1
|
McLellan MM, Aerne BL, Banerjee Dhoul JJ, Holder MV, Auchynnikava T, Tapon N. Meru co-ordinates spindle orientation with cell polarity and cell cycle progression. EMBO J 2025; 44:2949-2975. [PMID: 40169811 DOI: 10.1038/s44318-025-00420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
Correct mitotic spindle alignment is essential for tissue architecture and plays an important role in cell fate specification through asymmetric cell division. Spindle tethering factors such as Drosophila Mud (NuMA in mammals) are recruited to the cell cortex and capture astral microtubules, pulling the spindle in the correct orientation. However, how spindle tethering complexes read the cell polarity axis and how spindle attachment is coupled to mitotic progression remains poorly understood. We explore these questions in Drosophila sensory organ precursors (SOPs), which divide asymmetrically to give rise to epidermal mechanosensory bristles. We show that the scaffold protein Meru, which is enriched at the posterior cortex by the Frizzled/Dishevelled planar cell polarity complex, in turn recruits Mud, linking the spindle tethering and polarity machineries. Furthermore, Cyclin A/Cdk1 associates with Meru at the posterior cortex, promoting the formation of the Mud/Meru/Dsh complex via Meru and Dsh phosphorylation. Thus, Meru couples spindle orientation with cell polarity and provides a cell cycle-dependent cue for spindle tethering.
Collapse
Affiliation(s)
- Melissa M McLellan
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Birgit L Aerne
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jennifer J Banerjee Dhoul
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Maxine V Holder
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Tania Auchynnikava
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
2
|
Colombo S, Michel C, Speroni S, Ruhnow F, Gili M, Brito C, Surrey T. NuMA is a mitotic adaptor protein that activates dynein and connects it to microtubule minus ends. J Cell Biol 2025; 224:e202408118. [PMID: 39932518 PMCID: PMC11812572 DOI: 10.1083/jcb.202408118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/16/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Nuclear mitotic apparatus protein (NuMA) is indispensable for the mitotic functions of the major microtubule minus-end directed motor cytoplasmic dynein 1. NuMA and dynein are both essential for correct spindle pole organization. How these proteins cooperate to gather microtubule minus ends at spindle poles remains unclear. Here, we use microscopy-based in vitro reconstitutions to demonstrate that NuMA is a dynein adaptor, activating processive dynein motility together with dynein's cofactors dynactin and Lissencephaly-1 (Lis1). Additionally, we find that NuMA binds and stabilizes microtubule minus ends, allowing dynein/dynactin/NuMA to transport microtubule minus ends as cargo to other minus ends. We further show that the microtubule-nucleating γ-tubulin ring complex (γTuRC) hinders NuMA binding and that NuMA only caps minus ends of γTuRC-nucleated microtubules after γTuRC release. These results provide new mechanistic insight into how dynein, dynactin, NuMA, and Lis1 together with γTuRC and uncapping proteins cooperate to organize spindle poles in cells.
Collapse
Affiliation(s)
- Sabina Colombo
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Christel Michel
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Speroni
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Felix Ruhnow
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria Gili
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Cláudia Brito
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Thomas Surrey
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
3
|
Aslan M, d'Amico EA, Cho NH, Taheri A, Zhao Y, Zhong X, Blaauw M, Carter AP, Dumont S, Yildiz A. Structural and functional insights into activation and regulation of the dynein-dynactin-NuMA complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625568. [PMID: 39651296 PMCID: PMC11623564 DOI: 10.1101/2024.11.26.625568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
During cell division, NuMA orchestrates the focusing of microtubule minus-ends in spindle poles and cortical force generation on astral microtubules by interacting with dynein motors, microtubules, and other cellular factors. Here we used in vitro reconstitution, cryo-electron microscopy, and live cell imaging to understand the mechanism and regulation of NuMA. We determined the structure of the processive dynein/dynactin/NuMA complex (DDN) and showed that the NuMA N-terminus drives dynein motility in vitro and facilitates dynein-mediated transport in live cells. The C-terminus of NuMA directly binds to and suppresses the dynamics of the microtubule minus-end. Full-length NuMA is autoinhibited, but mitotically phosphorylated NuMA activates dynein in vitro and interphase cells. Together with dynein, activated full-length NuMA focuses microtubule minus-ends into aster-like structures. The binding of the cortical protein LGN to the NuMA C-terminus results in preferential binding of NuMA to the microtubule plus-end. These results provide critical insights into the activation of NuMA and dynein for their functions in the spindle body and the cell cortex.
Collapse
|
4
|
Neahring L, Cho NH, He Y, Liu G, Fernandes J, Rux CJ, Nakos K, Subramanian R, Upadhyayula S, Yildiz A, Dumont S. Torques within and outside the human spindle balance twist at anaphase. J Cell Biol 2024; 223:e202312046. [PMID: 38869473 PMCID: PMC11176257 DOI: 10.1083/jcb.202312046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
At each cell division, nanometer-scale motors and microtubules give rise to the micron-scale spindle. Many mitotic motors step helically around microtubules in vitro, and most are predicted to twist the spindle in a left-handed direction. However, the human spindle exhibits only slight global twist, raising the question of how these molecular torques are balanced. Here, we find that anaphase spindles in the epithelial cell line MCF10A have a high baseline twist, and we identify factors that both increase and decrease this twist. The midzone motors KIF4A and MKLP1 are together required for left-handed twist at anaphase, and we show that KIF4A generates left-handed torque in vitro. The actin cytoskeleton also contributes to left-handed twist, but dynein and its cortical recruitment factor LGN counteract it. Together, our work demonstrates that force generators regulate twist in opposite directions from both within and outside the spindle, preventing strong spindle twist during chromosome segregation.
Collapse
Affiliation(s)
- Lila Neahring
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Nathan H. Cho
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Tetrad Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Yifei He
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Gaoxiang Liu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Jonathan Fernandes
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Caleb J. Rux
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- UC Berkeley/UC San Francisco Graduate Group in Bioengineering, Berkeley, CA, USA
| | - Konstantinos Nakos
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ahmet Yildiz
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Physics Department, University of California Berkeley, Berkeley, CA, USA
| | - Sophie Dumont
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Tetrad Graduate Program, University of California San Francisco, San Francisco, CA, USA
- UC Berkeley/UC San Francisco Graduate Group in Bioengineering, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Neahring L, He Y, Cho NH, Liu G, Fernandes J, Rux CJ, Nakos K, Subramanian R, Upadhyayula S, Yildiz A, Dumont S. Torques within and outside the human spindle balance twist at anaphase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.10.570990. [PMID: 38405786 PMCID: PMC10888964 DOI: 10.1101/2023.12.10.570990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
At each cell division, nanometer-scale motors and microtubules give rise to the micron-scale spindle. Many mitotic motors step helically around microtubules in vitro, and most are predicted to twist the spindle in a left-handed direction. However, the human spindle exhibits only slight global twist, raising the question of how these molecular torques are balanced. Here, using lattice light sheet microscopy, we find that anaphase spindles in the epithelial cell line MCF10A have a high baseline twist, and we identify factors that both increase and decrease this twist. The midzone motors KIF4A and MKLP1 are redundantly required for left-handed twist at anaphase, and we show that KIF4A generates left-handed torque in vitro. The actin cytoskeleton also contributes to left-handed twist, but dynein and its cortical recruitment factor LGN counteract it. Together, our work demonstrates that force generators regulate twist in opposite directions from both within and outside the spindle, preventing strong spindle twist during chromosome segregation.
Collapse
Affiliation(s)
- Lila Neahring
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Developmental & Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Yifei He
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Nathan H. Cho
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Tetrad Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Gaoxiang Liu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Jonathan Fernandes
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Caleb J. Rux
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- UC Berkeley/UC San Francisco Graduate Group in Bioengineering, Berkeley, CA, USA
| | - Konstantinos Nakos
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ahmet Yildiz
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Physics Department, University of California Berkeley, Berkeley, CA, USA
| | - Sophie Dumont
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Developmental & Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Tetrad Graduate Program, University of California San Francisco, San Francisco, CA, USA
- UC Berkeley/UC San Francisco Graduate Group in Bioengineering, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Hu X, Xu Y, Wang C, Liu Y, Zhang L, Zhang J, Wang W, Chen Q, Liu H. Combined prediction and design reveals the target recognition mechanism of an intrinsically disordered protein interaction domain. Proc Natl Acad Sci U S A 2023; 120:e2305603120. [PMID: 37722056 PMCID: PMC10523638 DOI: 10.1073/pnas.2305603120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023] Open
Abstract
An increasing number of protein interaction domains and their targets are being found to be intrinsically disordered proteins (IDPs). The corresponding target recognition mechanisms are mostly elusive because of challenges in performing detailed structural analysis of highly dynamic IDP-IDP complexes. Here, we show that by combining recently developed computational approaches with experiments, the structure of the complex between the intrinsically disordered C-terminal domain (CTD) of protein 4.1G and its target IDP region in NuMA can be dissected at high resolution. First, we carry out systematic mutational scanning using dihydrofolate reductase-based protein complementarity analysis to identify essential interaction regions and key residues. The results are found to be highly consistent with an α/β-type complex structure predicted by AlphaFold2 (AF2). We then design mutants based on the predicted structure using a deep learning protein sequence design method. The solved crystal structure of one mutant presents the same core structure as predicted by AF2. Further computational prediction and experimental assessment indicate that the well-defined core structure is conserved across complexes of 4.1G CTD with other potential targets. Thus, we reveal that an intrinsically disordered protein interaction domain uses an α/β-type structure module formed through synergistic folding to recognize broad IDP targets. Moreover, we show that computational prediction and experiment can be jointly applied to segregate true IDP regions from the core structural domains of IDP-IDP complexes and to uncover the structure-dependent mechanisms of some otherwise elusive IDP-IDP interactions.
