1
|
Ugorets V, Mendez PL, Zagrebin D, Russo G, Kerkhoff Y, Kotsaris G, Jatzlau J, Stricker S, Knaus P. Dynamic remodeling of septin structures fine-tunes myogenic differentiation. iScience 2024; 27:110630. [PMID: 39246450 PMCID: PMC11380178 DOI: 10.1016/j.isci.2024.110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/02/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Controlled myogenic differentiation is integral to the development, maintenance and repair of skeletal muscle, necessitating precise regulation of myogenic progenitors and resident stem cells. The transformation of proliferative muscle progenitors into multinuclear syncytia involves intricate cellular processes driven by cytoskeletal reorganization. While actin and microtubles have been extensively studied, we illuminate the role of septins, an essential yet still often overlooked cytoskeletal component, in myoblast architecture. Notably, Septin9 emerges as a critical regulator of myoblast differentiation during the initial commitment phase. Knock-down of Septin9 in C2C12 cells and primary mouse myoblasts accelerates the transition from proliferation to committed progenitor transcriptional programs. Furthermore, we unveil significant reorganization and downregulation of Septin9 during myogenic differentiation. Collectively, we propose that filmamentous septin structures and their orchestrated reorganization in myoblasts are part of a temporal regulatory mechanism governing the differentiation of myogenic progenitors. This study sheds light on the dynamic interplay between cytoskeletal components underlying controlled myogenic differentiation.
Collapse
Affiliation(s)
- Vladimir Ugorets
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Paul-Lennard Mendez
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, IMPRS-Biology and Computation, 14195 Berlin, Germany
| | - Dmitrii Zagrebin
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Giulia Russo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Yannic Kerkhoff
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Bionanointerfaces Group, 14195 Berlin, Germany
| | - Georgios Kotsaris
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, 14195 Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Sigmar Stricker
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, 14195 Berlin, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
2
|
Connors CQ, Mauro MS, Wiles JT, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by reducing septin and anillin levels at the cell division plane. Mol Biol Cell 2024; 35:ar94. [PMID: 38696255 PMCID: PMC11244169 DOI: 10.1091/mbc.e24-02-0096-t] [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: 03/08/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024] Open
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formincyk-1(ts) mutant Caenorhabditis elegans 4-cell embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide with greatly reduced F-actin levels at the cell division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septinUNC-59 and anillinANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into the regulation of cytokinesis in other cell types, especially in stem cells with high potency.
Collapse
Affiliation(s)
- Caroline Q. Connors
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Michael S. Mauro
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - J. Tristian Wiles
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | | - Sophia L. Martin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Benjamin Lacroix
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Université de Montpellier, CNRS, Centre de Recherche en Biologie Cellulaire de Montpellier, UMR 5237 Montpellier, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Karen E. Kasza
- Department of Mechanical Engineering, Columbia University, New York, NY 10027
| | - Timothy R. Davies
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Julie C. Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| |
Collapse
|
3
|
Varela Salgado M, Adriaans IE, Touati SA, Ibanes S, Lai-Kee-Him J, Ancelin A, Cipelletti L, Picas L, Piatti S. Phosphorylation of the F-BAR protein Hof1 drives septin ring splitting in budding yeast. Nat Commun 2024; 15:3383. [PMID: 38649354 PMCID: PMC11035697 DOI: 10.1038/s41467-024-47709-3] [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: 07/12/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
A double septin ring accompanies cytokinesis in yeasts and mammalian cells. In budding yeast, reorganisation of the septin collar at the bud neck into a dynamic double ring is essential for actomyosin ring constriction and cytokinesis. Septin reorganisation requires the Mitotic Exit Network (MEN), a kinase cascade essential for cytokinesis. However, the effectors of MEN in this process are unknown. Here we identify the F-BAR protein Hof1 as a critical target of MEN in septin remodelling. Phospho-mimicking HOF1 mutant alleles overcome the inability of MEN mutants to undergo septin reorganisation by decreasing Hof1 binding to septins and facilitating its translocation to the actomyosin ring. Hof1-mediated septin rearrangement requires its F-BAR domain, suggesting that it may involve a local membrane remodelling that leads to septin reorganisation. In vitro Hof1 can induce the formation of intertwined septin bundles, while a phosphomimetic Hof1 protein has impaired septin-bundling activity. Altogether, our data indicate that Hof1 modulates septin architecture in distinct ways depending on its phosphorylation status.
Collapse
Affiliation(s)
- Maritzaida Varela Salgado
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Ingrid E Adriaans
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Sandra A Touati
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Sandy Ibanes
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Joséphine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Luca Cipelletti
- L2C (Laboratoire Charles Coulomb), University of Montpellier, CNRS 34095, Montpellier, France
- IUF (Institut Universitaire de France, 75231, Paris, France
| | - Laura Picas
- IRIM (Institut de Recherche en Infectiologie de Montpellier), University of Montpellier, CNRS UMR 9004, 34293, Montpellier, France
| | - Simonetta Piatti
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France.
| |
Collapse
|
4
|
Connors CQ, Mauro MS, Tristian Wiles J, Countryman AD, Martin SL, Lacroix B, Shirasu-Hiza M, Dumont J, Kasza KE, Davies TR, Canman JC. Germ fate determinants protect germ precursor cell division by restricting septin and anillin levels at the division plane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.566773. [PMID: 38014027 PMCID: PMC10680835 DOI: 10.1101/2023.11.17.566773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Animal cell cytokinesis, or the physical division of one cell into two, is thought to be driven by constriction of an actomyosin contractile ring at the division plane. The mechanisms underlying cell type-specific differences in cytokinesis remain unknown. Germ cells are totipotent cells that pass genetic information to the next generation. Previously, using formin cyk-1 (ts) mutant C. elegans embryos, we found that the P2 germ precursor cell is protected from cytokinesis failure and can divide without detectable F-actin at the division plane. Here, we identified two canonical germ fate determinants required for P2-specific cytokinetic protection: PIE-1 and POS-1. Neither has been implicated previously in cytokinesis. These germ fate determinants protect P2 cytokinesis by reducing the accumulation of septin UNC-59 and anillin ANI-1 at the division plane, which here act as negative regulators of cytokinesis. These findings may provide insight into cytokinetic regulation in other cell types, especially in stem cells with high potency.
