1
|
Martins CS, Iv F, Suman SK, Panagiotou TC, Sidor C, Ruso-López M, Plancke CN, Omi S, Pagès R, Gomes M, Llewellyn A, Bandi SR, Ramond L, Arbizzani F, Rimoli CV, Schnorrer F, Robin F, Wilde A, LeGoff L, Pedelacq JD, Jégou A, Cabantous S, Rincon SA, Chandre C, Brasselet S, Mavrakis M. Genetically encoded reporters of actin filament organization in living cells and tissues. Cell 2025; 188:2540-2559.e27. [PMID: 40179884 DOI: 10.1016/j.cell.2025.03.003] [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: 03/04/2024] [Revised: 12/09/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
The cytoskeletal protein actin is crucial for cell shape and integrity throughout eukaryotes. Actin filaments perform essential biological functions, including muscle contraction, cell division, and tissue morphogenesis. These diverse activities are achieved through the ability of actin filaments to be arranged into precise architectures. Much progress has been made in defining the proteome of the actin cytoskeleton, but a detailed appreciation of the dynamic organizational state of the actin filaments themselves has been hindered by available tools. Fluorescence polarization microscopy is uniquely placed for measuring actin filament organization by exploiting the sensitivity of polarized light excitation to the orientation of fluorophores attached to actin filaments. By engineering fusions of five widely used actin localization reporters to fluorescent proteins with constrained mobility, we have succeeded in developing genetically encoded, green- and red-fluorescent-protein-based reporters for non-invasive, quantitative measurements of actin filament organization in living cells and tissues by fluorescence polarization microscopy.
Collapse
Affiliation(s)
- Carla Silva Martins
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - François Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Shashi Kumar Suman
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Thomas C Panagiotou
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Clara Sidor
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - María Ruso-López
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | - Camille N Plancke
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Rebecca Pagès
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Alexander Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Sourish Reddy Bandi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Laurie Ramond
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | | | - Caio Vaz Rimoli
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Frank Schnorrer
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - François Robin
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Andrew Wilde
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Loïc LeGoff
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Jean-Denis Pedelacq
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), Inserm, Université Paul Sabatier - Toulouse III, CNRS, 31037 Toulouse, France
| | - Sergio A Rincon
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | | | - Sophie Brasselet
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| |
Collapse
|
2
|
Shatskiy D, Sivan A, Wedlich-Söldner R, Belyy A. Structure of the F-tractin-F-actin complex. J Cell Biol 2025; 224:e202409192. [PMID: 39928047 PMCID: PMC11809415 DOI: 10.1083/jcb.202409192] [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: 09/27/2024] [Revised: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
F-tractin is a peptide widely used to visualize the actin cytoskeleton in live eukaryotic cells but has been reported to impair cell migration and induce actin bundling at high expression levels. To elucidate these effects, we determined the cryo-EM structure of the F-tractin-F-actin complex, revealing that F-tractin consists of a flexible N-terminal region and an amphipathic C-terminal helix. The N-terminal part is dispensable for F-actin binding but responsible for the bundling effect. Based on these insights, we developed an optimized F-tractin, which eliminates the N-terminal region and minimizes bundling while retaining strong actin labeling. The C-terminal helix interacts with a hydrophobic pocket formed by two neighboring actin subunits, an interaction region shared by many actin-binding polypeptides, including the popular actin-binding probe Lifeact. Thus, rather than contrasting F-tractin and Lifeact, our data indicate that these peptides have analogous modes of interaction with F-actin. Our study dissects the structural elements of F-tractin and provides a foundation for developing future actin probes.
Collapse
Affiliation(s)
- Dmitry Shatskiy
- Membrane Enzymology Group, Groningen Institute of Biomolecular Sciences and Biotechnology (GBB), Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Athul Sivan
- Institute of Cell Dynamics and Imaging, and Cells-in-Motion Interfaculty Center (CiMIC), University of Münster, Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, and Cells-in-Motion Interfaculty Center (CiMIC), University of Münster, Münster, Germany
| | - Alexander Belyy
- Membrane Enzymology Group, Groningen Institute of Biomolecular Sciences and Biotechnology (GBB), Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
3
|
Walker C, Chandrasekaran A, Mansour D, Graham K, Torres A, Wang L, Lafer EM, Rangamani P, Stachowiak JC. Liquid-like condensates that bind actin promote assembly and bundling of actin filaments. Dev Cell 2025:S1534-5807(25)00032-2. [PMID: 39914390 DOI: 10.1016/j.devcel.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/30/2024] [Accepted: 01/15/2025] [Indexed: 02/12/2025]
Abstract
Biomolecular condensates perform diverse physiological functions. Previous work showed that VASP, a processive actin polymerase, forms condensates that assemble and bundle actin. Here, we show that this behavior does not require proteins with specific polymerase activity. Specifically, condensates composed of Lamellipodin, a protein that binds actin but is not an actin polymerase, were also capable of assembling actin filaments. To probe the minimum requirements for condensate-mediated actin bundling, we developed an agent-based computational model. Guided by its predictions, we hypothesized that any condensate-forming protein that binds filamentous actin could bundle filaments through multivalent crosslinking. To test this, we added a filamentous-actin-binding motif to Eps15, a condensate-forming protein that does not normally bind actin. The resulting chimera formed condensates that facilitated efficient assembly and bundling of actin filaments. Collectively, these findings broaden the family of proteins that could organize cytoskeletal filaments to include any filamentous-actin-binding protein that participates in protein condensation.
Collapse
Affiliation(s)
- Caleb Walker
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Aravind Chandrasekaran
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Mansour
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Kristin Graham
- Cell and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Andrea Torres
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Liping Wang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Eileen M Lafer
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego School of Medicine, La Jolla, CA, USA.
| | - Jeanne C Stachowiak
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
4
|
Sun SY, Nie L, Zhang J, Fang X, Luo H, Fu C, Wei Z, Tang AH. The interaction between KIF21A and KANK1 regulates dendritic morphology and synapse plasticity in neurons. Neural Regen Res 2025; 20:209-223. [PMID: 38767486 PMCID: PMC11246154 DOI: 10.4103/1673-5374.391301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/12/2023] [Accepted: 11/07/2023] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00029/figure1/v/2024-05-14T021156Z/r/image-tiff Morphological alterations in dendritic spines have been linked to changes in functional communication between neurons that affect learning and memory. Kinesin-4 KIF21A helps organize the microtubule-actin network at the cell cortex by interacting with KANK1; however, whether KIF21A modulates dendritic structure and function in neurons remains unknown. In this study, we found that KIF21A was distributed in a subset of dendritic spines, and that these KIF21A-positive spines were larger and more structurally plastic than KIF21A-negative spines. Furthermore, the interaction between KIF21A and KANK1 was found to be critical for dendritic spine morphogenesis and synaptic plasticity. Knockdown of either KIF21A or KANK1 inhibited dendritic spine morphogenesis and dendritic branching, and these deficits were fully rescued by coexpressing full-length KIF21A or KANK1, but not by proteins with mutations disrupting direct binding between KIF21A and KANK1 or binding between KANK1 and talin1. Knocking down KIF21A in the hippocampus of rats inhibited the amplitudes of long-term potentiation induced by high-frequency stimulation and negatively impacted the animals' cognitive abilities. Taken together, our findings demonstrate the function of KIF21A in modulating spine morphology and provide insight into its role in synaptic function.
