1
|
Wu M, Marchando P, Meyer K, Tang Z, Woolfson DN, Weiner OD. The WAVE complex forms linear arrays at negative membrane curvature to instruct lamellipodia formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.600855. [PMID: 39026726 PMCID: PMC11257481 DOI: 10.1101/2024.07.08.600855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cells generate a wide range of actin-based membrane protrusions for various cell behaviors. These protrusions are organized by different actin nucleation promoting factors. For example, N-WASP controls finger-like filopodia, whereas the WAVE complex controls sheet-like lamellipodia. These different membrane morphologies likely reflect different patterns of nucleator self-organization. N-WASP phase separation has been successfully studied through biochemical reconstitutions, but how the WAVE complex self-organizes to instruct lamellipodia is unknown. Because WAVE complex self-organization has proven refractory to cell-free studies, we leverage in vivo biochemical approaches to investigate WAVE complex organization within its native cellular context. With single molecule tracking and molecular counting, we show that the WAVE complex forms highly regular multilayered linear arrays at the plasma membrane that are reminiscent of a microtubule-like organization. Similar to the organization of microtubule protofilaments in a curved array, membrane curvature is both necessary and sufficient for formation of these WAVE complex linear arrays, though actin polymerization is not. This dependency on negative membrane curvature could explain both the templating of lamellipodia and their emergent behaviors, including barrier avoidance. Our data uncover the key biophysical properties of mesoscale WAVE complex patterning and highlight an integral relationship between NPF self-organization and cell morphogenesis.
Collapse
Affiliation(s)
- Muziyue Wu
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute,University of California San Francisco, San Francisco, CA, USA
| | - Paul Marchando
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - Kirstin Meyer
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute,University of California San Francisco, San Francisco, CA, USA
| | - Ziqi Tang
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Derek N Woolfson
- School of Chemistry, University of Bristol, Bristol, UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol, Bristol, UK
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol, UK
- Bristol BioDesign Institute, University of Bristol, Bristol, UK
| | - Orion D Weiner
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute,University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Kastian RF, Baba K, Kaewkascholkul N, Sasaki H, Watanabe R, Toriyama M, Inagaki N. Dephosphorylation of neural wiring protein shootin1 by PP1 phosphatase regulates netrin-1-induced axon guidance. J Biol Chem 2023; 299:104687. [PMID: 37044214 DOI: 10.1016/j.jbc.2023.104687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Axon pathfinding is an essential step in neuronal network formation. Shootin1a is a clutch-linker molecule that is mechanically involved in axon outgrowth and guidance. It was previously shown that concentration gradients of axon guidance molecule netrin-1 in the extracellular environment elicit asymmetrically localized Pak1 kinase-mediated phosphorylation of shootin1a within axonal growth cones, which is higher on the netrin-1 source side. This asymmetric phosphorylation promotes shootin1a-mediated local actin-adhesion coupling within growth cones, thereby generating directional forces for turning the growth cone toward the netrin-1 source. However, how the spatial differences in netrin-1 concentration are transduced into the asymmetrically localized signaling within growth cones remains unclear. Moreover, the protein phosphatases that dephosphorylate shootin1a remain unidentified. Here, we report that protein phosphatase-1 (PP1) dephosphorylates shootin1a in growth cones. We found that PP1 overexpression abolished the netrin-1-induced asymmetric localization of phosphorylated-shootin1a as well as axon turning. In addition, we show PP1 inhibition reversed the asymmetrically localized shootin1a phosphorylation within growth cones under netrin-1 gradient, thereby changing the netrin-1-induced growth cone turning from attraction to repulsion. These data indicate that PP1-mediated shootin1a dephosphorylation plays a key role in organizing asymmetrically-localized phosphorylated shootin1a within growth cones, which regulates netrin-1-induced axon guidance.
Collapse
Affiliation(s)
- Ria Fajarwati Kastian
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan; Mammalian Cell Engineering and Signal Transduction Research Group, Research Center for Genetic Engineering, National Research and Innovation Agency, KST Soekarno, Jl. Raya Bogor, KM. 46, Cibinong, Bogor, West Java, 16911, Indonesia
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Napol Kaewkascholkul
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Hisashi Sasaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Rikiya Watanabe
- Molecular Physiology Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Michinori Toriyama
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
4
|
Medina-Villalobos N, Avila R, Marsal M, Andilla J, Loza-Álvarez P, Ojeda-Ramírez MM, Tamariz E. Infrared Laser Effects on Cell Projection Depend on Irradiation Intermittence and Cell Activity. Cells 2023; 12:540. [PMID: 36831208 PMCID: PMC9954793 DOI: 10.3390/cells12040540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/06/2023] [Accepted: 01/28/2023] [Indexed: 02/10/2023] Open
Abstract
Highly focused near-infrared (NIR) lasers have been used to induce fibroblast and neuron protrusions in a technique called optical guidance. However, little is known about the biochemical and biophysical effects that the laser provokes in the cell and optimal protocols of stimulation have not yet been established. Using intermittent NIR laser radiation and multivariate time series representations of cell leading edge movement, we analyzed the direction and velocity of cell protrusions. We found that the orientation and advance of PC12 neuron phenotype cells and 3T3 fibroblasts protrusions remain after the laser is turned off, but the observed increase in velocity stops when radiation ceases. For an increase in the speed and distance of cell protrusions by NIR laser irradiation, the cell leading edge needs to be advancing prior to the stimulation, and NIR irradiation does not enable the cell to switch between retracting and advancing states. Using timelapse imaging of actin-GFP, we observed that NIR irradiation induces a faster recruitment of actin, promoting filament formation at the induced cell protrusions. These results provide fresh evidence to understand the phenomenon of the optical guidance of cell protrusions.
