1
|
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
|
2
|
Aquino-Perez C, Safaralizade M, Podhajecky R, Wang H, Lansky Z, Grosse R, Macurek L. FAM110A promotes mitotic spindle formation by linking microtubules with actin cytoskeleton. Proc Natl Acad Sci U S A 2024; 121:e2321647121. [PMID: 38995965 PMCID: PMC11260166 DOI: 10.1073/pnas.2321647121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/06/2024] [Indexed: 07/14/2024] Open
Abstract
Precise segregation of chromosomes during mitosis requires assembly of a bipolar mitotic spindle followed by correct attachment of microtubules to the kinetochores. This highly spatiotemporally organized process is controlled by various mitotic kinases and molecular motors. We have recently shown that Casein Kinase 1 (CK1) promotes timely progression through mitosis by phosphorylating FAM110A leading to its enrichment at spindle poles. However, the mechanism by which FAM110A exerts its function in mitosis is unknown. Using structure prediction and a set of deletion mutants, we mapped here the interaction of the N- and C-terminal domains of FAM110A with actin and tubulin, respectively. Next, we found that the FAM110A-Δ40-61 mutant deficient in actin binding failed to rescue defects in chromosomal alignment caused by depletion of endogenous FAM110A. Depletion of FAM110A impaired assembly of F-actin in the proximity of spindle poles and was rescued by expression of the wild-type FAM110A, but not the FAM110A-Δ40-61 mutant. Purified FAM110A promoted binding of F-actin to microtubules as well as bundling of actin filaments in vitro. Finally, we found that the inhibition of CK1 impaired spindle actin formation and delayed progression through mitosis. We propose that CK1 and FAM110A promote timely progression through mitosis by mediating the interaction between spindle microtubules and filamentous actin to ensure proper mitotic spindle formation.
Collapse
Affiliation(s)
- Cecilia Aquino-Perez
- Cancer Cell Biology, Institute of Molecular Genetics, Czech Academy of Sciences, PragueCZ14220, Czech Republic
| | - Mahira Safaralizade
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
| | - Roman Podhajecky
- Institute of Biotechnology, Czech Academy of Sciences, Biocev, VestecCZ25250, Czech Republic
| | - Hong Wang
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
- Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg79104, Germany
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, Biocev, VestecCZ25250, Czech Republic
| | - Robert Grosse
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
- Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg79104, Germany
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics, Czech Academy of Sciences, PragueCZ14220, Czech Republic
| |
Collapse
|
3
|
Brito C, Serna M, Guerra P, Llorca O, Surrey T. Transition of human γ-tubulin ring complex into a closed conformation during microtubule nucleation. Science 2024; 383:870-876. [PMID: 38305685 DOI: 10.1126/science.adk6160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Microtubules are essential for intracellular organization and chromosome segregation. They are nucleated by the γ-tubulin ring complex (γTuRC). However, isolated vertebrate γTuRC adopts an open conformation that deviates from the microtubule structure, raising the question of the nucleation mechanism. In this study, we determined cryo-electron microscopy structures of human γTuRC bound to a nascent microtubule. Structural changes of the complex into a closed conformation ensure that γTuRC templates the 13-protofilament microtubules that exist in human cells. Closure is mediated by a latch that interacts with incorporating tubulin, making it part of the closing mechanism. Further rearrangements involve all γTuRC subunits and the removal of the actin-containing luminal bridge. Our proposed mechanism of microtubule nucleation by human γTuRC relies on large-scale structural changes that are likely the target of regulation in cells.
Collapse
Affiliation(s)
- Cláudia Brito
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marina Serna
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Pablo Guerra
- Cryo-Electron Microscopy Platform-IBMB CSIC, Joint Electron Microscopy Center at ALBA (JEMCA), Barcelona, Spain
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, 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
|
4
|
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
|
5
|
Prakash A, Paunikar S, Webber M, McDermott E, Vellanki SH, Thompson K, Dockery P, Jahns H, Brown JAL, Hopkins AM, Bourke E. Centrosome amplification promotes cell invasion via cell-cell contact disruption and Rap-1 activation. J Cell Sci 2023; 136:jcs261150. [PMID: 37772773 PMCID: PMC10629695 DOI: 10.1242/jcs.261150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
Centrosome amplification (CA) is a prominent feature of human cancers linked to tumorigenesis in vivo. Here, we report mechanistic contributions of CA induction alone to tumour architecture and extracellular matrix (ECM) remodelling. CA induction in non-tumorigenic breast cells MCF10A causes cell migration and invasion, with underlying disruption of epithelial cell-cell junction integrity and dysregulation of expression and subcellular localisation of cell junction proteins. CA also elevates expression of integrin β-3, its binding partner fibronectin-1 and matrix metalloproteinase enzymes, promoting cell-ECM attachment, ECM degradation, and a migratory and invasive cell phenotype. Using a chicken embryo xenograft model for in vivo validation, we show that CA-induced (+CA) MCF10A cells invade into the chick mesodermal layer, with inflammatory cell infiltration and marked focal reactions between chorioallantoic membrane and cell graft. We also demonstrate a key role of small GTPase Rap-1 signalling through inhibition using GGTI-298, which blocked various CA-induced effects. These insights reveal that in normal cells, CA induction alone (without additional oncogenic alterations) is sufficient to confer early pro-tumorigenic changes within days, acting through Rap-1-dependent signalling to alter cell-cell contacts and ECM disruption.
Collapse
Affiliation(s)
- Anu Prakash
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Shishir Paunikar
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Mark Webber
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Emma McDermott
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Sri H. Vellanki
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Kerry Thompson
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Peter Dockery
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Hanne Jahns
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - James A. L. Brown
- Department of Biological Sciences, University of Limerick, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) and Health Research Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Ann M. Hopkins
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Emer Bourke
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| |
Collapse
|
6
|
Cazzagon G, Roubinet C, Baum B. Polarized SCAR and the Arp2/3 complex regulate apical cortical remodeling in asymmetrically dividing neuroblasts. iScience 2023; 26:107129. [PMID: 37434695 PMCID: PMC10331462 DOI: 10.1016/j.isci.2023.107129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/03/2023] [Accepted: 06/10/2023] [Indexed: 07/13/2023] Open
Abstract
Although the formin-nucleated actomyosin cortex has been shown to drive the changes in cell shape that accompany animal cell division in both symmetric and asymmetric cell divisions, the mitotic role of cortical Arp2/3-nucleated actin networks remain unclear. Here using asymmetrically dividing Drosophila neural stem cells as a model system, we identify a pool of membrane protrusions that form at the apical cortex of neuroblasts as they enter mitosis. Strikingly, these apically localized protrusions are enriched in SCAR, and depend on SCAR and Arp2/3 complexes for their formation. Because compromising SCAR or the Arp2/3 complex delays the apical clearance of Myosin II at the onset of anaphase and induces cortical instability at cytokinesis, these data point to a role for an apical branched actin filament network in fine-tuning the actomyosin cortex to enable the precise control of cell shape changes during an asymmetric cell division.