Collapse
Affiliation(s)
- Shi-Yan Sun
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| | - Lingyun Nie
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- CAS Center for Excellence in Molecular Cell Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Jing Zhang
- Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xue Fang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Hongmei Luo
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| | - Chuanhai Fu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- CAS Center for Excellence in Molecular Cell Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zhiyi Wei
- Department of Neurobiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Ai-Hui Tang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
| |
Collapse
|
5
|
Gadsby JR, Ioannou PS, Butler R, Mason J, Smith AJ, Dobramysl U, Chin SE, Dobson C, Gallop JL. The open to closed D-loop conformational switch determines length in filopodia-like actin bundles. Biochem J 2024; 481:1977-1995. [PMID: 39621444 DOI: 10.1042/bcj20240367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Filopodia, microspikes and cytonemes are implicated in sensing the environment and in dissemination of morphogens, organelles and pathogens across tissues. Their major structural component is parallel bundles of actin filaments that assemble from the cell membrane. Whilst the length of filopodia is central to their function, it is not known how their lengths are determined by actin bundle dynamics. Here, we identified a set of monoclonal antibodies that lengthen filopodia-like structures formed in a cell-free reconstitution system, and used them to uncover a key molecular switch governing length regulation. Using immunolabelling, enzyme-linked immunosorbent assays, immunoprecipitation and immunoblock experiments, we identified four antibodies that lengthen actin bundles by selectively binding the open DNase 1-binding loop (D-loop) of actin filaments. The antibodies inhibit actin disassembly and their effects can be alleviated by providing additional actin or cofilin. This work indicates that maintaining an open state of the actin filament D-loop is a mechanism of generating long filopodia-like actin bundles.
Collapse
Affiliation(s)
- Jonathan R Gadsby
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Pantelis Savvas Ioannou
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Richard Butler
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
| | - Julia Mason
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Alison J Smith
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, U.K
| | - Ulrich Dobramysl
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Stacey E Chin
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Claire Dobson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Jennifer L Gallop
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| |
Collapse
|
6
|
Ono S. Overexpression of Lifeact in the C. elegans body wall muscle causes sarcomere disorganization and embryonic or larval lethality. Front Cell Dev Biol 2024; 12:1504980. [PMID: 39605982 PMCID: PMC11599240 DOI: 10.3389/fcell.2024.1504980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Lifeact is a short peptide that is widely utilized as a probe for actin filaments in live imaging. However, high concentrations of Lifeact can alter actin filament dynamics and cause artificial modifications to the actin cytoskeleton. Here, I evaluated Caenorhabditis elegans strains expressing Lifeact fused to fluorescent proteins in the body wall muscle. I found that, while low-level expression of Lifeact from a single-copy transgene was appropriate for labeling sarcomeric actin filaments, overexpression of Lifeact from an extrachromosomal array causes severe disorganization of muscle sarcomeres and lethality at an embryonic or larval stage. Therefore, for imaging studies in C. elegans, Lifeact needs to be kept at a low level by proper management of the expression system.
Collapse
Affiliation(s)
- Shoichiro Ono
- Departments of Pathology and Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
7
|
Zsolnay V, Gardel ML, Kovar DR, Voth GA. Cracked actin filaments as mechanosensitive receptors. Biophys J 2024; 123:3283-3294. [PMID: 38894540 PMCID: PMC11480757 DOI: 10.1016/j.bpj.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/10/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
Actin filament networks are exposed to mechanical stimuli, but the effect of strain on actin filament structure has not been well established in molecular detail. This is a critical gap in understanding because the activity of a variety of actin-binding proteins has recently been determined to be altered by actin filament strain. We therefore used all-atom molecular dynamics simulations to apply tensile strains to actin filaments and find that changes in actin subunit organization are minimal in mechanically strained, but intact, actin filaments. However, a conformational change disrupts the critical D-loop to W-loop connection between longitudinal neighboring subunits, which leads to a metastable cracked conformation of the actin filament whereby one protofilament is broken prior to filament severing. We propose that the metastable crack presents a force-activated binding site for actin regulatory factors that specifically associate with strained actin filaments. Through protein-protein docking simulations, we find that 43 evolutionarily diverse members of the dual zinc-finger-containing LIM-domain family, which localize to mechanically strained actin filaments, recognize two binding sites exposed at the cracked interface. Furthermore, through its interactions with the crack, LIM domains increase the length of time damaged filaments remain stable. Our findings propose a new molecular model for mechanosensitive binding to actin filaments.
Collapse
Affiliation(s)
- Vilmos Zsolnay
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, Illinois
| | - Margaret L Gardel
- Department of Physics & Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois; Institute for Biophysical Dynamics and James Franck Institute, University of Chicago, Chicago, Illinois
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois; Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois.
| | - Gregory A Voth
- Institute for Biophysical Dynamics and James Franck Institute, University of Chicago, Chicago, Illinois; Department of Chemistry and Chicago Center for Theoretical Chemistry, University of Chicago, Chicago, Illinois.
| |
Collapse
|
8
|
Majumdar S, Liu ST. Spatiotemporal regulation of MELK during mitosis. Front Cell Dev Biol 2024; 12:1406940. [PMID: 39355119 PMCID: PMC11443572 DOI: 10.3389/fcell.2024.1406940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
Maternal Embryonic Leucine Zipper Kinase (MELK) has been studied intensively in recent years due to its overexpression in multiple cancers. However, the cell biology of MELK remains less characterized despite its well-documented association with mitosis. Here we report a distinctive pattern of human MELK that translocates from the cytoplasm to cell cortex within 3 min of anaphase onset. The cortex association lasts about 30 min till telophase. The spatiotemporal specific localization of MELK depends on the interaction between its Threonine-Proline (TP) rich domain and kinase associated 1 (KA1) domain, which is regulated by CDK1 kinase and PP4 protein phosphatase. KA1 domains are known to regulate kinase activities through various intramolecular interactions. Our results revealed a new role for KA1 domain to control subcellular localization of a protein kinase.
Collapse
Affiliation(s)
| | - Song-Tao Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
9
|
Wirshing ACE, Goode BL. Improved tools for live imaging of F-actin structures in yeast. Mol Biol Cell 2024; 35:mr7. [PMID: 39024291 PMCID: PMC11449393 DOI: 10.1091/mbc.e24-05-0212-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: 05/17/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
For over 20 years, the most effective probe for live imaging of yeast actin cables has been Abp140-GFP. Here, we report that endogenously-tagged Abp140-GFP poorly decorates actin patches and cables in the bud compartment of yeast cells, while robustly decorating these structures in the mother cell. Using mutagenesis, we found that asymmetric decoration by Abp140 requires F-actin binding. By expressing integrated Bni1-Bnr1 and Bnr1-Bni1 chimeras, we demonstrate that asymmetric cable decoration by Abp140 also does not depend on which formin assembles the cables in each compartment. In contrast, the short actin-binding fragment of Abp140 (known as "Lifeact"), fused to 1x or 3xmNeonGreen and expressed from the endogenous ABP140 promoter, uniformly decorates patches and cables in both compartments. Further, this probe dramatically improves live imaging detection of cables (and patches) without altering their in vivo dynamics or cell growth. Improved detection allows us to visualize cables growing inward from the cell cortex and dynamically interacting with the vacuole. This probe also robustly decorates the cytokinetic actomyosin ring. Because Lifeact-3xmNeon expressed at relatively low levels provides intense labeling of cellular F-actin structures, this tool may improve live imaging in other organisms where higher levels of Lifeact expression are detrimental.
Collapse
Affiliation(s)
- Alison C. E. Wirshing
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454
| | - Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454
| |
Collapse
|
10
|
Effiong UM, Khairandish H, Ramirez-Velez I, Wang Y, Belardi B. Turn-on protein switches for controlling actin binding in cells. Nat Commun 2024; 15:5840. [PMID: 38992021 PMCID: PMC11239668 DOI: 10.1038/s41467-024-49934-2] [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: 12/07/2023] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
Within a shared cytoplasm, filamentous actin (F-actin) plays numerous and critical roles across the cell body. Cells rely on actin-binding proteins (ABPs) to organize F-actin and to integrate its polymeric characteristics into diverse cellular processes. Yet, the multitude of ABPs that engage with and shape F-actin make studying a single ABP's influence on cellular activities a significant challenge. Moreover, without a means of manipulating actin-binding subcellularly, harnessing the F-actin cytoskeleton for synthetic biology purposes remains elusive. Here, we describe a suite of designed proteins, Controllable Actin-binding Switch Tools (CASTs), whose actin-binding behavior can be controlled with external stimuli. CASTs were developed that respond to different external inputs, providing options for turn-on kinetics and enabling orthogonality and multiplexing. Being genetically encoded, we show that CASTs can be inserted into native protein sequences to control F-actin association locally and engineered into structures to control cell and tissue shape and behavior.