Collapse
|
5
|
Gorry RL, Brennan K, Lavin PTM, Mazurski T, Mary C, Matallanas D, Guichou JF, Mc Gee MM. Cyclophilin A Isomerisation of Septin 2 Mediates Abscission during Cytokinesis. Int J Mol Sci 2023; 24:11084. [PMID: 37446263 PMCID: PMC10341793 DOI: 10.3390/ijms241311084] [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: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The isomerase activity of Cyclophilin A is important for midbody abscission during cell division, however, to date, midbody substrates remain unknown. In this study, we report that the GTP-binding protein Septin 2 interacts with Cyclophilin A. We highlight a dynamic series of Septin 2 phenotypes at the midbody, previously undescribed in human cells. Furthermore, Cyclophilin A depletion or loss of isomerase activity is sufficient to induce phenotypic Septin 2 defects at the midbody. Structural and molecular analysis reveals that Septin 2 proline 259 is important for interaction with Cyclophilin A. Moreover, an isomerisation-deficient EGFP-Septin 2 proline 259 mutant displays defective midbody localisation and undergoes impaired abscission, which is consistent with data from cells with loss of Cyclophilin A expression or activity. Collectively, these data reveal Septin 2 as a novel interacting partner and isomerase substrate of Cyclophilin A at the midbody that is required for abscission during cytokinesis in cancer cells.
Collapse
Affiliation(s)
- Rebecca L. Gorry
- School of Biomolecular and Biomedical Science (SBBS), Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland (K.B.)
| | - Kieran Brennan
- School of Biomolecular and Biomedical Science (SBBS), Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland (K.B.)
| | - Paul T. M. Lavin
- School of Biomolecular and Biomedical Science (SBBS), Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland (K.B.)
| | - Tayler Mazurski
- School of Biomolecular and Biomedical Science (SBBS), Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland (K.B.)
| | - Charline Mary
- Centre de Biologie Structurale, CNRS, INSERM, University Montpellier, 34090 Montpellier, France
| | - David Matallanas
- Systems Biology Ireland (SBI), School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, University Montpellier, 34090 Montpellier, France
| | - Margaret M. Mc Gee
- School of Biomolecular and Biomedical Science (SBBS), Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland (K.B.)
| |
Collapse
|
6
|
Carim SC, Hickson GR. The Rho1 GTPase controls anillo-septin assembly to facilitate contractile ring closure during cytokinesis. iScience 2023; 26:106903. [PMID: 37378349 PMCID: PMC10291328 DOI: 10.1016/j.isci.2023.106903] [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: 07/14/2021] [Revised: 03/20/2023] [Accepted: 05/12/2023] [Indexed: 06/29/2023] Open
Abstract
Animal cell cytokinesis requires activation of the GTPase RhoA (Rho1 in Drosophila), which assembles an F-actin- and myosin II-dependent contractile ring (CR) at the equatorial plasma membrane. CR closure is poorly understood, but involves the multidomain scaffold protein, Anillin. Anillin binds many CR components including F-actin and myosin II (collectively actomyosin), RhoA and the septins. Anillin recruits septins to the CR but the mechanism is unclear. Live imaging of Drosophila S2 cells and HeLa cells revealed that the Anillin N-terminus, which scaffolds actomyosin, cannot recruit septins to the CR. Rather, septin recruitment required the ability of the Anillin C-terminus to bind Rho1-GTP and the presence of the Anillin PH domain, in a sequential mechanism occurring at the plasma membrane, independently of F-actin. Anillin mutations that blocked septin recruitment, but not actomyosin scaffolding, slowed CR closure and disrupted cytokinesis. Thus, CR closure requires coordination of two Rho1-dependent networks: actomyosin and anillo-septin.
Collapse
Affiliation(s)
- Sabrya C. Carim
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Gilles R.X. Hickson
- CHU Sainte-Justine Research Center, 3175 Chemin de la Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
7
|
Benoit B, Poüs C, Baillet A. Septins as membrane influencers: direct play or in association with other cytoskeleton partners. Front Cell Dev Biol 2023; 11:1112319. [PMID: 36875762 PMCID: PMC9982393 DOI: 10.3389/fcell.2023.1112319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
The cytoskeleton comprises three polymerizing structures that have been studied for a long time, actin microfilaments, microtubules and intermediate filaments, plus more recently investigated dynamic assemblies like septins or the endocytic-sorting complex required for transport (ESCRT) complex. These filament-forming proteins control several cell functions through crosstalks with each other and with membranes. In this review, we report recent works that address how septins bind to membranes, and influence their shaping, organization, properties and functions, either by binding to them directly or indirectly through other cytoskeleton elements.
Collapse
Affiliation(s)
- Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France.,Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Hôpitaux Universitaires Paris-Saclay, Clamart, France
| | - Anita Baillet
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| |
Collapse
|
8
|
Kumari A, Ghosh A, Kolay S, Raghu P. Septins tune lipid kinase activity and PI(4,5)P 2 turnover during G-protein–coupled PLC signalling in vivo. Life Sci Alliance 2022; 5:5/6/e202101293. [PMID: 35277468 PMCID: PMC8921834 DOI: 10.26508/lsa.202101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/24/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] hydrolysis by phospholipase C (PLC) is a conserved mechanism of signalling. Given the low abundance of PI(4,5)P2, its hydrolysis needs to be coupled to resynthesis to ensure continued PLC activity; however, the mechanism by which depletion is coupled to resynthesis remains unknown. PI(4,5)P2 synthesis is catalyzed by the phosphorylation of phosphatidylinositol 4 phosphate (PI4P) by phosphatidylinositol 4 phosphate 5 kinase (PIP5K). In Drosophila photoreceptors, photon absorption is transduced into PLC activity and during this process, PI(4,5)P2 is resynthesized by a PIP5K. However, the mechanism by which PIP5K activity is coupled to PI(4,5)P2 hydrolysis is unknown. In this study, we identify a unique isoform dPIP5KL, that is both necessary and sufficient to mediate PI(4,5)P2 synthesis during phototransduction. Depletion of PNUT, a non-redundant subunit of the septin family, enhances dPIP5KL activity in vitro and PI(4,5)P2 resynthesis in vivo; co-depletion of dPIP5KL reverses the enhanced rate of PI(4,5)P2 resynthesis in vivo. Thus, our work defines a septin-mediated mechanism through which PIP5K activity is coupled to PLC-mediated PI(4,5)P2 hydrolysis.