Collapse
Affiliation(s)
- Giulia Cazzagon
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Chantal Roubinet
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Buzz Baum
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| |
Collapse
|
7
|
Zhao G, Liu S, Arun S, Renda F, Khodjakov A, Pellman D. A tubule-sheet continuum model for the mechanism of nuclear envelope assembly. Dev Cell 2023; 58:847-865.e10. [PMID: 37098350 PMCID: PMC10205699 DOI: 10.1016/j.devcel.2023.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/25/2023] [Accepted: 04/01/2023] [Indexed: 04/27/2023]
Abstract
Nuclear envelope (NE) assembly defects cause chromosome fragmentation, cancer, and aging. However, major questions about the mechanism of NE assembly and its relationship to nuclear pathology are unresolved. In particular, how cells efficiently assemble the NE starting from vastly different, cell type-specific endoplasmic reticulum (ER) morphologies is unclear. Here, we identify a NE assembly mechanism, "membrane infiltration," that defines one end of a continuum with another NE assembly mechanism, "lateral sheet expansion," in human cells. Membrane infiltration involves the recruitment of ER tubules or small sheets to the chromatin surface by mitotic actin filaments. Lateral sheet expansion involves actin-independent envelopment of peripheral chromatin by large ER sheets that then extend over chromatin within the spindle. We propose a "tubule-sheet continuum" model that explains the efficient NE assembly from any starting ER morphology, the cell type-specific patterns of nuclear pore complex (NPC) assembly, and the obligatory NPC assembly defect of micronuclei.
Collapse
Affiliation(s)
- Gengjing Zhao
- Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Shiwei Liu
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sanjana Arun
- Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Fioranna Renda
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Alexey Khodjakov
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - David Pellman
- Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Oda H, Sato Y, Kawashima SA, Fujiwara Y, Pálfy M, Wu E, Vastenhouw NL, Kanai M, Kimura H. Actin filaments accumulated in the nucleus remain in the vicinity of condensing chromosomes in the zebrafish early embryo. Biol Open 2023; 12:bio059783. [PMID: 37071022 PMCID: PMC10214854 DOI: 10.1242/bio.059783] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
In the cytoplasm, filamentous actin (F-actin) plays a critical role in cell regulation, including cell migration, stress fiber formation, and cytokinesis. Recent studies have shown that actin filaments that form in the nucleus are associated with diverse functions. Here, using live imaging of an F-actin-specific probe, superfolder GFP-tagged utrophin (UtrCH-sfGFP), we demonstrated the dynamics of nuclear actin in zebrafish (Danio rerio) embryos. In early zebrafish embryos up to around the high stage, UtrCH-sfGFP increasingly accumulated in nuclei during the interphase and reached a peak during the prophase. After nuclear envelope breakdown (NEBD), patches of UtrCH-sfGFP remained in the vicinity of condensing chromosomes during the prometaphase to metaphase. When zygotic transcription was inhibited by injecting α-amanitin, the nuclear accumulation of UtrCH-sfGFP was still observed at the sphere and dome stages, suggesting that zygotic transcription may induce a decrease in nuclear F-actin. The accumulation of F-actin in nuclei may contribute to proper mitotic progression of large cells with rapid cell cycles in zebrafish early embryos, by assisting in NEBD, chromosome congression, and/or spindle assembly.
Collapse
Affiliation(s)
- Haruka Oda
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Yuko Sato
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Shigehiro A. Kawashima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yusuke Fujiwara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Máté Pálfy
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden-01307, Germany
| | - Edlyn Wu
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden-01307, Germany
- University of Lausanne, Center for Integrative Genomics, Lausanne 1015, Switzerland
| | - Nadine L. Vastenhouw
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden-01307, Germany
- University of Lausanne, Center for Integrative Genomics, Lausanne 1015, Switzerland
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
9
|
Jeruzalska E, Mazur AJ. The Role of non-muscle actin paralogs in cell cycle progression and proliferation. Eur J Cell Biol 2023; 102:151315. [PMID: 37099935 DOI: 10.1016/j.ejcb.2023.151315] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Uncontrolled cell proliferation leads to several pathologies, including cancer. Thus, this process must be tightly regulated. The cell cycle accounts for cell proliferation, and its progression is coordinated with changes in cell shape, for which cytoskeleton reorganization is responsible. Rearrangement of the cytoskeleton allows for its participation in the precise division of genetic material and cytokinesis. One of the main cytoskeletal components is filamentous actin-based structures. Mammalian cells have at least six actin paralogs, four of which are muscle-specific, while two, named β- and γ-actin, are abundantly present in all types of cells. This review summarizes the findings that establish the role of non-muscle actin paralogs in regulating cell cycle progression and proliferation. We discuss studies showing that the level of a given non-muscle actin paralog in a cell influences the cell's ability to progress through the cell cycle and, thus, proliferation. Moreover, we elaborate on the non-muscle actins' role in regulating gene transcription, interactions of actin paralogs with proteins involved in controlling cell proliferation, and the contribution of non-muscle actins to different structures in a dividing cell. The data cited in this review show that non-muscle actins regulate the cell cycle and proliferation through varying mechanisms. We point to the need for further studies addressing these mechanisms.
Collapse
Affiliation(s)
- Estera Jeruzalska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| |
Collapse
|
10
|
Cao M, Zou X, Li C, Lin Z, Wang N, Zou Z, Ye Y, Seemann J, Levine B, Tang Z, Zhong Q. An actin filament branching surveillance system regulates cell cycle progression, cytokinesis and primary ciliogenesis. Nat Commun 2023; 14:1687. [PMID: 36973243 PMCID: PMC10042869 DOI: 10.1038/s41467-023-37340-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Dysfunction of cell cycle control and defects of primary ciliogenesis are two features of many cancers. Whether these events are interconnected and the driving mechanism coordinating them remains elusive. Here, we identify an actin filament branching surveillance system that alerts cells of actin branching insufficiency and regulates cell cycle progression, cytokinesis and primary ciliogenesis. We find that Oral-Facial-Digital syndrome 1 functions as a class II Nucleation promoting factor to promote Arp2/3 complex-mediated actin branching. Perturbation of actin branching promotes OFD1 degradation and inactivation via liquid-to-gel transition. Elimination of OFD1 or disruption of OFD1-Arp2/3 interaction drives proliferating, non-transformed cells into quiescence with ciliogenesis by an RB-dependent mechanism, while it leads oncogene-transformed/cancer cells to incomplete cytokinesis and irreversible mitotic catastrophe via actomyosin ring malformation. Inhibition of OFD1 leads to suppression of multiple cancer cell growth in mouse xenograft models. Thus, targeting OFD1-mediated actin filament branching surveillance system provides a direction for cancer therapy.
Collapse
Affiliation(s)
- Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| | - Xiaoxiao Zou
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Chaoyi Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Zaisheng Lin
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Ni Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Zhongju Zou
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Joachim Seemann
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zaiming Tang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| |
Collapse
|
11
|
Campellone KG, Lebek NM, King VL. Branching out in different directions: Emerging cellular functions for the Arp2/3 complex and WASP-family actin nucleation factors. Eur J Cell Biol 2023; 102:151301. [PMID: 36907023 DOI: 10.1016/j.ejcb.2023.151301] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The actin cytoskeleton impacts practically every function of a eukaryotic cell. Historically, the best-characterized cytoskeletal activities are in cell morphogenesis, motility, and division. The structural and dynamic properties of the actin cytoskeleton are also crucial for establishing, maintaining, and changing the organization of membrane-bound organelles and other intracellular structures. Such activities are important in nearly all animal cells and tissues, although distinct anatomical regions and physiological systems rely on different regulatory factors. Recent work indicates that the Arp2/3 complex, a broadly expressed actin nucleator, drives actin assembly during several intracellular stress response pathways. These newly described Arp2/3-mediated cytoskeletal rearrangements are coordinated by members of the Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation-promoting factors. Thus, the Arp2/3 complex and WASP-family proteins are emerging as crucial players in cytoplasmic and nuclear activities including autophagy, apoptosis, chromatin dynamics, and DNA repair. Characterizations of the functions of the actin assembly machinery in such stress response mechanisms are advancing our understanding of both normal and pathogenic processes, and hold great promise for providing insights into organismal development and interventions for disease.