Collapse
Affiliation(s)
| | - Remy Avila
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), A.P. 1-1010, Juriquilla 76000, Querétaro, Mexico
| | - María Marsal
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Jordi Andilla
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | - Pablo Loza-Álvarez
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Barcelona, Spain
| | | | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
| |
Collapse
|
5
|
Gardeta SR, García-Cuesta EM, D’Agostino G, Soler Palacios B, Quijada-Freire A, Lucas P, Bernardino de la Serna J, Gonzalez-Riano C, Barbas C, Rodríguez-Frade JM, Mellado M. Sphingomyelin Depletion Inhibits CXCR4 Dynamics and CXCL12-Mediated Directed Cell Migration in Human T Cells. Front Immunol 2022; 13:925559. [PMID: 35903108 PMCID: PMC9315926 DOI: 10.3389/fimmu.2022.925559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022] Open
Abstract
Sphingolipids, ceramides and cholesterol are integral components of cellular membranes, and they also play important roles in signal transduction by regulating the dynamics of membrane receptors through their effects on membrane fluidity. Here, we combined biochemical and functional assays with single-particle tracking analysis of diffusion in the plasma membrane to demonstrate that the local lipid environment regulates CXCR4 organization and function and modulates chemokine-triggered directed cell migration. Prolonged treatment of T cells with bacterial sphingomyelinase promoted the complete and sustained breakdown of sphingomyelins and the accumulation of the corresponding ceramides, which altered both membrane fluidity and CXCR4 nanoclustering and dynamics. Under these conditions CXCR4 retained some CXCL12-mediated signaling activity but failed to promote efficient directed cell migration. Our data underscore a critical role for the local lipid composition at the cell membrane in regulating the lateral mobility of chemokine receptors, and their ability to dynamically increase receptor density at the leading edge to promote efficient cell migration.
Collapse
Affiliation(s)
- Sofía R. Gardeta
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eva M. García-Cuesta
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Gianluca D’Agostino
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Blanca Soler Palacios
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Adriana Quijada-Freire
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pilar Lucas
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jorge Bernardino de la Serna
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Central Laser Facility, Rutherford Appleton Laboratory, Medical Research Council-Research Complex at Harwell, Science and Technology Facilities Council, Harwell, United Kingdom
- National Institute for Health and Care Research Imperial Biomedical Research Center, London, United Kingdom
| | - Carolina Gonzalez-Riano
- Metabolomic and Bioanalysis Center (CEMBIO), Pharmacy Faculty, Centro de Estudios Universitarios Universities, Madrid, Spain
| | - Coral Barbas
- Metabolomic and Bioanalysis Center (CEMBIO), Pharmacy Faculty, Centro de Estudios Universitarios Universities, Madrid, Spain
| | - José Miguel Rodríguez-Frade
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mario Mellado
- Chemokine Signaling Group, Department of Immunology and Oncology, National Center for Biotechnology/Consejo Superior de Investigaciones Científicas, Madrid, Spain
- *Correspondence: Mario Mellado,
| |
Collapse
|
6
|
Microtopographical guidance of macropinocytic signaling patches. Proc Natl Acad Sci U S A 2021; 118:2110281118. [PMID: 34876521 PMCID: PMC8685668 DOI: 10.1073/pnas.2110281118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/28/2022] Open
Abstract
Morphologies of amoebae and immune cells are highly deformable and dynamic, which facilitates migration in various terrains, as well as ingestion of extracellular solutes and particles. It remains largely unexplored whether and how the underlying membrane protrusions are triggered and guided by the geometry of the surface in contact. In this study, we show that in Dictyostelium, the precursor of a structure called macropinocytic cup, which has been thought to be a constitutive process for the uptake of extracellular fluid, is triggered by micrometer-scale surface features. Imaging analysis and computational simulations demonstrate how the topographical dependence of the self-organizing dynamics supports efficient guidance and capturing of the membrane protrusion and hence movement of an entire cell along such surface features. In fast-moving cells such as amoeba and immune cells, dendritic actin filaments are spatiotemporally regulated to shape large-scale plasma membrane protrusions. Despite their importance in migration, as well as in particle and liquid ingestion, how their dynamics are affected by micrometer-scale features of the contact surface is still poorly understood. Here, through quantitative image analysis of Dictyostelium on microfabricated surfaces, we show that there is a distinct mode of topographical guidance directed by the macropinocytic membrane cup. Unlike other topographical guidance known to date that depends on nanometer-scale curvature sensing protein or stress fibers, the macropinocytic membrane cup is driven by the Ras/PI3K/F-actin signaling patch and its dependency on the micrometer-scale topographical features, namely PI3K/F-actin–independent accumulation of Ras-GTP at the convex curved surface, PI3K-dependent patch propagation along the convex edge, and its actomyosin-dependent constriction at the concave edge. Mathematical model simulations demonstrate that the topographically dependent initiation, in combination with the mutually defining patch patterning and the membrane deformation, gives rise to the topographical guidance. Our results suggest that the macropinocytic cup is a self-enclosing structure that can support liquid ingestion by default; however, in the presence of structured surfaces, it is directed to faithfully trace bent and bifurcating ridges for particle ingestion and cell guidance.