Collapse
Affiliation(s)
- Aastha Kumari
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
| | - Avishek Ghosh
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sourav Kolay
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
- UT Southwestern Medical Center, Dallas, TX, USA
| | - Padinjat Raghu
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
| |
Collapse
|
9
|
Menon MB, Yakovleva T, Ronkina N, Suwandi A, Odak I, Dhamija S, Sandrock I, Hansmann F, Baumgärtner W, Förster R, Kotlyarov A, Gaestel M. Lyz2-Cre-Mediated Genetic Deletion of Septin7 Reveals a Role of Septins in Macrophage Cytokinesis and Kras-Driven Tumorigenesis. Front Cell Dev Biol 2022; 9:795798. [PMID: 35071236 PMCID: PMC8772882 DOI: 10.3389/fcell.2021.795798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
By crossing septin7-floxed mice with Lyz2-Cre mice carrying the Cre recombinase inserted in the Lysozyme-M (Lyz2) gene locus we aimed the specific deletion of septin7 in myeloid cells, such as monocytes, macrophages and granulocytes. Septin7 flox/flox :Lyz2-Cre mice show no alterations in the myeloid compartment. Septin7-deleted macrophages (BMDMs) were isolated and analyzed. The lack of Septin7 expression was confirmed and a constitutive double-nucleation was detected in Septin7-deficient BMDMs indicating a defect in macrophage cytokinesis. However, phagocytic function of macrophages as judged by uptake of labelled E. coli particles and LPS-stimulated macrophage activation as judged by induction of TNF mRNA expression and TNF secretion were not compromised. In addition to myeloid cells, Lyz2-Cre is also active in type II pneumocytes (AT2 cells). We monitored lung adenocarcinoma formation in these mice by crossing them with the conditional knock-in Kras-LSL-G12D allele. Interestingly, we found that control mice without septin7 depletion die after 3-5 weeks, while the Septin7-deficient animals survived 11 weeks or even longer. Control mice sacrificed in the age of 4 weeks display a bronchiolo-alveolar hyperplasia with multiple adenomas, whereas the Septin7-deficient animals of the same age are normal or show only a weak multifocal brochiolo-alveolar hyperplasia. Our findings indicate an essential role of Septin7 in macrophage cytokinesis but not in macrophage function. Furthermore, septin7 seems absolutely essential for oncogenic Kras-driven lung tumorigenesis making it a potential target for anti-tumor interventions.
Collapse
Affiliation(s)
- Manoj B Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany.,Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Tatiana Yakovleva
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Natalia Ronkina
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Abdulhadi Suwandi
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Sonam Dhamija
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany.,Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Florian Hansmann
- Institute of Pathology, Stiftung Tierärztliche Hochschule, Hannover, Germany.,Institute of Veterinary Pathology, Veterinary Faculty of Leipzig University, Leipzig, Germany
| | | | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Alexey Kotlyarov
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Schuster T, Geiger H. Septins in Stem Cells. Front Cell Dev Biol 2021; 9:801507. [PMID: 34957123 PMCID: PMC8695968 DOI: 10.3389/fcell.2021.801507] [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: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 12/01/2022] Open
Abstract
Septins were first described in yeast. Due to extensive research in non-yeast cells, Septins are now recognized across all species as important players in the regulation of the cytoskeleton, in the establishment of polarity, for migration, vesicular trafficking and scaffolding. Stem cells are primarily quiescent cells, and this actively maintained quiescent state is critical for proper stem cell function. Equally important though, stem cells undergo symmetric or asymmetric division, which is likely linked to the level of symmetry found in the mother stem cell. Due to the ability to organize barriers and be able to break symmetry in cells, Septins are thought to have a significant impact on organizing quiescence as well as the mode (symmetric vs asymmetric) of stem cell division to affect self-renewal versus differentiation. Mechanisms of regulating mammalian quiescence and symmetry breaking by Septins are though still somewhat elusive. Within this overview article, we summarize current knowledge on the role of Septins in stem cells ranging from yeast to mice especially with respect to quiescence and asymmetric division, with a special focus on hematopoietic stem cells.
Collapse
Affiliation(s)
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
11
|
Kandi R, Senger K, Grigoryan A, Soller K, Sakk V, Schuster T, Eiwen K, Menon MB, Gaestel M, Zheng Y, Florian MC, Geiger H. Cdc42-Borg4-Septin7 axis regulates HSC polarity and function. EMBO Rep 2021; 22:e52931. [PMID: 34661963 PMCID: PMC8647144 DOI: 10.15252/embr.202152931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/03/2023] Open
Abstract
Aging of hematopoietic stem cells (HSCs) is caused by the elevated activity of the small RhoGTPase Cdc42 and an apolar distribution of proteins. Mechanisms by which Cdc42 activity controls polarity of HSCs are not known. Binder of RhoGTPases proteins (Borgs) are known effector proteins of Cdc42 that are able to regulate the cytoskeletal Septin network. Here, we show that Cdc42 interacts with Borg4, which in turn interacts with Septin7 to regulate the polar distribution of Cdc42, Borg4, and Septin7 within HSCs. Genetic deletion of either Borg4 or Septin7 results in a reduced frequency of HSCs polar for Cdc42 or Borg4 or Septin7, a reduced engraftment potential and decreased lymphoid‐primed multipotent progenitor (LMPP) frequency in the bone marrow. Taken together, our data identify a Cdc42‐Borg4‐Septin7 axis essential for the maintenance of polarity within HSCs and for HSC function and provide a rationale for further investigating the role of Borgs and Septins in the regulation of compartmentalization within stem cells.