Collapse
Affiliation(s)
- Kenneth G Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA.
| | - Nadine M Lebek
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| | - Virginia L King
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| |
Collapse
|
12
|
Genomic instability caused by Arp2/3 complex inactivation results in micronucleus biogenesis and cellular senescence. PLoS Genet 2023; 19:e1010045. [PMID: 36706133 PMCID: PMC9907832 DOI: 10.1371/journal.pgen.1010045] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/08/2023] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
The Arp2/3 complex is an actin nucleator with well-characterized activities in cell morphogenesis and movement, but its roles in nuclear processes are relatively understudied. We investigated how the Arp2/3 complex affects genomic integrity and cell cycle progression using mouse fibroblasts containing an inducible knockout (iKO) of the ArpC2 subunit. We show that permanent Arp2/3 complex ablation results in DNA damage, the formation of cytosolic micronuclei, and cellular senescence. Micronuclei arise in ArpC2 iKO cells due to chromatin segregation defects during mitosis and premature mitotic exits. Such phenotypes are explained by the presence of damaged DNA fragments that fail to attach to the mitotic spindle, abnormalities in actin assembly during metaphase, and asymmetric microtubule architecture during anaphase. In the nuclei of Arp2/3-depleted cells, the tumor suppressor p53 is activated and the cell cycle inhibitor Cdkn1a/p21 mediates a G1 arrest. In the cytosol, micronuclei are recognized by the DNA sensor cGAS, which is important for stimulating a STING- and IRF3-associated interferon response. These studies establish functional requirements for the mammalian Arp2/3 complex in mitotic spindle organization and genome stability. They also expand our understanding of the mechanisms leading to senescence and suggest that cytoskeletal dysfunction is an underlying factor in biological aging.
Collapse
|
13
|
Moreno-Andrés D, Holl K, Antonin W. The second half of mitosis and its implications in cancer biology. Semin Cancer Biol 2023; 88:1-17. [PMID: 36436712 DOI: 10.1016/j.semcancer.2022.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022]
Abstract
The nucleus undergoes dramatic structural and functional changes during cell division. With the entry into mitosis, in human cells the nuclear envelope breaks down, chromosomes rearrange into rod-like structures which are collected and segregated by the spindle apparatus. While these processes in the first half of mitosis have been intensively studied, much less is known about the second half of mitosis, when a functional nucleus reforms in each of the emerging cells. Here we review our current understanding of mitotic exit and nuclear reformation with spotlights on the links to cancer biology.
Collapse
Affiliation(s)
- Daniel Moreno-Andrés
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Kristin Holl
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Wolfram Antonin
- Institute of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
14
|
Nguyen A, Goetz SC. TTBK2 controls cilium stability by regulating distinct modules of centrosomal proteins. Mol Biol Cell 2022; 34:ar8. [PMID: 36322399 PMCID: PMC9816645 DOI: 10.1091/mbc.e22-08-0373] [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] [Indexed: 11/06/2022] Open
Abstract
The serine-threonine kinase tau tubulin kinase 2 (TTBK2) is a key regulator of the assembly of primary cilia, which are vital signaling organelles. TTBK2 is also implicated in the stability of the assembled cilium through mechanisms that remain to be defined. Here we use mouse embryonic fibroblasts derived from Ttbk2fl/fl, UBC-CreERT+ embryos (hereafter Ttbk2cmut) to dissect the role of TTBK2 in cilium stability. This system depletes TTBK2 levels after cilia formation, allowing us to assess the molecular changes to the assembled cilium over time. As a consequence of Ttbk2 deletion, the ciliary axoneme is destabilized and primary cilia are lost within 48-72 h following recombination. Axoneme destabilization involves an increased frequency of cilia breaks and a reduction in axonemal microtubule modifications. Cilia loss was delayed by using inhibitors that affect actin-based trafficking. At the same time, we find that TTBK2 is required to regulate the composition of the centriolar satellites and to maintain the basal body pools of intraflagellar transport proteins. Altogether, our results reveal parallel pathways by which TTBK2 maintains cilium stability.
Collapse
Affiliation(s)
- Abraham Nguyen
- Molecular Cancer Biology Program, Duke University School of Medicine, Durham, NC 27710,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710
| | - Sarah C. Goetz
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710,*Address correspondence to: Sarah C. Goetz ()
| |
Collapse
|
15
|
Guo M, Lian J, Liu Y, Dong B, He Q, Zhao Q, Zhang H, Qi Y, Zhang Y, Huang L. Loss of miR-637 promotes cancer cell stemness via WASH/IL-8 pathway and serves as a novel prognostic marker in esophageal squamous cell carcinoma. Biomark Res 2022; 10:77. [PMID: 36329557 PMCID: PMC9635169 DOI: 10.1186/s40364-022-00424-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Esophageal carcinoma is the highly lethal cancer in the world, predominantly in some areas of East Asia. We previously reported that overexpression of cytoskeleton regulator Wiskott-Aldrich syndrome protein and SCAR Homolog (WASH) associates with poor prognosis of patients with esophageal squamous cell carcinoma (ESCC). However, the molecular mechanism and clinical significance involved in WASH overexpression have not been fully elucidated. METHODS Bioinformatics analysis and luciferase reporter assay were used to predict and validate miR-637 as a regulator of WASH in ESCC cell lines. qRT-PCR, Western blotting and ELISA assays were performed to examine RNA expression and protein levels, respectively. Next, the biological functions of miR-637 were explored by tumor sphere formation assay in vitro and nude mouse tumor xenograft in vivo. Finally, we evaluated the association of miR-637 levels with clinical features in ESCC patients. RESULTS We identified miR-637 as a WASH-targeting miRNA. miR-637 mimic strongly attenuated the downstream IL-8 production and tumor sphere formation in esophageal cancer cells, whereas miR-637 inhibitor displayed an opposite effect. IL-8 could facilitate stem-like properties and partially rescue the phenotypes induced by miR-637 mimic. Furthermore, miR-637 inhibitor dramatically promoted IL-8 expression and cancer stemness properties in a WASH-dependent manner. Ectopic expression of miR-637 also inhibited tumor growth in a mouse model. Clinically, low expression of miR-637 was observed in tumor tissues and the low expression levels of miR-637 were correlated with poor survival of ESCC patients. In particular, plasma miR-637 could be used as a noninvasive biomarker for ESCC patients. CONCLUSIONS These results implicate the potential application of miR-637 for diagnosis and prognosis of esophageal cancer.