Collapse
|
7
|
Mac Donald K, Iulianella A. The actin-cytoskeleton associating protein BASP1 regulates neural progenitor localization in the neural tube. Genesis 2021; 60:e23464. [PMID: 34897971 DOI: 10.1002/dvg.23464] [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: 08/06/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 11/10/2022]
Abstract
Brain acid soluble protein 1 (BASP1; previously NAP22 or CAP23) is an actin-associating protein that is highly expressed in the nervous system throughout development. While its roles at the neuromuscular junction and in certain non-neuronal tissues have been previously characterized, its function in the early neural tube is unclear. Using in ovo electroporation in the chicken (Gallus gallus) embryonic neural tube, we show that BASP1 overexpression resulted in the appearance of ectopic neural progenitor cells within the marginal zone of the neural tube. BASP1 knockdown did not affect the position of neural progenitors but did alter the complexity of axons developing from differentiated neurons. This suggests a role for BASP1 in regulating the apical polarity of progenitor cells and axon trajectories from developing neurons.
Collapse
Affiliation(s)
- Kaitlin Mac Donald
- Department of Medical Neuroscience, and Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Science Research Institute, Halifax, Nova Scotia, Canada
| | - Angelo Iulianella
- Department of Medical Neuroscience, and Brain Repair Centre, Faculty of Medicine, Dalhousie University, Life Science Research Institute, Halifax, Nova Scotia, Canada
| |
Collapse
|
8
|
Studying Neuronal Biology Using Spinning Disc Confocal Microscopy. Methods Mol Biol 2021. [PMID: 34028722 DOI: 10.1007/978-1-0716-1402-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Cytoskeletal integrity is essential for neuronal complexity and functionality. Certain inherited neurological diseases are associated with mutated genes that directly or indirectly compromise cytoskeletal stability. While the large size and complexity of the neurons grown in culture poses certain challenges for imaging, live-cell imaging is an excellent approach to determine the morphological consequences of such mutants. This protocol details the use of spinning disk confocal microscopy and image analysis tools to evaluate branching and neurite length of healthy iPSC-derived glutamatergic neurons that express specific fluorescent proteins. The protocols can be adapted to neuronal cell lines of choice by the investigator.
Collapse
|
9
|
Matyszewski M, Zheng W, Lueck J, Mazanek Z, Mohideen N, Lau AY, Egelman EH, Sohn J. Distinct axial and lateral interactions within homologous filaments dictate the signaling specificity and order of the AIM2-ASC inflammasome. Nat Commun 2021; 12:2735. [PMID: 33980849 PMCID: PMC8115694 DOI: 10.1038/s41467-021-23045-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/14/2021] [Indexed: 02/03/2023] Open
Abstract
Inflammasomes are filamentous signaling platforms integral to innate immunity. Currently, little is known about how these structurally similar filaments recognize and distinguish one another. A cryo-EM structure of the AIM2PYD filament reveals that the architecture of the upstream filament is essentially identical to that of the adaptor ASCPYD filament. In silico simulations using Rosetta and molecular dynamics followed by biochemical and cellular experiments consistently demonstrate that individual filaments assemble bidirectionally. By contrast, the recognition between AIM2 and ASC requires at least one to be oligomeric and occurs in a head-to-tail manner. Using in silico mutagenesis as a guide, we also identify specific axial and lateral interfaces that dictate the recognition and distinction between AIM2 and ASC filaments. Together, the results here provide a robust framework for delineating the signaling specificity and order of inflammasomes.
Collapse
Affiliation(s)
- Mariusz Matyszewski
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Weili Zheng
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jacob Lueck
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary Mazanek
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Naveen Mohideen
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Albert Y Lau
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jungsan Sohn
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Kastian RF, Minegishi T, Baba K, Saneyoshi T, Katsuno-Kambe H, Saranpal S, Hayashi Y, Inagaki N. Shootin1a-mediated actin-adhesion coupling generates force to trigger structural plasticity of dendritic spines. Cell Rep 2021; 35:109130. [PMID: 34010643 DOI: 10.1016/j.celrep.2021.109130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/09/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022] Open
Abstract
Dendritic spines constitute the major compartments of excitatory post-synapses. They undergo activity-dependent enlargement, which is thought to increase the synaptic efficacy underlying learning and memory. The activity-dependent spine enlargement requires activation of signaling pathways leading to promotion of actin polymerization within the spines. However, the molecular machinery that suffices for that structural plasticity remains unclear. Here, we demonstrate that shootin1a links polymerizing actin filaments in spines with the cell-adhesion molecules N-cadherin and L1-CAM, thereby mechanically coupling the filaments to the extracellular environment. Synaptic activation enhances shootin1a-mediated actin-adhesion coupling in spines. Promotion of actin polymerization is insufficient for the plasticity; the enhanced actin-adhesion coupling is required for polymerizing actin filaments to push against the membrane for spine enlargement. By integrating cell signaling, cell adhesion, and force generation into the current model of actin-based machinery, we propose molecular machinery that is sufficient to trigger the activity-dependent spine structural plasticity.