Collapse
Affiliation(s)
- Xiuhong Hu
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui230001, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Yang Xu
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui230001, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Chenchen Wang
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Yufeng Liu
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui230001, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Lu Zhang
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui230001, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Jiahai Zhang
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Wenning Wang
- Department of Chemistry, Institutes of Biomedical Sciences and Multiscale Research Institute of Complex Systems, Fudan University, Shanghai200438, China
| | - Quan Chen
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui230001, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui230027, China
| | - Haiyan Liu
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui230027, China
- School of Data Science, University of Science and Technology of China, Hefei, Anhui230027, China
| |
Collapse
|
7
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Myosin 10 uses its MyTH4 and FERM domains differentially to support two aspects of spindle pole biology required for mitotic spindle bipolarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545002. [PMID: 37398378 PMCID: PMC10312724 DOI: 10.1101/2023.06.15.545002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myosin 10 (Myo10) has the ability to link actin filaments to integrin-based adhesions and to microtubules by virtue of its integrin-binding FERM domain and microtubule-binding MyTH4 domain, respectively. Here we used Myo10 knockout cells to define Myo10's contribution to the maintenance of spindle bipolarity, and complementation to quantitate the relative contributions of its MyTH4 and FERM domains. Myo10 knockout HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in knockout MEFs and knockout HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates γ-tubulin-positive acentriolar foci that serve as additional spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both integrins and microtubules to promote PCM/pole integrity. Conversely, Myo10's ability to promote the clustering of supernumerary centrosomes only requires that it interact with integrins. Importantly, images of Halo-Myo10 knock-in cells show that the myosin localizes exclusively within adhesive retraction fibers during mitosis. Based on these and other results, we conclude that Myo10 promotes PCM/pole integrity at a distance, and that it facilitates supernumerary centrosome clustering by promoting retraction fiber-based cell adhesion, which likely provides an anchor for the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
8
|
Neville KE, Finegan TM, Lowe N, Bellomio PM, Na D, Bergstralh DT. The Drosophila mitotic spindle orientation machinery requires activation, not just localization. EMBO Rep 2023; 24:e56074. [PMID: 36629398 PMCID: PMC9986814 DOI: 10.15252/embr.202256074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
The orientation of the mitotic spindle at metaphase determines the placement of the daughter cells. Spindle orientation in animals typically relies on an evolutionarily conserved biological machine comprised of at least four proteins - called Pins, Gαi, Mud, and Dynein in flies - that exerts a pulling force on astral microtubules and reels the spindle into alignment. The canonical model for spindle orientation holds that the direction of pulling is determined by asymmetric placement of this machinery at the cell cortex. In most cell types, this placement is thought to be mediated by Pins, and a substantial body of literature is therefore devoted to identifying polarized cues that govern localized cortical enrichment of Pins. In this study we revisit the canonical model and find that it is incomplete. Spindle orientation in the Drosophila follicular epithelium and embryonic ectoderm requires not only Pins localization but also direct interaction between Pins and the multifunctional protein Discs large. This requirement can be over-ridden by interaction with another Pins interacting protein, Inscuteable.
Collapse
Affiliation(s)
| | - Tara M Finegan
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Nicholas Lowe
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | | | - Daxiang Na
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Dan T Bergstralh
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
- Department of Physics & AstronomyUniversity of RochesterRochesterNew YorkUSA
- Department of Biomedical GeneticsUniversity of Rochester Medical CenterRochesterNew YorkUSA
| |
Collapse
|
9
|
Sana S, Rajeevan A, Kotak S. Membrane compartmentalization of Ect2/Cyk4/Mklp1 and NuMA/dynein regulates cleavage furrow formation. J Biophys Biochem Cytol 2022; 221:213522. [PMID: 36197340 PMCID: PMC9539458 DOI: 10.1083/jcb.202203127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/09/2022] [Accepted: 09/02/2022] [Indexed: 12/13/2022] Open
Abstract
In animal cells, spindle elongation during anaphase is temporally coupled with cleavage furrow formation. Spindle elongation during anaphase is regulated by NuMA/dynein/dynactin complexes that occupy the polar region of the cell membrane and are excluded from the equatorial membrane. How NuMA/dynein/dynactin are excluded from the equatorial membrane and the biological significance of this exclusion remains unknown. Here, we show that the centralspindlin (Cyk4/Mklp1) and its interacting partner RhoGEF Ect2 are required for NuMA/dynein/dynactin exclusion from the equatorial cell membrane. The Ect2-based (Ect2/Cyk4/Mklp1) and NuMA-based (NuMA/dynein/dynactin) complexes occupy mutually exclusive membrane surfaces during anaphase. The equatorial membrane enrichment of Ect2-based complexes is essential for NuMA/dynein/dynactin exclusion and proper spindle elongation. Conversely, NuMA-based complexes at the polar region of the cell membrane ensure spatially confined localization of Ect2-based complexes and thus RhoA. Overall, our work establishes that membrane compartmentalization of NuMA-based and Ect2-based complexes at the two distinct cell surfaces restricts dynein/dynactin and RhoA for coordinating spindle elongation with cleavage furrow formation.
Collapse
Affiliation(s)
- Shrividya Sana
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India
| | - Ashwathi Rajeevan
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India,Correspondence to Sachin Kotak:
| |
Collapse
|
10
|
Tarannum N, Singh R, Woolner S. Sculpting an Embryo: The Interplay between Mechanical Force and Cell Division. J Dev Biol 2022; 10:37. [PMID: 36135370 PMCID: PMC9502278 DOI: 10.3390/jdb10030037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022] Open
Abstract
The journey from a single fertilised cell to a multicellular organism is, at the most fundamental level, orchestrated by mitotic cell divisions. Both the rate and the orientation of cell divisions are important in ensuring the proper development of an embryo. Simultaneous with cell proliferation, embryonic cells constantly experience a wide range of mechanical forces from their surrounding tissue environment. Cells must be able to read and respond correctly to these forces since they are known to affect a multitude of biological functions, including cell divisions. The interplay between the mechanical environment and cell divisions is particularly crucial during embryogenesis when tissues undergo dynamic changes in their shape, architecture, and overall organisation to generate functional tissues and organs. Here we review our current understanding of the cellular mechanisms by which mechanical force regulates cell division and place this knowledge within the context of embryogenesis and tissue morphogenesis.
Collapse
Affiliation(s)
- Nawseen Tarannum
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | | - Sarah Woolner
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
11
|
Darnat P, Burg A, Sallé J, Lacoste J, Louvet-Vallée S, Gho M, Audibert A. Cortical Cyclin A controls spindle orientation during asymmetric cell divisions in Drosophila. Nat Commun 2022; 13:2723. [PMID: 35581185 PMCID: PMC9114397 DOI: 10.1038/s41467-022-30182-1] [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: 04/21/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
The coordination between cell proliferation and cell polarity is crucial to orient the asymmetric cell divisions to generate cell diversity in epithelia. In many instances, the Frizzled/Dishevelled planar cell polarity pathway is involved in mitotic spindle orientation, but how this is spatially and temporally coordinated with cell cycle progression has remained elusive. Using Drosophila sensory organ precursor cells as a model system, we show that Cyclin A, the main Cyclin driving the transition to M-phase of the cell cycle, is recruited to the apical-posterior cortex in prophase by the Frizzled/Dishevelled complex. This cortically localized Cyclin A then regulates the orientation of the division by recruiting Mud, a homologue of NuMA, the well-known spindle-associated protein. The observed non-canonical subcellular localization of Cyclin A reveals this mitotic factor as a direct link between cell proliferation, cell polarity and spindle orientation. The Frizzled/Dishevelled planar cell polarity pathway is involved in mitotic spindle orientation, but how this is coordinated with the cell cycle is unclear. Here, the authors show with Drosophila sensory organ precursor cells that Cyclin A is recruited in prophase by Frizzled/Dishevelled, regulating division orientation.
Collapse
Affiliation(s)
- Pénélope Darnat
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Angélique Burg
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Jérémy Sallé
- Institut Jacques Monod, Université Paris Diderot/CNRS, Cellular Spatial Organization Team, F-75005, Paris, France
| | - Jérôme Lacoste
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Sophie Louvet-Vallée
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Michel Gho
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France.
| | - Agnès Audibert
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France.
| |
Collapse
|
12
|
Donà F, Eli S, Mapelli M. Insights Into Mechanisms of Oriented Division From Studies in 3D Cellular Models. Front Cell Dev Biol 2022; 10:847801. [PMID: 35356279 PMCID: PMC8959941 DOI: 10.3389/fcell.2022.847801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs, that ensure the correct organ formation and functioning. In these processes, mitotic rates and division orientation are crucial in regulating the velocity and the timing of the forming tissue. Division orientation, specified by mitotic spindle placement with respect to epithelial apico-basal polarity, controls not only the partitioning of cellular components but also the positioning of the daughter cells within the tissue, and hence the contacts that daughter cells retain with the surrounding microenvironment. Daughter cells positioning is important to determine signal sensing and fate, and therefore the final function of the developing organ. In this review, we will discuss recent discoveries regarding the mechanistics of planar divisions in mammalian epithelial cells, summarizing technologies and model systems used to study oriented cell divisions in vitro such as three-dimensional cysts of immortalized cells and intestinal organoids. We also highlight how misorientation is corrected in vivo and in vitro, and how it might contribute to the onset of pathological conditions.
Collapse
Affiliation(s)
- Federico Donà
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Susanna Eli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
13
|
Wang D, Wu S, Wang D, Song X, Yang M, Zhang W, Huang S, Weng J, Liu Z, Wang W. The importance of the compact disordered state in the fuzzy interactions between intrinsically disordered proteins. Chem Sci 2022; 13:2363-2377. [PMID: 35310482 PMCID: PMC8864705 DOI: 10.1039/d1sc06825c] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Abstract
The intrinsically disordered C-terminal domain (CTD) of protein 4.1G is able to specifically bind a 26-residue intrinsically disordered region of NuMA, forming a dynamic fuzzy complex. As one of a few cases of extremely fuzzy interactions between two intrinsically disordered proteins/regions (IDPs/IDRs) without induced folding, the principle of the binding is unknown. Here, we combined experimental and computational methods to explore the detailed mechanism of the interaction between 4.1G-CTD and NuMA. MD simulations suggest that the kinetic hub states in the structure ensemble of 4.1G-CTD are favorable in the fuzzy complex. The feature of these hub states is that the binding 'hot spot' motifs βA and βB exhibit β strand propensities and are well packed to each other. The binding between 4.1G-CTD and NuMA is disrupted at low pH, which changes the intramolecular packing of 4.1G-CTD and weakens the packing between βA and βB motifs. Low pH conditions also lead to increased hydrodynamic radius and acceleration of backbone dynamics of 4.1G-CTD. All these results underscore the importance of tertiary structural arrangements and overall compactness of 4.1G-CTD in its binding to NuMA, i.e. the compact disordered state of 4.1G-CTD is crucial for binding. Different from the short linear motifs (SLiMs) that are often found to mediate IDP interactions, 4.1G-CTD functions as an intrinsically disordered domain (IDD), which is a functional and structural unit similar to conventional protein domains. This work sheds light on the molecular recognition mechanism of IDPs/IDRs and expands the conventional structure-function paradigm in protein biochemistry.