Collapse
Affiliation(s)
- Mengxing Guo
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Jingyao Lian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Yaqing Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Bo Dong
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qianyi He
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Qitai Zhao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Hongyan Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China.
| | - Lan Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China.
| |
Collapse
|
16
|
Cheng H, Kao Y, Chen T, Sharma L, Yang W, Chuang Y, Huang S, Lin H, Huang Y, Kao C, Yang L, Bearon R, Cheng H, Hsia K, Lin Y. Actin filaments form a size-dependent diffusion barrier around centrosomes. EMBO Rep 2022; 24:e54935. [PMID: 36314725 PMCID: PMC9827556 DOI: 10.15252/embr.202254935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 01/10/2023] Open
Abstract
The centrosome, a non-membranous organelle, constrains various soluble molecules locally to execute its functions. As the centrosome is surrounded by various dense components, we hypothesized that it may be bordered by a putative diffusion barrier. After quantitatively measuring the trapping kinetics of soluble proteins of varying size at centrosomes by a chemically inducible diffusion trapping assay, we find that centrosomes are highly accessible to soluble molecules with a Stokes radius of less than 5.8 nm, whereas larger molecules rarely reach centrosomes, indicating the existence of a size-dependent diffusion barrier at centrosomes. The permeability of this barrier is tightly regulated by branched actin filaments outside of centrosomes and it decreases during anaphase when branched actin temporally increases. The actin-based diffusion barrier gates microtubule nucleation by interfering with γ-tubulin ring complex recruitment. We propose that actin filaments spatiotemporally constrain protein complexes at centrosomes in a size-dependent manner.
Collapse
Affiliation(s)
- Hsuan Cheng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Yu‐Lin Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ting Chen
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan
| | - Lohitaksh Sharma
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Wen‐Ting Yang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Yi‐Chien Chuang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Han Huang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hong‐Rui Lin
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan
| | - Yao‐Shen Huang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Chi‐Ling Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Lee‐Wei Yang
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan,Physics DivisionNational Center for Theoretical SciencesTaipeiTaiwan
| | - Rachel Bearon
- Department of Mathematical ScienceUniversity of LiverpoolLiverpoolUK
| | - Hui‐Chun Cheng
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan
| | | | - Yu‐Chun Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan,Department of Medical ScienceNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
17
|
Abstract
The centrosome, consisting of centrioles and the associated pericentriolar material, is the main microtubule-organizing centre (MTOC) in animal cells. During most of interphase, the two centrosomes of a cell are joined together by centrosome cohesion into one MTOC. The most dominant element of centrosome cohesion is the centrosome linker, an interdigitating, fibrous network formed by the protein C-Nap1 anchoring a number of coiled-coil proteins including rootletin to the proximal end of centrioles. Alternatively, centrosomes can be kept together by the action of the minus end directed kinesin motor protein KIFC3 that works on interdigitating microtubules organized by both centrosomes and probably by the actin network. Although cells connect the two interphase centrosomes by several mechanisms into one MTOC, the general importance of centrosome cohesion, particularly for an organism, is still largely unclear. In this article, we review the functions of the centrosome linker and discuss how centrosome cohesion defects can lead to diseases.
Collapse
Affiliation(s)
- Hairuo Dang
- Zentrum für Molekulare Biologie der Universität Heidelberg, Deutsches Krebsforschungszentrum-ZMBH Allianz, and,Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg 69120, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg, Deutsches Krebsforschungszentrum-ZMBH Allianz, and
| |
Collapse
|
18
|
Sulimenko V, Dráberová E, Dráber P. γ-Tubulin in microtubule nucleation and beyond. Front Cell Dev Biol 2022; 10:880761. [PMID: 36158181 PMCID: PMC9503634 DOI: 10.3389/fcell.2022.880761] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubules composed of αβ-tubulin dimers are dynamic cytoskeletal polymers that play key roles in essential cellular processes such as cell division, organelle positioning, intracellular transport, and cell migration. γ-Tubulin is a highly conserved member of the tubulin family that is required for microtubule nucleation. γ-Tubulin, together with its associated proteins, forms the γ-tubulin ring complex (γ-TuRC), that templates microtubules. Here we review recent advances in the structure of γ-TuRC, its activation, and centrosomal recruitment. This provides new mechanistic insights into the molecular mechanism of microtubule nucleation. Accumulating data suggest that γ-tubulin also has other, less well understood functions. We discuss emerging evidence that γ-tubulin can form oligomers and filaments, has specific nuclear functions, and might be involved in centrosomal cross-talk between microtubules and microfilaments.
Collapse
Affiliation(s)
| | | | - Pavel Dráber
- *Correspondence: Vadym Sulimenko, ; Pavel Dráber,
| |
Collapse
|
19
|
Nugues C, Helassa N, Haynes LP. Mitosis, Focus on Calcium. Front Physiol 2022; 13:951979. [PMID: 35784871 PMCID: PMC9247304 DOI: 10.3389/fphys.2022.951979] [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: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
The transformation of a single fertilised egg into an adult human consisting of tens of trillions of highly diverse cell types is a marvel of biology. The expansion is largely achieved by cell duplication through the process of mitosis. Mitosis is essential for normal growth, development, and tissue repair and is one of the most tightly regulated biological processes studied. This regulation is designed to ensure accurate segregation of chromosomes into each new daughter cell since errors in this process can lead to genetic imbalances, aneuploidy, that can lead to diseases including cancer. Understanding how mitosis operates and the molecular mechanisms that ensure its fidelity are therefore not only of significant intellectual value but provide unique insights into disease pathology. The purpose of this review is to revisit historical evidence that mitosis can be influenced by the ubiquitous second messenger calcium and to discuss this in the context of new findings revealing exciting new information about its role in cell division.
Collapse
Affiliation(s)
- Charlotte Nugues
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
20
|
Sun J, Zhong X, Fu X, Miller H, Lee P, Yu B, Liu C. The Actin Regulators Involved in the Function and Related Diseases of Lymphocytes. Front Immunol 2022; 13:799309. [PMID: 35371070 PMCID: PMC8965893 DOI: 10.3389/fimmu.2022.799309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Actin is an important cytoskeletal protein involved in signal transduction, cell structure and motility. Actin regulators include actin-monomer-binding proteins, Wiskott-Aldrich syndrome (WAS) family of proteins, nucleation proteins, actin filament polymerases and severing proteins. This group of proteins regulate the dynamic changes in actin assembly/disassembly, thus playing an important role in cell motility, intracellular transport, cell division and other basic cellular activities. Lymphocytes are important components of the human immune system, consisting of T-lymphocytes (T cells), B-lymphocytes (B cells) and natural killer cells (NK cells). Lymphocytes are indispensable for both innate and adaptive immunity and cannot function normally without various actin regulators. In this review, we first briefly introduce the structure and fundamental functions of a variety of well-known and newly discovered actin regulators, then we highlight the role of actin regulators in T cell, B cell and NK cell, and finally provide a landscape of various diseases associated with them. This review provides new directions in exploring actin regulators and promotes more precise and effective treatments for related diseases.