Collapse
Affiliation(s)
- Ria Fajarwati Kastian
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takunori Minegishi
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takeo Saneyoshi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Hiroko Katsuno-Kambe
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Singh Saranpal
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
11
|
Rubfiaro AS, Tsegay PS, Lai Y, Cabello E, Shaver M, Hutcheson J, Liu Y, He J. Scanning Ion Conductance Microscopy Study Reveals the Disruption of the Integrity of the Human Cell Membrane Structure by Oxidative DNA Damage. ACS APPLIED BIO MATERIALS 2021; 4:1632-1639. [PMID: 34430802 DOI: 10.1021/acsabm.0c01461] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oxidative stress can damage organs, tissues, and cells through reactive oxygen species (ROS) by oxidizing DNA, proteins, and lipids, thereby resulting in diseases. However, the underlying molecular mechanisms remain to be elucidated. In this study, employing scanning ion conductance microscopy (SICM), we explored the early responses of human embryonic kidney (HEK293H) cells to oxidative DNA damage induced by potassium chromate (K2CrO4). We found that the short term (1-2 h) exposure to a low concentration (10 μM) of K2CrO4 damaged the lipid membrane of HEK293H cells, resulting in structural defects and depolarization of the cell membrane and reducing cellular secretion activity shortly after the treatment. We further demonstrated that the K2CrO4 treatment decreased the expression of the cytoskeleton protein, β-actin, by inducing oxidative DNA damage in the exon 4 of the β-actin gene. These results suggest that K2CrO4 caused oxidative DNA damage in cytoskeleton genes such as β-actin and reduced their expression, thereby disrupting the organization of the cytoskeleton beneath the cell membrane and inducing cell membrane damages. Our study provides direct evidence that oxidative DNA damage disrupted human cell membrane integrity by deregulating cytoskeleton gene expression.
Collapse
Affiliation(s)
- Alberto S Rubfiaro
- Physics Department, Florida International University, Miami, Florida 33199, United States
| | - Pawlos S Tsegay
- Biochemistry Ph.D. Program, Florida International University, Miami, Florida 33199, United States
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Emmanuel Cabello
- Physics Department, Florida International University, Miami, Florida 33199, United States
| | - Mohammad Shaver
- Department of Biomedical Engineering, Florida International University, Miami, Florida 33199, United States
| | - Joshua Hutcheson
- Department of Biomedical Engineering and Biomolecular Science Institute, Florida International University, Miami, Florida 33199, United States
| | - Yuan Liu
- Biochemistry Ph.D. Program, Department of Chemistry and Biochemistry, and Biomolecular Science Institute, Florida International University, Miami, Florida 33199, United States
| | - Jin He
- Physics Department and Biomolecular Science Institute, Florida International University, Miami, Florida 33199, United States
| |
Collapse
|
12
|
Braïni C, Bugnicourt G, Villard C. Neuronal growth from a volume perspective. Phys Biol 2021; 18:016007. [PMID: 33147573 DOI: 10.1088/1478-3975/abc79c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Microfluidic-based fluorescent exclusion method allows to tackle the issue of neuronal growth from a volume perspective. Based on this technology, we studied the two main actin-rich structures accompanying the early stages of neuron development, i.e. growth cones, located at the tip of growing neuronal processes, and propagative actin waves. Our work reveals that growth cones tend to loose volume during their forward motion, as do actin waves during their journey from the cell body to the tip of neuronal processes, before the total transfer of their remaining volume to the growth cone. Actin waves seem thus to supply material to increasingly distant growth cones as neurons develop. In addition, our work may suggest the existence of a membrane recycling phenomena associated to actin waves as a pulsatile anterograde source of material and by a continuous retrograde transport.
Collapse
Affiliation(s)
- Céline Braïni
- Physico-Chimie Curie, CNRS UMR 168, Université PSL, Sorbonne Université, Paris, France
| | | | | |
Collapse
|
13
|
van Deventer S, Arp AB, van Spriel AB. Dynamic Plasma Membrane Organization: A Complex Symphony. Trends Cell Biol 2020; 31:119-129. [PMID: 33248874 DOI: 10.1016/j.tcb.2020.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 01/20/2023]
Abstract
Membrane protein organization is essential for proper cellular functioning and the result of a dynamic exchange between protein monomers, nanoscale protein clusters, and microscale higher-order structures. This exchange is affected by both lipid bilayer intrinsic factors, such as lipid rafts and tetraspanins, and extrinsic factors, such as cortical actin and galectins. Because membrane organizers act jointly like instruments in a symphony, it is challenging to define the 'key' organizers. Here, we posit, for the first time, definitions of key intrinsic and extrinsic membrane organizers. Tetraspanin nanodomains are key organizers that are often overlooked. We discuss how different key organizers can collaborate, which is important to get a full grasp of plasma membrane biology.
Collapse
Affiliation(s)
- Sjoerd van Deventer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Abbey B Arp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Su M, Zhuang Y, Miao X, Zeng Y, Gao W, Zhao W, Wu M. Comparative Study of Curvature Sensing Mediated by F-BAR and an Intrinsically Disordered Region of FBP17. iScience 2020; 23:101712. [PMID: 33205024 PMCID: PMC7649350 DOI: 10.1016/j.isci.2020.101712] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/11/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Membrane curvature has emerged as an intriguing physical principle underlying biological signaling and membrane trafficking. The CIP4/FBP17/Toca-1 F-BAR subfamily is unique in the BAR family because its structurally folded F-BAR domain does not contain any hydrophobic motifs that insert into membrane. Although widely assumed so, whether the banana-shaped F-BAR domain alone can sense curvature has never been experimentally demonstrated. Using a nanobar-supported lipid bilayer system, we found that the F-BAR domain of FBP17 displayed minimal curvature sensing in vitro. In comparison, an alternatively spliced intrinsically disordered region (IDR) adjacent to the F-BAR domain has the membrane curvature-sensing ability greatly exceeding that of F-BAR domain alone. In living cells, the presence of the IDR delayed the recruitment of FBP17 in curvature-coupled cortical waves. Collectively, we propose that contrary to the common belief, FBP17's curvature-sensing capability largely originates from IDR, and not the F-BAR domain alone.