Collapse
Affiliation(s)
- Ravinder Kandi
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | | | - Ani Grigoryan
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Karin Soller
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Tanja Schuster
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Karina Eiwen
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| | - Manoj B Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany.,Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
12
|
McNeely KC, Dwyer ND. Cytokinetic Abscission Regulation in Neural Stem Cells and Tissue Development. CURRENT STEM CELL REPORTS 2021; 7:161-173. [PMID: 36303610 PMCID: PMC9603694 DOI: 10.1007/s40778-021-00193-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Purpose of Review How stem cells balance proliferation with differentiation, giving rise to specific daughter cells during development to build an embryo or tissue, remains an open question. Here, we discuss recent evidence that cytokinetic abscission regulation in stem cells, particularly neural stem cells (NSCs), is part of the answer. Abscission is a multi-step process mediated by the midbody, a microtubule-based structure formed in the intercellular bridge between daughter cells after mitosis. Recent Findings Human mutations and mouse knockouts in abscission genes reveal that subtle disruptions of NSC abscission can cause brain malformations. Experiments in several epithelial systems have shown that midbodies serve as scaffolds for apical junction proteins and are positioned near apical membrane fate determinants. Abscission timing is tightly controlled and developmentally regulated in stem cells, with delayed abscission in early embryos and faster abscission later. Midbody remnants (MBRs) contain over 400 proteins and may influence polarity, fate, and ciliogenesis. Summary As NSCs and other stem cells build tissues, they tightly regulate three aspects of abscission: midbody positioning, duration, and MBR handling. Midbody positioning and remnants establish or maintain cell polarity. MBRs are deposited on the apical membranes of epithelia, can be released or internalized by surrounding cells, and may sequester fate determinants or transfer information between cells. Work in cell lines and simpler systems has shown multiple roles for abscission regulation influencing stem cell polarity, potency, and daughter fates during development. Elucidating how the abscission process influences cell fate and tissue growth is important for our continued understanding of brain development and stem cell biology.
Collapse
|
13
|
Septin4 promotes cardiomyocytes apoptosis by enhancing the VHL-mediated degradation of HIF-1α. Cell Death Discov 2021; 7:172. [PMID: 34230460 PMCID: PMC8260662 DOI: 10.1038/s41420-021-00563-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/12/2021] [Accepted: 06/13/2021] [Indexed: 11/09/2022] Open
Abstract
Septin4, a protein localized at mitochondrion, can promote cells apoptosis mainly by binding XIAP (X-linked inhibitors of apoptosis), however, nothing is known about the role and mechanism of Septin4 in cardiomyocytes apoptosis. Here in the current study, we report that HIF-1α (hypoxia-inducible factor 1 alpha) is a novel interacting protein with Septin4 at Septin4-GTPase domain. In addition, Septin4 enhances the binding between HIF-1α and the E3 ubiquitin ligase VHL (von Hippel-Lindau protein) to down-regulate HIF-1α, and by reducing cardio-protective factor HIF-1α levels, Septin4 aggravated the hypoxia-induced cardiomyocytes apoptosis. We believe these findings will be beneficial to provide effective strategies for clinical treatment of myocardial ischemia and the subsequent injury caused by myocardial hypoxia.
Collapse
|
14
|
Horváth P, Müller-Reichert T. A Structural View on ESCRT-Mediated Abscission. Front Cell Dev Biol 2020; 8:586880. [PMID: 33240884 PMCID: PMC7680848 DOI: 10.3389/fcell.2020.586880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/16/2020] [Indexed: 11/25/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) mediates cellular processes that are related to membrane remodeling, such as multivesicular body (MVB) formation, viral budding and cytokinesis. Abscission is the final stage of cytokinesis that results in the physical separation of the newly formed two daughter cells. Although abscission has been investigated for decades, there are still fundamental open questions related to the spatio-temporal organization of the molecular machinery involved in this process. Reviewing knowledge obtained from in vitro as well as in vivo experiments, we give a brief overview on the role of ESCRT components in abscission mainly focussing on mammalian cells.
Collapse
Affiliation(s)
- Péter Horváth
- Experimental Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thomas Müller-Reichert
- Experimental Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
15
|
Castillo-Badillo JA, Bandi AC, Harlalka S, Gautam N. SRRF-Stream Imaging of Optogenetically Controlled Furrow Formation Shows Localized and Coordinated Endocytosis and Exocytosis Mediating Membrane Remodeling. ACS Synth Biol 2020; 9:902-919. [PMID: 32155337 DOI: 10.1021/acssynbio.9b00521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cleavage furrow formation during cytokinesis involves extensive membrane remodeling. In the absence of methods to exert dynamic control over these processes, it has been a challenge to examine the basis of this remodeling. Here we used a subcellular optogenetic approach to induce this at will and found that furrow formation is mediated by actomyosin contractility, retrograde plasma membrane flow, localized decrease in membrane tension, and endocytosis. FRAP, 4-D imaging, and inhibition or upregulation of endocytosis or exocytosis show that ARF6 and Exo70 dependent localized exocytosis supports a potential model for intercellular bridge elongation. TIRF and Super Resolution Radial Fluctuation (SRRF) stream microscopy show localized VAMP2-mediated exocytosis and incorporation of membrane lipids from vesicles into the plasma membrane at the front edge of the nascent daughter cell. Thus, spatially separated but coordinated plasma membrane depletion and addition are likely contributors to membrane remodeling during cytokinetic processes.
