1
|
Debadarshini J, LeGoff L, Mavrakis M. Septins in animal tissue architecture: more than just peanuts. Curr Opin Cell Biol 2025; 94:102525. [PMID: 40311264 DOI: 10.1016/j.ceb.2025.102525] [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: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
Septins are cytoskeletal guanosine triphosphate (GTP)-binding proteins that were discovered in budding yeast and are conserved from algae and protists to mammals. Septins assemble into heteromeric complexes, which can polymerize into filaments and higher-order filament architectures, and perform functions in a wide range of biological processes, including cell division and motility and tissue morphogenesis. Although septin dysfunction in animals is linked to infertility, defective organogenesis, neurodegenerative diseases, and cancer, the molecular mechanisms underlying septin function are not clear. Studies of septins in vivo in whole animals provide a powerful approach for gaining insights into the role of septins in animal pathophysiology and unraveling the molecular and cell biological basis of septin function.
Collapse
Affiliation(s)
- Jyotirmayee Debadarshini
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013, Marseille, France
| | - Loïc LeGoff
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013, Marseille, France.
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013, Marseille, France.
| |
Collapse
|
2
|
Perry JA, Werner ME, Omi S, Heck BW, Maddox PS, Mavrakis M, Maddox AS. Animal septins contain functional transmembrane domains. Curr Biol 2025; 35:1910-1917.e5. [PMID: 40157353 DOI: 10.1016/j.cub.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/17/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Septins are a highly conserved family of proteins that form palindromic hetero-oligomeric rods, which anneal into non-polar filaments. Via association with the plasma membrane, septin filaments recognize micron-scale membrane curvature, create diffusion barriers, and regulate cell morphogenic events via scaffolding other cytoskeletal polymers (i.e., filamentous actin [F-actin] and microtubules) and biochemical regulators of cell division, cell migration, and polarity establishment.1,2 Although interaction with cellular membranes is thought to be crucial for septin polymer dynamics and function, how septins associate with membranes is not understood. Three polybasic regions (PB1, PB2, and PB3) and an amphipathic helix (AH) are each sufficient for membrane interaction in vitro, and while the AH domain has been implicated in conferring membrane curvature sensing in vivo in the filamentous fungus Ashbya, the functionality of these domains in the context of intact septin complexes in vivo is still incompletely defined.3,4,5,6,7,8,9 We identified and characterized an isoform of Caenorhabditis elegans septin UNC-61 that was predicted to contain a transmembrane domain (TMD; UNC-61a). UNC-61a was expressed in a subset of tissues where the known septins act, and the TMD was required for tissue integrity of the egg-laying apparatus. We found predicted TMD-containing septins across much of opisthokont phylogeny and demonstrated that the TMD-containing sequence of a primate TMD-septin is sufficient for localization to cellular membranes. Together, our findings reveal a novel mechanism of septin-membrane association with profound implications for septin dynamics and regulation.
Collapse
Affiliation(s)
- Jenna A Perry
- Department of Biology, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Michael E Werner
- Department of Biology, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille University, Centrale Med, 13013 Marseille, France
| | - Bryan W Heck
- Department of Biology, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul S Maddox
- Department of Biology, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille University, Centrale Med, 13013 Marseille, France
| | - Amy S Maddox
- Department of Biology, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Chen R, Grill S, Lin B, Saiduddin M, Lehmann R. Origin and establishment of the germline in Drosophila melanogaster. Genetics 2025; 229:iyae217. [PMID: 40180587 PMCID: PMC12005264 DOI: 10.1093/genetics/iyae217] [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: 05/14/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
The continuity of a species depends on germ cells. Germ cells are different from all the other cell types of the body (somatic cells) as they are solely destined to develop into gametes (sperm or egg) to create the next generation. In this review, we will touch on 4 areas of embryonic germ cell development in Drosophila melanogaster: the assembly and function of germplasm, which houses the determinants for germ cell specification and fate and the mitochondria of the next generation; the process of pole cell formation, which will give rise to primordial germ cells (PGCs); the specification of pole cells toward the PGC fate; and finally, the migration of PGCs to the somatic gonadal precursors, where they, together with somatic gonadal precursors, form the embryonic testis and ovary.
Collapse
Affiliation(s)
- Ruoyu Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Sherilyn Grill
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mariyah Saiduddin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Ruth Lehmann
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
4
|
Craig Z, Arnold TR, Walworth K, Walkon A, Miller AL. Anillin tunes contractility and regulates barrier function during Rho flare-mediated tight junction remodeling. Mol Biol Cell 2025; 36:ar31. [PMID: 39841565 PMCID: PMC11974952 DOI: 10.1091/mbc.e24-11-0513] [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: 11/22/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
To preserve barrier function, cell-cell junctions must dynamically remodel during cell shape changes. We have previously described a rapid tight junction repair pathway characterized by local, transient activations of RhoA, termed "Rho flares," which repair leaks in tight junctions via promoting local actomyosin-mediated junction remodeling. In this pathway, junction elongation is a mechanical trigger that initiates RhoA activation through an influx of intracellular calcium and recruitment of p115RhoGEF. However, mechanisms that tune the level of RhoA activation and Myosin II contractility during the process remain uncharacterized. Here, we show that the scaffolding protein Anillin localizes to Rho flares and regulates RhoA activity and actomyosin contraction at flares. Knocking down Anillin results in Rho flares with increased intensity but shorter duration. These changes in active RhoA dynamics weaken downstream F-actin and Myosin II accumulation at the site of Rho flares, resulting in decreased junction contraction. Consequently, tight junction breaks are not reinforced following Rho flares. We show that Anillin-driven RhoA regulation is necessary for successfully repairing tight junction leaks and protecting junctions from repeated barrier damage. Together, these results uncover a novel regulatory role for Anillin during tight junction repair and barrier function maintenance.
Collapse
Affiliation(s)
- Zie Craig
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Torey R. Arnold
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Kelsey Walworth
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Alexander Walkon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L. Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
5
|
Perry JA, Werner ME, Omi S, Heck BW, Maddox PS, Mavrakis M, Maddox AS. Animal septins contain functional transmembrane domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.20.567915. [PMID: 38045322 PMCID: PMC10690161 DOI: 10.1101/2023.11.20.567915] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Septins, a conserved family of filament-forming proteins, contribute to eukaryotic cell division, polarity, and membrane trafficking. Septins scaffold other proteins to cellular membranes, but it is not fully understood how septins associate with membranes. We identified and characterized an isoform of Caenorhabditis elegans septin UNC-61 that was predicted to contain a transmembrane domain (TMD). The TMD isoform is expressed in a subset of tissues where the known septins were known to act, and TMD function was required for tissue integrity of the egg-laying apparatus. We found predicted TMD-containing septins across much of opisthokont phylogeny and demonstrated that the TMD-containing sequence of a primate TMD-septin is sufficient for localization to cellular membranes. Together, our findings reveal a novel mechanism of septin-membrane association with profound implications for septin dynamics and regulation.
Collapse
Affiliation(s)
- Jenna A. Perry
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Michael E. Werner
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Bryan W. Heck
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Paul S. Maddox
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Amy S. Maddox
- Department of Biology, The University of North Carolina at Chapel Hill; Chapel Hill, North Carolina, 27599 USA
| |
Collapse
|
6
|
Huynh MA, Thi Phuong Thao D, Yoshida H. The anillin knockdown in the Drosophila nervous system shows locomotor and learning defects. Exp Cell Res 2025; 444:114364. [PMID: 39622466 DOI: 10.1016/j.yexcr.2024.114364] [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: 05/20/2024] [Revised: 10/29/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
Anillin (Ani) is an evolutionarily conserved protein with a multi-domain structure that cross-links cytoskeletal proteins and plays an essential role in the formation of the contractile ring during cytokinesis. However, Ani is highly expressed in the human central nervous system (CNS), and it scaffolds myelin in the CNS of mice and modulates neuronal migration and growth in Caenorhabditis elegans. Although Ani is also highly expressed in the Drosophila CNS, its role remains unclear. In the present study, we showed that Ani is not only highly expressed in larval neuroblasts of the CNS, but also weakly expressed in the neuromuscular junction (NMJ) and axons. In addition, the ani knockdown in the nervous system led to pupal lethality, larval locomotor defects, and learning disability, along with abnormal morphology of the NMJ and distribution patterns of the mature neuropil in the CNS. These results show that Ani plays an important role also in the Drosophila nervous system.
Collapse
Affiliation(s)
- Man Anh Huynh
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Dang Thi Phuong Thao
- Faculty of Biology and Biotechnology, VNUHCM-University of Science, 700000, Viet Nam
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan.
| |
Collapse
|
7
|
Goldner AN, Cheikh MI, Osterfield M, Doubrovinski K. Viscous shear is a key force in Drosophila ventral furrow morphogenesis. Development 2024; 151:dev202892. [PMID: 39428993 PMCID: PMC11586522 DOI: 10.1242/dev.202892] [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: 03/20/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Ventral furrow (VF) formation in Drosophila melanogaster is an important model of epithelial folding. Previous models of VF formation require cell volume conservation to convert apically localized constriction forces into lateral cell elongation and tissue folding. Here, we have investigated embryonic morphogenesis in anillin knockdown (scra RNAi) embryos, where basal cell membranes fail to form and therefore cells can lose cytoplasmic volume through their basal side. Surprisingly, the mesoderm elongation and subsequent folding that comprise VF formation occurred essentially normally. We hypothesized that the effects of viscous shear may be sufficient to drive membrane elongation, providing effective volume conservation, and thus driving tissue folding. Since this hypothesis may not be possible to test experimentally, we turned to a computational approach. To test whether viscous shear is a dominant force for morphogenesis in vivo, we developed a 3D computational model incorporating both accurate cell and tissue geometry, and experimentally measured material parameters. Results from this model demonstrate that viscous shear generates sufficient force to drive cell elongation and tissue folding in vivo.