Collapse
Affiliation(s)
- Maohan Su
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8002, USA.,Centre for BioImaging Sciences, Mechanobiology Institute, Department of Biological Sciences, National University of Singapore, Singapore, 117411
| | - Yinyin Zhuang
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8002, USA.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457
| | - Xinwen Miao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457
| | - Yongpeng Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457
| | - Weibo Gao
- School of Physics and Mathematical Science, Nanyang Technological University, Singapore, 637371
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457
| | - Min Wu
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8002, USA.,Centre for BioImaging Sciences, Mechanobiology Institute, Department of Biological Sciences, National University of Singapore, Singapore, 117411
| |
Collapse
|
15
|
Beta C, Gov NS, Yochelis A. Why a Large-Scale Mode Can Be Essential for Understanding Intracellular Actin Waves. Cells 2020; 9:cells9061533. [PMID: 32585983 PMCID: PMC7349605 DOI: 10.3390/cells9061533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 01/18/2023] Open
Abstract
During the last decade, intracellular actin waves have attracted much attention due to their essential role in various cellular functions, ranging from motility to cytokinesis. Experimental methods have advanced significantly and can capture the dynamics of actin waves over a large range of spatio-temporal scales. However, the corresponding coarse-grained theory mostly avoids the full complexity of this multi-scale phenomenon. In this perspective, we focus on a minimal continuum model of activator-inhibitor type and highlight the qualitative role of mass conservation, which is typically overlooked. Specifically, our interest is to connect between the mathematical mechanisms of pattern formation in the presence of a large-scale mode, due to mass conservation, and distinct behaviors of actin waves.
Collapse
Affiliation(s)
- Carsten Beta
- Institute of Physics and Astronomy, University of Potsdam, 14476 Potsdam, Germany;
| | - Nir S. Gov
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Arik Yochelis
- Department of Solar Energy and Environmental Physics, Blaustein Institutes for Desert Research (BIDR), Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion 8499000, Israel
- Department of Physics, Ben-Gurion University of the Negev, Be’er Sheva 8410501, Israel
- Correspondence:
| |
Collapse
|
16
|
Actin Alpha 2 (ACTA2) Downregulation Inhibits Neural Stem Cell Migration through Rho GTPase Activation. Stem Cells Int 2020; 2020:4764012. [PMID: 32508931 PMCID: PMC7246394 DOI: 10.1155/2020/4764012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023] Open
Abstract
Although neural stem cells (NSCs) could migrate towards lesions after central nervous system (CNS) injury, the migration ability always is restricted due to the disturbed composition and density of the adhesion ligands and extracellular matrix (ECM) gradient after CNS injury. To date, various methods have been developed to enhance NSC migration and a number of factors, which are affecting NSC migration potential, have been identified. Here, primary NSCs were cultured and the expression of actin alpha 2 (ACTA2) in NSCs was determined using reverse transcription polymerase chain reaction (RT-PCR) and immunostaining. Next, the role of ACTA2 in regulating NSC migration and the potential mechanism was explored. Our results demonstrated that ACTA2 expressed in NSCs. Meanwhile, downregulated ACTA2 using siRNA inhibited NSC migration through hindering actin filament polymerization via increasing RhoA expression and decreasing Rac1 expression. The present study might enrich the basic knowledge of ACTA2 in NSC migration and open an avenue for enhancing NSC migration potential, subsequently providing an intervention target for functional recovery after CNS injury.
Collapse
|
17
|
Huang YA, Hsu CH, Chiu HC, Hsi PY, Ho CT, Lo WL, Hwang E. Actin waves transport RanGTP to the neurite tip to regulate non-centrosomal microtubules in neurons. J Cell Sci 2020; 133:jcs241992. [PMID: 32253322 DOI: 10.1242/jcs.241992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/17/2020] [Indexed: 12/18/2022] Open
Abstract
Microtubules (MTs) are the most abundant cytoskeleton in neurons, and control multiple facets of their development. While the MT-organizing center (MTOC) in mitotic cells is typically located at the centrosome, the MTOC in neurons switches to non-centrosomal sites. A handful of cellular components have been shown to promote non-centrosomal MT (ncMT) formation in neurons, yet the regulation mechanism remains unknown. Here, we demonstrate that the small GTPase Ran is a key regulator of ncMTs in neurons. Using an optogenetic tool that enables light-induced local production of RanGTP, we demonstrate that RanGTP promotes ncMT plus-end growth along the neurite. Additionally, we discovered that actin waves drive the anterograde transport of RanGTP. Pharmacological disruption of actin waves abolishes the enrichment of RanGTP and reduces growing ncMT plus-ends at the neurite tip. These observations identify a novel regulation mechanism for ncMTs and pinpoint an indirect connection between the actin and MT cytoskeletons in neurons.