Collapse
Affiliation(s)
- Unyime M Effiong
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hannah Khairandish
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Yanran Wang
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
11
|
Walker C, Chandrasekaran A, Mansour D, Graham K, Torres A, Wang L, Lafer EM, Rangamani P, Stachowiak JC. Liquid-like condensates that bind actin drive filament polymerization and bundling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592527. [PMID: 38826190 PMCID: PMC11142076 DOI: 10.1101/2024.05.04.592527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Liquid-like protein condensates perform diverse physiological functions. Previous work showed that VASP, a processive actin polymerase, forms condensates that polymerize and bundle actin. To minimize their curvature, filaments accumulated at the inner condensate surface, ultimately deforming the condensate into a rod-like shape, filled with a bundle of parallel filaments. Here we show that this behavior does not require proteins with specific polymerase activity. Specifically, we found that condensates composed of Lamellipodin, a protein that binds actin but is not an actin polymerase, were also capable of polymerizing and bundling actin filaments. To probe the minimum requirements for condensate-mediated actin bundling, we developed an agent-based computational model. Guided by its predictions, we hypothesized that any condensate-forming protein that binds actin could bundle filaments through multivalent crosslinking. To test this idea, we added an actin-binding motif to Eps15, a condensate-forming protein that does not normally bind actin. The resulting chimera formed condensates that drove efficient actin polymerization and bundling. Collectively, these findings broaden the family of proteins that could organize cytoskeletal filaments to include any actin-binding protein that participates in protein condensation.
Collapse
Affiliation(s)
- Caleb Walker
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Aravind Chandrasekaran
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, United States
| | - Daniel Mansour
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, United States
| | - Kristin Graham
- Cell and Molecular Biology, The University of Texas at Austin, Austin, TX, United States
| | - Andrea Torres
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Liping Wang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Eileen M. Lafer
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, United States
| | - Jeanne C. Stachowiak
- Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Chemical Engineering, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
12
|
Kong SG, Yamazaki Y, Shimada A, Kijima ST, Hirose K, Katoh K, Ahn J, Song HG, Han JW, Higa T, Takano A, Nakamura Y, Suetsugu N, Kohda D, Uyeda TQP, Wada M. CHLOROPLAST UNUSUAL POSITIONING 1 is a plant-specific actin polymerization factor regulating chloroplast movement. THE PLANT CELL 2024; 36:1159-1181. [PMID: 38134410 PMCID: PMC10980345 DOI: 10.1093/plcell/koad320] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/09/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Plants have unique responses to fluctuating light conditions. One such response involves chloroplast photorelocation movement, which optimizes photosynthesis under weak light by the accumulation of chloroplasts along the periclinal side of the cell, which prevents photodamage under strong light by avoiding chloroplast positioning toward the anticlinal side of the cell. This light-responsive chloroplast movement relies on the reorganization of chloroplast actin (cp-actin) filaments. Previous studies have suggested that CHLOROPLAST UNUSUAL POSITIONING 1 (CHUP1) is essential for chloroplast photorelocation movement as a regulator of cp-actin filaments. In this study, we conducted comprehensive analyses to understand CHUP1 function. Functional, fluorescently tagged CHUP1 colocalized with and was coordinately reorganized with cp-actin filaments on the chloroplast outer envelope during chloroplast movement in Arabidopsis thaliana. CHUP1 distribution was reversibly regulated in a blue light- and phototropin-dependent manner. X-ray crystallography revealed that the CHUP1-C-terminal domain shares structural homology with the formin homology 2 (FH2) domain, despite lacking sequence similarity. Furthermore, the CHUP1-C-terminal domain promoted actin polymerization in the presence of profilin in vitro. Taken together, our findings indicate that CHUP1 is a plant-specific actin polymerization factor that has convergently evolved to assemble cp-actin filaments and enables chloroplast photorelocation movement.
Collapse
Affiliation(s)
- Sam-Geun Kong
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Chungnam 32588, Korea
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | - Yosuke Yamazaki
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Department of Physics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Atsushi Shimada
- Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Saku T Kijima
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8562, Japan
| | - Keiko Hirose
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8562, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8562, Japan
| | - Jeongsu Ahn
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Chungnam 32588, Korea
| | - Hyun-Geun Song
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Chungnam 32588, Korea
| | - Jae-Woo Han
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Chungnam 32588, Korea
| | - Takeshi Higa
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Akira Takano
- Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuki Nakamura
- Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Noriyuki Suetsugu
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | - Daisuke Kohda
- Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Taro Q P Uyeda
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki 305-8562, Japan
| | - Masamitsu Wada
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan
- Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
13
|
Joo EE, Olson MF. BioID Analysis of Actin-Binding Proteins. Methods Mol Biol 2024; 2794:95-104. [PMID: 38630223 DOI: 10.1007/978-1-0716-3810-1_9] [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: 04/19/2024]
Abstract
Proteins often exist and function as part of higher-order complexes or networks. A challenge is to identify the universe of proximal and interacting partners for a given protein. We describe how the high-activity promiscuous biotin ligase called TurboID is fused to the actin-binding peptide LifeAct to label by biotinylation proteins that bind, or are in close proximity, to actin. The rapid enzyme kinetics of TurboID allows the profiles of actin-binding proteins to be compared under different conditions, such as acute disruption of filamentous actin structures with cytochalasin D.
Collapse
Affiliation(s)
- E Emily Joo
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Michael F Olson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada.
| |
Collapse
|
14
|
Torii T, Sugimoto W, Itoh K, Kinoshita N, Gessho M, Goto T, Uehara I, Nakajima W, Budirahardja Y, Miyoshi D, Nishikata T, Tanaka N, Hirata H, Kawauchi K. Loss of p53 function promotes DNA damage-induced formation of nuclear actin filaments. Cell Death Dis 2023; 14:766. [PMID: 38001089 PMCID: PMC10674001 DOI: 10.1038/s41419-023-06310-0] [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: 02/25/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
Tumor suppressor p53 plays a central role in response to DNA damage. DNA-damaging agents modulate nuclear actin dynamics, influencing cell behaviors; however, whether p53 affects the formation of nuclear actin filaments remains unclear. In this study, we found that p53 depletion promoted the formation of nuclear actin filaments in response to DNA-damaging agents, such as doxorubicin (DOXO) and etoposide (VP16). Even though the genetic probes used for the detection of nuclear actin filaments exerted a promotive effect on actin polymerization, the detected formation of nuclear actin filaments was highly dependent on both p53 depletion and DNA damage. Whilst active p53 is known to promote caspase-1 expression, the overexpression of caspase-1 reduced DNA damage-induced formation of nuclear actin filaments in p53-depleted cells. In contrast, co-treatment with DOXO and the pan-caspase inhibitor Q-VD-OPh or the caspase-1 inhibitor Z-YVAD-FMK induced the formation of nuclear actin filament formation even in cells bearing wild-type p53. These results suggest that the p53-caspase-1 axis suppresses DNA damage-induced formation of nuclear actin filaments. In addition, we found that the expression of nLifeact-GFP, the filamentous-actin-binding peptide Lifeact fused with the nuclear localization signal (NLS) and GFP, modulated the structure of nuclear actin filaments to be phalloidin-stainable in p53-depleted cells treated with the DNA-damaging agent, altering the chromatin structure and reducing the transcriptional activity. The level of phosphorylated H2AX (γH2AX), a marker of DNA damage, in these cells also reduced upon nLifeact-GFP expression, whilst details of the functional relationship between the formation of nLifeact-GFP-decorated nuclear actin filaments and DNA repair remained to be elucidated. Considering that the loss of p53 is associated with cancer progression, the results of this study raise a possibility that the artificial reinforcement of nuclear actin filaments by nLifeact-GFP may enhance the cytotoxic effect of DNA-damaging agents in aggressive cancer cells through a reduction in gene transcription.