Collapse
Affiliation(s)
- Dan Wang
- Department of Chemistry, Multiscale Research Institute of Complex Systems and Institute of Biomedical Sciences, Fudan University Shanghai 200438 China
| | - Shaowen Wu
- Guangdong Key Laboratory for Crop Germplasm Resources Preservation and Utilization, Agro-biological Gene Research Center, Guangdong Academy of Agricultural Sciences Guangzhou 510640 Guangdong China
| | | | - Xingyu Song
- Department of Chemistry, Multiscale Research Institute of Complex Systems and Institute of Biomedical Sciences, Fudan University Shanghai 200438 China
| | - Maohua Yang
- Department of Chemistry, Multiscale Research Institute of Complex Systems and Institute of Biomedical Sciences, Fudan University Shanghai 200438 China
| | - Wolun Zhang
- LightEdge Technologies Limited Zhongshan 528403 China
| | - Shaohui Huang
- Institute of Biophysics, Chinese Academy of Sciences Beijing 100101 China
- University of Chinese Academy of Science Beijing 101408 China
| | - Jingwei Weng
- Department of Chemistry, Multiscale Research Institute of Complex Systems and Institute of Biomedical Sciences, Fudan University Shanghai 200438 China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences Shanghai 201210 China
| | - Wenning Wang
- Department of Chemistry, Multiscale Research Institute of Complex Systems and Institute of Biomedical Sciences, Fudan University Shanghai 200438 China
| |
Collapse
|
14
|
Moreci RS, Lechler T. KIF18B is a cell type-specific regulator of spindle orientation in the epidermis. Mol Biol Cell 2021; 32:ar29. [PMID: 34432485 PMCID: PMC8693959 DOI: 10.1091/mbc.e21-06-0291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023] Open
Abstract
Proper spindle orientation is required for asymmetric cell division and the establishment of complex tissue architecture. In the developing epidermis, spindle orientation requires a conserved cortical protein complex of LGN/NuMA/dynein-dynactin. However, how microtubule dynamics are regulated to interact with this machinery and properly position the mitotic spindle is not fully understood. Furthermore, our understanding of the processes that link spindle orientation during asymmetric cell division to cell fate specification in distinct tissue contexts remains incomplete. We report a role for the microtubule catastrophe factor KIF18B in regulating microtubule dynamics to promote spindle orientation in keratinocytes. During mitosis, KIF18B accumulates at the cell cortex, colocalizing with the conserved spindle orientation machinery. In vivo we find that KIF18B is required for oriented cell divisions within the hair placode, the first stage of hair follicle morphogenesis, but is not essential in the interfollicular epidermis. Disrupting spindle orientation in the placode, using mutations in either KIF18B or NuMA, results in aberrant cell fate marker expression of hair follicle progenitor cells. These data functionally link spindle orientation to cell fate decisions during hair follicle morphogenesis. Taken together, our data demonstrate a role for regulated microtubule dynamics in spindle orientation in epidermal cells. This work also highlights the importance of spindle orientation during asymmetric cell division to dictate cell fate specification.
Collapse
Affiliation(s)
- Rebecca S. Moreci
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| | - Terry Lechler
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| |
Collapse
|
15
|
Lechler T, Mapelli M. Spindle positioning and its impact on vertebrate tissue architecture and cell fate. Nat Rev Mol Cell Biol 2021; 22:691-708. [PMID: 34158639 PMCID: PMC10544824 DOI: 10.1038/s41580-021-00384-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/18/2022]
Abstract
In multicellular systems, oriented cell divisions are essential for morphogenesis and homeostasis as they determine the position of daughter cells within the tissue and also, in many cases, their fate. Early studies in invertebrates led to the identification of conserved core mechanisms of mitotic spindle positioning centred on the Gαi-LGN-NuMA-dynein complex. In recent years, much has been learnt about the way this complex functions in vertebrate cells. In particular, studies addressed how the Gαi-LGN-NuMA-dynein complex dynamically crosstalks with astral microtubules and the actin cytoskeleton, and how it is regulated to orient the spindle according to cellular and tissue-wide cues. We have also begun to understand how dynein motors and actin regulators interact with mechanosensitive adhesion molecules sensing extracellular mechanical stimuli, such as cadherins and integrins, and with signalling pathways so as to respond to extracellular cues instructing the orientation of the division axis in vivo. In this Review, with the focus on epithelial tissues, we discuss the molecular mechanisms of mitotic spindle orientation in vertebrate cells, and how this machinery is regulated by epithelial cues and extracellular signals to maintain tissue cohesiveness during mitosis. We also outline recent knowledge of how spindle orientation impacts tissue architecture in epithelia and its emerging links to the regulation of cell fate decisions. Finally, we describe how defective spindle orientation can be corrected or its effects eliminated in tissues under physiological conditions, and the pathological implications associated with spindle misorientation.
Collapse
Affiliation(s)
- Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
16
|
Yang Q, Liu J, Wang Z. 4.1N-Mediated Interactions and Functions in Nerve System and Cancer. Front Mol Biosci 2021; 8:711302. [PMID: 34589518 PMCID: PMC8473747 DOI: 10.3389/fmolb.2021.711302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/16/2021] [Indexed: 01/05/2023] Open
Abstract
Scaffolding protein 4.1N is a neuron-enriched 4.1 homologue. 4.1N contains three conserved domains, including the N-terminal 4.1-ezrin-radixin-moesin (FERM) domain, internal spectrin–actin–binding (SAB) domain, and C-terminal domain (CTD). Interspersed between the three domains are nonconserved domains, including U1, U2, and U3. The role of 4.1N was first reported in the nerve system. Then, extensive studies reported the role of 4.1N in cancers and other diseases. 4.1N performs numerous vital functions in signaling transduction by interacting, locating, supporting, and coordinating different partners and is involved in the molecular pathogenesis of various diseases. In this review, recent studies on the interactions between 4.1N and its contactors (including the α7AChr, IP3R1, GluR1/4, GluK1/2/3, mGluR8, KCC2, D2/3Rs, CASK, NuMA, PIKE, IP6K2, CAM 1/3, βII spectrin, flotillin-1, pp1, and 14-3-3) and the 4.1N-related biological functions in the nerve system and cancers are specifically and comprehensively discussed. This review provides critical detailed mechanistic insights into the role of 4.1N in disease relationships.
Collapse
Affiliation(s)
- Qin Yang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,School of Medical Laboratory, Shao Yang University, Shaoyang, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zi Wang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
17
|
Vukušić K, Tolić IM. Anaphase B: Long-standing models meet new concepts. Semin Cell Dev Biol 2021; 117:127-139. [PMID: 33849764 PMCID: PMC8406420 DOI: 10.1016/j.semcdb.2021.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022]
Abstract
Mitotic cell divisions ensure stable transmission of genetic information from a mother to daughter cells in a series of generations. To ensure this crucial task is accomplished, the cell forms a bipolar structure called the mitotic spindle that divides sister chromatids to the opposite sides of the dividing mother cell. After successful establishment of stable attachments of microtubules to chromosomes and inspection of connections between them, at the heart of mitosis, the cell starts the process of segregation. This spectacular moment in the life of a cell is termed anaphase, and it involves two distinct processes: depolymerization of microtubules bound to chromosomes, which is also known as anaphase A, and elongation of the spindle or anaphase B. Both processes ensure physical separation of disjointed sister chromatids. In this chapter, we review the mechanisms of anaphase B spindle elongation primarily in mammalian systems, combining different pioneering ideas and concepts with more recent findings that shed new light on the force generation and regulation of biochemical modules operating during spindle elongation. Finally, we present a comprehensive model of spindle elongation that includes structural, biophysical, and molecular aspects of anaphase B.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
18
|
Huang SC, Vu LV, Yu FH, Nguyen DT, Benz EJ. Multifunctional protein 4.1R regulates the asymmetric segregation of Numb during terminal erythroid maturation. J Biol Chem 2021; 297:101051. [PMID: 34364872 PMCID: PMC8408529 DOI: 10.1016/j.jbc.2021.101051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 10/25/2022] Open
Abstract
The asymmetric cell division of stem or progenitor cells generates daughter cells with distinct fates that balance proliferation and differentiation. Asymmetric segregation of Notch signaling regulatory protein Numb plays a crucial role in cell diversification. However, the molecular mechanism remains unclear. Here, we examined the unequal distribution of Numb in the daughter cells of murine erythroleukemia cells (MELCs) that undergo DMSO-induced erythroid differentiation. In contrast to the cytoplasmic localization of Numb during uninduced cell division, Numb is concentrated at the cell boundary in interphase, near the one-spindle pole in metaphase, and is unequally distributed to one daughter cell in anaphase in induced cells. The inheritance of Numb guides this daughter cell toward erythroid differentiation while the other cell remains a progenitor cell. Mitotic spindle orientation, critical for distribution of cell fate determinants, requires complex communication between the spindle microtubules and the cell cortex mediated by the NuMA-LGN-dynein/dynactin complex. Depletion of each individual member of the complex randomizes the position of Numb relative to the mitotic spindle. Gene replacement confirms that multifunctional erythrocyte protein 4.1R (4.1R) functions as a member of the NuMA-LGN-dynein/dynactin complex and is necessary for regulating spindle orientation, in which interaction between 4.1R and NuMA plays an important role. These results suggest that mispositioning of Numb is the result of spindle misorientation. Finally, disruption of the 4.1R-NuMA-LGN complex increases Notch signaling and decreases the erythroblast population. Together, our results identify a critical role for 4.1R in regulating the asymmetric segregation of Numb to mediate erythropoiesis.