Collapse
Affiliation(s)
- Amanda Nicole Goldner
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Mohamad Ibrahim Cheikh
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Miriam Osterfield
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Konstantin Doubrovinski
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
8
|
Willet AH, Park JS, Snider CE, Huang JJ, Chen JS, Gould KL. Fission yeast Duc1 links to ER-PM contact sites and influences PM lipid composition and cytokinetic ring anchoring. J Cell Sci 2024; 137:jcs262347. [PMID: 39239853 PMCID: PMC11449445 DOI: 10.1242/jcs.262347] [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: 06/13/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Cytokinesis is the final stage of the cell cycle that results in the physical separation of daughter cells. To accomplish cytokinesis, many organisms build an actin- and myosin-based cytokinetic ring (CR) that is anchored to the plasma membrane (PM). Defects in CR-PM anchoring can arise when the PM lipid phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2] is depleted. In Schizosaccharomyces pombe, reduced PM PI(4,5)P2 results in a CR that cannot maintain a medial position and slides toward one cell end, resulting in two differently sized daughter cells. S. pombe PM PI(4,5)P2 is synthesized by the phosphatidylinositol 4-phosphate 5-kinase (PI5-kinase) Its3, but what regulates this enzyme to maintain appropriate PM PI(4,5)P2 levels in S. pombe is not known. To identify Its3 regulators, we used proximity-based biotinylation, and the uncharacterized protein Duc1 was specifically detected. We discovered that Duc1 decorates the PM except at the cell division site and that its unique localization pattern is dictated by binding to the endoplasmic reticulum (ER)-PM contact site proteins Scs2 and Scs22. Our evidence suggests that Duc1 also binds PI(4,5)P2 and helps enrich Its3 at the lateral PM, thereby promoting PM PI(4,5)P2 synthesis and robust CR-PM anchoring.
Collapse
Affiliation(s)
- Alaina H. Willet
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Joshua S. Park
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Chloe E. Snider
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Jingdian Jamie Huang
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Jun-Song Chen
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Kathleen L. Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
9
|
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
|
10
|
Stjepić V, Nakamura M, Hui J, Parkhurst SM. Two Septin complexes mediate actin dynamics during cell wound repair. Cell Rep 2024; 43:114215. [PMID: 38728140 PMCID: PMC11203717 DOI: 10.1016/j.celrep.2024.114215] [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: 11/16/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Cells have robust wound repair systems to prevent further damage or infection and to quickly restore cell cortex integrity when exposed to mechanical and chemical stress. Actomyosin ring formation and contraction at the wound edge are major events during closure of the plasma membrane and underlying cytoskeleton during cell wound repair. Here, we show that all five Drosophila Septins are required for efficient cell wound repair. Based on their different recruitment patterns and knockdown/mutant phenotypes, two distinct Septin complexes, Sep1/Sep2/Pnut and Sep4/Sep5/Pnut, are assembled to regulate actin ring assembly, contraction, and remodeling during the repair process. Intriguingly, we find that these two Septin complexes have different F-actin bending activities. In addition, we find that Anillin regulates the recruitment of only one of two Septin complexes upon wounding. Our results demonstrate that two functionally distinct Septin complexes work side by side to discretely regulate actomyosin ring dynamics during cell wound repair.
Collapse
Affiliation(s)
- Viktor Stjepić
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Justin Hui
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA.
| |
Collapse
|
11
|
Countryman AD, Doherty CA, Herrera-Perez RM, Kasza KE. Endogenous OptoRhoGEFs reveal biophysical principles of epithelial tissue furrowing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593711. [PMID: 38766210 PMCID: PMC11100791 DOI: 10.1101/2024.05.12.593711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
During development, epithelia function as malleable substrates that undergo extensive remodeling to shape developing embryos. Optogenetic control of Rho signaling provides an avenue to investigate the mechanisms of epithelial morphogenesis, but transgenic optogenetic tools can be limited by variability in tool expression levels and deleterious effects of transgenic overexpression on development. Here, we use CRISPR/Cas9 to tag Drosophila RhoGEF2 and Cysts/Dp114RhoGEF with components of the iLID/SspB optogenetic heterodimer, permitting light-dependent control over endogenous protein activities. Using quantitative optogenetic perturbations, we uncover a dose-dependence of tissue furrow depth and bending behavior on RhoGEF recruitment, revealing mechanisms by which developing embryos can shape tissues into particular morphologies. We show that at the onset of gastrulation, furrows formed by cell lateral contraction are oriented and size-constrained by a stiff basal actomyosin layer. Our findings demonstrate the use of quantitative, 3D-patterned perturbations of cell contractility to precisely shape tissue structures and interrogate developmental mechanics.
Collapse
|
12
|
Tam R, Harris TJC. Reshaping the Syncytial Drosophila Embryo with Cortical Actin Networks: Four Main Steps of Early Development. Results Probl Cell Differ 2024; 71:67-90. [PMID: 37996673 DOI: 10.1007/978-3-031-37936-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Drosophila development begins as a syncytium. The large size of the one-cell embryo makes it ideal for studying the structure, regulation, and effects of the cortical actin cytoskeleton. We review four main steps of early development that depend on the actin cortex. At each step, dynamic remodelling of the cortex has specific effects on nuclei within the syncytium. During axial expansion, a cortical actomyosin network assembles and disassembles with the cell cycle, generating cytoplasmic flows that evenly distribute nuclei along the ovoid cell. When nuclei move to the cell periphery, they seed Arp2/3-based actin caps which grow into an array of dome-like compartments that house the nuclei as they divide at the cell cortex. To separate germline nuclei from the soma, posterior germ plasm induces full cleavage of mono-nucleated primordial germ cells from the syncytium. Finally, zygotic gene expression triggers formation of the blastoderm epithelium via cellularization and simultaneous division of ~6000 mono-nucleated cells from a single internal yolk cell. During these steps, the cortex is regulated in space and time, gains domain and sub-domain structure, and undergoes mesoscale interactions that lay a structural foundation of animal development.
Collapse
Affiliation(s)
- Rebecca Tam
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Tony J C Harris
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
13
|
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
|
14
|
Stjepić V, Nakamura M, Hui J, Parkhurst SM. Two Septin Complexes Mediate Actin Dynamics During Cell Wound Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567084. [PMID: 38014090 PMCID: PMC10680708 DOI: 10.1101/2023.11.14.567084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cells have robust wound repair systems to prevent further damage or infection and to quickly restore cell cortex integrity when exposed to mechanical and chemical stress. Actomyosin ring formation and contraction at the wound edge are major events during closure of the plasma membrane and underlying cytoskeleton during cell wound repair. Here, we show that all five Drosophila Septins are required for efficient cell wound repair. Based on their different recruitment patterns and knockdown/mutant phenotypes, two distinct Septin complexes, Sep1-Sep2-Pnut and Sep4-Sep5-Pnut, are assembled to regulate actin ring assembly, contraction, and remodeling during the repair process. Intriguingly, we find that these two Septin complexes have different F-actin bending activities. In addition, we find that Anillin regulates the recruitment of only one of two Septin complexes upon wounding. Our results demonstrate that two functionally distinct Septin complexes work side-by-side to discretely regulate actomyosin ring dynamics during cell wound repair.
Collapse
Affiliation(s)
- Viktor Stjepić
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Justin Hui
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| |
Collapse
|
15
|
Cheikh MI, Tchoufag J, Osterfield M, Dean K, Bhaduri S, Zhang C, Mandadapu KK, Doubrovinski K. A comprehensive model of Drosophila epithelium reveals the role of embryo geometry and cell topology in mechanical responses. eLife 2023; 12:e85569. [PMID: 37782009 PMCID: PMC10584372 DOI: 10.7554/elife.85569] [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: 12/14/2022] [Accepted: 09/29/2023] [Indexed: 10/03/2023] Open
Abstract
In order to understand morphogenesis, it is necessary to know the material properties or forces shaping the living tissue. In spite of this need, very few in vivo measurements are currently available. Here, using the early Drosophila embryo as a model, we describe a novel cantilever-based technique which allows for the simultaneous quantification of applied force and tissue displacement in a living embryo. By analyzing data from a series of experiments in which embryonic epithelium is subjected to developmentally relevant perturbations, we conclude that the response to applied force is adiabatic and is dominated by elastic forces and geometric constraints, or system size effects. Crucially, computational modeling of the experimental data indicated that the apical surface of the epithelium must be softer than the basal surface, a result which we confirmed experimentally. Further, we used the combination of experimental data and comprehensive computational model to estimate the elastic modulus of the apical surface and set a lower bound on the elastic modulus of the basal surface. More generally, our investigations revealed important general features that we believe should be more widely addressed when quantitatively modeling tissue mechanics in any system. Specifically, different compartments of the same cell can have very different mechanical properties; when they do, they can contribute differently to different mechanical stimuli and cannot be merely averaged together. Additionally, tissue geometry can play a substantial role in mechanical response, and cannot be neglected.