Collapse
Affiliation(s)
- Takeru Torii
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Wataru Sugimoto
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Katsuhiko Itoh
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Natsuki Kinoshita
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Masaya Gessho
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Toshiyuki Goto
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Ikuno Uehara
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Wataru Nakajima
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Yemima Budirahardja
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Daisuke Miyoshi
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Takahito Nishikata
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan
| | - Nobuyuki Tanaka
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, 113-8602, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, 924-0838, Japan.
| | - Keiko Kawauchi
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, 650-0047, Japan.
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, 113-8602, Japan.
| |
Collapse
|
15
|
Oosterheert W, Blanc FEC, Roy A, Belyy A, Sanders MB, Hofnagel O, Hummer G, Bieling P, Raunser S. Molecular mechanisms of inorganic-phosphate release from the core and barbed end of actin filaments. Nat Struct Mol Biol 2023; 30:1774-1785. [PMID: 37749275 PMCID: PMC10643162 DOI: 10.1038/s41594-023-01101-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/18/2023] [Indexed: 09/27/2023]
Abstract
The release of inorganic phosphate (Pi) from actin filaments constitutes a key step in their regulated turnover, which is fundamental to many cellular functions. The mechanisms underlying Pi release from the core and barbed end of actin filaments remain unclear. Here, using human and bovine actin isoforms, we combine cryo-EM with molecular-dynamics simulations and in vitro reconstitution to demonstrate how actin releases Pi through a 'molecular backdoor'. While constantly open at the barbed end, the backdoor is predominantly closed in filament-core subunits and opens only transiently through concerted amino acid rearrangements. This explains why Pi escapes rapidly from the filament end but slowly from internal subunits. In a nemaline-myopathy-associated actin variant, the backdoor is predominantly open in filament-core subunits, resulting in accelerated Pi release and filaments with drastically shortened ADP-Pi caps. Our results provide the molecular basis for Pi release from actin and exemplify how a disease-linked mutation distorts the nucleotide-state distribution and atomic structure of the filament.
Collapse
Affiliation(s)
- Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Florian E C Blanc
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Ankit Roy
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
- Institute for Biophysics, Goethe University, Frankfurt am Main, Germany.
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
16
|
Effiong UM, Khairandish H, Ramirez-Velez I, Wang Y, Belardi B. Turn-On Protein Switches for Controlling Actin Binding in Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.561921. [PMID: 37961502 PMCID: PMC10634840 DOI: 10.1101/2023.10.26.561921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Within a shared cytoplasm, filamentous actin (F-actin) plays numerous and critical roles across the cell body. Cells rely on actin-binding proteins (ABPs) to organize F-actin and to integrate its polymeric characteristics into diverse cellular processes. Yet, the multitude of ABPs that engage with and shape F-actin make studying a single ABP's influence on cellular activities a significant challenge. Moreover, without a means of manipulating actin-binding subcellularly, harnessing the F-actin cytoskeleton for synthetic biology purposes remains elusive. Here, we describe a suite of designed proteins, Controllable Actin-binding Switch Tools (CASTs), whose actin-binding behavior can be controlled with external stimuli. CASTs were developed that respond to different external inputs, providing options for turn-on kinetics and enabling orthogonality. Being genetically encoded, we show that CASTs can be inserted into native protein sequences to control F-actin association locally and engineered into new structures to control cell and tissue shape and behavior.
Collapse
Affiliation(s)
- Unyime M. Effiong
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Hannah Khairandish
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Yanran Wang
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712
| |
Collapse
|
17
|
Zsolnay V, Gardel ML, Kovar DR, Voth GA. Cracked actin filaments as mechanosensitive receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546553. [PMID: 37425801 PMCID: PMC10327158 DOI: 10.1101/2023.06.26.546553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Actin filament networks are exposed to mechanical stimuli, but the effect of strain on actin filament structure has not been well-established in molecular detail. This is a critical gap in understanding because the activity of a variety of actin-binding proteins have recently been determined to be altered by actin filament strain. We therefore used all-atom molecular dynamics simulations to apply tensile strains to actin filaments and find that changes in actin subunit organization are minimal in mechanically strained, but intact, actin filaments. However, a conformational change disrupts the critical D-loop to W-loop connection between longitudinal neighboring subunits, which leads to a metastable cracked conformation of the actin filament, whereby one protofilament is broken prior to filament severing. We propose that the metastable crack presents a force-activated binding site for actin regulatory factors that specifically associate with strained actin filaments. Through protein-protein docking simulations, we find that 43 evolutionarily-diverse members of the dual zinc finger containing LIM domain family, which localize to mechanically strained actin filaments, recognize two binding sites exposed at the cracked interface. Furthermore, through its interactions with the crack, LIM domains increase the length of time damaged filaments remain stable. Our findings propose a new molecular model for mechanosensitive binding to actin filaments.
Collapse
Affiliation(s)
- Vilmos Zsolnay
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL
| | - Margaret L. Gardel
- Department of Physics & Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL
- Institute for Biophysical Dynamics and James Franck Institute, University of Chicago, Chicago, IL
| | - David R. Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL
| | - Gregory A. Voth
- Institute for Biophysical Dynamics and James Franck Institute, University of Chicago, Chicago, IL
- Department of Chemistry and Chicago Center for Theoretical Chemistry, University of Chicago, Chicago, IL
| |
Collapse
|
18
|
Ignácz A, Nagy-Herczeg D, Hausser A, Schlett K. Dendritic effects of genetically encoded actin-labeling probes in cultured hippocampal neurons. Mol Biol Cell 2023; 34:br8. [PMID: 36989034 PMCID: PMC10295473 DOI: 10.1091/mbc.e22-08-0331] [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: 08/15/2022] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Actin cytoskeleton predominantly regulates the formation and maintenance of synapses by controlling dendritic spine morphology and motility. To visualize actin dynamics, actin molecules can be labeled by genetically fusing fluorescent proteins to actin monomers, actin-binding proteins, or single-chain anti-actin antibodies. In the present study, we compared the dendritic effect of EGFP-actin, LifeAct-TagGFP2 (LifeAct-GFP), and Actin-Chromobody-TagGFP2 (AC-GFP) in mouse cultured hippocampal neurons using unbiased quantitative methods. The actin-binding probes LifeAct-GFP and AC-GFP showed similar affinity to F-actin, but in contrast to EGFP-actin, they did not reveal subtle changes in actin remodeling between mushroom-shaped spines and filopodia. All tested actin probes colocalized with phalloidin similarly; however, the enrichment of LifeAct-GFP in dendritic spines was remarkably lower compared with the other constructs. LifeAct-GFP expression was tolerated at a higher expression level compared with EGFP-actin and AC-GFP with only subtle differences identified in dendritic spine morphology and protrusion density. While EGFP-actin and LifeAct-GFP expression did not alter dendritic arborization, AC-GFP-expressing neurons displayed a reduced dendritic tree. Thus, although all tested actin probes may be suitable for actin imaging studies, certain limitations should be considered before performing experiments with a particular actin-labeling probe in primary neurons.