Collapse
Affiliation(s)
- Yung-An Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Chih-Hsuan Hsu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Ho-Chieh Chiu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Pei-Yu Hsi
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Chris T Ho
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Wei-Lun Lo
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan 30068
| | - Eric Hwang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan 30068
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan 30068
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan 30068
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan 30068
| |
Collapse
|
18
|
|
19
|
Muñoz-Lasso DC, Mollá B, Calap-Quintana P, García-Giménez JL, Pallardo FV, Palau F, Gonzalez-Cabo P. Cofilin dysregulation alters actin turnover in frataxin-deficient neurons. Sci Rep 2020; 10:5207. [PMID: 32251310 PMCID: PMC7090085 DOI: 10.1038/s41598-020-62050-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 03/04/2020] [Indexed: 01/04/2023] Open
Abstract
Abnormalities in actin cytoskeleton have been linked to Friedreich's ataxia (FRDA), an inherited peripheral neuropathy characterised by an early loss of neurons in dorsal root ganglia (DRG) among other clinical symptoms. Despite all efforts to date, we still do not fully understand the molecular events that contribute to the lack of sensory neurons in FRDA. We studied the adult neuronal growth cone (GC) at the cellular and molecular level to decipher the connection between frataxin and actin cytoskeleton in DRG neurons of the well-characterised YG8R Friedreich's ataxia mouse model. Immunofluorescence studies in primary cultures of DRG from YG8R mice showed neurons with fewer and smaller GCs than controls, associated with an inhibition of neurite growth. In frataxin-deficient neurons, we also observed an increase in the filamentous (F)-actin/monomeric (G)-actin ratio (F/G-actin ratio) in axons and GCs linked to dysregulation of two crucial modulators of filamentous actin turnover, cofilin-1 and the actin-related protein (ARP) 2/3 complex. We show how the activation of cofilin is due to the increase in chronophin (CIN), a cofilin-activating phosphatase. Thus cofilin emerges, for the first time, as a link between frataxin deficiency and actin cytoskeleton alterations.
Collapse
Affiliation(s)
- Diana C Muñoz-Lasso
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - Belén Mollá
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Instituto de Biomedicina de Valencia (IBV), CSIC, Valencia, 46010, Spain
| | - Pablo Calap-Quintana
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - José Luis García-Giménez
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - Federico V Pallardo
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - Francesc Palau
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Institut de Recerca Sant Joan de Déu and Department of Genetic & Molecular Medicine and IPER, Hospital Sant Joan de Déu, Barcelona, 08950, Spain
- Hospital Clínic and Division of Pediatrics, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain
| | - Pilar Gonzalez-Cabo
- CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain.
- Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain.
| |
Collapse
|
20
|
Dai J, Wang Y, Gong J, Yao Y. Biointerface anisotropy modulates migration of breast cancer cell. Colloids Surf B Biointerfaces 2020; 190:110973. [PMID: 32199258 DOI: 10.1016/j.colsurfb.2020.110973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/24/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
Migration of cancer cell is a cyclic process, which involves dynamic interaction between extracellular biointerface and cellular responds. In tumors, collagen as extracellular matrix reorganizes biointerface from curl and isotropic fibers to straightened and anisotropic fibers during tumorigenesis, yet how cell migration respond to topography of biointerface is unknown. In this research, we introduced a facile fabrication method on nanofibers of varying topography, which was mimicking the alignment of extracellular nanofibers, to examine the change of cytoskeleton during cell migration. We took advantage of breast carcinoma cell line (MDA-MB-231) for time-lapse imaging analysis. We found that biointerface anisotropy modulated morphology of cell and mediated the pattern of migration. Morphologically, cells on anisotropic nanofiber showed extending spindle shape. The trajectories of migration templated the topographic pattern on biointerface. Besides, aligned nanofiber induced caterpillar-like model of migration through protrusion - retraction cycle, which was indicated by periodical variation of aspect ratio and velocity of cells. The biointerface anisotropy triggered vimentin filaments and microtubule networks preferentially oriented along the alignment of nanofibers. And the velocity of cell mobility by vimentin, β-catenin or CDC42 knockdown was significantly enhanced on aligned nanofibers. Thus, we implied that biointerface anisotropy modulated migration of breast cancer cell and it associated with reorganization of cytoskeleton filaments.
Collapse
Affiliation(s)
- Jing Dai
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Changning, Shanghai, 200050, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan, Beijing, 100049, China
| | - Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Jinkang Gong
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China.
| |
Collapse
|
21
|
Wang Y, Yao Y. Nanofiber Alignment Mediates the Pattern of Single Cell Migration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2129-2135. [PMID: 32040329 DOI: 10.1021/acs.langmuir.9b03314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In natural tissue, nanofibrils in extracellular matrix (ECM), such as collagen fibrils, direct cell migration through contacting guidance. The contacting nanofibers on cell-ECM interface are reorganized from curl fibers to straightened fibers. However, how these nanofibers regulate single cell migration remains obscure. To investigate this issue, we fabricated collagen/polymer based biomimetic nanofiber sheets of varying topography. And we selected tumorigenic cell KGN and nontumorigenic cell 293T for comparison. We found KGN showed higher sensitivity to the nanofiber alignment rather than the nontumorigenic cell 293T, in morphological change, trajectory adaptation, and velocity variation. We also found aligned nanofibers shaped both KGN and 293T into elongated spindle morphology. Comparatively, KGN had greater perimeter and lower roundness than 293T. To study the dynamics of single cell migration of KGN and 293T, we conducted trajectory tracking and siRNA validation on regulatory proteins. We found nanofibers of varying topography regulated the patterns of single cell migration differently. For KGN cell, β-catenin, Rac1, and Cdc42 participated in its directional migration, but it was impervious to vimentin. Comparatively, epithelial cell 293T involved vimentin in its directional migration.