Collapse
|
16
|
Qiu R, Runxiang Q, Geng A, Liu J, Xu CW, Menon MB, Gaestel M, Lu Q. SEPT7 Interacts with KIF20A and Regulates the Proliferative State of Neural Progenitor Cells During Cortical Development. Cereb Cortex 2019; 30:3030-3043. [PMID: 31813992 DOI: 10.1093/cercor/bhz292] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/01/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Balanced proliferation and differentiation of neural progenitor cells (NPCs) are critical for brain development, but how the process is regulated and what components of the cell division machinery is involved are not well understood. Here we report that SEPT7, a cell division regulator originally identified in Saccharomyces cerevisiae, interacts with KIF20A in the intercellular bridge of dividing NPCs and plays an essential role in maintaining the proliferative state of NPCs during cortical development. Knockdown of SEPT7 in NPCs results in displacement of KIF20A from the midbody and early neuronal differentiation. NPC-specific inducible knockout of Sept7 causes early cell cycle exit, precocious neuronal differentiation, and ventriculomegaly in the cortex, but surprisingly does not lead to noticeable cytokinesis defect. Our data uncover an interaction of SEPT7 and KIF20A during NPC divisions and demonstrate a crucial role of SEPT7 in cell fate determination. In addition, this study presents a functional approach for identifying additional cell fate regulators of the mammalian brain.
Collapse
Affiliation(s)
- Runxiang Qiu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Qiu Runxiang
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Anqi Geng
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.,Institute of Medical Research, Northwestern Polytechnical University, Xian, Shaanxi Province, China
| | - Jiancheng Liu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - C Wilson Xu
- Balto Pharmaceuticals, Inc., South Pasadena, CA 91030, USA
| | - Manoj B Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany.,Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New-Delhi 110016, India
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany
| | - Qiang Lu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
17
|
He H, Li J, Xu M, Kan Z, Gao Y, Yuan C. Expression of septin 2 and association with clinicopathological parameters in colorectal cancer. Oncol Lett 2019; 18:2376-2383. [PMID: 31402940 PMCID: PMC6676678 DOI: 10.3892/ol.2019.10528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/07/2019] [Indexed: 12/16/2022] Open
Abstract
Septin 2 (SEPT2) is a tumor-related gene belonging to the SEPT family that affects the cellular processes of hepatoma carcinoma cells, glioblastoma cells and mesangial cells and is highly expressed in breast cancer, biliary tract cancer and acute myeloid leukemia. Colorectal cancer (CRC) is the third most common type of malignancy in humans. In the present study, Oncomine database was used to compare the expression pattern of SEPT2 mRNA between CRC and normal tissues. Additionally, protein expression in 90 pairs of CRC and paracancerous tissues was analyzed by western blotting and immunohistochemistry (IHC). The results showed that SEPT2 was highly expressed in CRC tissues at the mRNA and protein levels. SEPT2 expression quantified by IHC was associated with lymph node metastasis, the degree of differentiation and TNM staging. Increased SEPT2 wass associated with reduced overall survival (OS) according to Kaplan-Meier analysis. COX proportional hazard analysis indicated that SEPT2 was an independent factor that influenced the OS of patients with CRC. Therefore, SEPT2 was associated with the occurrence, progression and prognosis of CRC and thus, may be a marker and prognostic indicator of CRC.
Collapse
Affiliation(s)
- Haoyu He
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Junjun Li
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Meng Xu
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Ziliang Kan
- Graduate School, Singapore Management University, Singapore 178903, Republic of Singapore
| | - Yang Gao
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Caijun Yuan
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
18
|
Falk J, Boubakar L, Castellani V. Septin functions during neuro-development, a yeast perspective. Curr Opin Neurobiol 2019; 57:102-109. [DOI: 10.1016/j.conb.2019.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/24/2022]
|
19
|
Katz ZB, Zhang C, Quintana A, Lillemeier BF, Hogan PG. Septins organize endoplasmic reticulum-plasma membrane junctions for STIM1-ORAI1 calcium signalling. Sci Rep 2019; 9:10839. [PMID: 31346209 PMCID: PMC6658532 DOI: 10.1038/s41598-019-46862-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/28/2019] [Indexed: 12/21/2022] Open
Abstract
ORAI1 Ca2+ channels in the plasma membrane (PM) are gated by STIM1 at endoplasmic reticulum (ER)-PM junctions to effect store-dependent Ca2+ entry into cells, but little is known about how local STIM-ORAI signalling at junctions is coordinated with overall cellular architecture. Filamentous septins can specify cytoskeletal rearrangements and have been found recently to modulate STIM-ORAI signalling. Here we show by super-resolution imaging of ORAI1, STIM1, and septin 4 in living cells that septins facilitate Ca2+ signalling indirectly. Septin 4 does not colocalize preferentially with ORAI1 in resting or stimulated cells, assemble stably at ER-PM junctions, or specify a boundary that directs or confines ORAI1 to junctions. Rather, ORAI1 is recruited to junctions solely through interaction with STIM proteins, while septins regulate the number of ER-PM junctions and enhance STIM1-ORAI1 interactions within junctions. Thus septins communicate with STIM1 and ORAI1 through protein or lipid intermediaries, and are favorably positioned to coordinate Ca2+ signalling with rearrangements in cellular architecture.
Collapse
Affiliation(s)
- Zachary B Katz
- Division of Signalling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis & Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Chen Zhang
- Division of Signalling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Ariel Quintana
- Division of Signalling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
- Translational Science Division, Clinical Science Department, Moffitt Cancer Center Magnolia Campus, Tampa, FL, 33612, USA
| | - Björn F Lillemeier
- NOMIS Center for Immunobiology and Microbial Pathogenesis & Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| | - Patrick G Hogan
- Division of Signalling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.
