanism induced by Mps1 point mutations is therefore vital for the development of novel inhibitors against malignancies. Methods In this study, conventional molecular dynamics (MD) simulation and Gaussian accelerated MD (GaMD) simulation were performed to elucidate the resistance mechanisms of Cpd-5, a potent Mps1 inhibitor, induced by the four representative mutations I531M, I598F, C604Y, S611R. Results Our results from conventional MD simulation combined with structural analysis and free energy calculation indicated that the four mutations weaken the binding affinity of Cpd-5 and the major variations in structural were the conformational changes of the P-loop, A-loop and αC-helix. Energetic differences of per-residue between the WT system and the mutant systems indicated the mutations may allosterically regulate the conformational ensemble and the major variations were residues of Ile-663 and Gln-683, which located in the key loops of catalytic loop and A-loop, respectively. The large conformational and energetic differences were further supported by the GaMD simulations. Overall, these obtained molecular mechanisms will aid rational design of novel Mps1 inhibitors to combat inhibitor resistance.
Collapse
Affiliation(s)
- Yan Han
- Department of TCM Orthopedics & Traumatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yungang Wu
- Department of TCM Orthopedics & Traumatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Xu
- Department of Pharmacy, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wentao Guo
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Zhang
- Department of TCM Orthopedics & Traumatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoyi Wang
- Department of TCM Orthopedics & Traumatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
93
|
Zhao G, Cheng Y, Gui P, Cui M, Liu W, Wang W, Wang X, Ali M, Dou Z, Niu L, Liu H, Anderson L, Ruan K, Hong J, Yao X. Dynamic acetylation of the kinetochore-associated protein HEC1 ensures accurate microtubule-kinetochore attachment. J Biol Chem 2019; 294:576-592. [PMID: 30409912 PMCID: PMC6333894 DOI: 10.1074/jbc.ra118.003844] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 10/18/2018] [Indexed: 11/06/2022] Open
Abstract
Faithful chromosome segregation during mitosis is critical for maintaining genome integrity in cell progeny and relies on accurate and robust kinetochore-microtubule attachments. The NDC80 complex, a tetramer comprising kinetochore protein HEC1 (HEC1), NDC80 kinetochore complex component NUF2 (NUF2), NDC80 kinetochore complex component SPC24 (SPC24), and SPC25, plays a critical role in kinetochore-microtubule attachment. Mounting evidence indicates that phosphorylation of HEC1 is important for regulating the binding of the NDC80 complex to microtubules. However, it remains unclear whether other post-translational modifications, such as acetylation, regulate NDC80-microtubule attachment during mitosis. Here, using pulldown assays with HeLa cell lysates and site-directed mutagenesis, we show that HEC1 is a bona fide substrate of the lysine acetyltransferase Tat-interacting protein, 60 kDa (TIP60) and that TIP60-mediated acetylation of HEC1 is essential for accurate chromosome segregation in mitosis. We demonstrate that TIP60 regulates the dynamic interactions between NDC80 and spindle microtubules during mitosis and observed that TIP60 acetylates HEC1 at two evolutionarily conserved residues, Lys-53 and Lys-59. Importantly, this acetylation weakened the phosphorylation of the N-terminal HEC1(1-80) region at Ser-55 and Ser-62, which is governed by Aurora B and regulates NDC80-microtubule dynamics, indicating functional cross-talk between these two post-translation modifications of HEC1. Moreover, the TIP60-mediated acetylation was specifically reversed by sirtuin 1 (SIRT1). Taken together, our results define a conserved signaling hierarchy, involving HEC1, TIP60, Aurora B, and SIRT1, that integrates dynamic HEC1 acetylation and phosphorylation for accurate kinetochore-microtubule attachment in the maintenance of genomic stability during mitosis.