Collapse
Affiliation(s)
- Mohamad Ibrahim Cheikh
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Joel Tchoufag
- Department of Chemical and Biomolecular Engineering, University of California, BerkeleyBerkeleyUnited States
- Chemical Sciences Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Miriam Osterfield
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Kevin Dean
- Department of Bioinformatics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Swayamdipta Bhaduri
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Chuzhong Zhang
- Department of Material Science and Engineering, University of Texas at ArlingtonArlingtonUnited States
| | - Kranthi Kiran Mandadapu
- Department of Chemical and Biomolecular Engineering, University of California, BerkeleyBerkeleyUnited States
- Chemical Sciences Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Konstantin Doubrovinski
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
16
|
Lebedev M, Chan FY, Lochner A, Bellessem J, Osório DS, Rackles E, Mikeladze-Dvali T, Carvalho AX, Zanin E. Anillin forms linear structures and facilitates furrow ingression after septin and formin depletion. Cell Rep 2023; 42:113076. [PMID: 37665665 PMCID: PMC10548094 DOI: 10.1016/j.celrep.2023.113076] [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: 12/24/2022] [Revised: 07/13/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
During cytokinesis, a contractile ring consisting of unbranched filamentous actin (F-actin) and myosin II constricts at the cell equator. Unbranched F-actin is generated by formin, and without formin no cleavage furrow forms. In Caenorhabditis elegans, depletion of septin restores furrow ingression in formin mutants. How the cleavage furrow ingresses without a detectable unbranched F-actin ring is unknown. We report that, in this setting, anillin (ANI-1) forms a meshwork of circumferentially aligned linear structures decorated by non-muscle myosin II (NMY-2). Analysis of ANI-1 deletion mutants reveals that its disordered N-terminal half is required for linear structure formation and sufficient for furrow ingression. NMY-2 promotes the circumferential alignment of the linear ANI-1 structures and interacts with various lipids, suggesting that NMY-2 links the ANI-1 network with the plasma membrane. Collectively, our data reveal a compensatory mechanism, mediated by ANI-1 linear structures and membrane-bound NMY-2, that promotes furrowing when unbranched F-actin polymerization is compromised.
Collapse
Affiliation(s)
- Mikhail Lebedev
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department Biologie, 91058 Erlangen, Germany; Department Biologie, Ludwig-Maximilians University, Munich, 82152 Planegg-Martinsried, Germany
| | - Fung-Yi Chan
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Anna Lochner
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department Biologie, 91058 Erlangen, Germany
| | - Jennifer Bellessem
- Department Biologie, Ludwig-Maximilians University, Munich, 82152 Planegg-Martinsried, Germany
| | - Daniel S Osório
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Elisabeth Rackles
- Department Biologie, Ludwig-Maximilians University, Munich, 82152 Planegg-Martinsried, Germany
| | - Tamara Mikeladze-Dvali
- Department Biologie, Ludwig-Maximilians University, Munich, 82152 Planegg-Martinsried, Germany
| | - Ana Xavier Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Esther Zanin
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department Biologie, 91058 Erlangen, Germany; Department Biologie, Ludwig-Maximilians University, Munich, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
17
|
Chen J, Verissimo AF, Kull AR, He B. Early zygotic gene product Dunk interacts with anillin to regulate Myosin II during Drosophila cleavage. Mol Biol Cell 2023; 34:ar102. [PMID: 37494082 PMCID: PMC10551699 DOI: 10.1091/mbc.e22-02-0046] [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: 02/15/2022] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
Drosophila melanogaster cellularization is a special form of cleavage that converts syncytial embryos into cellular blastoderms by partitioning the peripherally localized nuclei into individual cells. An early event in cellularization is the recruitment of nonmuscle myosin II ("myosin") to the leading edge of cleavage furrows, where myosin forms an interconnected basal array before reorganizing into individual cytokinetic rings. The initial recruitment and organization of basal myosin are regulated by a cellularization-specific gene, dunk, but the underlying mechanism is unclear. Through a genome-wide yeast two-hybrid screen, we identified anillin (Scraps in Drosophila), a conserved scaffolding protein in cytokinesis, as the primary binding partner of Dunk. Dunk colocalizes with anillin and regulates its cortical localization during the formation of cleavage furrows, while the localization of Dunk is independent of anillin. Furthermore, Dunk genetically interacts with anillin to regulate the basal myosin array during cellularization. Similar to Dunk, anillin colocalizes with myosin since the very early stage of cellularization and is required for myosin retention at the basal array, before the well-documented function of anillin in regulating cytokinetic ring assembly. Based on these results, we propose that Dunk regulates myosin recruitment and spatial organization during early cellularization by interacting with and regulating anillin.
Collapse
Affiliation(s)
- Jiayang Chen
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755
| | - Andreia F. Verissimo
- Institute for Biomolecular Targeting (bioMT), Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Angela R. Kull
- Institute for Biomolecular Targeting (bioMT), Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Bing He
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755
| |
Collapse
|
18
|
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
|
19
|
Sokac AM, Biel N, De Renzis S. Membrane-actin interactions in morphogenesis: Lessons learned from Drosophila cellularization. Semin Cell Dev Biol 2023; 133:107-122. [PMID: 35396167 PMCID: PMC9532467 DOI: 10.1016/j.semcdb.2022.03.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/12/2023]
Abstract
During morphogenesis, changes in the shapes of individual cells are harnessed to mold an entire tissue. These changes in cell shapes require the coupled remodeling of the plasma membrane and underlying actin cytoskeleton. In this review, we highlight cellularization of the Drosophila embryo as a model system to uncover principles of how membrane and actin dynamics are co-regulated in space and time to drive morphogenesis.
Collapse
Affiliation(s)
- Anna Marie Sokac
- Department of Cell and Developmental Biology, University of Illinois at Urbana Champaign, Urbana, IL 61801, USA; Graduate Program in Integrative and Molecular Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Natalie Biel
- Department of Cell and Developmental Biology, University of Illinois at Urbana Champaign, Urbana, IL 61801, USA; Graduate Program in Integrative and Molecular Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stefano De Renzis
- European Molecular Biology Laboratory Heidelberg, 69117 Heidelberg, Germany
| |
Collapse
|
20
|
Van Itallie ES, Field CM, Mitchison TJ, Kirschner MW. Dorsal lip maturation and initial archenteron extension depend on Wnt11 family ligands. Dev Biol 2023; 493:67-79. [PMID: 36334838 PMCID: PMC10194025 DOI: 10.1016/j.ydbio.2022.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/05/2022]
Abstract
Wnt11 family proteins are ligands that activate a type of Dishevelled-mediated, non-canonical Wnt signaling pathway. Loss of function causes defects in gastrulation and/or anterior-posterior axis extension in all vertebrates. Non-mammalian vertebrate genomes encode two Wnt11 family proteins whose distinct functions have been unclear. We knocked down Wnt11b and Wnt11, separately and together, in Xenopus laevis. Single morphants exhibited very similar phenotypes of delayed blastopore closure, but they had different phenotypes during the tailbud period. In response to their very similar gastrulation phenotypes, we chose to characterize dual morphants. Using dark field illuminated time-lapse imaging and kymograph analysis, we identified a failure of dorsal blastopore lip maturation that correlated with slower blastopore closure and failure to internalize the endoderm at the dorsal blastopore lip. We connected these externally visible phenotypes to cellular events in the internal tissues by imaging intact fixed embryos stained for anillin and microtubules. We found that the initial extension of the archenteron is correlated with blastopore lip maturation, and archenteron extension is dramatically disrupted by decreased Wnt11 family signaling. We were aided in our interpretation of the immunofluorescence by the novel, membrane proximal location of the cleavage furrow protein anillin in the epithelium of the blastopore lip and early archenteron.
Collapse
Affiliation(s)
| | - Christine M Field
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
21
|
Baldauf L, van Buren L, Fanalista F, Koenderink GH. Actomyosin-Driven Division of a Synthetic Cell. ACS Synth Biol 2022; 11:3120-3133. [PMID: 36164967 PMCID: PMC9594324 DOI: 10.1021/acssynbio.2c00287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 01/24/2023]
Abstract
One of the major challenges of bottom-up synthetic biology is rebuilding a minimal cell division machinery. From a reconstitution perspective, the animal cell division apparatus is mechanically the simplest and therefore attractive to rebuild. An actin-based ring produces contractile force to constrict the membrane. By contrast, microbes and plant cells have a cell wall, so division requires concerted membrane constriction and cell wall synthesis. Furthermore, reconstitution of the actin division machinery helps in understanding the physical and molecular mechanisms of cytokinesis in animal cells and thus our own cells. In this review, we describe the state-of-the-art research on reconstitution of minimal actin-mediated cytokinetic machineries. Based on the conceptual requirements that we obtained from the physics of the shape changes involved in cell division, we propose two major routes for building a minimal actin apparatus capable of division. Importantly, we acknowledge both the passive and active roles that the confining lipid membrane can play in synthetic cytokinesis. We conclude this review by identifying the most pressing challenges for future reconstitution work, thereby laying out a roadmap for building a synthetic cell equipped with a minimal actin division machinery.