- Program in Immunology, University of California San Diego, La Jolla, CA, 92037, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Akhmetova KA, Chesnokov IN, Fedorova SA. [Functional Characterization of Septin Complexes]. Mol Biol (Mosk) 2018; 52:155-171. [PMID: 29695686 DOI: 10.7868/s0026898418020015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/27/2016] [Indexed: 12/28/2022]
Abstract
Septins belong to a family of conserved GTP-binding proteins found in majority of eukaryotic species except for higher plants. Septins form nonpolar complexes that further polymerize into filaments and associate with cell membranes, thus comprising newly acknowledged cytoskeletal system. Septins participate in a variety of cell processes and contribute to various pathophysiological states, including tumorigenesis and neurodegeneration. Here, we review the structural and functional properties of septins and the regulation of their dynamics with special emphasis on the role of septin filaments as a cytoskeletal system and its interaction with actin and microtubule cytoskeletons. We also discuss how septins compartmentalize the cell by forming local protein-anchoring scaffolds and by providing barriers for the lateral diffusion of the membrane proteins.
Collapse
Affiliation(s)
- K A Akhmetova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia.,University of Alabama at Birmingham, Birmingham, 35294 USA.,Novosibirsk National Research State University, Novosibirsk, 630090 Russia
| | - I N Chesnokov
- University of Alabama at Birmingham, Birmingham, 35294 USA
| | - S A Fedorova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia.,Novosibirsk National Research State University, Novosibirsk, 630090 Russia.,
| |
Collapse
|
21
|
|
22
|
Simi AK, Anlaş AA, Stallings-Mann M, Zhang S, Hsia T, Cichon M, Radisky DC, Nelson CM. A Soft Microenvironment Protects from Failure of Midbody Abscission and Multinucleation Downstream of the EMT-Promoting Transcription Factor Snail. Cancer Res 2018; 78:2277-2289. [PMID: 29483094 DOI: 10.1158/0008-5472.can-17-2899] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 02/08/2018] [Accepted: 02/20/2018] [Indexed: 02/07/2023]
Abstract
Multinucleation is found in more than one third of tumors and is linked to increased tolerance for mutation, resistance to chemotherapy, and invasive potential. The integrity of the genome depends on proper execution of the cell cycle, which can be altered through mechanotransduction pathways as the tumor microenvironment stiffens during tumorigenesis. Here, we show that signaling downstream of matrix metalloproteinase-3 (MMP3) or TGFβ, known inducers of epithelial-mesenchymal transition (EMT), also promotes multinucleation in stiff microenvironments through Snail-dependent expression of the filament-forming protein septin-6, resulting in midbody persistence, abscission failure, and multinucleation. Consistently, we observed elevated expression of Snail and septin-6 as well as multinucleation in a human patient sample of metaplastic carcinoma of the breast, a rare classification characterized by deposition of collagen fibers and active EMT. In contrast, a soft microenvironment protected mammary epithelial cells from becoming multinucleated by preventing Snail-induced upregulation of septin-6. Our data suggest that tissue stiffening during tumorigenesis synergizes with oncogenic signaling to promote genomic abnormalities that drive cancer progression.Significance: These findings reveal tissue stiffening during tumorigenesis synergizes with oncogenic signaling to promote genomic abnormalities that drive cancer progression. Cancer Res; 78(9); 2277-89. ©2018 AACR.
Collapse
Affiliation(s)
- Allison K Simi
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Alişya A Anlaş
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | | | - Sherry Zhang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Tiffaney Hsia
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Magdalena Cichon
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, Florida
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, Florida
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey. .,Department of Molecular Biology, Princeton University, Princeton, New Jersey
| |
Collapse
|
23
|
Spiliotis ET. Spatial effects - site-specific regulation of actin and microtubule organization by septin GTPases. J Cell Sci 2018; 131:jcs207555. [PMID: 29326311 PMCID: PMC5818061 DOI: 10.1242/jcs.207555] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The actin and microtubule cytoskeletons comprise a variety of networks with distinct architectures, dynamics and protein composition. A fundamental question in eukaryotic cell biology is how these networks are spatially and temporally controlled, so they are positioned in the right intracellular places at the right time. While significant progress has been made in understanding the self-assembly of actin and microtubule networks, less is known about how they are patterned and regulated in a site-specific manner. In mammalian systems, septins are a large family of GTP-binding proteins that multimerize into higher-order structures, which associate with distinct subsets of actin filaments and microtubules, as well as membranes of specific curvature and lipid composition. Recent studies have shed more light on how septins interact with actin and microtubules, and raised the possibility that the cytoskeletal topology of septins is determined by their membrane specificity. Importantly, new functions have emerged for septins regarding the generation, maintenance and positioning of cytoskeletal networks with distinct organization and biochemical makeup. This Review presents new and past findings, and discusses septins as a unique regulatory module that instructs the local differentiation and positioning of distinct actin and microtubule networks.
Collapse
Affiliation(s)
- Elias T Spiliotis
- Drexel University, Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Ribet D, Boscaini S, Cauvin C, Siguier M, Mostowy S, Echard A, Cossart P. SUMOylation of human septins is critical for septin filament bundling and cytokinesis. J Cell Biol 2017; 216:4041-4052. [PMID: 29051266 PMCID: PMC5716278 DOI: 10.1083/jcb.201703096] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/25/2017] [Accepted: 08/23/2017] [Indexed: 01/22/2023] Open
Abstract
Septins are cytoskeletal proteins that assemble into nonpolar filaments. They are critical in diverse cellular functions, acting as scaffolds for protein recruitment and as diffusion barriers for subcellular compartmentalization. Human septins are encoded by 13 different genes and are classified into four groups based on sequence homology (SEPT2, SEPT3, SEPT6, and SEPT7 groups). In yeast, septins were among the first proteins reported to be modified by SUMOylation, a ubiquitin-like posttranslational modification. However, whether human septins could be modified by small ubiquitin-like modifiers (SUMOs) and what roles this modification may have in septin function remains unknown. In this study, we first show that septins from all four human septin groups can be covalently modified by SUMOs. We show in particular that endogenous SEPT7 is constitutively SUMOylated during the cell cycle. We then map SUMOylation sites to the C-terminal domain of septins belonging to the SEPT6 and SEPT7 groups and to the N-terminal domain of septins from the SEPT3 group. We finally demonstrate that expression of non-SUMOylatable septin variants from the SEPT6 and SEPT7 groups leads to aberrant septin bundle formation and defects in cytokinesis after furrow ingression. Altogether, our results demonstrate a pivotal role for SUMOylation in septin filament bundling and cell division.