Collapse
Affiliation(s)
- Attila Ignácz
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Domonkos Nagy-Herczeg
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
19
|
Oh SC, Kim SE, Jang IH, Kim SM, Lee SY, Lee S, Chu IS, Yoon SR, Jung H, Choi I, Doh J, Kim TD. NgR1 is an NK cell inhibitory receptor that destabilizes the immunological synapse. Nat Immunol 2023; 24:463-473. [PMID: 36624164 DOI: 10.1038/s41590-022-01394-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/22/2022] [Indexed: 01/10/2023]
Abstract
The formation of an immunological synapse (IS) is essential for natural killer (NK) cells to eliminate target cells. Despite an advanced understanding of the characteristics of the IS and its formation processes, the mechanisms that regulate its stability via the cytoskeleton are unclear. Here, we show that Nogo receptor 1 (NgR1) has an important function in modulating NK cell-mediated killing by destabilization of IS formation. NgR1 deficiency or blockade resulted in improved tumor control of NK cells by enhancing NK-to-target cell contact stability and regulating F-actin dynamics during IS formation. Patients with tumors expressing abundant NgR1 ligand had poor prognosis despite high levels of NK cell infiltration. Thus, our study identifies NgR1 as an immune checkpoint in IS formation and indicates a potential approach to improve the cytolytic function of NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Se-Chan Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seong-Eun Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In-Hwan Jang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seok-Min Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Soo Yun Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - In-Sun Chu
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Suk Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Haiyoung Jung
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX Institute, Soft Foundry Institute, Seoul National University, Seoul, Republic of Korea.
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea. .,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea. .,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Republic of Korea. .,Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
20
|
Deciphering the molecular mechanisms of actin cytoskeleton regulation in cell migration using cryo-EM. Biochem Soc Trans 2023; 51:87-99. [PMID: 36695514 PMCID: PMC9987995 DOI: 10.1042/bst20220221] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The actin cytoskeleton plays a key role in cell migration and cellular morphodynamics in most eukaryotes. The ability of the actin cytoskeleton to assemble and disassemble in a spatiotemporally controlled manner allows it to form higher-order structures, which can generate forces required for a cell to explore and navigate through its environment. It is regulated not only via a complex synergistic and competitive interplay between actin-binding proteins (ABP), but also by filament biochemistry and filament geometry. The lack of structural insights into how geometry and ABPs regulate the actin cytoskeleton limits our understanding of the molecular mechanisms that define actin cytoskeleton remodeling and, in turn, impact emerging cell migration characteristics. With the advent of cryo-electron microscopy (cryo-EM) and advanced computational methods, it is now possible to define these molecular mechanisms involving actin and its interactors at both atomic and ultra-structural levels in vitro and in cellulo. In this review, we will provide an overview of the available cryo-EM methods, applicable to further our understanding of the actin cytoskeleton, specifically in the context of cell migration. We will discuss how these methods have been employed to elucidate ABP- and geometry-defined regulatory mechanisms in initiating, maintaining, and disassembling cellular actin networks in migratory protrusions.
Collapse
|
21
|
Actin in action. Nat Methods 2023; 20:178-182. [PMID: 36765135 DOI: 10.1038/s41592-022-01762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
22
|
Bhaskar H, Kleinjan D, Oi C, Gidden Z, Rosser SJ, Horrocks MH, Regan L. Live-cell super-resolution imaging of actin using LifeAct-14 with a PAINT-based approach. Protein Sci 2023; 32:e4558. [PMID: 36585831 PMCID: PMC9878614 DOI: 10.1002/pro.4558] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
We present direct-LIVE-PAINT, an easy-to-implement approach for the nanoscopic imaging of protein structures in live cells using labeled binding peptides. We demonstrate the feasibility of direct-LIVE-PAINT with an actin-binding peptide fused to EGFP, the location of which can be accurately determined as it transiently binds to actin filaments. We show that direct-LIVE-PAINT can be used to image actin structures below the diffraction-limit of light and have used it to observe the dynamic nature of actin in live cells. We envisage a similar approach could be applied to imaging other proteins within live mammalian cells.
Collapse
Affiliation(s)
- Haresh Bhaskar
- The School of Biological SciencesUniversity of EdinburghEdinburghUK
- EaStCHEM School of ChemistryUniversity of EdinburghEdinburghUK
| | - Dirk‐Jan Kleinjan
- Centre for Synthetic and Systems Biology and UK Centre for Mammalian Synthetic Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Curran Oi
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Zoe Gidden
- The School of Biological SciencesUniversity of EdinburghEdinburghUK
- EaStCHEM School of ChemistryUniversity of EdinburghEdinburghUK
| | - Susan J. Rosser
- Centre for Synthetic and Systems Biology and UK Centre for Mammalian Synthetic Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | | | - Lynne Regan
- The School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
23
|
Ali MF, Shin JM, Fatema U, Kurihara D, Berger F, Yuan L, Kawashima T. Cellular dynamics of coenocytic endosperm development in Arabidopsis thaliana. NATURE PLANTS 2023; 9:330-342. [PMID: 36646830 DOI: 10.1038/s41477-022-01331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
After double fertilization, the endosperm in the seeds of many flowering plants undergoes repeated mitotic nuclear divisions without cytokinesis, resulting in a large coenocytic endosperm that then cellularizes. Growth during the coenocytic phase is strongly associated with the final seed size; however, a detailed description of the cellular dynamics controlling the unique coenocytic development in flowering plants has remained elusive. By integrating confocal microscopy live-cell imaging and genetics, we have characterized the entire development of the coenocytic endosperm of Arabidopsis thaliana including nuclear divisions, their timing intervals, nuclear movement and cytoskeleton dynamics. Around each nucleus, microtubules organize into aster-shaped structures that drive actin filament (F-actin) organization. Microtubules promote nuclear movement after division, while F-actin restricts it. F-actin is also involved in controlling the size of both the coenocytic endosperm and the mature seed. The characterization of cytoskeleton dynamics in real time throughout the entire coenocyte endosperm period provides foundational knowledge of plant coenocytic development, insights into the coordination of F-actin and microtubules in nuclear dynamics, and new opportunities to increase seed size and our food security.
Collapse
Affiliation(s)
- Mohammad Foteh Ali
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
| | - Ji Min Shin
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
- Kentucky Tobacco Research and Development Center, University of Kentucky, Lexington, KY, USA
| | - Umma Fatema
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
| | - Daisuke Kurihara
- Institute of Transformative Bio-Molecules (ITbM), Nagoya University, Nagoya, Japan
- Institute for Advanced Research (IAR), Nagoya University, Nagoya, Japan
| | - Frédéric Berger
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Ling Yuan
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
- Kentucky Tobacco Research and Development Center, University of Kentucky, Lexington, KY, USA
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Tomokazu Kawashima
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
24
|
Kroll KL, French AR, Sosnick TR, Rock RS. LILAC: enhanced actin imaging with an optogenetic Lifeact. Nat Methods 2023; 20:214-217. [PMID: 36717692 PMCID: PMC10986358 DOI: 10.1038/s41592-022-01761-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
Lifeact is a popular peptide-based label of actin filaments in live cells. We have designed an improved Lifeact variant, LILAC, that binds to actin in light using the LOV2 protein. Light control allows the user to modulate actin labeling, enabling image analysis that leverages modulation for an enhanced view of F-actin dynamics in cells. Furthermore, the tool reduces actin perturbations and cell sickness caused by Lifeact overexpression.
Collapse
Affiliation(s)
- Kourtney L Kroll
- Biophysical Sciences Graduate Program, The University of Chicago, Chicago, IL, USA
| | - Alexander R French
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Tobin R Sosnick
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA.