Collapse
Affiliation(s)
- Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Zhong
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Modification of BRCA1-associated breast cancer risk by HMMR overexpression. Nat Commun 2022; 13:1895. [PMID: 35393420 PMCID: PMC8989921 DOI: 10.1038/s41467-022-29335-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer risk for carriers of BRCA1 pathological variants is modified by genetic factors. Genetic variation in HMMR may contribute to this effect. However, the impact of risk modifiers on cancer biology remains undetermined and the biological basis of increased risk is poorly understood. Here, we depict an interplay of molecular, cellular, and tissue microenvironment alterations that increase BRCA1-associated breast cancer risk. Analysis of genome-wide association results suggests that diverse biological processes, including links to BRCA1-HMMR profiles, influence risk. HMMR overexpression in mouse mammary epithelium increases Brca1-mutant tumorigenesis by modulating the cancer cell phenotype and tumor microenvironment. Elevated HMMR activates AURKA and reduces ARPC2 localization in the mitotic cell cortex, which is correlated with micronucleation and activation of cGAS-STING and non-canonical NF-κB signaling. The initial tumorigenic events are genomic instability, epithelial-to-mesenchymal transition, and tissue infiltration of tumor-associated macrophages. The findings reveal a biological foundation for increased risk of BRCA1-associated breast cancer.
Collapse
|
22
|
Würtz M, Zupa E, Atorino ES, Neuner A, Böhler A, Rahadian AS, Vermeulen BJA, Tonon G, Eustermann S, Schiebel E, Pfeffer S. Modular assembly of the principal microtubule nucleator γ-TuRC. Nat Commun 2022; 13:473. [PMID: 35078983 PMCID: PMC8789826 DOI: 10.1038/s41467-022-28079-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractThe gamma-tubulin ring complex (γ-TuRC) is the principal microtubule nucleation template in vertebrates. Recent cryo-EM reconstructions visualized the intricate quaternary structure of the γ-TuRC, containing more than thirty subunits, raising fundamental questions about γ-TuRC assembly and the role of actin as an integral part of the complex. Here, we reveal the structural mechanism underlying modular γ-TuRC assembly and identify a functional role of actin in microtubule nucleation. During γ-TuRC assembly, a GCP6-stabilized core comprising GCP2-3-4-5-4-6 is expanded by stepwise recruitment, selective stabilization and conformational locking of four pre-formed GCP2-GCP3 units. Formation of the lumenal bridge specifies incorporation of the terminal GCP2-GCP3 unit and thereby leads to closure of the γ-TuRC ring in a left-handed spiral configuration. Actin incorporation into the complex is not relevant for γ-TuRC assembly and structural integrity, but determines γ-TuRC geometry and is required for efficient microtubule nucleation and mitotic chromosome alignment in vivo.
Collapse
|
23
|
Magistrati E, Maestrini G, Niño CA, Lince-Faria M, Beznoussenko G, Mironov A, Maspero E, Bettencourt-Dias M, Polo S. Myosin VI regulates ciliogenesis by promoting the turnover of the centrosomal/satellite protein OFD1. EMBO Rep 2021; 23:e54160. [PMID: 34957672 PMCID: PMC8892233 DOI: 10.15252/embr.202154160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022] Open
Abstract
The actin motor protein myosin VI is a multivalent protein with diverse functions. Here, we identified and characterised a myosin VI ubiquitous interactor, the oral‐facial‐digital syndrome 1 (OFD1) protein, whose mutations cause malformations of the face, oral cavity, digits and polycystic kidney disease. We found that myosin VI regulates the localisation of OFD1 at the centrioles and, as a consequence, the recruitment of the distal appendage protein Cep164. Myosin VI depletion in non‐tumoural cell lines causes an aberrant localisation of OFD1 along the centriolar walls, which is due to a reduction in the OFD1 mobile fraction. Finally, loss of myosin VI triggers a severe defect in ciliogenesis that could be, at least partially, ascribed to an impairment in the autophagic removal of OFD1 from satellites. Altogether, our results highlight an unprecedent layer of regulation of OFD1 and a pivotal role of myosin VI in coordinating the formation of the distal appendages and primary cilium with important implications for the genetic disorders known as ciliopathies.
Collapse
Affiliation(s)
- Elisa Magistrati
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Giorgia Maestrini
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Carlos A Niño
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | | | | | - Alexandre Mironov
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Elena Maspero
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | | | - Simona Polo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy.,Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
24
|
Tilwani S, Gandhi K, Narayan S, Ainavarapu SRK, Dalal SN. Disruption of desmosome function leads to increased centrosome clustering in 14-3-3γ-knockout cells with supernumerary centrosomes. FEBS Lett 2021; 595:2675-2690. [PMID: 34626438 DOI: 10.1002/1873-3468.14204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/09/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023]
Abstract
14-3-3 proteins are conserved, dimeric, acidic proteins that regulate multiple cellular pathways. Loss of either 14-3-3ε or 14-3-3γ leads to centrosome amplification. However, we find that while the knockout of 14-3-3ε leads to multipolar mitoses, the knockout of 14-3-3γ results in centrosome clustering and pseudo-bipolar mitoses. 14-3-3γ knockouts demonstrate compromised desmosome function and a decrease in keratin levels, leading to decreased cell stiffness and an increase in centrosome clustering. Restoration of desmosome function increased multipolar mitoses, whereas knockdown of either plakoglobin or keratin 5 led to decreased cell stiffness and increased pseudo-bipolar mitoses. These results suggest that the ability of the desmosome to anchor keratin filaments maintains cell stiffness, thus inhibiting centrosome clustering, and that phenotypes observed upon 14-3-3 loss reflect the dysregulation of multiple pathways.
Collapse
Affiliation(s)
- Sarika Tilwani
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Karan Gandhi
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Satya Narayan
- Department of Chemical Sciences, TIFR, Mumbai, India
| | | | - Sorab Nariman Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| |
Collapse
|
25
|
POLArIS, a versatile probe for molecular orientation, revealed actin filaments associated with microtubule asters in early embryos. Proc Natl Acad Sci U S A 2021; 118:2019071118. [PMID: 33674463 DOI: 10.1073/pnas.2019071118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Biomolecular assemblies govern the physiology of cells. Their function often depends on the changes in molecular arrangements of constituents, both in the positions and orientations. While recent advancements of fluorescence microscopy including super-resolution microscopy have enabled us to determine the positions of fluorophores with unprecedented accuracy, monitoring the orientation of fluorescently labeled molecules within living cells in real time is challenging. Fluorescence polarization microscopy (FPM) reports the orientation of emission dipoles and is therefore a promising solution. For imaging with FPM, target proteins need labeling with fluorescent probes in a sterically constrained manner, but because of difficulties in the rational three-dimensional design of protein connection, a universal method for constrained tagging with fluorophore was not available. Here, we report POLArIS, a genetically encoded and versatile probe for molecular orientation imaging. Instead of using a direct tagging approach, we used a recombinant binder connected to a fluorescent protein in a sterically constrained manner that can target specific biomolecules of interest by combining with phage display screening. As an initial test case, we developed POLArISact, which specifically binds to F-actin in living cells. We confirmed that the orientation of F-actin can be monitored by observing cells expressing POLArISact with FPM. In living starfish early embryos expressing POLArISact, we found actin filaments radially extending from centrosomes in association with microtubule asters during mitosis. By taking advantage of the genetically encoded nature, POLArIS can be used in a variety of living specimens, including whole bodies of developing embryos and animals, and also be expressed in a cell type/tissue specific manner.