Collapse
Affiliation(s)
| | | | - Federico Fanalista
- Department of Bionanoscience,
Kavli Institute of Nanoscience Delft, Delft
University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje Hendrika Koenderink
- Department of Bionanoscience,
Kavli Institute of Nanoscience Delft, Delft
University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
22
|
Cruz JVR, Batista C, Afonso BDH, Alexandre-Moreira MS, Dubois LG, Pontes B, Moura Neto V, Mendes FDA. Obstacles to Glioblastoma Treatment Two Decades after Temozolomide. Cancers (Basel) 2022; 14:cancers14133203. [PMID: 35804976 PMCID: PMC9265128 DOI: 10.3390/cancers14133203] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Glioblastomas are the most common and aggressive brain tumors in adults, with a median survival of 15 months. Treatment is surgical removal, followed by chemotherapy and/or radiotherapy. Current chemotherapeutics do not kill all the tumor cells and some cells survive, leading to the appearance of a new tumor resistant to the treatment. These treatment-resistant cells are called tumor stem cells. In addition, glioblastoma cells have a high capacity for migration, forming new tumors in areas distant from the original tumor. Studies are now focused on understanding the molecular mechanisms of chemoresistance and controlling drug entry into the brain to improve drug performance. Another promising therapeutic approach is the use of viruses that specifically destroy glioblastoma cells, preserving the neural tissue around the tumor. In this review, we summarize the main biological features of glioblastoma and the therapeutic targets that are currently under study for new clinical trials. Abstract Glioblastomas are considered the most common and aggressive primary brain tumor in adults, with an average of 15 months’ survival rate. The treatment is surgery resection, followed by chemotherapy with temozolomide, and/or radiotherapy. Glioblastoma must have wild-type IDH gene and some characteristics, such as TERT promoter mutation, EGFR gene amplification, microvascular proliferation, among others. Glioblastomas have great heterogeneity at cellular and molecular levels, presenting distinct phenotypes and diversified molecular signatures in each tumor mass, making it difficult to define a specific therapeutic target. It is believed that the main responsibility for the emerge of these distinct patterns lies in subcellular populations of tumor stem cells, capable of tumor initiation and asymmetric division. Studies are now focused on understanding molecular mechanisms of chemoresistance, the tumor microenvironment, due to hypoxic and necrotic areas, cytoskeleton and extracellular matrix remodeling, and in controlling blood brain barrier permeabilization to improve drug delivery. Another promising therapeutic approach is the use of oncolytic viruses that are able to destroy specifically glioblastoma cells, preserving the neural tissue around the tumor. In this review, we summarize the main biological characteristics of glioblastoma and the cutting-edge therapeutic targets that are currently under study for promising new clinical trials.
Collapse
Affiliation(s)
- João Victor Roza Cruz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Carolina Batista
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Bernardo de Holanda Afonso
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Instituto Estadual do Cérebro Paulo Niemeyer, Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil
| | - Magna Suzana Alexandre-Moreira
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Campus A.C. Simões, Avenida Lourival Melo Mota, Maceio 57072-970, Brazil;
| | - Luiz Gustavo Dubois
- UFRJ Campus Duque de Caxias Professor Geraldo Cidade, Rodovia Washington Luiz, n. 19.593, km 104.5, Santa Cruz da Serra, Duque de Caxias 25240-005, Brazil;
| | - Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
| | - Vivaldo Moura Neto
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Instituto Estadual do Cérebro Paulo Niemeyer, Rua do Rezende 156, Rio de Janeiro 20231-092, Brazil
| | - Fabio de Almeida Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco F, Ilha do Fundão, Cidade Universitária, Rio de Janeiro 21941-590, Brazil; (J.V.R.C.); (C.B.); (B.d.H.A.); (B.P.); (V.M.N.)
- Correspondence:
| |
Collapse
|
23
|
Role of the anillin-like protein in growth of Cryptococcus neoformans at human host temperature. Fungal Genet Biol 2022; 160:103697. [DOI: 10.1016/j.fgb.2022.103697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
|
24
|
Microtubule and Actin Cytoskeletal Dynamics in Male Meiotic Cells of Drosophila melanogaster. Cells 2022; 11:cells11040695. [PMID: 35203341 PMCID: PMC8870657 DOI: 10.3390/cells11040695] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 01/12/2023] Open
Abstract
Drosophila dividing spermatocytes offer a highly suitable cell system in which to investigate the coordinated reorganization of microtubule and actin cytoskeleton systems during cell division of animal cells. Like male germ cells of mammals, Drosophila spermatogonia and spermatocytes undergo cleavage furrow ingression during cytokinesis, but abscission does not take place. Thus, clusters of primary and secondary spermatocytes undergo meiotic divisions in synchrony, resulting in cysts of 32 secondary spermatocytes and then 64 spermatids connected by specialized structures called ring canals. The meiotic spindles in Drosophila males are substantially larger than the spindles of mammalian somatic cells and exhibit prominent central spindles and contractile rings during cytokinesis. These characteristics make male meiotic cells particularly amenable to immunofluorescence and live imaging analysis of the spindle microtubules and the actomyosin apparatus during meiotic divisions. Moreover, because the spindle assembly checkpoint is not robust in spermatocytes, Drosophila male meiosis allows investigating of whether gene products required for chromosome segregation play additional roles during cytokinesis. Here, we will review how the research studies on Drosophila male meiotic cells have contributed to our knowledge of the conserved molecular pathways that regulate spindle microtubules and cytokinesis with important implications for the comprehension of cancer and other diseases.
Collapse
|
25
|
Garno C, Irons ZH, Gamache CM, McKim Q, Reyes G, Wu X, Shuster CB, Henson JH. Building the cytokinetic contractile ring in an early embryo: Initiation as clusters of myosin II, anillin and septin, and visualization of a septin filament network. PLoS One 2021; 16:e0252845. [PMID: 34962917 PMCID: PMC8714119 DOI: 10.1371/journal.pone.0252845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/24/2021] [Indexed: 01/15/2023] Open
Abstract
The cytokinetic contractile ring (CR) was first described some 50 years ago, however our understanding of the assembly and structure of the animal cell CR remains incomplete. We recently reported that mature CRs in sea urchin embryos contain myosin II mini-filaments organized into aligned concatenated arrays, and that in early CRs myosin II formed discrete clusters that transformed into the linearized structure over time. The present study extends our previous work by addressing the hypothesis that these myosin II clusters also contain the crucial scaffolding proteins anillin and septin, known to help link actin, myosin II, RhoA, and the membrane during cytokinesis. Super-resolution imaging of cortices from dividing embryos indicates that within each cluster, anillin and septin2 occupy a centralized position relative to the myosin II mini-filaments. As CR formation progresses, the myosin II, septin and anillin containing clusters enlarge and coalesce into patchy and faintly linear patterns. Our super-resolution images provide the initial visualization of anillin and septin nanostructure within an animal cell CR, including evidence of a septin filament-like network. Furthermore, Latrunculin-treated embryos indicated that the localization of septin or anillin to the myosin II clusters in the early CR was not dependent on actin filaments. These results highlight the structural progression of the CR in sea urchin embryos from an array of clusters to a linearized purse string, the association of anillin and septin with this process, and provide the visualization of an apparent septin filament network with the CR structure of an animal cell.
Collapse
Affiliation(s)
- Chelsea Garno
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
| | - Zoe H. Irons
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
- Department of Biology, Dickinson College, Carlisle, Pennsylvania, United States of America
| | - Courtney M. Gamache
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
- Department of Biology, Dickinson College, Carlisle, Pennsylvania, United States of America
| | - Quenelle McKim
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
- Department of Biology, Dickinson College, Carlisle, Pennsylvania, United States of America
| | - Gabriela Reyes
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
| | - Xufeng Wu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Charles B. Shuster
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
| | - John H. Henson
- Friday Harbor Laboratories, University of Washington, Friday Harbor, Washington, United States of America
- Department of Biology, Dickinson College, Carlisle, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Russo G, Krauss M. Septin Remodeling During Mammalian Cytokinesis. Front Cell Dev Biol 2021; 9:768309. [PMID: 34805175 PMCID: PMC8600141 DOI: 10.3389/fcell.2021.768309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/18/2021] [Indexed: 01/22/2023] Open
Abstract
Cytokinesis mediates the final separation of a mother cell into two daughter cells. Septins are recruited to the cleavage furrow at an early stage. During cytokinetic progression the septin cytoskeleton is constantly rearranged, ultimately leading to a concentration of septins within the intercellular bridge (ICB), and to the formation of two rings adjacent to the midbody that aid ESCRT-dependent abscission. The molecular mechanisms underlying this behavior are poorly understood. Based on observations that septins can associate with actin, microtubules and associated motors, we review here established roles of septins in mammalian cytokinesis, and discuss, how septins may support cytokinetic progression by exerting their functions at particular sites. Finally, we discuss how this might be assisted by phosphoinositide-metabolizing enzymes.
Collapse
Affiliation(s)
- Giulia Russo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Michael Krauss
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
27
|
Nomura S, Fujisawa T, Sota T. Role of sex-concordant gene expression in the coevolution of exaggerated male and female genitalia in a beetle group. Mol Biol Evol 2021; 38:3593-3605. [PMID: 33905498 PMCID: PMC8382896 DOI: 10.1093/molbev/msab122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Some sexual traits, including genitalia, have undergone coevolutionary diversification
toward exaggerated states in both sexes among closely related species, but the underlying
genetic mechanisms that allow correlated character evolution between the sexes are poorly
understood. Here, we studied interspecific differences in gene expression timing profiles
involved in the correlated evolution of corresponding male and female genital parts in
three species of ground beetle in Carabus (Ohomopterus).