Collapse
Affiliation(s)
- Gangyin Zhao
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Yubao Cheng
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Ping Gui
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Meiying Cui
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Wei Liu
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Wenwen Wang
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Xueying Wang
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Mahboob Ali
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Zhen Dou
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Liwen Niu
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Haiyan Liu
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Leonard Anderson
- the Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Ke Ruan
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Jingjun Hong
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| | - Xuebiao Yao
- From the Division of Molecular and Cell Biophysics, Hefei National Science Center for Physical Sciences at the Microscale, University of Science and Technology of the China School of Life Sciences, Chinese Academy of Sciences Center of Excellence on Molecular Cell Sciences, Hefei 230027, China and
| |
Collapse
|
94
|
Chen C, Whitney IP, Banerjee A, Sacristan C, Sekhri P, Kern DM, Fontan A, Kops GJPL, Tyson JJ, Cheeseman IM, Joglekar AP. Ectopic Activation of the Spindle Assembly Checkpoint Signaling Cascade Reveals Its Biochemical Design. Curr Biol 2018; 29:104-119.e10. [PMID: 30595520 DOI: 10.1016/j.cub.2018.11.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 11/27/2022]
Abstract
Switch-like activation of the spindle assembly checkpoint (SAC) is critical for accurate chromosome segregation and for cell division in a timely manner. To determine the mechanisms that achieve this, we engineered an ectopic, kinetochore-independent SAC activator: the "eSAC." The eSAC stimulates SAC signaling by artificially dimerizing Mps1 kinase domain and a cytosolic KNL1 phosphodomain, the kinetochore signaling scaffold. By exploiting variable eSAC expression in a cell population, we defined the dependence of the eSAC-induced mitotic delay on eSAC concentration in a cell to reveal the dose-response behavior of the core signaling cascade of the SAC. These quantitative analyses and subsequent mathematical modeling of the dose-response data uncover two crucial properties of the core SAC signaling cascade: (1) a cellular limit on the maximum anaphase-inhibitory signal that the cascade can generate due to the limited supply of SAC proteins and (2) the ability of the KNL1 phosphodomain to produce the anaphase-inhibitory signal synergistically, when it recruits multiple SAC proteins simultaneously. We propose that these properties together achieve inverse, non-linear scaling between the signal output per kinetochore and the number of signaling kinetochores. When the number of kinetochores is low, synergistic signaling by KNL1 enables each kinetochore to produce a disproportionately strong signal output. However, when many kinetochores signal concurrently, they compete for a limited supply of SAC proteins. This frustrates synergistic signaling and lowers their signal output. Thus, the signaling activity of unattached kinetochores will adapt to the changing number of signaling kinetochores to enable the SAC to approximate switch-like behavior.
Collapse
Affiliation(s)
- Chu Chen
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ian P Whitney
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Anand Banerjee
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Carlos Sacristan
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences), and Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Palak Sekhri
- Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David M Kern
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Adrienne Fontan
- Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Geert J P L Kops
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences), and Molecular Cancer Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research and Department of Biology, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Ajit P Joglekar
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
95
|
Yi Q, Chen Q, Yan H, Zhang M, Liang C, Xiang X, Pan X, Wang F. Aurora B kinase activity-dependent and -independent functions of the chromosomal passenger complex in regulating sister chromatid cohesion. J Biol Chem 2018; 294:2021-2035. [PMID: 30523151 DOI: 10.1074/jbc.ra118.005978] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/28/2018] [Indexed: 11/06/2022] Open
Abstract
The chromosomal passenger complex (CPC) is a master regulator of mitosis. CPC consists of inner centromere protein (INCENP), Survivin, Borealin, and the kinase Aurora B and plays key roles in regulating kinetochore-microtubule attachments and spindle assembly checkpoint signaling. However, the role of CPC in sister chromatid cohesion, mediated by the cohesin complex, remains incompletely understood. Here, we show that Aurora B kinase activity contributes to centromeric cohesion protection partly through promoting kinetochore localization of the kinase Bub1. Interestingly, disrupting the interaction of INCENP with heterochromatin protein 1 (HP1) in HeLa cells selectively weakens cohesion at mitotic centromeres without detectably reducing the kinase activity of Aurora B. Thus, through this INCENP-HP1 interaction, the CPC also protects centromeric cohesion independently of Aurora B kinase activity. Moreover, the requirement for the INCENP-HP1 interaction in centromeric cohesion protection can be bypassed by tethering HP1 to centromeres or by depleting the cohesin release factor Wapl. We provide further evidence suggesting that the INCENP-HP1 interaction protects centromeric cohesion by promoting the centromere localization of Haspin, a protein kinase that antagonizes Wapl activity at centromeres. Taken together, this study identifies Aurora B kinase activity-dependent and -independent roles for the CPC in regulating centromeric cohesion during mitosis in human cells.