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
25
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
26
|
Niu H, Bi F, Zhao W, Xu Y, Han Q, Guo W, Chen Y. Smurf1 regulates ameloblast polarization by ubiquitination-mediated degradation of RhoA. Cell Prolif 2022; 56:e13387. [PMID: 36579844 PMCID: PMC10068949 DOI: 10.1111/cpr.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022] Open
Abstract
Cell polarity is essential for ameloblast differentiation and enamel formation. Smurf1 can mediate cell polarization through ubiquitination degradation of specific substrates. But it remains unclear whether Smurf1 could regulate ameloblast polarity and the underlying mechanism. Here, immuno-fluorescence staining and RT-qPCR were applied to detect the expression of Smurf1 and F-actin. A mouse lower incisor defect model was constructed. Scanning electron microscope, rat lower incisor culture, western blot, wound healing assay and trans-well migration assay were performed to detect the influence of Smurf1 knockdown on ameloblast. IF double staining, western blot and co-immunoprecipitation were conducted to detect the interaction between Smurf1 and RhoA. The in vivo experiment was also performed. We found that Smurf1 was mainly expressed in the membrane and cell cortex of ameloblast, similar to F-actin. Smurf1 expression increased along ameloblast polarization and differentiation. After knocking down Smurf1, the cytoskeleton and cell morphology changed and the cell polarity was damaged. Smurf1 regulated ameloblast polarity through ubiquitination degradation of activated RhoA in vitro. Local knockdown of Smurf1 in rat lower incisor ameloblast resulted in ameloblast polarity loss, enamel matrix secretion disorder and chalky enamel, but RhoA inhibitor Y-27632 could reverse this effect. Collectively, Smurf1 could regulate the polarization of ameloblast through ubiquitination degradation of activated RhoA, which contributed to the knowledge of tooth development and provided new research ideas for cell polarity.
Collapse
Affiliation(s)
- Haoman Niu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Fei Bi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wenjun Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yuchan Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qi Han
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Oral Pathology, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Chou SZ, Chatterjee M, Pollard TD. Mechanism of actin filament branch formation by Arp2/3 complex revealed by a high-resolution cryo-EM structureof the branch junction. Proc Natl Acad Sci U S A 2022; 119:e2206722119. [PMID: 36442092 PMCID: PMC9894260 DOI: 10.1073/pnas.2206722119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 10/17/2022] [Indexed: 11/29/2022] Open
Abstract
We reconstructed the structure of actin filament branch junctions formed by fission yeast Arp2/3 complex at 3.5 Å resolution from images collected by electron cryo-microscopy. During specimen preparation, all of the actin subunits and Arp3 hydrolyzed their bound adenosine triphosphate (ATP) and dissociated the γ-phosphate, but Arp2 retained the γ-phosphate. Binding tightly to the side of the mother filament and nucleating the daughter filament growing as a branch requires Arp2/3 complex to undergo a dramatic conformational change where two blocks of structure rotate relative to each other about 25° to align Arp2 and Arp3 as the first two subunits in the branch. During branch formation, Arp2/3 complex acquires more than 8,000 Å2 of new buried surface, accounting for the stability of the branch. Inactive Arp2/3 complex binds only transiently to the side of an actin filament, because its conformation allows only a subset of the interactions found in the branch junction.
Collapse
Affiliation(s)
- Steven Z. Chou
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT06520
| | - Moon Chatterjee
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT06520
| | - Thomas D. Pollard
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT06520
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520
- Department of Cell Biology, Yale University, New Haven, CT06520
| |
Collapse
|
28
|
Oosterheert W, Klink BU, Belyy A, Pospich S, Raunser S. Structural basis of actin filament assembly and aging. Nature 2022; 611:374-379. [DOI: 10.1038/s41586-022-05241-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022]
Abstract
AbstractThe dynamic turnover of actin filaments (F-actin) controls cellular motility in eukaryotes and is coupled to changes in the F-actin nucleotide state1–3. It remains unclear how F-actin hydrolyses ATP and subsequently undergoes subtle conformational rearrangements that ultimately lead to filament depolymerization by actin-binding proteins. Here we present cryo-electron microscopy structures of F-actin in all nucleotide states, polymerized in the presence of Mg2+ or Ca2+ at approximately 2.2 Å resolution. The structures show that actin polymerization induces the relocation of water molecules in the nucleotide-binding pocket, activating one of them for the nucleophilic attack of ATP. Unexpectedly, the back door for the subsequent release of inorganic phosphate (Pi) is closed in all structures, indicating that Pi release occurs transiently. The small changes in the nucleotide-binding pocket after ATP hydrolysis and Pi release are sensed by a key amino acid, amplified and transmitted to the filament periphery. Furthermore, differences in the positions of water molecules in the nucleotide-binding pocket explain why Ca2+-actin shows slower polymerization rates than Mg2+-actin. Our work elucidates the solvent-driven rearrangements that govern actin filament assembly and aging and lays the foundation for the rational design of drugs and small molecules for imaging and therapeutic applications.
Collapse
|
29
|
Belyy A, Lindemann F, Roderer D, Funk J, Bardiaux B, Protze J, Bieling P, Oschkinat H, Raunser S. Mechanism of threonine ADP-ribosylation of F-actin by a Tc toxin. Nat Commun 2022; 13:4202. [PMID: 35858890 PMCID: PMC9300711 DOI: 10.1038/s41467-022-31836-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022] Open
Abstract
Tc toxins deliver toxic enzymes into host cells by a unique injection mechanism. One of these enzymes is the actin ADP-ribosyltransferase TccC3, whose activity leads to the clustering of the cellular cytoskeleton and ultimately cell death. Here, we show in atomic detail how TccC3 modifies actin. We find that the ADP-ribosyltransferase does not bind to G-actin but interacts with two consecutive actin subunits of F-actin. The binding of TccC3 to F-actin occurs via an induced-fit mechanism that facilitates access of NAD+ to the nucleotide binding pocket. The following nucleophilic substitution reaction results in the transfer of ADP-ribose to threonine-148 of F-actin. We demonstrate that this site-specific modification of F-actin prevents its interaction with depolymerization factors, such as cofilin, which impairs actin network turnover and leads to steady actin polymerization. Our findings reveal in atomic detail a mechanism of action of a bacterial toxin through specific targeting and modification of F-actin. Entomopathogenic bacteria used for pest control secrete potent Tc toxins. Here, the authors combine biochemistry, solution and solid-state NMR spectroscopy and cryo-EM to show in atomic detail how the toxin disrupts the host cell cytoskeleton and kills the target cell.
Collapse
Affiliation(s)
- Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Florian Lindemann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Daniel Roderer
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.,Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Johanna Funk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Benjamin Bardiaux
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, 25-28 Rue du Docteur Roux, F-75015, Paris, France
| | - Jonas Protze
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.
| |
Collapse
|
30
|
Cordell P, Carrington G, Curd A, Parker F, Tomlinson D, Peckham M. Affimers and nanobodies as molecular probes and their applications in imaging. J Cell Sci 2022; 135:276020. [PMID: 35848463 PMCID: PMC9450889 DOI: 10.1242/jcs.259168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibodies are the most widely used, traditional tool for labelling molecules in cells. In the past five to ten years, many new labelling tools have been developed with significant advantages over the traditional antibody. Here, we focus on nanobodies and the non-antibody binding scaffold proteins called Affimers. We explain how they are generated, selected and produced, and we describe how their small size, high binding affinity and specificity provides them with many advantages compared to antibodies. Of particular importance, their small size enables them to better penetrate dense cytoskeletal regions within cells, as well as tissues, providing them with specific advantage for super-resolution imaging, as they place the fluorophore with a few nanometres of the target protein being imaged. We expect these novel tools to be of broad interest to many cell biologists and anticipate them becoming the tools of choice for super-resolution imaging.