Collapse
Affiliation(s)
- Shu-Ching Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| | - Long V Vu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Faye H Yu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Dan T Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Edward J Benz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts, USA; Leukemia Program, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Aquino Perez C, Burocziova M, Jenikova G, Macurek L. CK1-mediated phosphorylation of FAM110A promotes its interaction with mitotic spindle and controls chromosomal alignment. EMBO Rep 2021; 22:e51847. [PMID: 34080749 DOI: 10.15252/embr.202051847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
Progression through the cell cycle is driven by cyclin-dependent kinases that control gene expression, orchestration of mitotic spindle, and cell division. To identify new regulators of the cell cycle, we performed transcriptomic analysis of human non-transformed cells expressing a fluorescent ubiquitination-based cell cycle indicator and identified 701 transcripts differentially expressed in G1 and G2 cells. Family with sequence similarity 110 member A (FAM110A) protein is highly expressed in G2 cells and localized at mitotic spindle and spindle poles during mitosis. Depletion of FAM110A impairs chromosomal alignment, delays metaphase-to-anaphase transition, and affects spindle positioning. Using mass spectrometry and immunoprecipitation, we identified casein kinase I (CK1) in complex with FAM110A during mitosis. CK1 phosphorylates the C-terminal domain of FAM110A in vitro, and inhibition of CK1 reduces phosphorylation of mitotic FAM110A. Wild-type FAM110A, but not the FAM110A-S252-S255A mutant deficient in CK1 phosphorylation, rescues the chromosomal alignment, duration of mitosis, and orientation of the mitotic spindle after depletion of endogenous FAM110A. We propose that CK1 regulates chromosomal alignment by phosphorylating FAM110A and promoting its interaction with mitotic spindle.
Collapse
Affiliation(s)
- Cecilia Aquino Perez
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Monika Burocziova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Gabriela Jenikova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
20
|
Kiyomitsu T, Boerner S. The Nuclear Mitotic Apparatus (NuMA) Protein: A Key Player for Nuclear Formation, Spindle Assembly, and Spindle Positioning. Front Cell Dev Biol 2021; 9:653801. [PMID: 33869212 PMCID: PMC8047419 DOI: 10.3389/fcell.2021.653801] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/10/2021] [Indexed: 01/10/2023] Open
Abstract
The nuclear mitotic apparatus (NuMA) protein is well conserved in vertebrates, and dynamically changes its subcellular localization from the interphase nucleus to the mitotic/meiotic spindle poles and the mitotic cell cortex. At these locations, NuMA acts as a key structural hub in nuclear formation, spindle assembly, and mitotic spindle positioning, respectively. To achieve its variable functions, NuMA interacts with multiple factors, including DNA, microtubules, the plasma membrane, importins, and cytoplasmic dynein. The binding of NuMA to dynein via its N-terminal domain drives spindle pole focusing and spindle positioning, while multiple interactions through its C-terminal region define its subcellular localizations and functions. In addition, NuMA can self-assemble into high-ordered structures which likely contribute to spindle positioning and nuclear formation. In this review, we summarize recent advances in NuMA’s domains, functions and regulations, with a focus on human NuMA, to understand how and why vertebrate NuMA participates in these functions in comparison with invertebrate NuMA-related proteins.
Collapse
Affiliation(s)
- Tomomi Kiyomitsu
- Cell Division Dynamics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Susan Boerner
- Cell Division Dynamics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| |
Collapse
|
21
|
Polverino F, Naso FD, Asteriti IA, Palmerini V, Singh D, Valente D, Bird AW, Rosa A, Mapelli M, Guarguaglini G. The Aurora-A/TPX2 Axis Directs Spindle Orientation in Adherent Human Cells by Regulating NuMA and Microtubule Stability. Curr Biol 2020; 31:658-667.e5. [PMID: 33275894 DOI: 10.1016/j.cub.2020.10.096] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/16/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022]
Abstract
Mitotic spindle orientation is a crucial process that defines the axis of cell division, contributing to daughter cell positioning and fate, and hence to tissue morphogenesis and homeostasis.1,2 The trimeric NuMA/LGN/Gαi complex, the major determinant of spindle orientation, exerts pulling forces on the spindle poles by anchoring astral microtubules (MTs) and dynein motors to the cell cortex.3,4 Mitotic kinases contribute to correct spindle orientation by regulating nuclear mitotic apparatus protein (NuMA) localization,5-7 among which the Aurora-A centrosomal kinase regulates NuMA targeting to the cell cortex in metaphase.8,9 Aurora-A and its activator targeting protein for Xklp2 (TPX2) are frequently overexpressed in cancer,10-12 raising the question as to whether spindle orientation is among the processes downstream the Aurora-A/TPX2 signaling axis altered under pathological conditions. Here, we investigated the role of TPX2 in the Aurora-A- and NuMA-dependent spindle orientation. We show that, in cultured adherent human cells, the interaction with TPX2 is required for Aurora-A to exert this function. We also show that Aurora-A, TPX2, and NuMA are part of a complex at spindle MTs, where TPX2 acts as a platform for Aurora-A regulation of NuMA. Interestingly, excess TPX2 does not influence NuMA localization but induces a "super-alignment" of the spindle axis with respect to the substrate, although an excess of Aurora-A induces spindle misorientation. These opposite effects are both linked to altered MT stability. Overall, our results highlight the importance of TPX2 for spindle orientation and suggest that spindle orientation is differentially sensitive to unbalanced levels of Aurora-A, TPX2, or the Aurora-A/TPX2 complex.
Collapse
Affiliation(s)
- Federica Polverino
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy; Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy
| | - Francesco D Naso
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Italia A Asteriti
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Valentina Palmerini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy
| | - Divya Singh
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Davide Valente
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Alexander W Bird
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Alessandro Rosa
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; Department of Biology and Biotechnology "C. Darwin," Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marina Mapelli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy.
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy.
| |
Collapse
|
22
|
Rajeevan A, Keshri R, Kapoor S, Kotak S. NuMA interaction with chromatin is vital for proper chromosome decondensation at the mitotic exit. Mol Biol Cell 2020; 31:2437-2451. [PMID: 32845810 PMCID: PMC7851854 DOI: 10.1091/mbc.e20-06-0415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
NuMA is an abundant long coiled-coil protein that plays a prominent role in spindle organization during mitosis. In interphase, NuMA is localized to the nucleus and hypothesized to control gene expression and chromatin organization. However, because of the prominent mitotic phenotype upon NuMA loss, its precise function in the interphase nucleus remains elusive. Here, we report that NuMA is associated with chromatin in interphase and prophase but released upon nuclear envelope breakdown (NEBD) by the action of Cdk1. We uncover that NuMA directly interacts with DNA via evolutionarily conserved sequences in its C-terminus. Notably, the expression of the DNA-binding-deficient mutant of NuMA affects chromatin decondensation at the mitotic exit, and nuclear shape in interphase. We show that the nuclear shape defects observed upon mutant NuMA expression are due to its potential to polymerize into higher-order fibrillar structures. Overall, this work establishes the spindle-independent function of NuMA in choreographing proper chromatin decompaction and nuclear shape by directly associating with the DNA.
Collapse
Affiliation(s)
- Ashwathi Rajeevan
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Riya Keshri
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Sukriti Kapoor
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| |
Collapse
|
23
|
Mohamadalizadeh-Hanjani Z, Shahbazi S, Geranpayeh L. Investigation of the SPAG5 gene expression and amplification related to the NuMA mRNA levels in breast ductal carcinoma. World J Surg Oncol 2020; 18:225. [PMID: 32838814 PMCID: PMC7445913 DOI: 10.1186/s12957-020-02001-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
Background The cell proliferative markers are very important in breast cancer. Since SPAG5 and NuMA proteins play a significant role in the mitosis regulatory network and cell division, we aimed to study their mRNA levels as well as SPAG5 gene amplification correlated to clinicopathological status in ductal carcinoma of the breast. Methods SPAG5 and NuMA gene expressions were investigated in 40 breast cancer tissues and normal adjacent tissues via real-time PCR. PUM1 was selected as the reference gene. QMF PCR method was applied to study SPAG5 gene amplification and AGBL2, BOD1L, and POR were designated as internal control genes. Gene amplification was determined by calculating a dosage quotient for each DNA fragment. Results Increased SPAG5 mRNA expression was detected in breast cancer tissues (p = 0.005) and related to tumor size. No significant difference was observed between NuMA gene expression level in tumor tissue and the normal adjacent tissue (p = 0.56). However, we observed that NuMA expression was significantly increased in ER-positive tumor tissues. There was no clear correlation pattern between SPAG5 and NuMA mRNA levels (r = 0.33). Seventeen percent of tissues showed complete amplification in SPAG5 gene fragments. Conclusion Our results were consistent with the previous publications regarding SPAG5 gene expression and amplification in breast cancer with an emphasis on the prominent role of this protein in tumor pathogenesis. Our results failed to yield any correlation between SPAG5 and NuMA mRNA levels which implies independence of these genes in breast cancer pathogenesis.
Collapse
Affiliation(s)
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Loabat Geranpayeh
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Keshri R, Rajeevan A, Kotak S. PP2A--B55γ counteracts Cdk1 and regulates proper spindle orientation through the cortical dynein adaptor NuMA. J Cell Sci 2020; 133:jcs243857. [PMID: 32591484 PMCID: PMC7406356 DOI: 10.1242/jcs.243857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
Proper orientation of the mitotic spindle is critical for accurate development and morphogenesis. In human cells, spindle orientation is regulated by the evolutionarily conserved protein NuMA, which interacts with dynein and enriches it at the cell cortex. Pulling forces generated by cortical dynein orient the mitotic spindle. Cdk1-mediated phosphorylation of NuMA at threonine 2055 (T2055) negatively regulates its cortical localization. Thus, only NuMA not phosphorylated at T2055 localizes at the cell cortex. However, the identity and the mechanism of action of the phosphatase complex involved in T2055 dephosphorylation remains elusive. Here, we characterized the PPP2CA-B55γ (PPP2R2C)-PPP2R1B complex that counteracts Cdk1 to orchestrate cortical NuMA for proper spindle orientation. In vitro reconstitution experiments revealed that this complex is sufficient for T2055 dephosphorylation. Importantly, we identified polybasic residues in NuMA that are critical for T2055 dephosphorylation, and for maintaining appropriate cortical NuMA levels for accurate spindle elongation. Furthermore, we found that Cdk1-mediated phosphorylation and PP2A-B55γ-mediated dephosphorylation at T2055 are reversible events. Altogether, this study uncovers a novel mechanism by which Cdk1 and its counteracting PP2A-B55γ complex orchestrate spatiotemporal levels of cortical force generators for flawless mitosis.