Collapse
Affiliation(s)
- Qi Yi
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qinfu Chen
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Haiyan Yan
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Miao Zhang
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Cai Liang
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xingfeng Xiang
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xuan Pan
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Fangwei Wang
- From the Ministry of Education (MOE) Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
96
|
Loss of Kif18A Results in Spindle Assembly Checkpoint Activation at Microtubule-Attached Kinetochores. Curr Biol 2018; 28:2685-2696.e4. [DOI: 10.1016/j.cub.2018.06.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 11/18/2022]
|
97
|
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: 4.1] [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
|
98
|
Schleicher K, Porter M, Ten Have S, Sundaramoorthy R, Porter IM, Swedlow JR. The Ndc80 complex targets Bod1 to human mitotic kinetochores. Open Biol 2018; 7:rsob.170099. [PMID: 29142109 PMCID: PMC5717335 DOI: 10.1098/rsob.170099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022] Open
Abstract
Regulation of protein phosphatase activity by endogenous protein inhibitors is an important mechanism to control protein phosphorylation in cells. We recently identified Biorientation defective 1 (Bod1) as a small protein inhibitor of protein phosphatase 2A containing the B56 regulatory subunit (PP2A-B56). This phosphatase controls the amount of phosphorylation of several kinetochore proteins and thus the establishment of load-bearing chromosome-spindle attachments in time for accurate separation of sister chromatids in mitosis. Like PP2A-B56, Bod1 directly localizes to mitotic kinetochores and is required for correct segregation of mitotic chromosomes. In this report, we have probed the spatio-temporal regulation of Bod1 during mitotic progression. Kinetochore localization of Bod1 increases from nuclear envelope breakdown until metaphase. Phosphorylation of Bod1 at threonine 95 (T95), which increases Bod1's binding to and inhibition of PP2A-B56, peaks in prometaphase when PP2A-B56 localization to kinetochores is highest. We demonstrate here that kinetochore targeting of Bod1 depends on the outer kinetochore protein Ndc80 and not PP2A-B56. Crucially, Bod1 depletion functionally affects Ndc80 phosphorylation at the N-terminal serine 55 (S55), as well as a number of other phosphorylation sites within the outer kinetochore, including Knl1 at serine 24 and 60 (S24, S60), and threonine T943 and T1155 (T943, T1155). Therefore, Ndc80 recruits a phosphatase inhibitor to kinetochores which directly feeds forward to regulate Ndc80, and Knl1 phosphorylation, including sites that mediate the attachment of microtubules to kinetochores.
Collapse
Affiliation(s)
- Katharina Schleicher
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Michael Porter
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Sara Ten Have
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | | | - Iain M Porter
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Jason R Swedlow
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
99
|
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: 94] [Impact Index Per Article: 13.4] [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
|
100
|
Zhu D, Xu S, Deyanat-Yazdi G, Peng SX, Barnes LA, Narla RK, Tran T, Mikolon D, Ning Y, Shi T, Jiang N, Raymon HK, Riggs JR, Boylan JF. Synthetic Lethal Strategy Identifies a Potent and Selective TTK and CLK1/2 Inhibitor for Treatment of Triple-Negative Breast Cancer with a Compromised G 1-S Checkpoint. Mol Cancer Ther 2018; 17:1727-1738. [PMID: 29866747 DOI: 10.1158/1535-7163.mct-17-1084] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/13/2018] [Accepted: 05/08/2018] [Indexed: 11/16/2022]
Abstract
Historically, phenotypic-based drug discovery has yielded a high percentage of novel drugs while uncovering new tumor biology. CC-671 was discovered using a phenotypic screen for compounds that preferentially induced apoptosis in triple-negative breast cancer cell lines while sparing luminal breast cancer cell lines. Detailed in vitro kinase profiling shows CC-671 potently and selectively inhibits two kinases-TTK and CLK2. Cellular mechanism of action studies demonstrate that CC-671 potently inhibits the phosphorylation of KNL1 and SRp75, direct TTK and CLK2 substrates, respectively. Furthermore, CC-671 causes mitotic acceleration and modification of pre-mRNA splicing leading to apoptosis, consistent with cellular TTK and CLK inhibition. Correlative analysis of genomic and potency data against a large panel of breast cancer cell lines identifies breast cancer cells with a dysfunctional G1-S checkpoint as more sensitive to CC-671, suggesting synthetic lethality between G1-S checkpoint and TTK/CLK2 inhibition. Furthermore, significant in vivo CC-671 efficacy was demonstrated in two cell line-derived and one patient tumor-derived xenograft models of triple-negative breast cancer (TNBC) following weekly dosing. These findings are the first to demonstrate the unique inhibitory combination activity of a dual TTK/CLK2 inhibitor that preferably kills TNBC cells and shows synthetic lethality with a compromised G1-S checkpoint in breast cancer cell lines. On the basis of these data, CC-671 was moved forward for clinical development as a potent and selective TTK/CLK2 inhibitor in a subset of patients with TNBC. Mol Cancer Ther; 17(8); 1727-38. ©2018 AACR.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Oncology Research, Celgene Corporation, San Diego, California.
| | - Shuichan Xu
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | | | - Sophie X Peng
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | - Leo A Barnes
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | | | - Tam Tran
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - David Mikolon
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - Yuhong Ning
- Informatics and Knowledge Utilization Department, Celgene Corporation, San Diego, California
| | - Tao Shi
- Informatics and Knowledge Utilization Department, Celgene Corporation, San Diego, California
| | - Ning Jiang
- Department of Oncology Research, Celgene Corporation, San Diego, California
| | - Heather K Raymon
- Department of Pharmacology, Celgene Corporation, San Diego, California
| | - Jennifer R Riggs
- Department of Chemistry, Celgene Corporation, San Diego, California
| | - John F Boylan
- Department of Oncology Research, Celgene Corporation, San Diego, California
| |
Collapse
|