Collapse
Affiliation(s)
- Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| |
Collapse
|
22
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
23
|
Takahashi Y, Zhou Y, Miyamoto T, Higashi H, Nakamichi N, Takeda Y, Kato Y, Korchev Y, Fukuma T. High-Speed SICM for the Visualization of Nanoscale Dynamic Structural Changes in Hippocampal Neurons. Anal Chem 2019; 92:2159-2167. [PMID: 31840491 DOI: 10.1021/acs.analchem.9b04775] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Dynamic reassembly of the cytoskeleton and structural changes represented by dendritic spines, cargo transport, and synapse formation are closely related to memory. However, the visualization of the nanoscale topography is challenging because of the diffraction limit of optical microscopy. Scanning ion conductance microscopy (SICM) is an effective tool for visualizing the nanoscale topography changes of the cell surface without labeling. The temporal resolution of SICM is a critical issue of live-cell time-lapse imaging. Here, we developed a new scanning method, automation region of interest (AR)-mode SICM, to select the next imaging region by predicting the location of a cell, thus improving the scanning speed of time-lapse imaging. The newly developed algorithm reduced the scanning time by half. The time-lapse images provided not only novel information about nanoscale structural changes but also quantitative information on the dendritic spine and synaptic bouton volume changes and formation process of the neural network that are closely related to memory. Furthermore, translocation of plasmalemmal precursor vesicles (ppvs), for which fluorescent labeling has not been established, were also visualized along with the rearrangement of the cytoskeleton at the growth cone.
Collapse
Affiliation(s)
- Yasufumi Takahashi
- WPI Nano Life Science Institute (WPI-NanoLSI) , Kanazawa University , Kanazawa 920-1192 , Japan.,Precursory Research for Embryonic Science and Technology (PRESTO) , Japan Science and Technology Agency (JST) , Saitama 332-0012 , Japan
| | - Yuanshu Zhou
- WPI Nano Life Science Institute (WPI-NanoLSI) , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Takafumi Miyamoto
- Department Division of Electrical Engineering and Computer Science , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Hiroki Higashi
- Department Division of Electrical Engineering and Computer Science , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Yuka Takeda
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences , Kanazawa University , Kanazawa 920-1192 , Japan
| | - Yuri Korchev
- WPI Nano Life Science Institute (WPI-NanoLSI) , Kanazawa University , Kanazawa 920-1192 , Japan.,Department of Medicine , Imperial College London , London W12 0NN , United Kingdom.,National University of Science and Technology (MISiS) , Leninskiy prospect 4 , Moscow 119049 , Russia
| | - Takeshi Fukuma
- WPI Nano Life Science Institute (WPI-NanoLSI) , Kanazawa University , Kanazawa 920-1192 , Japan
| |
Collapse
|
24
|
Vohnoutka RB, Gulvady AC, Goreczny G, Alpha K, Handelman SK, Sexton JZ, Turner CE. The focal adhesion scaffold protein Hic-5 regulates vimentin organization in fibroblasts. Mol Biol Cell 2019; 30:3037-3056. [PMID: 31644368 PMCID: PMC6880880 DOI: 10.1091/mbc.e19-08-0442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion (FA)-stimulated reorganization of the F-actin cytoskeleton regulates cellular size, shape, and mechanical properties. However, FA cross-talk with the intermediate filament cytoskeleton is poorly understood. Genetic ablation of the FA-associated scaffold protein Hic-5 in mouse cancer-associated fibroblasts (CAFs) promoted a dramatic collapse of the vimentin network, which was rescued following EGFP-Hic-5 expression. Vimentin collapse correlated with a loss of detergent-soluble vimentin filament precursors and decreased vimentin S72/S82 phosphorylation. Additionally, fluorescence recovery after photobleaching analysis indicated impaired vimentin dynamics. Microtubule (MT)-associated EB1 tracking and Western blotting of MT posttranslational modifications indicated no change in MT dynamics that could explain the vimentin collapse. However, pharmacological inhibition of the RhoGTPase Cdc42 in Hic-5 knockout CAFs rescued the vimentin collapse, while pan-formin inhibition with SMIFH2 promoted vimentin collapse in Hic-5 heterozygous CAFs. Our results reveal novel regulation of vimentin organization/dynamics by the FA scaffold protein Hic-5 via modulation of RhoGTPases and downstream formin activity.
Collapse
Affiliation(s)
- Rishel B Vohnoutka
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Anushree C Gulvady
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Gregory Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kyle Alpha
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Samuel K Handelman
- Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109
| | - Jonathan Z Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
25
|
Urasaki A, Morishita S, Naka K, Uozumi M, Abe K, Huang L, Watase E, Nakagawa O, Kawakami K, Matsui T, Bessho Y, Inagaki N. Shootins mediate collective cell migration and organogenesis of the zebrafish posterior lateral line system. Sci Rep 2019; 9:12156. [PMID: 31434971 PMCID: PMC6704158 DOI: 10.1038/s41598-019-48585-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2019] [Indexed: 01/23/2023] Open
Abstract
The zebrafish sensory posterior lateral line is an excellent model system to study collective cell migration and organogenesis. Shootin1 is a cytoplasmic protein involved in neuronal polarization and axon guidance. Previous studies have shown that shootin1 couples actin filament retrograde flow with extracellular adhesive substrates at the leading edge of axonal growth cones, thereby producing mechanical force for the migration and guidance of axonal growth cones. However, the functions of shootin in peripheral cells remain unknown. Here we identified two novel shootin family members, shootin2 and shootin3. In zebrafish, shootin1 and shootin3 are expressed in the posterior lateral line primordium (PLLP) and neuromasts during embryonic development. A shootin1 mutant displayed a reduced speed of PLLP migration, while shootin1;shootin3 double mutation inhibited cell proliferation in the PLLP. Furthermore, our results suggest that shootin1 and shootin3 positively regulate the number of neuromasts and the number of cells in deposited neuromasts. Our study demonstrates that shootins mediate collective cell migration of the posterior lateral line primordium and formation of neuromasts in zebrafish.