Collapse
Affiliation(s)
- Riya Keshri
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Ashwathi Rajeevan
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| |
Collapse
|
25
|
Sharma A, Dagar S, Mylavarapu SVS. Transgelin-2 and phosphoregulation of the LIC2 subunit of dynein govern mitotic spindle orientation. J Cell Sci 2020; 133:jcs239673. [PMID: 32467330 DOI: 10.1242/jcs.239673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/29/2020] [Indexed: 08/31/2023] Open
Abstract
The molecular motor dynein is essential for mitotic spindle orientation, which defines the axis of cell division. The light intermediate chain subunits, LIC1 and LIC2, define biochemically and functionally distinct vertebrate dynein complexes, with LIC2-dynein playing a crucial role in ensuring spindle orientation. We reveal a novel, mitosis-specific interaction of LIC2-dynein with the cortical actin-bundling protein transgelin-2. Transgelin-2 is required for maintaining proper spindle length, equatorial metaphase chromosome alignment, spindle orientation and timely anaphase onset. We show that transgelin-2 stabilizes the cortical recruitment of LGN-NuMA, which together with dynein is required for spindle orientation. The opposing actions of transgelin-2 and LIC2-dynein maintain optimal cortical levels of LGN-NuMA. In addition, we show that the highly conserved serine 194 phosphorylation of LIC2 is required for proper spindle orientation, by maintaining mitotic centrosome integrity to ensure optimal astral microtubule nucleation. The work reveals two specific mechanisms through which LIC2-dynein regulates mitotic spindle orientation; namely, through a new interactor transgelin-2, which is required for engagement of LGN-NuMA with the actin cortex, and through mitotic phosphoregulation of LIC2 to control microtubule nucleation from the poles.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Amit Sharma
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sunayana Dagar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
26
|
Renna C, Rizzelli F, Carminati M, Gaddoni C, Pirovano L, Cecatiello V, Pasqualato S, Mapelli M. Organizational Principles of the NuMA-Dynein Interaction Interface and Implications for Mitotic Spindle Functions. Structure 2020; 28:820-829.e6. [PMID: 32413290 DOI: 10.1016/j.str.2020.04.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/18/2020] [Accepted: 04/22/2020] [Indexed: 01/01/2023]
Abstract
Mitotic progression is orchestrated by the microtubule-based motor dynein, which sustains all mitotic spindle functions. During cell division, cytoplasmic dynein acts with the high-molecular-weight complex dynactin and nuclear mitotic apparatus (NuMA) to organize and position the spindle. Here, we analyze the interaction interface between NuMA and the light intermediate chain (LIC) of eukaryotic dynein. Structural studies show that NuMA contains a hook domain contacting directly LIC1 and LIC2 chains through a conserved hydrophobic patch shared among other Hook adaptors. In addition, we identify a LIC-binding motif within the coiled-coil region of NuMA that is homologous to CC1-boxes. Analysis of mitotic cells revealed that both LIC-binding sites of NuMA are essential for correct spindle placement and cell division. Collectively, our evidence depicts NuMA as the dynein-activating adaptor acting in the mitotic processes of spindle organization and positioning.
Collapse
Affiliation(s)
- Cristina Renna
- IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | | | | | - Chiara Gaddoni
- IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Laura Pirovano
- IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | | | | | - Marina Mapelli
- IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| |
Collapse
|
27
|
Decoupling the Roles of Cell Shape and Mechanical Stress in Orienting and Cueing Epithelial Mitosis. Cell Rep 2020; 26:2088-2100.e4. [PMID: 30784591 PMCID: PMC6381790 DOI: 10.1016/j.celrep.2019.01.102] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/11/2018] [Accepted: 01/28/2019] [Indexed: 01/08/2023] Open
Abstract
Distinct mechanisms involving cell shape and mechanical force are known to influence the rate and orientation of division in cultured cells. However, uncoupling the impact of shape and force in tissues remains challenging. Combining stretching of Xenopus tissue with mathematical methods of inferring relative mechanical stress, we find separate roles for cell shape and mechanical stress in orienting and cueing division. We demonstrate that division orientation is best predicted by an axis of cell shape defined by the position of tricellular junctions (TCJs), which align with local cell stress rather than tissue-level stress. The alignment of division to cell shape requires functional cadherin and the localization of the spindle orientation protein, LGN, to TCJs but is not sensitive to relative cell stress magnitude. In contrast, proliferation rate is more directly regulated by mechanical stress, being correlated with relative isotropic stress and decoupled from cell shape when myosin II is depleted. Tissue stretching increases division rate and reorients divisions with stretch Division orientation is regulated by cell shape defined by tricellular junctions Cadherin and LGN localize to tricellular junctions aligning division to cell shape Division rate is linked to mechanical stress and can be decoupled from cell shape
Collapse
|
28
|
Rizzelli F, Malabarba MG, Sigismund S, Mapelli M. The crosstalk between microtubules, actin and membranes shapes cell division. Open Biol 2020; 10:190314. [PMID: 32183618 PMCID: PMC7125961 DOI: 10.1098/rsob.190314] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Mitotic progression is orchestrated by morphological and mechanical changes promoted by the coordinated activities of the microtubule (MT) cytoskeleton, the actin cytoskeleton and the plasma membrane (PM). MTs assemble the mitotic spindle, which assists sister chromatid separation, and contact the rigid and tensile actomyosin cortex rounded-up underneath the PM. Here, we highlight the dynamic crosstalk between MTs, actin and cell membranes during mitosis, and discuss the molecular connections between them. We also summarize recent views on how MT traction forces, the actomyosin cortex and membrane trafficking contribute to spindle positioning in isolated cells in culture and in epithelial sheets. Finally, we describe the emerging role of membrane trafficking in synchronizing actomyosin tension and cell shape changes with cell-substrate adhesion, cell-cell contacts and extracellular signalling events regulating proliferation.
Collapse
Affiliation(s)
| | - Maria Grazia Malabarba
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | - Sara Sigismund
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
29
|
Takayanagi H, Hayase J, Kamakura S, Miyano K, Chishiki K, Yuzawa S, Sumimoto H. Intramolecular interaction in LGN, an adaptor protein that regulates mitotic spindle orientation. J Biol Chem 2019; 294:19655-19666. [PMID: 31732560 DOI: 10.1074/jbc.ra119.011457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/07/2019] [Indexed: 12/11/2022] Open
Abstract
Proper mitotic spindle orientation requires that astral microtubules are connected to the cell cortex by the microtubule-binding protein NuMA, which is recruited from the cytoplasm. Cortical recruitment of NuMA is at least partially mediated via direct binding to the adaptor protein LGN. LGN normally adopts a closed conformation via an intramolecular interaction between its N-terminal NuMA-binding domain and its C-terminal region that contains four GoLoco (GL) motifs, each capable of binding to the membrane-anchored Gαi subunit of heterotrimeric G protein. Here we show that the intramolecular association with the N-terminal domain in LGN involves GL3, GL4, and a region between GL2 and GL3, whereas GL1 and GL2 do not play a major role. This conformation renders GL1 but not the other GL motifs in a state easily accessible to Gαi To interact with full-length LGN in a closed state, NuMA requires the presence of Gαi; both NuMA and Gαi are essential for cortical recruitment of LGN in mitotic cells. In contrast, mInsc, a protein that competes with NuMA for binding to LGN and regulates mitotic spindle orientation in asymmetric cell division, efficiently binds to full-length LGN without Gαi and induces its conformational change, enhancing its association with Gαi In nonpolarized symmetrically dividing HeLa cells, disruption of the LGN-NuMA interaction by ectopic expression of mInsc results in a loss of cortical localization of NuMA during metaphase and anaphase and promotes mitotic spindle misorientation and a delayed anaphase progression. These findings highlight a specific role for LGN-mediated cell cortex recruitment of NuMA.
Collapse
Affiliation(s)
- Hiroki Takayanagi
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Junya Hayase
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Sachiko Kamakura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kei Miyano
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kanako Chishiki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Satoru Yuzawa
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|
30
|
Abstract
Mechanical forces drive the remodeling of tissues during morphogenesis. This relies on the transmission of forces between cells by cadherin-based adherens junctions, which couple the force-generating actomyosin cytoskeletons of neighboring cells. Moreover, components of cadherin adhesions adopt force-dependent conformations that induce changes in the composition of adherens junctions, enabling transduction of mechanical forces into an intracellular response. Cadherin mechanotransduction can mediate reinforcement of cell–cell adhesions to withstand forces but also induce biochemical signaling to regulate cell behavior or direct remodeling of cell–cell adhesions to enable cell rearrangements. By transmission and transduction of mechanical forces, cadherin adhesions coordinate cellular behaviors underlying morphogenetic processes of collective cell migration, cell division, and cell intercalation. Here, we review recent advances in our understanding of this central role of cadherin adhesions in force-dependent regulation of morphogenesis.
Collapse
Affiliation(s)
- Willem-Jan Pannekoek
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn Gloerich
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Wolf B, Busso C, Gönczy P. Live imaging screen reveals that TYRO3 and GAK ensure accurate spindle positioning in human cells. Nat Commun 2019; 10:2859. [PMID: 31253758 PMCID: PMC6599018 DOI: 10.1038/s41467-019-10446-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
Proper spindle positioning is crucial for spatial cell division control. Spindle positioning in human cells relies on a ternary complex comprising Gαi1-3, LGN and NuMA, which anchors dynein at the cell cortex, thus enabling pulling forces to be exerted on astral microtubules. We develop a live imaging siRNA-based screen using stereotyped fibronectin micropatterns to uncover components modulating spindle positioning in human cells, testing 1280 genes, including all kinases and phosphatases. We thus discover 16 components whose inactivation dramatically perturbs spindle positioning, including tyrosine receptor kinase 3 (TYRO3) and cyclin G associated kinase (GAK). TYRO3 depletion results in excess NuMA and dynein at the cortex during metaphase, similar to the effect of blocking the TYRO3 downstream target phosphatidylinositol 3-kinase (PI3K). Furthermore, depletion of GAK leads to impaired astral microtubules, similar to the effect of downregulating the GAK-interactor Clathrin. Overall, our work uncovers components and mechanisms governing spindle positioning in human cells.