Collapse
|
26
|
Abstract
Actin filaments and microtubules are cytoskeletal polymers that participate in many vital cell functions including division, morphogenesis, phagocytosis, and motility. Despite the persistent dogma that actin filament and microtubule networks are distinct in localization, structure, and function, a growing body of evidence shows that these elements are choreographed through intricate mechanisms sensitive to either polymer. Many proteins and cellular signals that mediate actin–microtubule interactions have already been identified. However, the impact of these regulators is typically assessed with actin filament or microtubule polymers alone, independent of the other system. Further, unconventional modes and regulators coordinating actin–microtubule interactions are still being discovered. Here we examine several methods of actin–microtubule crosstalk with an emphasis on the molecular links between both polymer systems and their higher-order interactions.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210.,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
27
|
Pereira SG, Dias Louro MA, Bettencourt-Dias M. Biophysical and Quantitative Principles of Centrosome Biogenesis and Structure. Annu Rev Cell Dev Biol 2021; 37:43-63. [PMID: 34314592 DOI: 10.1146/annurev-cellbio-120219-051400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The centrosome is a main orchestrator of the animal cellular microtubule cytoskeleton. Dissecting its structure and assembly mechanisms has been a goal of cell biologists for over a century. In the last two decades, a good understanding of the molecular constituents of centrosomes has been achieved. Moreover, recent breakthroughs in electron and light microscopy techniques have enabled the inspection of the centrosome and the mapping of its components with unprecedented detail. However, we now need a profound and dynamic understanding of how these constituents interact in space and time. Here, we review the latest findings on the structural and molecular architecture of the centrosome and how its biogenesis is regulated, highlighting how biophysical techniques and principles as well as quantitative modeling are changing our understanding of this enigmatic cellular organelle. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
|
28
|
Karlsson R, Dráber P. Profilin-A master coordinator of actin and microtubule organization in mammalian cells. J Cell Physiol 2021; 236:7256-7265. [PMID: 33821475 DOI: 10.1002/jcp.30379] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022]
Abstract
The last two decades have witnessed a tremendous increase in cell biology data. Not least is this true for studies of the dynamic organization of the microfilament and microtubule systems in animal cells where analyses of the molecular components and their interaction patterns have deepened our understanding of these complex force-generating machineries. Previous observations of a molecular cross-talk between the two systems have now led to the realization of the existence of several intricate mechanisms operating to maintain their coordinated cellular organization. In this short review, we relate to this development by discussing new results concerning the function of the actin regulator profilin 1 as a control component of microfilament-microtubule cross-talk.
Collapse
Affiliation(s)
- Roger Karlsson
- Department of Molecular Biosciences, WGI, Stockholm University, Stockholm, Sweden
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
29
|
Fokin AI, Gautreau AM. Assembly and Activity of the WASH Molecular Machine: Distinctive Features at the Crossroads of the Actin and Microtubule Cytoskeletons. Front Cell Dev Biol 2021; 9:658865. [PMID: 33869225 PMCID: PMC8047104 DOI: 10.3389/fcell.2021.658865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/12/2021] [Indexed: 01/10/2023] Open
Abstract
The Arp2/3 complex generates branched actin networks at different locations of the cell. The WASH and WAVE Nucleation Promoting Factors (NPFs) activate the Arp2/3 complex at the surface of endosomes or at the cell cortex, respectively. In this review, we will discuss how these two NPFs are controlled within distinct, yet related, multiprotein complexes. These complexes are not spontaneously assembled around WASH and WAVE, but require cellular assembly factors. The centrosome, which nucleates microtubules and branched actin, appears to be a privileged site for WASH complex assembly. The actin and microtubule cytoskeletons are both responsible for endosome shape and membrane remodeling. Motors, such as dynein, pull endosomes and extend membrane tubules along microtubule tracks, whereas branched actin pushes onto the endosomal membrane. It was recently uncovered that WASH assembles a super complex with dynactin, the major dynein activator, where the Capping Protein (CP) is exchanged from dynactin to the WASH complex. This CP swap initiates the first actin filament that primes the autocatalytic nucleation of branched actin at the surface of endosomes. Possible coordination between pushing and pulling forces in the remodeling of endosomal membranes is discussed.
Collapse
Affiliation(s)
- Artem I. Fokin
- Laboratoire de Biologie Structurale de la Cellule, CNRS, Ecole Polytechnique, IP Paris, Palaiseau, France
| | - Alexis M. Gautreau
- Laboratoire de Biologie Structurale de la Cellule, CNRS, Ecole Polytechnique, IP Paris, Palaiseau, France
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
30
|
Dimchev V, Lahmann I, Koestler SA, Kage F, Dimchev G, Steffen A, Stradal TEB, Vauti F, Arnold HH, Rottner K. Induced Arp2/3 Complex Depletion Increases FMNL2/3 Formin Expression and Filopodia Formation. Front Cell Dev Biol 2021; 9:634708. [PMID: 33598464 PMCID: PMC7882613 DOI: 10.3389/fcell.2021.634708] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/07/2021] [Indexed: 01/12/2023] Open
Abstract
The Arp2/3 complex generates branched actin filament networks operating in cell edge protrusion and vesicle trafficking. Here we employ a conditional knockout mouse model permitting tissue- or cell-type specific deletion of the murine Actr3 gene (encoding Arp3). A functional Actr3 gene appeared essential for fibroblast viability and growth. Thus, we developed cell lines for exploring the consequences of acute, tamoxifen-induced Actr3 deletion causing near-complete loss of functional Arp2/3 complex expression as well as abolished lamellipodia formation and membrane ruffling, as expected. Interestingly, Arp3-depleted cells displayed enhanced rather than reduced cell spreading, employing numerous filopodia, and showed little defects in the rates of random cell migration. However, both exploration of new space by individual cells and collective migration were clearly compromised by the incapability to efficiently maintain directionality of migration, while the principal ability to chemotax was only moderately affected. Examination of actin remodeling at the cell periphery revealed reduced actin turnover rates in Arp2/3-deficient cells, clearly deviating from previous sequestration approaches. Most surprisingly, induced removal of Arp2/3 complexes reproducibly increased FMNL formin expression, which correlated with the explosive induction of filopodia formation. Our results thus highlight both direct and indirect effects of acute Arp2/3 complex removal on actin cytoskeleton regulation.
Collapse
Affiliation(s)
- Vanessa Dimchev
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ines Lahmann
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Stefan A Koestler
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frieda Kage
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Georgi Dimchev
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Franz Vauti
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Hans-Henning Arnold
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Klemens Rottner
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Braunschweig Integrated Centre of Systems Biology (BRICS), Braunschweig, Germany
| |
Collapse
|
31
|
Luthold C, Varlet AA, Lambert H, Bordeleau F, Lavoie JN. Chaperone-Assisted Mitotic Actin Remodeling by BAG3 and HSPB8 Involves the Deacetylase HDAC6 and Its Substrate Cortactin. Int J Mol Sci 2020; 22:ijms22010142. [PMID: 33375626 PMCID: PMC7795263 DOI: 10.3390/ijms22010142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The fidelity of actin dynamics relies on protein quality control, but the underlying molecular mechanisms are poorly defined. During mitosis, the cochaperone BCL2-associated athanogene 3 (BAG3) modulates cell rounding, cortex stability, spindle orientation, and chromosome segregation. Mitotic BAG3 shows enhanced interactions with its preferred chaperone partner HSPB8, the autophagic adaptor p62/SQSTM1, and HDAC6, a deacetylase with cytoskeletal substrates. Here, we show that depletion of BAG3, HSPB8, or p62/SQSTM1 can recapitulate the same inhibition of mitotic cell rounding. Moreover, depletion of either of these proteins also interfered with the dynamic of the subcortical actin cloud that contributes to spindle positioning. These phenotypes were corrected by drugs that limit the Arp2/3 complex or HDAC6 activity, arguing for a role for BAG3 in tuning branched actin network assembly. Mechanistically, we found that cortactin acetylation/deacetylation is mitotically regulated and is correlated with a reduced association of cortactin with HDAC6 in situ. Remarkably, BAG3 depletion hindered the mitotic decrease in cortactin–HDAC6 association. Furthermore, expression of an acetyl-mimic cortactin mutant in BAG3-depleted cells normalized mitotic cell rounding and the subcortical actin cloud organization. Together, these results reinforce a BAG3′s function for accurate mitotic actin remodeling, via tuning cortactin and HDAC6 spatial dynamics.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada; (C.L.); (A.-A.V.); (H.L.)
- Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Alice-Anaïs Varlet
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada; (C.L.); (A.-A.V.); (H.L.)
- Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Herman Lambert
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada; (C.L.); (A.-A.V.); (H.L.)
- Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada; (C.L.); (A.-A.V.); (H.L.)
- Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (F.B.); (J.N.L.)
| | - Josée N. Lavoie
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada; (C.L.); (A.-A.V.); (H.L.)
- Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (F.B.); (J.N.L.)
| |
Collapse
|
32
|
Juanes MA. Cytoskeletal Control and Wnt Signaling-APC's Dual Contributions in Stem Cell Division and Colorectal Cancer. Cancers (Basel) 2020; 12:E3811. [PMID: 33348689 PMCID: PMC7766042 DOI: 10.3390/cancers12123811] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal epithelium architecture is sustained by stem cell division. In principle, stem cells can divide symmetrically to generate two identical copies of themselves or asymmetrically to sustain tissue renewal in a balanced manner. The choice between the two helps preserve stem cell and progeny pools and is crucial for tissue homeostasis. Control of spindle orientation is a prime contributor to the specification of symmetric versus asymmetric cell division. Competition for space within the niche may be another factor limiting the stem cell pool. An integrative view of the multiple links between intracellular and extracellular signals and molecular determinants at play remains a challenge. One outstanding question is the precise molecular roles of the tumour suppressor Adenomatous polyposis coli (APC) for sustaining gut homeostasis through its respective functions as a cytoskeletal hub and a down regulator in Wnt signalling. Here, we review our current understanding of APC inherent activities and partners in order to explore novel avenues by which APC may act as a gatekeeper in colorectal cancer and as a therapeutic target.
Collapse
Affiliation(s)
- M. Angeles Juanes
- School of Health and Life Science, Teesside University, Middlesbrough TS1 3BX, UK;
- National Horizons Centre, Teesside University, 38 John Dixon Lane, Darlington DL1 1HG, UK
| |
Collapse
|
33
|
Bornens M. Centrosome organization and functions. Curr Opin Struct Biol 2020; 66:199-206. [PMID: 33338884 DOI: 10.1016/j.sbi.2020.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
The centrosome, discovered near 1875, was named by Boveri when proposing the chromosomal theory of heredity. After a long eclipse, a considerable amount of molecular data has been accumulated on the centrosome and its biogenesis in the last 30 years, summarized regularly in excellent reviews. Major questions are still at stake in 2021 however, as we lack a comprehensive view of the centrosome functions. I will first try to see how progress towards a unified view of the role of centrosomes during evolution is possible, and then review recent data on only some of the many important questions raised by this organelle.
Collapse
Affiliation(s)
- Michel Bornens
- Institut Curie, PSL University, CNRS - UMR 144, 75005 Paris, France.
| |
Collapse
|
34
|
Nejedlá M, Klebanovych A, Sulimenko V, Sulimenko T, Dráberová E, Dráber P, Karlsson R. The actin regulator profilin 1 is functionally associated with the mammalian centrosome. Life Sci Alliance 2020; 4:4/1/e202000655. [PMID: 33184056 PMCID: PMC7668531 DOI: 10.26508/lsa.202000655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
The actin regulator profilin 1 recently shown to control microtubule elongation at the cell periphery is found to interact with the γ-tubulin ring complex and tune centrosomal microtubule nucleation. Profilin 1 is a crucial actin regulator, interacting with monomeric actin and several actin-binding proteins controlling actin polymerization. Recently, it has become evident that this profilin isoform associates with microtubules via formins and interferes with microtubule elongation at the cell periphery. Recruitment of microtubule-associated profilin upon extensive actin polymerizations, for example, at the cell edge, enhances microtubule growth, indicating that profilin contributes to the coordination of actin and microtubule organization. Here, we provide further evidence for the profilin-microtubule connection by demonstrating that it also functions in centrosomes where it impacts on microtubule nucleation.
Collapse
Affiliation(s)
- Michaela Nejedlá
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anastasiya Klebanovych
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vadym Sulimenko
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tetyana Sulimenko
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eduarda Dráberová
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Roger Karlsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
35
|
Pimm ML, Hotaling J, Henty-Ridilla JL. Profilin choreographs actin and microtubules in cells and cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:155-204. [PMID: 32859370 PMCID: PMC7461721 DOI: 10.1016/bs.ircmb.2020.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Actin and microtubules play essential roles in aberrant cell processes that define and converge in cancer including: signaling, morphology, motility, and division. Actin and microtubules do not directly interact, however shared regulators coordinate these polymers. While many of the individual proteins important for regulating and choreographing actin and microtubule behaviors have been identified, the way these molecules collaborate or fail in normal or disease contexts is not fully understood. Decades of research focus on Profilin as a signaling molecule, lipid-binding protein, and canonical regulator of actin assembly. Recent reports demonstrate that Profilin also regulates microtubule dynamics and polymerization. Thus, Profilin can coordinate both actin and microtubule polymer systems. Here we reconsider the biochemical and cellular roles for Profilin with a focus on the essential cytoskeletal-based cell processes that go awry in cancer. We also explore how the use of model organisms has helped to elucidate mechanisms that underlie the regulatory essence of Profilin in vivo and in the context of disease.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica Hotaling
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Jessica L Henty-Ridilla
- Department of Cell and Developmental Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
36
|
Biber G, Ben-Shmuel A, Sabag B, Barda-Saad M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:131-196. [PMID: 33066873 DOI: 10.1016/bs.ircmb.2020.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.