Collapse
Affiliation(s)
- David Ribet
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Paris, France
| | - Serena Boscaini
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Paris, France
| | - Clothilde Cauvin
- Unité de Trafic Membranaire et Division Cellulaire, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique UMR3691, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Institut de Formation Doctorale, Paris, France
| | - Martin Siguier
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Paris, France
| | - Serge Mostowy
- Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, England, UK
| | - Arnaud Echard
- Unité de Trafic Membranaire et Division Cellulaire, Département de Biologie Cellulaire et Infection, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique UMR3691, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Institut de Formation Doctorale, Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Paris, France
| |
Collapse
|
25
|
Boubakar L, Falk J, Ducuing H, Thoinet K, Reynaud F, Derrington E, Castellani V. Molecular Memory of Morphologies by Septins during Neuron Generation Allows Early Polarity Inheritance. Neuron 2017; 95:834-851.e5. [DOI: 10.1016/j.neuron.2017.07.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 05/23/2017] [Accepted: 07/24/2017] [Indexed: 01/22/2023]
|
26
|
Thieleke-Matos C, Osório DS, Carvalho AX, Morais-de-Sá E. Emerging Mechanisms and Roles for Asymmetric Cytokinesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:297-345. [PMID: 28526136 DOI: 10.1016/bs.ircmb.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytokinesis completes cell division by physically separating the contents of the mother cell between the two daughter cells. This event requires the highly coordinated reorganization of the cytoskeleton within a precise window of time to ensure faithful genomic segregation. In addition, recent progress in the field highlighted the importance of cytokinesis in providing particularly important cues in the context of multicellular tissues. The organization of the cytokinetic machinery and the asymmetric localization or inheritance of the midbody remnants is critical to define the spatial distribution of mechanical and biochemical signals. After a brief overview of the conserved steps of animal cytokinesis, we review the mechanisms controlling polarized cytokinesis focusing on the challenges of epithelial cytokinesis. Finally, we discuss the significance of these asymmetries in defining embryonic body axes, determining cell fate, and ensuring the correct propagation of epithelial organization during proliferation.
Collapse
Affiliation(s)
- C Thieleke-Matos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - D S Osório
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A X Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - E Morais-de-Sá
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
27
|
Altamirano S, Chandrasekaran S, Kozubowski L. Mechanisms of Cytokinesis in Basidiomycetous Yeasts. FUNGAL BIOL REV 2017; 31:73-87. [PMID: 28943887 DOI: 10.1016/j.fbr.2016.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
While mechanisms of cytokinesis exhibit considerable plasticity, it is difficult to precisely define the level of conservation of this essential part of cell division in fungi, as majority of our knowledge is based on ascomycetous yeasts. However, in the last decade more details have been uncovered regarding cytokinesis in the second largest fungal phylum, basidiomycetes, specifically in two yeasts, Cryptococcus neoformans and Ustilago maydis. Based on these findings, and current sequenced genomes, we summarize cytokinesis in basidiomycetous yeasts, indicating features that may be unique to this phylum, species-specific characteristics, as well as mechanisms that may be common to all eukaryotes.
Collapse
Affiliation(s)
- Sophie Altamirano
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| | | | - Lukasz Kozubowski
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
28
|
Poüs C, Klipfel L, Baillet A. Cancer-Related Functions and Subcellular Localizations of Septins. Front Cell Dev Biol 2016; 4:126. [PMID: 27878118 PMCID: PMC5099157 DOI: 10.3389/fcell.2016.00126] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/24/2016] [Indexed: 12/16/2022] Open
Abstract
Since the initial discovery of septin family GTPases, the understanding of their molecular organization and cellular roles keeps being refined. Septins have been involved in many physiological processes and the misregulation of specific septin gene expression has been implicated in diverse human pathologies, including neurological disorders and cancer. In this minireview, we focus on the importance of the subunit composition and subcellular localization of septins relevant to tumor initiation, progression, and metastasis. We especially underline the importance of septin polymer composition and of their association with the plasma membrane, actin, or microtubules in cell functions involved in cancer and in resistance to cancer therapies. Through their scaffolding role, their function in membrane compartmentalization or through their protective function against protein degradation, septins also emerge as critical organizers of membrane-associated proteins and of signaling pathways implicated in cancer-associated angiogenesis, apoptosis, polarity, migration, proliferation, and in metastasis. Also, the question as to which of the free monomers, hetero-oligomers, or filaments is the functional form of mammalian septins is raised and the control over their spatial and temporal localization is discussed. The increasing amount of crosstalks identified between septins and cellular signaling mediators reinforces the exciting possibility that septins could be new targets in anti-cancer therapies or in therapeutic strategies to limit drug resistance.