Collapse
Affiliation(s)
- Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
32
|
Pirovano L, Culurgioni S, Carminati M, Alfieri A, Monzani S, Cecatiello V, Gaddoni C, Rizzelli F, Foadi J, Pasqualato S, Mapelli M. Hexameric NuMA:LGN structures promote multivalent interactions required for planar epithelial divisions. Nat Commun 2019; 10:2208. [PMID: 31101817 PMCID: PMC6525239 DOI: 10.1038/s41467-019-09999-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cortical force generators connect epithelial polarity sites with astral microtubules, allowing dynein movement to orient the mitotic spindle as astral microtubules depolymerize. Complexes of the LGN and NuMA proteins, fundamental components of force generators, are recruited to the cortex by Gαi-subunits of heterotrimeric G-proteins. They associate with dynein/dynactin and activate the motor activity pulling on astral microtubules. The architecture of cortical force generators is unknown. Here we report the crystal structure of NuMA:LGN hetero-hexamers, and unveil their role in promoting the assembly of active cortical dynein/dynactin motors that are required in orchestrating oriented divisions in polarized cells. Our work elucidates the basis for the structural organization of essential spindle orientation motors. LGN and NuMA link epithelial polarity sites with astral microtubules and associate with dynein, but the architecture of such cortical force-generating complexes is unknown. Here, the authors report the crystal structure of NuMA:LGN hetero-hexamers, and unveil their role in promoting the assembly of active cortical dynein/dynactin motors.
Collapse
Affiliation(s)
- Laura Pirovano
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy
| | - Simone Culurgioni
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy.,Exscientia Ltd., The Schröedinger Building, Heatley Road, Oxford Science Park, Oxford, OX4 4GE, UK
| | - Manuel Carminati
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy.,MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Andrea Alfieri
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy.,Department of Biosciences, Università degli Studi di Milano, 20133, Milan, Italy
| | - Silvia Monzani
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy
| | | | - Chiara Gaddoni
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy
| | | | - James Foadi
- Department of Mathematical Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | | | - Marina Mapelli
- IEO, European Institute of Oncology IRCCS, 20141, MILANO, Italy.
| |
Collapse
|
33
|
Extreme Fuzziness: Direct Interactions between Two IDPs. Biomolecules 2019; 9:biom9030081. [PMID: 30813629 PMCID: PMC6468500 DOI: 10.3390/biom9030081] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/10/2019] [Accepted: 02/18/2019] [Indexed: 01/06/2023] Open
Abstract
Protein interactions involving intrinsically disordered proteins (IDPs) greatly extend the range of binding mechanisms available to proteins. In interactions employing coupled folding and binding, IDPs undergo disorder-to-order transitions to form a complex with a well-defined structure. In many other cases, IDPs retain structural plasticity in the final complexes, which have been defined as the fuzzy complexes. While a large number of fuzzy complexes have been characterized with variety of fuzzy patterns, many of the interactions are between an IDP and a structured protein. Thus, whether two IDPs can interact directly to form a fuzzy complex without disorder-to-order transition remains an open question. Recently, two studies of interactions between IDPs (4.1G-CTD/NuMA and H1/ProTα) have found a definite answer to this question. Detailed characterizations combined with nuclear magnetic resonance (NMR), single-molecule Förster resonance energy transfer (smFRET) and molecular dynamics (MD) simulation demonstrate that direct interactions between these two pairs of IDPs do form fuzzy complexes while retaining the conformational dynamics of the isolated proteins, which we name as the extremely fuzzy complexes. Extreme fuzziness completes the full spectrum of protein-protein interaction modes, suggesting that a more generalized model beyond existing binding mechanisms is required. Previous models of protein interaction could be applicable to some aspects of the extremely fuzzy interactions, but in more general sense, the distinction between native and nonnative contacts, which was used to understand protein folding and binding, becomes obscure. Exploring the phenomenon of extreme fuzziness may shed new light on molecular recognition and drug design.
Collapse
|
34
|
Kotak S. Mechanisms of Spindle Positioning: Lessons from Worms and Mammalian Cells. Biomolecules 2019; 9:E80. [PMID: 30823600 PMCID: PMC6406873 DOI: 10.3390/biom9020080] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Proper positioning of the mitotic spindle is fundamental for specifying the site for cleavage furrow, and thus regulates the appropriate sizes and accurate distribution of the cell fate determinants in the resulting daughter cells during development and in the stem cells. The past couple of years have witnessed tremendous work accomplished in the area of spindle positioning, and this has led to the emergence of a working model unravelling in-depth mechanistic insight of the underlying process orchestrating spindle positioning. It is evident now that the correct positioning of the mitotic spindle is not only guided by the chemical cues (protein⁻protein interactions) but also influenced by the physical nature of the cellular environment. In metazoans, the key players that regulate proper spindle positioning are the actin-rich cell cortex and associated proteins, the ternary complex (Gα/GPR-1/2/LIN-5 in Caenorhabditis elegans, Gαi/Pins/Mud in Drosophila and Gαi1-3/LGN/NuMA in humans), minus-end-directed motor protein dynein and the cortical machinery containing myosin. In this review, I will mainly discuss how the abovementioned components precisely and spatiotemporally regulate spindle positioning by sensing the physicochemical environment for execution of flawless mitosis.
Collapse
Affiliation(s)
- Sachin Kotak
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore 560012, India.
| |
Collapse
|
35
|
Kakihana A, Oto Y, Saito Y, Nakayama Y. Heat shock-induced mitotic arrest requires heat shock protein 105 for the activation of spindle assembly checkpoint. FASEB J 2018; 33:3936-3953. [PMID: 30496702 DOI: 10.1096/fj.201801369r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heat shock causes proteotoxic stress that induces various cellular responses, including delayed mitotic progression and the generation of an aberrant number of chromosomes. In this study, heat shock delayed the onset of anaphase by increasing the number of misoriented cells, accompanied by the kinetochore localization of budding uninhibited by benzimidazole-related (BubR)1 in a monopolar spindle (Mps)1-dependent manner. The mitotic delay was canceled by knockdown of mitotic arrest defect (Mad)2. Knockdown of heat shock protein (Hsp)105 partially abrogated the mitotic delay with the loss of the kinetochore localization of BubR1 under heat shock conditions and accelerated mitotic progression under nonstressed conditions. Consistent with this result, Hsp105 knockdown increased the number of anaphase cells with lagging chromosomes, through mitotic slippage, and decreased taxol sensitivity more than Mad2 knockdown. Hsp105 was coprecipitated with cell division cycle (Cdc)20 in an Mps1-dependent manner; however, its knockdown did not affect coprecipitation of Mad2 and BubR1 with Cdc20. We propose that heat shock delays the onset of anaphase via the activation of the spindle assembly checkpoint (SAC). Hsp105 prevents abnormal cell division by contributing to SAC activation under heat shock and nonstressed conditions by interacting with Cdc20 but not affecting formation of the mitotic checkpoint complex.-Kakihana, A., Oto, Y., Saito, Y., Nakayama, Y. Heat shock-induced mitotic arrest requires heat shock protein 105 for the activation of spindle assembly checkpoint.
Collapse
Affiliation(s)
- Ayana Kakihana
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yui Oto
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Youhei Saito
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuji Nakayama
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
36
|
Sana S, Keshri R, Rajeevan A, Kapoor S, Kotak S. Plk1 regulates spindle orientation by phosphorylating NuMA in human cells. Life Sci Alliance 2018; 1:e201800223. [PMID: 30456393 PMCID: PMC6240335 DOI: 10.26508/lsa.201800223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 11/24/2022] Open
Abstract
Proper orientation of the mitotic spindle defines the correct division plane and is essential for accurate cell division and development. In metazoans, an evolutionarily conserved complex comprising of NuMA/LGN/Gαi regulates proper orientation of the mitotic spindle by orchestrating cortical dynein levels during metaphase. However, the molecular mechanisms that modulate the spatiotemporal dynamics of this complex during mitosis remain elusive. Here, we report that acute inactivation of Polo-like kinase 1 (Plk1) during metaphase enriches cortical levels of dynein/NuMA/LGN and thus influences spindle orientation. We establish that this impact of Plk1 on cortical levels of dynein/NuMA/LGN is through NuMA, but not via dynein/LGN. Moreover, we reveal that Plk1 inhibition alters the dynamic behavior of NuMA at the cell cortex. We further show that Plk1 directly interacts and phosphorylates NuMA. Notably, NuMA-phosphorylation by Plk1 impacts its cortical localization, and this is needed for precise spindle orientation during metaphase. Overall, our finding connects spindle-pole pool of Plk1 with cortical NuMA and answers a long-standing puzzle about how spindle-pole Plk1 gradient dictates proper spindle orientation for error-free mitosis.
Collapse
Affiliation(s)
- Shrividya Sana
- Department of Microbiology and Cell Biology, Indian Institute of Science (IISc), Bangalore, India
| | - Riya Keshri
- Department of Microbiology and Cell Biology, Indian Institute of Science (IISc), Bangalore, India
| | - Ashwathi Rajeevan
- Department of Microbiology and Cell Biology, Indian Institute of Science (IISc), Bangalore, India
| | - Sukriti Kapoor
- Department of Microbiology and Cell Biology, Indian Institute of Science (IISc), Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology, Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
37
|
Venkei ZG, Yamashita YM. Emerging mechanisms of asymmetric stem cell division. J Cell Biol 2018; 217:3785-3795. [PMID: 30232100 PMCID: PMC6219723 DOI: 10.1083/jcb.201807037] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/10/2023] Open
Abstract
Venkei and Yamashita summarize recent advances in our understanding of asymmetric stem cell division in tissue homeostasis. The asymmetric cell division of stem cells, which produces one stem cell and one differentiating cell, has emerged as a mechanism to balance stem cell self-renewal and differentiation. Elaborate cellular mechanisms that orchestrate the processes required for asymmetric cell divisions are often shared between stem cells and other asymmetrically dividing cells. During asymmetric cell division, cells must establish asymmetry/polarity, which is guided by varying degrees of intrinsic versus extrinsic cues, and use intracellular machineries to divide in a desired orientation in the context of the asymmetry/polarity. Recent studies have expanded our knowledge on the mechanisms of asymmetric cell divisions, revealing the previously unappreciated complexity in setting up the cellular and/or environmental asymmetry, ensuring binary outcomes of the fate determination. In this review, we summarize recent progress in understanding the mechanisms and regulations of asymmetric stem cell division.