Collapse
Affiliation(s)
- G Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - A Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - B Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
37
|
Nunes V, Dantas M, Castro D, Vitiello E, Wang I, Carpi N, Balland M, Piel M, Aguiar P, Maiato H, Ferreira JG. Centrosome-nuclear axis repositioning drives the assembly of a bipolar spindle scaffold to ensure mitotic fidelity. Mol Biol Cell 2020; 31:1675-1690. [PMID: 32348198 PMCID: PMC7521851 DOI: 10.1091/mbc.e20-01-0047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
During the initial stages of cell division, the cytoskeleton is extensively reorganized so that a bipolar mitotic spindle can be correctly assembled. This process occurs through the action of molecular motors, cytoskeletal networks, and the nucleus. How the combined activity of these different components is spatiotemporally regulated to ensure efficient spindle assembly remains unclear. To investigate how cell shape, cytoskeletal organization, and molecular motors cross-talk to regulate initial spindle assembly, we use a combination of micropatterning with high-resolution imaging and 3D cellular reconstruction. We show that during prophase, centrosomes and nucleus reorient so that centrosomes are positioned on the shortest nuclear axis at nuclear envelope (NE) breakdown. We also find that this orientation depends on a combination of centrosome movement controlled by Arp2/3-mediated regulation of microtubule dynamics and Dynein-generated forces on the NE that regulate nuclear reorientation. Finally, we observe this centrosome configuration favors the establishment of an initial bipolar spindle scaffold, facilitating chromosome capture and accurate segregation, without compromising division plane orientation.
Collapse
Affiliation(s)
- Vanessa Nunes
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), 4200-135 Porto, Portugal.,BiotechHealth PhD program, Instituto de Ciências Biomédicas (ICBAS), 4050-313 Porto, Portugal
| | - Margarida Dantas
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), 4200-135 Porto, Portugal.,BiotechHealth PhD program, Instituto de Ciências Biomédicas (ICBAS), 4050-313 Porto, Portugal
| | - Domingos Castro
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto Nacional de Engenharia Biomédica (INEB), 4200-135 Porto, Portugal
| | - Elisa Vitiello
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1) 38058, France
| | - Irène Wang
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1) 38058, France
| | - Nicolas Carpi
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Martial Balland
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1) 38058, France
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Paulo Aguiar
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto Nacional de Engenharia Biomédica (INEB), 4200-135 Porto, Portugal
| | - Helder Maiato
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), 4200-135 Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina do Porto, 4200-450 Porto, Portugal
| | - Jorge G Ferreira
- Instituto de Investigação e Inovação em Saúde (i3S), 4200-135 Porto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), 4200-135 Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina do Porto, 4200-450 Porto, Portugal
| |
Collapse
|
38
|
Artificially decreasing cortical tension generates aneuploidy in mouse oocytes. Nat Commun 2020; 11:1649. [PMID: 32245998 PMCID: PMC7125192 DOI: 10.1038/s41467-020-15470-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/10/2020] [Indexed: 01/28/2023] Open
Abstract
Human and mouse oocytes’ developmental potential can be predicted by their mechanical properties. Their development into blastocysts requires a specific stiffness window. In this study, we combine live-cell and computational imaging, laser ablation, and biophysical measurements to investigate how deregulation of cortex tension in the oocyte contributes to early developmental failure. We focus on extra-soft cells, the most common defect in a natural population. Using two independent tools to artificially decrease cortical tension, we show that chromosome alignment is impaired in extra-soft mouse oocytes, despite normal spindle morphogenesis and dynamics, inducing aneuploidy. The main cause is a cytoplasmic increase in myosin-II activity that could sterically hinder chromosome capture. We describe here an original mode of generation of aneuploidies that could be very common in oocytes and could contribute to the high aneuploidy rate observed during female meiosis, a leading cause of infertility and congenital disorders. The developmental potential of human and murine oocytes is predicted by their mechanical properties. Here the authors show that artificial reduction of cortex tension produces aneuploid mouse oocytes and speculate that this may contribute to the high aneuploidy rate typical of female meiosis.
Collapse
|
39
|
Actin-based regulation of ciliogenesis - The long and the short of it. Semin Cell Dev Biol 2019; 102:132-138. [PMID: 31862221 DOI: 10.1016/j.semcdb.2019.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/23/2019] [Accepted: 12/07/2019] [Indexed: 12/11/2022]
Abstract
The primary cilia is found on the mammalian cell surface where it serves as an antenna for the reception and transmission of a variety of cellular signaling pathways. At its core the cilium is a microtubule-based organelle, but it is clear that its assembly and function are dependent upon the coordinated regulation of both actin and microtubule dynamics. In particular, the discovery that the centrosome is able to act as both a microtubule and actin organizing centre implies that both cytoskeletal networks are acting directly on the process of cilia assembly. In this review, we set our recent results with the formin FHDC1 in the context of current reports that show each stage of ciliogenesis is impacted by changes in actin dynamics. These include direct effects of actin filament assembly on basal body positioning, vesicle trafficking to and entry into the cilium, cilia length, cilia membrane organization and cilia-dependent signaling.
Collapse
|
40
|
Helassa N, Nugues C, Rajamanoharan D, Burgoyne RD, Haynes LP. A centrosome-localized calcium signal is essential for mammalian cell mitosis. FASEB J 2019; 33:14602-14610. [PMID: 31682764 PMCID: PMC6910830 DOI: 10.1096/fj.201901662r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/23/2019] [Indexed: 02/02/2023]
Abstract
Mitosis defects can lead to premature ageing and cancer. Understanding mitosis regulation therefore has important implications for human disease. Early data suggested that calcium (Ca2+) signals could influence mitosis, but these have hitherto not been observed in mammalian cells. Here, we reveal a prolonged yet spatially restricted Ca2+ signal at the centrosomes of actively dividing cells. Local buffering of the centrosomal Ca2+ signals, by flash photolysis of the caged Ca2+ chelator diazo-2-acetoxymethyl ester, arrests mitosis. We also provide evidence that this Ca2+ signal emanates from the endoplasmic reticulum. In summary, we characterize a unique centrosomal Ca2+ signal as a functionally essential input into mitosis.-Helassa, N., Nugues, C., Rajamanoharan, D., Burgoyne, R. D., Haynes, L. P. A centrosome-localized calcium signal is essential for mammalian cell mitosis.
Collapse
Affiliation(s)
- Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Charlotte Nugues
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Dayani Rajamanoharan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Robert D. Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Kita AM, Swider ZT, Erofeev I, Halloran MC, Goryachev AB, Bement WM. Spindle-F-actin interactions in mitotic spindles in an intact vertebrate epithelium. Mol Biol Cell 2019; 30:1645-1654. [PMID: 31091161 PMCID: PMC6727749 DOI: 10.1091/mbc.e19-02-0126] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Mitotic spindles are well known to be assembled from and dependent on microtubules. In contrast, whether actin filaments (F-actin) are required for or are even present in mitotic spindles has long been controversial. Here we have developed improved methods for simultaneously preserving F-actin and microtubules in fixed samples and exploited them to demonstrate that F-actin is indeed associated with mitotic spindles in intact Xenopus laevis embryonic epithelia. We also find that there is an “F-actin cycle,” in which the distribution and organization of spindle F-actin changes over the course of the cell cycle. Live imaging using a probe for F-actin reveals that at least two pools of F-actin are associated with mitotic spindles: a relatively stable internal network of cables that moves in concert with and appears to be linked to spindles, and F-actin “fingers” that rapidly extend from the cell cortex toward the spindle and make transient contact with the spindle poles. We conclude that there is a robust endoplasmic F-actin network in normal vertebrate epithelial cells and that this network is also a component of mitotic spindles. More broadly, we conclude that there is far more internal F-actin in epithelial cells than is commonly believed.
Collapse
Affiliation(s)
- Angela M Kita
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Zachary T Swider
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Ivan Erofeev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3JD, United Kingdom
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53706
| | - Andrew B Goryachev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3JD, United Kingdom
| | - William M Bement
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|