Collapse
|
31
|
Morales EA, Arnaiz C, Krystofiak ES, Zanic M, Tyska MJ. Mitotic Spindle Positioning (MISP) is an actin bundler that selectively stabilizes the rootlets of epithelial microvilli. Cell Rep 2022; 39:110692. [PMID: 35443169 PMCID: PMC9097542 DOI: 10.1016/j.celrep.2022.110692] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/20/2022] [Accepted: 03/24/2022] [Indexed: 11/11/2022] Open
Abstract
Microvilli are conserved actin-based surface protrusions that have been repurposed throughout evolution to fulfill diverse cell functions. In the case of transporting epithelia, microvilli are supported by a core of actin filaments bundled in parallel by villin, fimbrin, and espin. Remarkably, microvilli biogenesis persists in mice lacking all three of these factors, suggesting the existence of unknown bundlers. We identified Mitotic Spindle Positioning (MISP) as an actin-binding factor that localizes specifically to the rootlet end of the microvillus. MISP promotes rootlet elongation in cells, and purified MISP exhibits potent filament bundling activity in vitro. MISP-bundled filaments also recruit fimbrin, which further elongates and stabilizes bundles. MISP confinement to the rootlet is enforced by ezrin, which prevents decoration of the membrane-wrapped distal end of the core bundle. These discoveries reveal how epithelial cells optimize apical membrane surface area and offer insight on the remarkable robustness of microvilli biogenesis. Morales et al. identify Mitotic Spindle Positioning (MISP) as an actin bundler in the rootlets of epithelial microvilli. MISP cooperates with other bundlers, and its rootlet-specific localization is enforced by membrane-actin linker ezrin. These findings illuminate mechanisms that drive the assembly and compartmentalization of actin bundle-supported protrusions.
Collapse
Affiliation(s)
- E Angelo Morales
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Cayetana Arnaiz
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Evan S Krystofiak
- Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN 37232, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
32
|
Ma OX, Chong WG, Lee JKE, Cai S, Siebert CA, Howe A, Zhang P, Shi J, Surana U, Gan L. Cryo-ET detects bundled triple helices but not ladders in meiotic budding yeast. PLoS One 2022; 17:e0266035. [PMID: 35421110 PMCID: PMC9009673 DOI: 10.1371/journal.pone.0266035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/13/2022] [Indexed: 11/19/2022] Open
Abstract
In meiosis, cells undergo two sequential rounds of cell division, termed meiosis I and meiosis II. Textbook models of the meiosis I substage called pachytene show that nuclei have conspicuous 100-nm-wide, ladder-like synaptonemal complexes and ordered chromatin loops. It remains unknown if these cells have any other large, meiosis-related intranuclear structures. Here we present cryo-ET analysis of frozen-hydrated budding yeast cells before, during, and after pachytene. We found no cryo-ET densities that resemble dense ladder-like structures or ordered chromatin loops. Instead, we found large numbers of 12-nm-wide triple-helices that pack into ordered bundles. These structures, herein called meiotic triple helices (MTHs), are present in meiotic cells, but not in interphase cells. MTHs are enriched in the nucleus but not enriched in the cytoplasm. Bundles of MTHs form at the same timeframe as synaptonemal complexes (SCs) in wild-type cells and in mutant cells that are unable to form SCs. These results suggest that in yeast, SCs coexist with previously unreported large, ordered assemblies.
Collapse
Affiliation(s)
- Olivia X. Ma
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Wen Guan Chong
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Joy K. E. Lee
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Shujun Cai
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - C. Alistair Siebert
- Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, Oxfordshire, United Kingdom
| | - Andrew Howe
- Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, Oxfordshire, United Kingdom
| | - Peijun Zhang
- Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, Oxfordshire, United Kingdom
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jian Shi
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Uttam Surana
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
- Bioprocessing Technology Institute, A*STAR, Singapore, Singapore
- Biotransformation Innovation Platform, A*STAR, Singapore, Singapore
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Lu Gan
- Department of Biological Sciences and Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
33
|
Erdener ŞE, Küreli G, Dalkara T. Contractile apparatus in CNS capillary pericytes. NEUROPHOTONICS 2022; 9:021904. [PMID: 35106320 PMCID: PMC8785978 DOI: 10.1117/1.nph.9.2.021904] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Significance: Whether or not capillary pericytes contribute to blood flow regulation in the brain and retina has long been debated. This was partly caused by failure of detecting the contractile protein α -smooth muscle actin ( α -SMA) in capillary pericytes. Aim: The aim of this review is to summarize recent developments in detecting α -SMA and contractility in capillary pericytes and the relevant literature on the biology of actin filaments. Results: Evidence suggests that for visualization of the small amounts of α -SMA in downstream mid-capillary pericytes, actin depolymerization must be prevented during tissue processing. Actin filaments turnover is mainly based on de/re-polymerization rather than transcription of the monomeric form, hence, small amounts of α -SMA mRNA may evade detection by transcriptomic studies. Similarly, transgenic mice expressing fluorescent reporters under the α -SMA promoter may yield low fluorescence due to limited transcriptional activity in mid-capillary pericytes. Recent studies show that pericytes including mid-capillary ones express several actin isoforms and myosin heavy chain type 11, the partner of α -SMA in mediating contraction. Emerging evidence also suggests that actin polymerization in pericytes may have a role in regulating the tone of downstream capillaries. Conclusions: With guidance of actin biology, innovative labeling and imaging techniques can reveal the molecular machinery of contraction in pericytes.
Collapse
Affiliation(s)
- Şefik E. Erdener
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Gülce Küreli
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Turgay Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| |
Collapse
|
34
|
Sing CN, Yang EJ, Swayne TC, Higuchi-Sanabria R, Tsang CA, Boldogh IR, Pon LA. Imaging the Actin Cytoskeleton in Live Budding Yeast Cells. Methods Mol Biol 2022; 2364:53-80. [PMID: 34542848 PMCID: PMC11060504 DOI: 10.1007/978-1-0716-1661-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Although budding yeast, Saccharomyces cerevisiae, is widely used as a model organism in biological research, studying cell biology in yeast was hindered due to its small size, rounded morphology, and cell wall. However, with improved techniques, researchers can acquire high-resolution images and carry out rapid multidimensional analysis of a yeast cell. As a result, imaging in yeast has emerged as an important tool to study cytoskeletal organization, function, and dynamics. This chapter describes techniques and approaches for visualizing the actin cytoskeleton in live yeast cells.
Collapse
Affiliation(s)
- Cierra N Sing
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
| | - Emily J Yang
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
| | - Theresa C Swayne
- Confocal and Specialized Microscopy Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Ryo Higuchi-Sanabria
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Catherine A Tsang
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
| | - Istvan R Boldogh
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
- Confocal and Specialized Microscopy Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Liza A Pon
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY, USA.
- Confocal and Specialized Microscopy Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| |
Collapse
|
35
|
Doran MH, Lehman W. The Central Role of the F-Actin Surface in Myosin Force Generation. BIOLOGY 2021; 10:1221. [PMID: 34943138 PMCID: PMC8698748 DOI: 10.3390/biology10121221] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022]
Abstract
Actin is one of the most abundant and versatile proteins in eukaryotic cells. As discussed in many contributions to this Special Issue, its transition from a monomeric G-actin to a filamentous F-actin form plays a critical role in a variety of cellular processes, including control of cell shape and cell motility. Once polymerized from G-actin, F-actin forms the central core of muscle-thin filaments and acts as molecular tracks for myosin-based motor activity. The ATP-dependent cross-bridge cycle of myosin attachment and detachment drives the sliding of myosin thick filaments past thin filaments in muscle and the translocation of cargo in somatic cells. The variation in actin function is dependent on the variation in muscle and non-muscle myosin isoform behavior as well as interactions with a plethora of additional actin-binding proteins. Extensive work has been devoted to defining the kinetics of actin-based force generation powered by the ATPase activity of myosin. In addition, over the past decade, cryo-electron microscopy has revealed the atomic-evel details of the binding of myosin isoforms on the F-actin surface. Most accounts of the structural interactions between myosin and actin are described from the perspective of the myosin molecule. Here, we discuss myosin-binding to actin as viewed from the actin surface. We then describe conserved structural features of actin required for the binding of all or most myosin isoforms while also noting specific interactions unique to myosin isoforms.