Collapse
Affiliation(s)
- Zsolt G Venkei
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Yukiko M Yamashita
- Life Sciences Institute, University of Michigan, Ann Arbor, MI .,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI
| |
Collapse
|
38
|
Combinatorial Contact Cues Specify Cell Division Orientation by Directing Cortical Myosin Flows. Dev Cell 2018; 46:257-270.e5. [PMID: 30032990 PMCID: PMC7695482 DOI: 10.1016/j.devcel.2018.06.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 04/13/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022]
Abstract
Cell division axes during development are specified in different orientations to establish multicellular assemblies, but the mechanisms that generate division axis diversity remain unclear. We show here that patterns of cell contact provide cues that diversify cell division orientation by modulating cortical non-muscle myosin flow. We reconstituted in vivo contact patterns using beads or isolated cells to show two findings. First, we identified three contact-dependent cues that pattern cell division orientation and myosin flow: physical contact, contact asymmetry, and a Wnt signal. Second, we experimentally demonstrated that myosin flow generates forces that trigger plasma membrane movements and propose that their anisotropy drives cell division orientation. Our data suggest that contact-dependent control of myosin specifies the division axes of Caenorhabditis elegans AB, ABa, EMS cells, and the mouse AB cell. The contact pattern-dependent generation of myosin flows, in concert with known microtubule/dynein pathways, may greatly expand division axis diversity during development.
Collapse
|
39
|
Malerød L, Le Borgne R, Lie-Jensen A, Eikenes ÅH, Brech A, Liestøl K, Stenmark H, Haglund K. Centrosomal ALIX regulates mitotic spindle orientation by modulating astral microtubule dynamics. EMBO J 2018; 37:embj.201797741. [PMID: 29858227 DOI: 10.15252/embj.201797741] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 04/08/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022] Open
Abstract
The orientation of the mitotic spindle (MS) is tightly regulated, but the molecular mechanisms are incompletely understood. Here we report a novel role for the multifunctional adaptor protein ALG-2-interacting protein X (ALIX) in regulating MS orientation in addition to its well-established role in cytokinesis. We show that ALIX is recruited to the pericentriolar material (PCM) of the centrosomes and promotes correct orientation of the MS in asymmetrically dividing Drosophila stem cells and epithelial cells, and symmetrically dividing Drosophila and human epithelial cells. ALIX-deprived cells display defective formation of astral microtubules (MTs), which results in abnormal MS orientation. Specifically, ALIX is recruited to the PCM via Drosophila Spindle defective 2 (DSpd-2)/Cep192, where ALIX promotes accumulation of γ-tubulin and thus facilitates efficient nucleation of astral MTs. In addition, ALIX promotes MT stability by recruiting microtubule-associated protein 1S (MAP1S), which stabilizes newly formed MTs. Altogether, our results demonstrate a novel evolutionarily conserved role of ALIX in providing robustness to the orientation of the MS by promoting astral MT formation during asymmetric and symmetric cell division.
Collapse
Affiliation(s)
- Lene Malerød
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Roland Le Borgne
- CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, Univ. Rennes, Rennes, France.,Equipe labélisée Ligue Contre Le Cancer, Rennes, France
| | - Anette Lie-Jensen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Åsmund Husabø Eikenes
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Andreas Brech
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Liestøl
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaisa Haglund
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway .,Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Heppert JK, Pani AM, Roberts AM, Dickinson DJ, Goldstein B. A CRISPR Tagging-Based Screen Reveals Localized Players in Wnt-Directed Asymmetric Cell Division. Genetics 2018; 208:1147-1164. [PMID: 29348144 PMCID: PMC5844328 DOI: 10.1534/genetics.117.300487] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/08/2018] [Indexed: 11/18/2022] Open
Abstract
Oriented cell divisions are critical to establish and maintain cell fates and tissue organization. Diverse extracellular and intracellular cues have been shown to provide spatial information for mitotic spindle positioning; however, the molecular mechanisms by which extracellular signals communicate with cells to direct mitotic spindle positioning are largely unknown. In animal cells, oriented cell divisions are often achieved by the localization of force-generating motor protein complexes to discrete cortical domains. Disrupting either these force-generating complexes or proteins that globally affect microtubule stability results in defects in mitotic positioning, irrespective of whether these proteins function as spatial cues for spindle orientation. This poses a challenge to traditional genetic dissection of this process. Therefore, as an alternative strategy to identify key proteins that act downstream of intercellular signaling, we screened the localization of many candidate proteins by inserting fluorescent tags directly into endogenous gene loci, without overexpressing the proteins. We tagged 23 candidate proteins in Caenorhabditis elegans and examined each protein's localization in a well-characterized, oriented cell division in the four-cell-stage embryo. We used cell manipulations and genetic experiments to determine which cells harbor key localized proteins and which signals direct these localizations in vivo We found that Dishevelled and adenomatous polyposis coli homologs are polarized during this oriented cell division in response to a Wnt signal, but two proteins typically associated with mitotic spindle positioning, homologs of NuMA and Dynein, were not detectably polarized. These results suggest an unexpected mechanism for mitotic spindle positioning in this system, they pinpoint key proteins of interest, and they highlight the utility of a screening approach based on analyzing the localization of endogenously tagged proteins.
Collapse
Affiliation(s)
- Jennifer K Heppert
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Ariel M Pani
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Allyson M Roberts
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Daniel J Dickinson
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina 27599
| |
Collapse
|
41
|
Kschonsak YT, Hoffmann I. Activated Ezrin controls MISP levels to ensure correct NuMA polarization and spindle orientation. J Cell Sci 2018; 131:jcs.214544. [DOI: 10.1242/jcs.214544] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Correct spindle orientation is achieved through signaling pathways that provide a molecular link between the cell cortex and spindle microtubules in an F-actin dependent manner. A conserved cortical protein complex, composed of LGN, NuMA, dynein-dynactin, plays a key role in establishing proper spindle orientation. It has also been shown that the actin-binding protein MISP and the ERM family, that are activated by LOK/SLK in mitosis, regulate spindle orientation. Here, we report that MISP functions between the ERM family member Ezrin and NuMA to allow optimal spindle positioning. We show that MISP directly interacts with Ezrin and that SLK/LOK-activated Ezrin ensures appropriate cortical MISP levels in mitosis by competing with MISP for actin-binding sites at the cell cortex. Furthermore, we found that regulation of proper cortical MISP levels by preventing its excessive accumulation is essential for crescent-like polarized NuMA localization at the cortex and as a consequence for highly dynamic astral microtubules. Our results uncover how appropriate MISP levels at the cortex are required for proper NuMA polarization and therefore an optimal placement of the mitotic spindle within the cell.
Collapse
Affiliation(s)
- Yvonne T. Kschonsak
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
- University of Heidelberg, F045, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, F045, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
Lee BH, Schwager F, Meraldi P, Gotta M. p37/UBXN2B regulates spindle orientation by limiting cortical NuMA recruitment via PP1/Repo-Man. J Cell Biol 2017; 217:483-493. [PMID: 29222185 PMCID: PMC5800812 DOI: 10.1083/jcb.201707050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/27/2017] [Accepted: 11/16/2017] [Indexed: 12/03/2022] Open
Abstract
The p97 adapter p37 was known to regulate spindle orientation in human cells, but the mechanism was unknown. In this study, we show that it limits the cortical recruitment of NuMA in a PP1–Repo-Man–dependent manner. This study identifies a novel pathway controlling cortical NuMA localization. Spindle orientation determines the axis of division and is crucial for cell fate, tissue morphogenesis, and the development of an organism. In animal cells, spindle orientation is regulated by the conserved Gαi–LGN–NuMA complex, which targets the force generator dynein–dynactin to the cortex. In this study, we show that p37/UBXN2B, a cofactor of the p97 AAA ATPase, regulates spindle orientation in mammalian cells by limiting the levels of cortical NuMA. p37 controls cortical NuMA levels via the phosphatase PP1 and its regulatory subunit Repo-Man, but it acts independently of Gαi, the kinase Aurora A, and the phosphatase PP2A. Our data show that in anaphase, when the spindle elongates, PP1/Repo-Man promotes the accumulation of NuMA at the cortex. In metaphase, p37 negatively regulates this function of PP1, resulting in lower cortical NuMA levels and correct spindle orientation.
Collapse
Affiliation(s)
- Byung Ho Lee
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Francoise Schwager
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.,Swiss National Centre for Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Monica Gotta
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland .,Swiss National Centre for Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
43
|
E-cadherin and LGN align epithelial cell divisions with tissue tension independently of cell shape. Proc Natl Acad Sci U S A 2017; 114:E5845-E5853. [PMID: 28674014 DOI: 10.1073/pnas.1701703114] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue morphogenesis requires the coordinated regulation of cellular behavior, which includes the orientation of cell division that defines the position of daughter cells in the tissue. Cell division orientation is instructed by biochemical and mechanical signals from the local tissue environment, but how those signals control mitotic spindle orientation is not fully understood. Here, we tested how mechanical tension across an epithelial monolayer is sensed to orient cell divisions. Tension across Madin-Darby canine kidney cell monolayers was increased by a low level of uniaxial stretch, which oriented cell divisions with the stretch axis irrespective of the orientation of the cell long axis. We demonstrate that stretch-induced division orientation required mechanotransduction through E-cadherin cell-cell adhesions. Increased tension on the E-cadherin complex promoted the junctional recruitment of the protein LGN, a core component of the spindle orientation machinery that binds the cytosolic tail of E-cadherin. Consequently, uniaxial stretch triggered a polarized cortical distribution of LGN. Selective disruption of trans engagement of E-cadherin in an otherwise cohesive cell monolayer, or loss of LGN expression, resulted in randomly oriented cell divisions in the presence of uniaxial stretch. Our findings indicate that E-cadherin plays a key role in sensing polarized tensile forces across the tissue and transducing this information to the spindle orientation machinery to align cell divisions.