The male and female genital parts maintain morphological matching, whereas large
interspecific variation in genital part size has occurred in the genital coevolution
between the sexes toward exaggeration. We analyzed differences in gene expression involved
in the interspecific differences in genital morphology using whole transcriptome data from
genital tissues during genital morphogenesis. We found that the gene expression variance
attributed to sex was negligible for the majority of differentially expressed genes, thus
exhibiting sex-concordant expression, although large variances were attributed to stage
and species differences. For each sex, we obtained co-expression gene networks and hub
genes from differentially expressed genes between species that might be involved in
interspecific differences in genital morphology. These gene networks were common to both
sexes, and both sex-discordant and sex-concordant gene expression were likely involved in
species-specific genital morphology. In particular, the gene expression related to
exaggerated genital size showed no significant intersexual differences, implying that the
genital sizes in both sexes are controlled by the same gene network with sex-concordant
expression patterns, thereby facilitating the coevolution of exaggerated genitalia between
the sexes while maintaining intersexual matching.
Collapse
Affiliation(s)
- Shota Nomura
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Tomochika Fujisawa
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan.,The Center for Data Science Education and Research, Shiga University, Hikone, Shiga, 522-8522, Japan
| | - Teiji Sota
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| |
Collapse
|
28
|
Zhan L, Li MG, Hays T, Bischof J. Cryopreservation method for Drosophila melanogaster embryos. Nat Commun 2021; 12:2412. [PMID: 33893303 PMCID: PMC8065140 DOI: 10.1038/s41467-021-22694-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/25/2021] [Indexed: 01/09/2023] Open
Abstract
The development of a widely adopted cryopreservation method remains a major challenge in Drosophila research. Here we report a robust and easily implemented cryopreservation protocol of Drosophila melanogaster embryos. We present innovations for embryo permeabilization, cryoprotectant agent loading, and rewarming. We show that the protocol is broadly applicable, successfully implemented in 25 distinct strains from different sources. We demonstrate that for most strains, >50% embryos hatch and >25% of the resulting larvae develop into adults after cryopreservation. We determine that survival can be significantly improved by outcrossing to mitigate the effect of genetic background for strains with low survival after cryopreservation. We show that flies retain normal sex ratio, fertility, and original mutation after successive cryopreservation of 5 generations and 6-month storage in liquid nitrogen. Lastly, we find that non-specialists are able to use this protocol to obtain consistent results, demonstrating potential for wide adoption.
Collapse
Affiliation(s)
- Li Zhan
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
- Center for Advanced Technologies for the Preservation of Biological Systems (ATP-Bio), University of Minnesota, Minneapolis, MN, USA
| | - Min-Gang Li
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Thomas Hays
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA.
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA.
- Center for Advanced Technologies for the Preservation of Biological Systems (ATP-Bio), University of Minnesota, Minneapolis, MN, USA.
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
29
|
Biophysical Analysis of Schistosoma mansoni Septins. Methods Mol Biol 2021; 2151:197-210. [PMID: 32452006 DOI: 10.1007/978-1-0716-0635-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Septins are dynamic filament-forming proteins that are recognized as important components of the cytoskeleton and are involved in numerous functions inside the cells, such as cytokinesis, exocytosis, and ciliogenesis and even in defense against pathogenic bacteria. Despite being highly conserved in eukaryotes, there is scarce literature on the role of septins in organisms other than humans and yeast. Therefore, septins from Schistosoma mansoni represent an interesting model to study an unexplored branch of this protein family. Here we described standard protocols for recombinant production and initial characterization of septins from S. mansoni. Septins are notably difficult to purify, mostly due to their tendency to assemble into filaments. Therefore, specific protocols to stabilize these proteins have been developed. In this chapter, we systematically describe protocols to clone, express, and purify schistosome septins. We also describe the use of circular dichroism to assess the folding and stability of septins and use of chromatography to characterize their oligomeric state, bound guanine nucleotide, and GTP hydrolysis. We expect that these protocols may help researchers involved in the study of schistosome septins as well as assist to establish protocols for septins from other organisms.
Collapse
|
30
|
Rominiyi O, Vanderlinden A, Clenton SJ, Bridgewater C, Al-Tamimi Y, Collis SJ. Tumour treating fields therapy for glioblastoma: current advances and future directions. Br J Cancer 2021; 124:697-709. [PMID: 33144698 PMCID: PMC7884384 DOI: 10.1038/s41416-020-01136-5] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumour in adults and continues to portend poor survival, despite multimodal treatment using surgery and chemoradiotherapy. The addition of tumour-treating fields (TTFields)-an approach in which alternating electrical fields exert biophysical force on charged and polarisable molecules known as dipoles-to standard therapy, has been shown to extend survival for patients with newly diagnosed GBM, recurrent GBM and mesothelioma, leading to the clinical approval of this approach by the FDA. TTFields represent a non-invasive anticancer modality consisting of low-intensity (1-3 V/cm), intermediate-frequency (100-300 kHz), alternating electric fields delivered via cutaneous transducer arrays configured to provide optimal tumour-site coverage. Although TTFields were initially demonstrated to inhibit cancer cell proliferation by interfering with mitotic apparatus, it is becoming increasingly clear that TTFields show a broad mechanism of action by disrupting a multitude of biological processes, including DNA repair, cell permeability and immunological responses, to elicit therapeutic effects. This review describes advances in our current understanding of the mechanisms by which TTFields mediate anticancer effects. Additionally, we summarise the landscape of TTFields clinical trials across various cancers and consider how emerging preclinical data might inform future clinical applications for TTFields.
Collapse
Affiliation(s)
- Ola Rominiyi
- Weston Park Cancer Centre, Department of Oncology & Metabolism, The University of Sheffield Medical School, Sheffield, UK.
- Department of Neurosurgery, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - Aurelie Vanderlinden
- Weston Park Cancer Centre, Department of Oncology & Metabolism, The University of Sheffield Medical School, Sheffield, UK
| | - Susan Jane Clenton
- Department of Clinical Oncology, Weston Park Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Caroline Bridgewater
- Department of Clinical Oncology, Weston Park Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Yahia Al-Tamimi
- Department of Neurosurgery, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Spencer James Collis
- Weston Park Cancer Centre, Department of Oncology & Metabolism, The University of Sheffield Medical School, Sheffield, UK.
| |
Collapse
|
31
|
Carim SC, Kechad A, Hickson GRX. Animal Cell Cytokinesis: The Rho-Dependent Actomyosin-Anilloseptin Contractile Ring as a Membrane Microdomain Gathering, Compressing, and Sorting Machine. Front Cell Dev Biol 2020; 8:575226. [PMID: 33117802 PMCID: PMC7575755 DOI: 10.3389/fcell.2020.575226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Cytokinesis is the last step of cell division that partitions the cellular organelles and cytoplasm of one cell into two. In animal cells, cytokinesis requires Rho-GTPase-dependent assembly of F-actin and myosin II (actomyosin) to form an equatorial contractile ring (CR) that bisects the cell. Despite 50 years of research, the precise mechanisms of CR assembly, tension generation and closure remain elusive. This hypothesis article considers a holistic view of the CR that, in addition to actomyosin, includes another Rho-dependent cytoskeletal sub-network containing the scaffold protein, Anillin, and septin filaments (collectively termed anillo-septin). We synthesize evidence from our prior work in Drosophila S2 cells that actomyosin and anillo-septin form separable networks that are independently anchored to the plasma membrane. This latter realization leads to a simple conceptual model in which CR assembly and closure depend upon the micro-management of the membrane microdomains to which actomyosin and anillo-septin sub-networks are attached. During CR assembly, actomyosin contractility gathers and compresses its underlying membrane microdomain attachment sites. These microdomains resist this compression, which builds tension. During CR closure, membrane microdomains are transferred from the actomyosin sub-network to the anillo-septin sub-network, with which they flow out of the CR as it advances. This relative outflow of membrane microdomains regulates tension, reduces the circumference of the CR and promotes actomyosin disassembly all at the same time. According to this hypothesis, the metazoan CR can be viewed as a membrane microdomain gathering, compressing and sorting machine that intrinsically buffers its own tension through coordination of actomyosin contractility and anillo-septin-membrane relative outflow, all controlled by Rho. Central to this model is the abandonment of the dogmatic view that the plasma membrane is always readily deformable by the underlying cytoskeleton. Rather, the membrane resists compression to build tension. The notion that the CR might generate tension through resistance to compression of its own membrane microdomain attachment sites, can account for numerous otherwise puzzling observations and warrants further investigation using multiple systems and methods.
Collapse
Affiliation(s)
- Sabrya C. Carim
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
| | - Amel Kechad
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
| | - Gilles R. X. Hickson
- CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
32
|
Chowdhary S, Madan S, Tomer D, Mavrakis M, Rikhy R. Mitochondrial morphology and activity regulate furrow ingression and contractile ring dynamics in Drosophila cellularization. Mol Biol Cell 2020; 31:2331-2347. [PMID: 32755438 PMCID: PMC7851960 DOI: 10.1091/mbc.e20-03-0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mitochondria are maternally inherited in many organisms. Mitochondrial morphology and activity regulation is essential for cell survival, differentiation, and migration. An analysis of mitochondrial dynamics and function in morphogenetic events in early metazoan embryogenesis has not been carried out. In our study we find a crucial role of mitochondrial morphology regulation in cell formation in Drosophila embryogenesis. We find that mitochondria are small and fragmented and translocate apically on microtubules and distribute progressively along the cell length during cellularization. Embryos mutant for the mitochondrial fission protein, Drp1 (dynamin-related protein 1), die in embryogenesis and show an accumulation of clustered mitochondria on the basal side in cellularization. Additionally, Drp1 mutant embryos contain lower levels of reactive oxygen species (ROS). ROS depletion was previously shown to decrease myosin II activity. Drp1 loss also leads to myosin II depletion at the membrane furrow, thereby resulting in decreased cell height and larger contractile ring area in cellularization similar to that in myosin II mutants. The mitochondrial morphology and cellularization defects in Drp1 mutants are suppressed by reducing mitochondrial fusion and increasing cytoplasmic ROS in superoxide dismutase mutants. Our data show a key role for mitochondrial morphology and activity in supporting the morphogenetic events that drive cellularization in Drosophila embryos.