Collapse
Affiliation(s)
- Christian Poüs
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1193, Université Paris-Sud, Université Paris-SaclayChâtenay-Malabry, France; Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HPClamart, France
| | - Laurence Klipfel
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1193, Université Paris-Sud, Université Paris-SaclayChâtenay-Malabry, France; Département de Génétique, Institut de la Vision, Université Pierre et Marie Curie Paris 06, Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale UMR-S 968, Centre National de la Recherche Scientifique UMR 7210Paris, France
| | - Anita Baillet
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1193, Université Paris-Sud, Université Paris-Saclay Châtenay-Malabry, France
| |
Collapse
|
29
|
Hu L, Su P, Li R, Yan K, Chen Z, Shang P, Qian A. Knockdown of microtubule actin crosslinking factor 1 inhibits cell proliferation in MC3T3-E1 osteoblastic cells. BMB Rep 2016; 48:583-8. [PMID: 26277981 PMCID: PMC4911186 DOI: 10.5483/bmbrep.2015.48.10.098] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 01/12/2023] Open
Abstract
Microtubule actin crosslinking factor 1 (MACF1), a widely expressed cytoskeletal linker, plays important roles in various cells by regulating cytoskeleton dynamics. However, its role in osteoblastic cells is not well understood. Based on our previous findings that the association of MACF1 with F-actin and microtubules in osteoblast-like cells was altered under magnetic force conditions, here, by adopting a stable MACF1-knockdown MC3T3-E1 osteoblastic cell line, we found that MACF1 knockdown induced large cells with a binuclear/multinuclear structure. Further, immunofluorescence staining showed disorganization of F-actin and microtubules in MACF1-knockdown cells. Cell counting revealed significant decrease of cell proliferation and cell cycle analysis showed an S phase cell cycle arrest in MACF1-knockdown cells. Moreover and interestingly, MACF1 knockdown showed a potential effect on cellular MTT reduction activity and mitochondrial content, suggesting an impact on cellular metabolic activity. These results together indicate an important role of MACF1 in regulating osteoblastic cell morphology and function. [BMB Reports 2015; 48(10): 583-588]
Collapse
Affiliation(s)
- Lifang Hu
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Peihong Su
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Runzhi Li
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Kun Yan
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Zhihao Chen
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Peng Shang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| | - Airong Qian
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, P. R. China
| |
Collapse
|
30
|
Jordan SN, Davies T, Zhuravlev Y, Dumont J, Shirasu-Hiza M, Canman JC. Cortical PAR polarity proteins promote robust cytokinesis during asymmetric cell division. J Cell Biol 2016; 212:39-49. [PMID: 26728855 PMCID: PMC4700484 DOI: 10.1083/jcb.201510063] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In asymmetrically dividing C. elegans embryos, the core cortical PAR proteins are required to retain septin and anillin at the anterior cortex away from the contractile ring and to promote normal F-actin levels at the contractile ring and successful cytokinesis. Cytokinesis, the physical division of one cell into two, is thought to be fundamentally similar in most animal cell divisions and driven by the constriction of a contractile ring positioned and controlled solely by the mitotic spindle. During asymmetric cell divisions, the core polarity machinery (partitioning defective [PAR] proteins) controls the unequal inheritance of key cell fate determinants. Here, we show that in asymmetrically dividing Caenorhabditis elegans embryos, the cortical PAR proteins (including the small guanosine triphosphatase CDC-42) have an active role in regulating recruitment of a critical component of the contractile ring, filamentous actin (F-actin). We found that the cortical PAR proteins are required for the retention of anillin and septin in the anterior pole, which are cytokinesis proteins that our genetic data suggest act as inhibitors of F-actin at the contractile ring. Collectively, our results suggest that the cortical PAR proteins coordinate the establishment of cell polarity with the physical process of cytokinesis during asymmetric cell division to ensure the fidelity of daughter cell formation.
Collapse
Affiliation(s)
- Shawn N Jordan
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Tim Davies
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Yelena Zhuravlev
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032 Department of Genetics and Development, Columbia University, New York, NY 10032
| | - Julien Dumont
- Institut Jacques Monod, Centre National de la Recherche Scientifique, Unites Mixtes de Recherche 7592, Universite Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University, New York, NY 10032
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| |
Collapse
|
31
|
Abstract
Septins are highly conserved and essential eukaryotic cytoskeletal proteins that interact with the inner plasma membrane. They are involved in essential functions requiring cell membrane remodeling and compartmentalization, such as cell division and dendrite morphogenesis, and have been implicated in numerous diseases. Depending on the organisms and on the type of tissue, a specific set of septins genes are expressed, ranging from 2 to 13. Septins self-assemble into linear, symmetric rods that can further organize into linear filaments several microns in length. Only a subset of human septins has been described at high resolution by X-ray crystallography (Sirajuddin et al., 2007). Electron microscopy (EM) has proven to be a method of choice for analyzing the molecular organization of septins. It is possible to localize each septin subunit within the rod complex using genetic tags, such as maltose-binding protein or green fluorescent protein, to generate a visible label of a specific septin subunit in EM images that are processed using single-particle EM methodology. In this chapter we present, in detail, the methods that we have used to analyze the molecular organization of budding yeast septins (Bertin et al., 2008). These methods include purification of septin complexes, sample preparation for EM, and image processing procedures. Such methods can be generalized to analyze the organization of septins from any organism.
Collapse
|
32
|
Affiliation(s)
- Tim Davies
- a Department of Pathology and Cell Biology , Columbia University , New York , NY , USA
| | - Shawn N Jordan
- a Department of Pathology and Cell Biology , Columbia University , New York , NY , USA
| | - Julie C Canman
- a Department of Pathology and Cell Biology , Columbia University , New York , NY , USA
| |
Collapse
|
33
|
GTPase domain driven dimerization of SEPT7 is dispensable for the critical role of septins in fibroblast cytokinesis. Sci Rep 2016; 6:20007. [PMID: 26818767 PMCID: PMC4730212 DOI: 10.1038/srep20007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022] Open
Abstract
Septin 7 (SEPT7) has been described to be essential for successful completion of cytokinesis in mouse fibroblasts, and Sept7-deficiency in fibroblasts constitutively results in multinucleated cells which stop proliferation. Using Sept7flox/floxfibroblasts we generated a cellular system, where the cytokinetic defects of Cre-mediated deletion of the Sept7 gene can be rescued by ectopically expressed doxycycline-inducible wild type SEPT7. Using this system, we analyzed the ability of SEPT7-mutants with alterations in their GTPase domain-dependent dimerization to prevent multinucleation and rescue proliferation. Although biochemical analysis of the mutants demonstrates differences in homo- and/or hetero-polymerization, in GTP-binding and/or GTPase activities, all analyzed mutants were able to rescue the cytokinesis phenotype of Sept7flox/floxfibroblasts associated with Cre-mediated deletion of endogenous Sept7. These findings indicate that the ability of septins to assemble into well-defined SEPT7-dimerization dependent native filaments is dispensable for cytokinesis in fibroblasts and opens the way to search for other mechanisms of the involvement of SEPT7 in cytokinesis.
Collapse
|