Collapse
|
44
|
Seldin L, Macara I. Epithelial spindle orientation diversities and uncertainties: recent developments and lingering questions. F1000Res 2017; 6:984. [PMID: 28713562 PMCID: PMC5490480 DOI: 10.12688/f1000research.11370.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2017] [Indexed: 01/09/2023] Open
Abstract
Mitotic spindle orientation is a conserved, dynamic, and highly complex process that plays a key role in dictating the cleavage plane, fate, and positioning of cells within a tissue, therefore laying the blueprint for tissue structure and function. While the spindle-positioning pathway has been extensively studied in lower-model organisms, research over the past several years has highlighted its relevance to mammalian epithelial tissues. Although we continue to gain critical insights into the mechanisms underlying spindle positioning, many uncertainties persist. In this commentary, we will review the protein interactions that modulate spindle orientation and we will present important recent findings that underscore epithelial tissue-specific requirements and variations in this important pathway, as well as its potential relevance to cancer.
Collapse
Affiliation(s)
- Lindsey Seldin
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Ian Macara
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
45
|
Wu S, Wang D, Liu J, Feng Y, Weng J, Li Y, Gao X, Liu J, Wang W. The Dynamic Multisite Interactions between Two Intrinsically Disordered Proteins. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201701883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shaowen Wu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Dongdong Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Jin Liu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Yitao Feng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Jingwei Weng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Yu Li
- King Abdullah University of Science and Technology (KAUST); Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division; Thuwal 23955 Saudi Arabia
| | - Xin Gao
- King Abdullah University of Science and Technology (KAUST); Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division; Thuwal 23955 Saudi Arabia
| | - Jianwei Liu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| | - Wenning Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials; Department of Chemistry, and Institutes of Biomedical Sciences; Fudan University; Shanghai 200433 China
| |
Collapse
|
46
|
Wu S, Wang D, Liu J, Feng Y, Weng J, Li Y, Gao X, Liu J, Wang W. The Dynamic Multisite Interactions between Two Intrinsically Disordered Proteins. Angew Chem Int Ed Engl 2017; 56:7515-7519. [PMID: 28493424 DOI: 10.1002/anie.201701883] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Indexed: 01/10/2023]
Abstract
Protein interactions involving intrinsically disordered proteins (IDPs) comprise a variety of binding modes, from the well-characterized folding upon binding to dynamic fuzzy complexes. To date, most studies concern the binding of an IDP to a structured protein, while the interaction between two IDPs is poorly understood. In this study, NMR, smFRET, and molecular dynamics (MD) simulation are combined to characterize the interaction between two IDPs, the C-terminal domain (CTD) of protein 4.1G and the nuclear mitotic apparatus (NuMA) protein. It is revealed that CTD and NuMA form a fuzzy complex with remaining structural disorder. Multiple binding sites on both proteins were identified by molecular dynamics and mutagenesis studies. This study provides an atomic scenario in which two IDPs bearing multiple binding sites interact with each other in dynamic equilibrium. The combined approach employed here could be widely applicable for investigating IDPs and their dynamic interactions.
Collapse
Affiliation(s)
- Shaowen Wu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Dongdong Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Jin Liu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Yitao Feng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Jingwei Weng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Yu Li
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, Thuwal, 23955, Saudi Arabia
| | - Xin Gao
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, Thuwal, 23955, Saudi Arabia
| | - Jianwei Liu
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Wenning Wang
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| |
Collapse
|
47
|
Stooke-Vaughan GA, Davidson LA, Woolner S. Xenopus as a model for studies in mechanical stress and cell division. Genesis 2017; 55. [PMID: 28095623 DOI: 10.1002/dvg.23004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 11/17/2016] [Accepted: 11/17/2016] [Indexed: 01/03/2023]
Abstract
We exist in a physical world, and cells within biological tissues must respond appropriately to both environmental forces and forces generated within the tissue to ensure normal development and homeostasis. Cell division is required for normal tissue growth and maintenance, but both the direction and rate of cell division must be tightly controlled to avoid diseases of over-proliferation such as cancer. Recent studies have shown that mechanical cues can cause mitotic entry and orient the mitotic spindle, suggesting that physical force could play a role in patterning tissue growth. However, to fully understand how mechanics guides cells in vivo, it is necessary to assess the interaction of mechanical strain and cell division in a whole tissue context. In this mini-review we first summarise the body of work linking mechanics and cell division, before looking at the advantages that the Xenopus embryo can offer as a model organism for understanding: (1) the mechanical environment during embryogenesis, and (2) factors important for cell division. Finally, we introduce a novel method for applying a reproducible strain to Xenopus embryonic tissue and assessing subsequent cell divisions.
Collapse
Affiliation(s)
- Georgina A Stooke-Vaughan
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Lance A Davidson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213.,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213.,Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Sarah Woolner
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
48
|
Bergstralh DT, Dawney NS, St Johnston D. Spindle orientation: a question of complex positioning. Development 2017; 144:1137-1145. [DOI: 10.1242/dev.140764] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The direction in which a cell divides is determined by the orientation of its mitotic spindle at metaphase. Spindle orientation is therefore important for a wide range of developmental processes, ranging from germline stem cell division to epithelial tissue homeostasis and regeneration. In multiple cell types in multiple animals, spindle orientation is controlled by a conserved biological machine that mediates a pulling force on astral microtubules. Restricting the localization of this machine to only specific regions of the cortex can thus determine how the mitotic spindle is oriented. As we review here, recent findings based on studies in tunicate, worm, fly and vertebrate cells have revealed that the mechanisms for mediating this restriction are surprisingly diverse.
Collapse
Affiliation(s)
- Dan T. Bergstralh
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Nicole S. Dawney
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
49
|
Santoro A, Vlachou T, Carminati M, Pelicci PG, Mapelli M. Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 2016; 17:1700-1720. [PMID: 27872203 DOI: 10.15252/embr.201643021] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 01/16/2023] Open
Abstract
Stem cells have the remarkable ability to undergo proliferative symmetric divisions and self-renewing asymmetric divisions. Balancing of the two modes of division sustains tissue morphogenesis and homeostasis. Asymmetric divisions of Drosophila neuroblasts (NBs) and sensory organ precursor (SOP) cells served as prototypes to learn what we consider now principles of asymmetric mitoses. They also provide initial evidence supporting the notion that aberrant symmetric divisions of stem cells could correlate with malignancy. However, transferring the molecular knowledge of circuits underlying asymmetry from flies to mammals has proven more challenging than expected. Several experimental approaches have been used to define asymmetry in mammalian systems, based on daughter cell fate, unequal partitioning of determinants and niche contacts, or proliferative potential. In this review, we aim to provide a critical evaluation of the assays used to establish the stem cell mode of division, with a particular focus on the mammary gland system. In this context, we will discuss the genetic alterations that impinge on the modality of stem cell division and their role in breast cancer development.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Thalia Vlachou
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Manuel Carminati
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
50
|
Portegijs V, Fielmich LE, Galli M, Schmidt R, Muñoz J, van Mourik T, Akhmanova A, Heck AJR, Boxem M, van den Heuvel S. Multisite Phosphorylation of NuMA-Related LIN-5 Controls Mitotic Spindle Positioning in C. elegans. PLoS Genet 2016; 12:e1006291. [PMID: 27711157 PMCID: PMC5053539 DOI: 10.1371/journal.pgen.1006291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 08/10/2016] [Indexed: 11/18/2022] Open
Abstract
During cell division, the mitotic spindle segregates replicated chromosomes to opposite poles of the cell, while the position of the spindle determines the plane of cleavage. Spindle positioning and chromosome segregation depend on pulling forces on microtubules extending from the centrosomes to the cell cortex. Critical in pulling force generation is the cortical anchoring of cytoplasmic dynein by a conserved ternary complex of Gα, GPR-1/2, and LIN-5 proteins in C. elegans (Gα–LGN–NuMA in mammals). Previously, we showed that the polarity kinase PKC-3 phosphorylates LIN-5 to control spindle positioning in early C. elegans embryos. Here, we investigate whether additional LIN-5 phosphorylations regulate cortical pulling forces, making use of targeted alteration of in vivo phosphorylated residues by CRISPR/Cas9-mediated genetic engineering. Four distinct in vivo phosphorylated LIN-5 residues were found to have critical functions in spindle positioning. Two of these residues form part of a 30 amino acid binding site for GPR-1, which we identified by reverse two-hybrid screening. We provide evidence for a dual-kinase mechanism, involving GSK3 phosphorylation of S659 followed by phosphorylation of S662 by casein kinase 1. These LIN-5 phosphorylations promote LIN-5–GPR-1/2 interaction and contribute to cortical pulling forces. The other two critical residues, T168 and T181, form part of a cyclin-dependent kinase consensus site and are phosphorylated by CDK1-cyclin B in vitro. We applied a novel strategy to characterize early embryonic defects in lethal T168,T181 knockin substitution mutants, and provide evidence for sequential LIN-5 N-terminal phosphorylation and dephosphorylation in dynein recruitment. Our data support that phosphorylation of multiple LIN-5 domains by different kinases contributes to a mechanism for spatiotemporal control of spindle positioning and chromosome segregation. Protein kinases control biological processes by phosphorylating specific amino acids of substrate proteins. It remains a major challenge to identify which phosphorylation events are critical in vivo and how phosphorylation affects protein function. Recent developments in CRISPR/Cas9-based genetic engineering make it possible to substitute individual amino acids, which allows investigating the role of single and multi-site phosphorylation of substrates in vivo. Here, we focus on an intensively phosphorylated cell division protein, LIN-5NuMA. C. elegans LIN-5 participates in chromosome segregation and is essential for positioning the spindle and cell cleavage plane during asymmetric cell division. Previously, we demonstrated that the polarity kinase PKC-3 phosphorylates LIN-5 to inhibit its function. Our current analysis reveals four additional phosphorylated residues that are critical for LIN-5 function. Two of these residues contribute to the interaction of LIN-5 with its binding partner GPR-1/2, whereas the other two residues are critical in dynein motor recruitment by LIN-5. Together, our results reveal that multisite phosphorylation of LIN-5 is essential to ensure proper chromosome segregation and spindle positioning.
Collapse
Affiliation(s)
- Vincent Portegijs
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Lars-Eric Fielmich
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Matilde Galli
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Ruben Schmidt
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Javier Muñoz
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Tim van Mourik
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Mike Boxem
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Sander van den Heuvel
- Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|