Collapse
Affiliation(s)
- Sayali Chowdhary
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Somya Madan
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Darshika Tomer
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | - Manos Mavrakis
- Aix Marseille University, CNRS, Centrale Marseille, Institut Fresnel, 13013 Marseille, France
| | - Richa Rikhy
- Department of Biology, Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| |
Collapse
|
33
|
Abstract
It is widely believed that cleavage-furrow formation during cytokinesis is driven by the contraction of a ring containing F-actin and type-II myosin. However, even in cells that have such rings, they are not always essential for furrow formation. Moreover, many taxonomically diverse eukaryotic cells divide by furrowing but have no type-II myosin, making it unlikely that an actomyosin ring drives furrowing. To explore this issue further, we have used one such organism, the green alga Chlamydomonas reinhardtii We found that although F-actin is associated with the furrow region, none of the three myosins (of types VIII and XI) is localized there. Moreover, when F-actin was eliminated through a combination of a mutation and a drug, furrows still formed and the cells divided, although somewhat less efficiently than normal. Unexpectedly, division of the large Chlamydomonas chloroplast was delayed in the cells lacking F-actin; as this organelle lies directly in the path of the cleavage furrow, this delay may explain, at least in part, the delay in cytokinesis itself. Earlier studies had shown an association of microtubules with the cleavage furrow, and we used a fluorescently tagged EB1 protein to show that microtubules are still associated with the furrows in the absence of F-actin, consistent with the possibility that the microtubules are important for furrow formation. We suggest that the actomyosin ring evolved as one way to improve the efficiency of a core process for furrow formation that was already present in ancestral eukaryotes.
Collapse
|
34
|
Tuan NM, Lee CH. Role of Anillin in Tumour: From a Prognostic Biomarker to a Novel Target. Cancers (Basel) 2020; 12:E1600. [PMID: 32560530 PMCID: PMC7353083 DOI: 10.3390/cancers12061600] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 01/21/2023] Open
Abstract
Anillin (ANLN), an actin-binding protein, reportedly plays a vital role in cell proliferation and migration, particularly in cytokinesis. Although there have been findings pointing to a contribution of ANLN to the development of cancer, the association of ANLN to cancer remains not fully understood. Here, we gather evidence to determine the applicability of ANLN as a prognostic tool for some types of cancer, and the impact that ANLN has on the hallmarks of cancer. We searched academic repositories including PubMed and Google Scholar to find and review studies related to cancer and ANLN. The conclusion is that ANLN could be a potent target for cancer treatment, but the roles ANLN, other than in cytokinesis and its influence on tumour microenvironment remodeling in cancer development, must be further elucidated, and specific ANLN inhibitors should be found.
Collapse
Affiliation(s)
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Korea;
| |
Collapse
|
35
|
Abstract
The Ran pathway has a well-described function in nucleocytoplasmic transport, where active Ran dissociates importin/karyopherin-bound cargo containing a nuclear localization signal (NLS) in the nucleus. As cells enter mitosis, the nuclear envelope breaks down and a gradient of active Ran forms where levels are highest near chromatin. This gradient plays a crucial role in regulating mitotic spindle assembly, where active Ran binds to and releases importins from NLS-containing spindle assembly factors. An emerging theme is that the Ran gradient also regulates the actomyosin cortex for processes including polar body extrusion during meiosis, and cytokinesis. For these events, active Ran could play an inhibitory role, where importin-binding may help promote or stabilize a conformation or interaction that favours the recruitment and function of cortical regulators. For either spindle assembly or cortical polarity, the gradient of active Ran determines the extent of importin-binding, the effects of which could vary for different proteins.
Collapse
Affiliation(s)
- Imge Ozugergin
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
36
|
Anillin regulates breast cancer cell migration, growth, and metastasis by non-canonical mechanisms involving control of cell stemness and differentiation. Breast Cancer Res 2020; 22:3. [PMID: 31910867 PMCID: PMC6947866 DOI: 10.1186/s13058-019-1241-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Breast cancer metastasis is driven by a profound remodeling of the cytoskeleton that enables efficient cell migration and invasion. Anillin is a unique scaffolding protein regulating major cytoskeletal structures, such as actin filaments, microtubules, and septin polymers. It is markedly overexpressed in breast cancer, and high anillin expression is associated with poor prognosis. The aim of this study was to investigate the role of anillin in breast cancer cell migration, growth, and metastasis. Methods CRISPR/Cas9 technology was used to deplete anillin in highly metastatic MDA-MB-231 and BT549 cells and to overexpress it in poorly invasive MCF10AneoT cells. The effects of anillin depletion and overexpression on breast cancer cell motility in vitro were examined by wound healing and Matrigel invasion assays. Assembly of the actin cytoskeleton and matrix adhesion were evaluated by immunofluorescence labeling and confocal microscopy. In vitro tumor development was monitored by soft agar growth assays, whereas cancer stem cells were examined using a mammosphere formation assay and flow cytometry. The effects of anillin knockout on tumor growth and metastasis in vivo were determined by injecting control and anillin-depleted breast cancer cells into NSG mice. Results Loss-of-function and gain-of-function studies demonstrated that anillin is necessary and sufficient to accelerate migration, invasion, and anchorage-independent growth of breast cancer cells in vitro. Furthermore, loss of anillin markedly attenuated primary tumor growth and metastasis of breast cancer in vivo. In breast cancer cells, anillin was localized in the nucleus; however, knockout of this protein affected the cytoplasmic/cortical events, e.g., the organization of actin cytoskeleton and cell-matrix adhesions. Furthermore, we observed a global transcriptional reprogramming of anillin-depleted breast cancer cells that resulted in suppression of their stemness and induction of the mesenchymal to epithelial trans-differentiation. Such trans-differentiation was manifested by the upregulation of basal keratins along with the increased expression of E-cadherin and P-cadherin. Knockdown of E-cadherin restored the impaired migration and invasion of anillin-deficient breast cancer cells. Conclusion Our study demonstrates that anillin plays essential roles in promoting breast cancer growth and metastatic dissemination in vitro and in vivo and unravels novel functions of anillin in regulating breast cancer stemness and differentiation.
Collapse
|
37
|
The midbody interactome reveals unexpected roles for PP1 phosphatases in cytokinesis. Nat Commun 2019; 10:4513. [PMID: 31586073 PMCID: PMC6778137 DOI: 10.1038/s41467-019-12507-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022] Open
Abstract
The midbody is an organelle assembled at the intercellular bridge between the two daughter cells at the end of mitosis. It controls the final separation of the daughter cells and has been involved in cell fate, polarity, tissue organization, and cilium and lumen formation. Here, we report the characterization of the intricate midbody protein-protein interaction network (interactome), which identifies many previously unknown interactions and provides an extremely valuable resource for dissecting the multiple roles of the midbody. Initial analysis of this interactome revealed that PP1β-MYPT1 phosphatase regulates microtubule dynamics in late cytokinesis and de-phosphorylates the kinesin component MKLP1/KIF23 of the centralspindlin complex. This de-phosphorylation antagonizes Aurora B kinase to modify the functions and interactions of centralspindlin in late cytokinesis. Our findings expand the repertoire of PP1 functions during mitosis and indicate that spatiotemporal changes in the distribution of kinases and counteracting phosphatases finely tune the activity of cytokinesis proteins. The midbody is an organelle present at the bridge connecting two cells at the end of cell division. Here, the authors use mass spectrometry to define the midbody interactome and uncover a role for PP1 phosphatases in microtubule dynamics and regulation of cytokinesis.
Collapse
|
38
|
Lian YF, Huang YL, Wang JL, Deng MH, Xia TL, Zeng MS, Chen MS, Wang HB, Huang YH. Anillin is required for tumor growth and regulated by miR-15a/miR-16-1 in HBV-related hepatocellular carcinoma. Aging (Albany NY) 2019; 10:1884-1901. [PMID: 30103211 PMCID: PMC6128427 DOI: 10.18632/aging.101510] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/27/2018] [Indexed: 12/26/2022]
Abstract
Anillin (ANLN) is an actin-binding protein essential for assembly of cleavage furrow during cytokinesis. Although reportedly overexpressed in various human cancers, its role in hepatocellular carcinoma (HCC) is unclear. To address this issue, we confirmed that in 436 liver samples obtained from surgically removed HCC tissues, higher ANLN expression was detected in tumor tissues than in adjacent non-tumor tissues of HCC as measured by immunohistochemistry, quantitative real-time PCR and western blotting. Correlation and Kaplan-Meier analysis revealed that patients with higher ANLN expression were associated with worse clinical outcomes and a shorter survival time, respectively. Moreover, ANLN inhibition resulted in growth restraint, reduced colony formation, and a lower sphere number in suspension culture. Mechanistically, ANLN deficiency induced an increasing number of multinucleated cells along with the activation of apoptosis signaling and DNA damage checkpoints. Furthermore, HBV infection increased ANLN expression by inhibiting the expression of microRNA (miR)-15a and miR-16-1, both of which were identified as ANLN upstream repressors by targeting its 3’ untranslated region. Thus, we conclude that ANLN promotes tumor growth by ways of decreased apoptosis and DNA damage. Expression level of ANLN significantly influences the survival probability of HCC patients and may represent a promising prognostic biomarker.