Collapse
Affiliation(s)
- Matthew H. Doran
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | - William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
36
|
Belyy A, Merino F, Mechold U, Raunser S. Mechanism of actin-dependent activation of nucleotidyl cyclase toxins from bacterial human pathogens. Nat Commun 2021; 12:6628. [PMID: 34785651 PMCID: PMC8595890 DOI: 10.1038/s41467-021-26889-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial human pathogens secrete initially inactive nucleotidyl cyclases that become potent enzymes by binding to actin inside eukaryotic host cells. The underlying molecular mechanism of this activation is, however, unclear. Here, we report structures of ExoY from Pseudomonas aeruginosa and Vibrio vulnificus bound to their corresponding activators F-actin and profilin-G-actin. The structures reveal that in contrast to the apo-state, two flexible regions become ordered and interact strongly with actin. The specific stabilization of these regions results in an allosteric stabilization of the nucleotide binding pocket and thereby to an activation of the enzyme. Differences in the sequence and conformation of the actin-binding regions are responsible for the selective binding to either F- or G-actin. Other nucleotidyl cyclase toxins that bind to calmodulin rather than actin undergo a similar disordered-to-ordered transition during activation, suggesting that the allosteric activation-by-stabilization mechanism of ExoY is conserved in these enzymes, albeit the different activator.
Collapse
Affiliation(s)
- Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tübingen, Germany
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.
| |
Collapse
|
37
|
Xu R, Du S. Overexpression of Lifeact-GFP Disrupts F-Actin Organization in Cardiomyocytes and Impairs Cardiac Function. Front Cell Dev Biol 2021; 9:746818. [PMID: 34765602 PMCID: PMC8576398 DOI: 10.3389/fcell.2021.746818] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/07/2021] [Indexed: 11/28/2022] Open
Abstract
Lifeact-GFP is a frequently used molecular probe to study F-actin structure and dynamic assembly in living cells. In this study, we generated transgenic zebrafish models expressing Lifeact-GFP specifically in cardiac muscles to investigate the effect of Lifeact-GFP on heart development and its application to study cardiomyopathy. The data showed that transgenic zebrafish with low to moderate levels of Lifeact-GFP expression could be used as a good model to study contractile dynamics of actin filaments in cardiac muscles in vivo. Using this model, we demonstrated that loss of Smyd1b, a lysine methyltransferase, disrupted F-actin filament organization in cardiomyocytes of zebrafish embryos. Our studies, however, also demonstrated that strong Lifeact-GFP expression in cardiomyocytes was detrimental to actin filament organization in cardiomyocytes that led to pericardial edema and early embryonic lethality of zebrafish embryos. Collectively, these data suggest that although Lifeact-GFP is a good probe for visualizing F-actin dynamics, transgenic models need to be carefully evaluated to avoid artifacts induced by Lifeact-GFP overexpression.
Collapse
Affiliation(s)
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
38
|
Kilo L, Stürner T, Tavosanis G, Ziegler AB. Drosophila Dendritic Arborisation Neurons: Fantastic Actin Dynamics and Where to Find Them. Cells 2021; 10:2777. [PMID: 34685757 PMCID: PMC8534399 DOI: 10.3390/cells10102777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 01/27/2023] Open
Abstract
Neuronal dendrites receive, integrate, and process numerous inputs and therefore serve as the neuron's "antennae". Dendrites display extreme morphological diversity across different neuronal classes to match the neuron's specific functional requirements. Understanding how this structural diversity is specified is therefore important for shedding light on information processing in the healthy and diseased nervous system. Popular models for in vivo studies of dendrite differentiation are the four classes of dendritic arborization (c1da-c4da) neurons of Drosophila larvae with their class-specific dendritic morphologies. Using da neurons, a combination of live-cell imaging and computational approaches have delivered information on the distinct phases and the time course of dendrite development from embryonic stages to the fully developed dendritic tree. With these data, we can start approaching the basic logic behind differential dendrite development. A major role in the definition of neuron-type specific morphologies is played by dynamic actin-rich processes and the regulation of their properties. This review presents the differences in the growth programs leading to morphologically different dendritic trees, with a focus on the key role of actin modulatory proteins. In addition, we summarize requirements and technological progress towards the visualization and manipulation of such actin regulators in vivo.
Collapse
Affiliation(s)
- Lukas Kilo
- Dendrite Differentiation, German Center for Neurodegenerative Diseases, 53115 Bonn, Germany; (L.K.); (G.T.)
| | - Tomke Stürner
- Department of Zoology, University of Cambridge, Cambridge CB2 1TN, UK;
| | - Gaia Tavosanis
- Dendrite Differentiation, German Center for Neurodegenerative Diseases, 53115 Bonn, Germany; (L.K.); (G.T.)
- LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Anna B. Ziegler
- Institute of Neuro- and Behavioral Biology, University of Münster, 48149 Münster, Germany
| |
Collapse
|
39
|
Pospich S, Küllmer F, Nasufović V, Funk J, Belyy A, Bieling P, Arndt H, Raunser S. Cryo‐EM Resolves Molecular Recognition Of An Optojasp Photoswitch Bound To Actin Filaments In Both Switch States. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Sabrina Pospich
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Florian Küllmer
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Veselin Nasufović
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Johanna Funk
- Department of Systemic Cell Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Alexander Belyy
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Peter Bieling
- Department of Systemic Cell Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Hans‐Dieter Arndt
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Stefan Raunser
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| |
Collapse
|
40
|
Pospich S, Küllmer F, Nasufović V, Funk J, Belyy A, Bieling P, Arndt HD, Raunser S. Cryo-EM Resolves Molecular Recognition Of An Optojasp Photoswitch Bound To Actin Filaments In Both Switch States. Angew Chem Int Ed Engl 2021; 60:8678-8682. [PMID: 33449370 PMCID: PMC8048601 DOI: 10.1002/anie.202013193] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/15/2020] [Indexed: 12/16/2022]
Abstract
Actin is essential for key processes in all eukaryotic cells. Cellpermeable optojasps provide spatiotemporal control of the actin cytoskeleton, confining toxicity and potentially rendering F-actin druggable by photopharmacology. Here, we report cryo electron microscopy (cryo-EM) structures of both isomeric states of one optojasp bound to actin filaments. The high-resolution structures reveal for the first time the pronounced effects of photoswitching a functionalized azobenzene. By characterizing the optojasp binding site and identifying conformational changes within F-actin that depend on the optojasp isomeric state, we refine determinants for the design of functional F-actin photoswitches.
Collapse
Affiliation(s)
- Sabrina Pospich
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Florian Küllmer
- Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University, Humboldtstr. 10, 07743, Jena, Germany
| | - Veselin Nasufović
- Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University, Humboldtstr. 10, 07743, Jena, Germany
| | - Johanna Funk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Hans-Dieter Arndt
- Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University, Humboldtstr. 10, 07743, Jena, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| |
Collapse
|
41
|
Fäßler F, Dimchev G, Hodirnau VV, Wan W, Schur FKM. Cryo-electron tomography structure of Arp2/3 complex in cells reveals new insights into the branch junction. Nat Commun 2020; 11:6437. [PMID: 33353942 PMCID: PMC7755917 DOI: 10.1038/s41467-020-20286-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
The actin-related protein (Arp)2/3 complex nucleates branched actin filament networks pivotal for cell migration, endocytosis and pathogen infection. Its activation is tightly regulated and involves complex structural rearrangements and actin filament binding, which are yet to be understood. Here, we report a 9.0 Å resolution structure of the actin filament Arp2/3 complex branch junction in cells using cryo-electron tomography and subtomogram averaging. This allows us to generate an accurate model of the active Arp2/3 complex in the branch junction and its interaction with actin filaments. Notably, our model reveals a previously undescribed set of interactions of the Arp2/3 complex with the mother filament, significantly different to the previous branch junction model. Our structure also indicates a central role for the ArpC3 subunit in stabilizing the active conformation.
Collapse
Affiliation(s)
- Florian Fäßler
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Georgi Dimchev
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | | | - William Wan
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University, Nashville, United States of America
| | - Florian K M Schur
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria.
| |
Collapse
|