Collapse
Affiliation(s)
- Yi-Fan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan-Lin Huang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia-Liang Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mei-Hai Deng
- Department of Hepatobiliary Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tian-Liang Xia
- State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Min-Shan Chen
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong-Bo Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yue-Hua Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
39
|
Spracklen AJ, Thornton-Kolbe EM, Bonner AN, Florea A, Compton PJ, Fernandez-Gonzalez R, Peifer M. The Crk adapter protein is essential for Drosophila embryogenesis, where it regulates multiple actin-dependent morphogenic events. Mol Biol Cell 2019; 30:2399-2421. [PMID: 31318326 PMCID: PMC6741062 DOI: 10.1091/mbc.e19-05-0302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Small Src homology domain 2 (SH2) and 3 (SH3) adapter proteins regulate cell fate and behavior by mediating interactions between cell surface receptors and downstream signaling effectors in many signal transduction pathways. The CT10 regulator of kinase (Crk) family has tissue-specific roles in phagocytosis, cell migration, and neuronal development and mediates oncogenic signaling in pathways like that of Abelson kinase. However, redundancy among the two mammalian family members and the position of the Drosophila gene on the fourth chromosome precluded assessment of Crk's full role in embryogenesis. We circumvented these limitations with short hairpin RNA and CRISPR technology to assess Crk's function in Drosophila morphogenesis. We found that Crk is essential beginning in the first few hours of development, where it ensures accurate mitosis by regulating orchestrated dynamics of the actin cytoskeleton to keep mitotic spindles in syncytial embryos from colliding. In this role, it positively regulates cortical localization of the actin-related protein 2/3 complex (Arp2/3), its regulator suppressor of cAMP receptor (SCAR), and filamentous actin to actin caps and pseudocleavage furrows. Crk loss leads to the loss of nuclei and formation of multinucleate cells. We also found roles for Crk in embryonic wound healing and in axon patterning in the nervous system, where it localizes to the axons and midline glia. Thus, Crk regulates diverse events in embryogenesis that require orchestrated cytoskeletal dynamics.
Collapse
Affiliation(s)
- Andrew J Spracklen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Emma M Thornton-Kolbe
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Alison N Bonner
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Alexandru Florea
- Institute of Biomaterials and Biomedical Engineering, Ted Rogers Centre for Heart Research, and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Peter J Compton
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, Ted Rogers Centre for Heart Research, and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Mark Peifer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
40
|
Wang P, Qian W, Wang W, Guo M, Xia Q, Cheng D. Identification and Characterization of the Anillin Gene in Silkworm. DNA Cell Biol 2019; 38:532-540. [PMID: 30985224 DOI: 10.1089/dna.2019.4660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Anillin is an actin binding protein and plays crucial roles during mitotic cell cycle progression in metazoan. However, the sequence and functions of the Anillin gene have not been yet characterized in the silkworm, Bombyx mori. In this study, we cloned the full-length cDNA sequence of the silkworm Anillin (BmAnillin) gene. The deduced amino acid sequence for BmAnillin protein comprises an Anillin homology region (AHR) covering an Anillin homology domain and a pleckstrin homology domain. Phylogenetic analysis and multiple alignments of the Anillin genes from silkworm and other organisms indicated evolutionary conservation in the AHR containing conserved phosphorylation sites. Reverse transcription-PCR experiments confirmed that the BmAnillin gene was highly expressed during larval development of gonads in which cells undergo mitotic cycles and exhibited an unexpected high expression in silk gland with endocycle during larval molting. RNA interference-mediated knockdown of the BmAnillin gene in silkworm BmN4-SID1 cells derived from ovary disrupted chromosome separation and resulted in a loss of the F-actin filament at cleavage furrow during anaphase, suggesting that the BmAnillin gene is essential for cytokinesis in silkworm.
Collapse
Affiliation(s)
- Peng Wang
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
| | - Wenliang Qian
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
| | - Weina Wang
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
| | - Mengpei Guo
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China
| | - Qingyou Xia
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China.,2 Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| | - Daojun Cheng
- 1 State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing, China.,2 Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, China
| |
Collapse
|
41
|
Erwig MS, Patzig J, Steyer AM, Dibaj P, Heilmann M, Heilmann I, Jung RB, Kusch K, Möbius W, Jahn O, Nave KA, Werner HB. Anillin facilitates septin assembly to prevent pathological outfoldings of central nervous system myelin. eLife 2019; 8:43888. [PMID: 30672734 PMCID: PMC6344079 DOI: 10.7554/elife.43888] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
Myelin serves as an axonal insulator that facilitates rapid nerve conduction along axons. By transmission electron microscopy, a healthy myelin sheath comprises compacted membrane layers spiraling around the cross-sectioned axon. Previously we identified the assembly of septin filaments in the innermost non-compacted myelin layer as one of the latest steps of myelin maturation in the central nervous system (CNS) (Patzig et al., 2016). Here we show that loss of the cytoskeletal adaptor protein anillin (ANLN) from oligodendrocytes disrupts myelin septin assembly, thereby causing the emergence of pathological myelin outfoldings. Since myelin outfoldings are a poorly understood hallmark of myelin disease and brain aging we assessed axon/myelin-units in Anln-mutant mice by focused ion beam-scanning electron microscopy (FIB-SEM); myelin outfoldings were three-dimensionally reconstructed as large sheets of multiple compact membrane layers. We suggest that anillin-dependent assembly of septin filaments scaffolds mature myelin sheaths, facilitating rapid nerve conduction in the healthy CNS.
Collapse
Affiliation(s)
- Michelle S Erwig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Julia Patzig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anna M Steyer
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Payam Dibaj
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Mareike Heilmann
- Department of Cellular Biochemistry, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ingo Heilmann
- Department of Cellular Biochemistry, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
42
|
Blake-Hedges C, Megraw TL. Coordination of Embryogenesis by the Centrosome in Drosophila melanogaster. Results Probl Cell Differ 2019; 67:277-321. [PMID: 31435800 PMCID: PMC11725063 DOI: 10.1007/978-3-030-23173-6_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The first 3 h of Drosophila melanogaster embryo development are exemplified by rapid nuclear divisions within a large syncytium, transforming the zygote to the cellular blastoderm after 13 successive cleavage divisions. As the syncytial embryo develops, it relies on centrosomes and cytoskeletal dynamics to transport nuclei, maintain uniform nuclear distribution throughout cleavage cycles, ensure generation of germ cells, and coordinate cellularization. For the sake of this review, we classify six early embryo stages that rely on processes coordinated by the centrosome and its regulation of the cytoskeleton. The first stage features migration of one of the female pronuclei toward the male pronucleus following maturation of the first embryonic centrosomes. Two subsequent stages distribute the nuclei first axially and then radially in the embryo. The remaining three stages involve centrosome-actin dynamics that control cortical plasma membrane morphogenesis. In this review, we highlight the dynamics of the centrosome and its role in controlling the six stages that culminate in the cellularization of the blastoderm embryo.
Collapse
Affiliation(s)
- Caitlyn Blake-Hedges
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA.
| | - Timothy L Megraw
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
43
|
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
|
44
|
Chen AS, Wardwell-Ozgo J, Shah NN, Wright D, Appin CL, Vigneswaran K, Brat DJ, Kornblum HI, Read RD. Drak/STK17A Drives Neoplastic Glial Proliferation through Modulation of MRLC Signaling. Cancer Res 2018; 79:1085-1097. [PMID: 30530503 DOI: 10.1158/0008-5472.can-18-0482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/21/2018] [Accepted: 12/05/2018] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) and lower grade gliomas (LGG) are the most common primary malignant brain tumors and are resistant to current therapies. Genomic analyses reveal that signature genetic lesions in GBM and LGG include copy gain and amplification of chromosome 7, amplification, mutation, and overexpression of receptor tyrosine kinases (RTK) such as EGFR, and activating mutations in components of the PI3K pathway. In Drosophila melanogaster, constitutive co-activation of RTK and PI3K signaling in glial progenitor cells recapitulates key features of human gliomas. Here we use this Drosophila glioma model to identify death-associated protein kinase (Drak), a cytoplasmic serine/threonine kinase orthologous to the human kinase STK17A, as a downstream effector of EGFR and PI3K signaling pathways. Drak was necessary for glial neoplasia, but not for normal glial proliferation and development, and Drak cooperated with EGFR to promote glial cell transformation. Drak phosphorylated Sqh, the Drosophila ortholog of nonmuscle myosin regulatory light chain (MRLC), which was necessary for transformation. Moreover, Anillin, which is a binding partner of phosphorylated Sqh, was upregulated in a Drak-dependent manner in mitotic cells and colocalized with phosphorylated Sqh in neoplastic cells undergoing mitosis and cytokinesis, consistent with their known roles in nonmuscle myosin-dependent cytokinesis. These functional relationships were conserved in human GBM. Our results indicate that Drak/STK17A, its substrate Sqh/MRLC, and the effector Anillin/ANLN regulate mitosis and cytokinesis in gliomas. This pathway may provide a new therapeutic target for gliomas.Significance: These findings reveal new insights into differential regulation of cell proliferation in malignant brain tumors, which will have a broader impact on research regarding mechanisms of oncogene cooperation and dependencies in cancer.See related commentary by Lathia, p. 1036.
Collapse
Affiliation(s)
- Alexander S Chen
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Joanna Wardwell-Ozgo
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Nilang N Shah
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Deidre Wright
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Christina L Appin
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Daniel J Brat
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Harley I Kornblum
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California.,Department of Psychiatry and Behavioral Sciences, and Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, California
| | - Renee D Read
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia. .,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
45
|
Branter J, Basu S, Smith S. Tumour treating fields in a combinational therapeutic approach. Oncotarget 2018; 9:36631-36644. [PMID: 30564303 PMCID: PMC6290966 DOI: 10.18632/oncotarget.26344] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 10/24/2018] [Indexed: 12/15/2022] Open
Abstract
The standard of care for patients with newly diagnosed Glioblastoma multiforme (GBM) has remained unchanged since 2005, with patients undergoing maximal surgical resection, followed by radiotherapy plus concomitant and maintenance Temozolomide. More recently, Tumour treating fields (TTFields) therapy has become FDA approved for adult recurrent and adult newly-diagnosed GBM following the EF-11 and EF-14 trials, respectively. TTFields is a non-invasive anticancer treatment which utilizes medium frequency alternating electric fields to target actively dividing cancerous cells. TTFields selectively targets cells within mitosis through interacting with key mitotic proteins to cause mitotic arrest and cell death. TTFields therapy presents itself as a candidate for the combinational therapy route due to the lack of overlapping toxicities associated with electric fields. Here we review current literature pertaining to TTFields in combination with alkylating agents, radiation, anti-angiogenics, mitotic inhibitors, immunotherapies, and also with novel agents. This review highlights the observed synergistic and additive effects of combining TTFields with various other therapies, as well highlighting the strategies relating to combinations with electric fields.
Collapse
Affiliation(s)
- Joshua Branter
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Queen's Medical Centre, Department of Neurosurgery, Nottingham, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
46
|
Subcellular Specialization and Organelle Behavior in Germ Cells. Genetics 2018; 208:19-51. [PMID: 29301947 DOI: 10.1534/genetics.117.300184] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 08/17/2017] [Indexed: 11/18/2022] Open
Abstract
Gametes, eggs and sperm, are the highly specialized cell types on which the development of new life solely depends. Although all cells share essential organelles, such as the ER (endoplasmic reticulum), Golgi, mitochondria, and centrosomes, germ cells display unique regulation and behavior of organelles during gametogenesis. These germ cell-specific functions of organelles serve critical roles in successful gamete production. In this chapter, I will review the behaviors and roles of organelles during germ cell differentiation.
Collapse
|
47
|
The ABD on the nascent polypeptide and PH domain are required for the precise Anillin localization in Drosophila syncytial blastoderm. Sci Rep 2018; 8:12910. [PMID: 30150713 PMCID: PMC6110771 DOI: 10.1038/s41598-018-31106-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/03/2018] [Indexed: 01/19/2023] Open
Abstract
Targeting proteins to regions where they are required is essential for proper development of organisms. For achievement of this, subcellular mRNA localization is one of the critical mechanisms. Subcellular mRNA localization is an evolutionarily conserved phenomenon from E. coli to human and contributes to limiting the regions at which its products function and efficiently supplies substrates for protein translation. During early Drosophila embryogenesis, while 71% of the 3370 mRNAs analyzed have shown prominent subcellular localization, the underlying molecular mechanisms have not been elucidated. Here, we reveal that anillin mRNA, one of the localized mRNAs in early Drosophila embryo, localizes to the tip of the pseudo-cleavage furrow in the Drosophila syncytial blastoderm using in situ hybridization combined with immunohistochemistry. Localization analyses with transgenic fly lines carrying a series of deletion mRNAs indicate that this localization is dependent on its own nascent polypeptides including the actin binding domain (ABD). In addition to the mRNA localization, it is revealed that the pleckstrin homology (PH) domain of Anillin protein is also required for its proper localization. Thus, we indicate that the precise localization of Anillin protein is tightly regulated by the ABD on the nascent polypeptide and PH domain in the Drosophila syncytial blastoderm.
Collapse
|
48
|
Davies T, Kim HX, Romano Spica N, Lesea-Pringle BJ, Dumont J, Shirasu-Hiza M, Canman JC. Cell-intrinsic and -extrinsic mechanisms promote cell-type-specific cytokinetic diversity. eLife 2018; 7:36204. [PMID: 30028292 PMCID: PMC6054530 DOI: 10.7554/elife.36204] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/10/2018] [Indexed: 01/05/2023] Open
Abstract
Cytokinesis, the physical division of one cell into two, is powered by constriction of an actomyosin contractile ring. It has long been assumed that all animal cells divide by a similar molecular mechanism, but growing evidence suggests that cytokinetic regulation in individual cell types has more variation than previously realized. In the four-cell Caenorhabditis elegans embryo, each blastomere has a distinct cell fate, specified by conserved pathways. Using fast-acting temperature-sensitive mutants and acute drug treatment, we identified cell-type-specific variation in the cytokinetic requirement for a robust forminCYK-1-dependent filamentous-actin (F-actin) cytoskeleton. In one cell (P2), this cytokinetic variation is cell-intrinsically regulated, whereas in another cell (EMS) this variation is cell-extrinsically regulated, dependent on both SrcSRC-1 signaling and direct contact with its neighbor cell, P2. Thus, both cell-intrinsic and -extrinsic mechanisms control cytokinetic variation in individual cell types and can protect against division failure when the contractile ring is weakened. The successful division of one cell into two is essential for all organisms to live, grow and reproduce. For an animal cell, the nucleus – the compartment containing the genetic material – must divide before the surrounding material. The rest of the cell, called the cytoplasm, physically separates later in a process known as cytokinesis. Cytokinesis in animal cells is driven by the formation of a ring in the middle of the dividing cell. The ring is composed of myosin motor proteins and filaments made of a protein called actin. The movements of the motor proteins along the filaments cause the ring to contract and tighten. This pulls the cell membrane inward and physically pinches the cell into two. For a long time, the mechanism of cytokinesis was assumed to be same across different types of animal cell, but later evidence suggested otherwise. For example, in liver, heat and bone cells, cytokinesis naturally fails during development to create cells with two or more nuclei. If a similar ‘failure’ happened in other cell types, it could lead to diseases such as cancers or blood disorders. This raised the question: what are the molecular mechanisms that allow cytokinesis to happen differently in different cell types? Davies et al. investigated this question using embryos of the worm Caenorhabditis elegans at a stage in their development when they consist of just four cells. The proteins forming the contractile ring in this worm are the same as those in humans. However, in the worm, the contractile ring can easily be damaged using chemical inhibitors or by mutating the genes that encode its proteins. Davies et al. show that when the contractile ring was damaged, two of the four cells in the worm embryo still divided successfully. This result indicates the existence of new mechanisms to divide the cytoplasm that allow division even with a weak contractile ring. In a further experiment, the embryos were dissected to isolate each of the four cells. Davies et al. saw that one of the two dividing cells could still divide on its own, while the other cell could not. This shows that this new method of cytokinesis is regulated both by factors inherent to the dividing cell and by external signals from other cells. Moreover, one of these extrinsic signals was found to be a signaling protein that had previously been implicated in human cancers. Future work will determine if these variations in cytokinesis between the different cell types found in the worm apply to humans too; and, more importantly from a therapeutic standpoint, if these new mechanisms exist in human cancers.
Collapse
Affiliation(s)
- Tim Davies
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Han X Kim
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Natalia Romano Spica
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Benjamin J Lesea-Pringle
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Julien Dumont
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Paris, France
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, United States
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| |
Collapse
|
49
|
Zhang Y, Yu JC, Jiang T, Fernandez-Gonzalez R, Harris TJC. Collision of Expanding Actin Caps with Actomyosin Borders for Cortical Bending and Mitotic Rounding in a Syncytium. Dev Cell 2018; 45:551-564.e4. [PMID: 29804877 DOI: 10.1016/j.devcel.2018.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/30/2018] [Accepted: 04/25/2018] [Indexed: 10/24/2022]
Abstract
The early Drosophila embryo is a large syncytial cell that compartmentalizes mitotic spindles with furrows. Before furrow ingression, an Arp2/3 actin cap forms above each nucleus and is encircled by actomyosin. We investigated how these networks transform a flat cortex into a honeycomb-like compartmental array. The growing caps circularize and ingress upon meeting their actomyosin borders, which become the furrow base. Genetic perturbations indicate that the caps physically displace their borders and, reciprocally, that the borders resist and circularize their caps. These interactions create an actomyosin cortex arrayed with circular caps. The Rac-GEF Sponge, Rac-GTP, Arp3, and actin coat the caps as a growing material that can drive cortical bending for initial furrow ingression. Additionally, laser ablations indicate that actomyosin contraction squeezes the cytoplasm, producing counterforces that swell the caps. Thus, Arp2/3 caps form clearances of the actomyosin cortex and control buckling and swelling of these clearances for metaphase compartmentalization.
Collapse
Affiliation(s)
- Yixie Zhang
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jessica C Yu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Tao Jiang
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Tony J C Harris
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
50
|
|