Collapse
Affiliation(s)
- Akihiro Urasaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.,Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Seiya Morishita
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kosuke Naka
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Minato Uozumi
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kouki Abe
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Liguo Huang
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Emiko Watase
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, 411-8540, Japan
| | - Takaaki Matsui
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yasumasa Bessho
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
26
|
Avota E, de Lira MN, Schneider-Schaulies S. Sphingomyelin Breakdown in T Cells: Role of Membrane Compartmentalization in T Cell Signaling and Interference by a Pathogen. Front Cell Dev Biol 2019; 7:152. [PMID: 31457008 PMCID: PMC6700246 DOI: 10.3389/fcell.2019.00152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Sphingolipids are major components of cellular membranes, and at steady-state level, their metabolic fluxes are tightly controlled. On challenge by external signals, they undergo rapid turnover, which substantially affects the biophysical properties of membrane lipid and protein compartments and, consequently, signaling and morphodynamics. In T cells, external cues translate into formation of membrane microdomains where proximal signaling platforms essential for metabolic reprograming and cytoskeletal reorganization are organized. This review will focus on sphingomyelinases, which mediate sphingomyelin breakdown and ensuing ceramide release that have been implicated in T-cell viability and function. Acting at the sphingomyelin pool at the extrafacial or cytosolic leaflet of cellular membranes, acid and neutral sphingomyelinases organize ceramide-enriched membrane microdomains that regulate T-cell homeostatic activity and, upon stimulation, compartmentalize receptors, membrane proximal signaling complexes, and cytoskeletal dynamics as essential for initiating T-cell motility and interaction with endothelia and antigen-presenting cells. Prominent examples to be discussed in this review include death receptor family members, integrins, CD3, and CD28 and their associated signalosomes. Progress made with regard to experimental tools has greatly aided our understanding of the role of bioactive sphingolipids in T-cell biology at a molecular level and of targets explored by a model pathogen (measles virus) to specifically interfere with their physiological activity.
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Maria Nathalia de Lira
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
27
|
Chakrabarty N, Jung P. Stochastic models of polymerization-based axonal actin transport. Phys Biol 2019; 16:056001. [PMID: 31195374 DOI: 10.1088/1478-3975/ab29cd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Recent advances in live cell imaging of F-actin structures, combined with pulse-chase imaging and computational modeling have suggested that actin is transported along the axon via biased polymerization of metastable actin fibers (actin trails). This mechanism is distinct from motor driven polymer transport, such as for neurofilaments and can be best described as molecular hitchhiking, where G-actin molecules are intermittently incorporated into actin fibers which grow preferentially in the anterograde direction. In this paper, we discuss how various axonal and actin trail parameters like axon diameter, trail nucleation rates, basal G-actin concentration, and trail length influence the transport rate. These predictions can help guide future experiments to verify this novel protein transport mechanism. We introduce a simplified, analytically solvable model of actin transport which relates these parameters to experimentally measurable quantities. We also discuss why a simple diffusion-based transport mechanism cannot explain bulk actin transport in the axon.
Collapse
Affiliation(s)
- Nilaj Chakrabarty
- Department of Physics and Astronomy, Neuroscience Program and Quantitative Biology Institute, Athens, OH 45701, United States of America
| | | |
Collapse
|
28
|
Long J, Hu Z, Xue H, Wang Y, Chen J, Tang F, Zhou J, Liu L, Qiu W, Zhang S, Ouyang Y, Ye Y, Xu G, Li L, Zeng Z. Vascular endothelial growth factor (VEGF) impairs the motility and immune function of human mature dendritic cells through the VEGF receptor 2-RhoA-cofilin1 pathway. Cancer Sci 2019; 110:2357-2367. [PMID: 31169331 PMCID: PMC6676124 DOI: 10.1111/cas.14091] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Dendritic cells (DCs) are potent and specialized antigen presenting cells, which play a crucial role in initiating and amplifying both the innate and adaptive immune responses against cancer. Tumor cells can escape from immune attack by secreting suppressive cytokines that solely or cooperatively impair the immune function of DCs. However, the underlying mechanisms are not fully defined. Vascular endothelial growth factor (VEGF) has been identified as a major cytokine in the tumor microenvironment. To elucidate the effects of VEGF on the motility and immune function of mature DCs (mDCs), the cells were treated with 50 ng/mL VEGF and investigated by proteomics and molecular biological technologies. The results showed that VEGF can impair the migration capacity and immune function of mDCs through the RhoA-cofilin1 pathway mediated by the VEGF receptor 2, suggesting impaired motility of mDCs by VEGF is one of the aspects of immune escape mechanisms of tumors. It is clinically important to understand the biological behavior of DCs and the immune escape mechanisms of tumor as well as how to improve the efficiency of antitumor therapy based on DCs.
Collapse
Affiliation(s)
- Jinhua Long
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Department of Head and Neck, Affiliated Tumor Hospital, Guizhou Medical University, Guiyang, China
| | - Zuquan Hu
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Hui Xue
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yun Wang
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Jin Chen
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Fuzhou Tang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Jing Zhou
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Lina Liu
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Wei Qiu
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Shichao Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yan Ouyang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yuannong Ye
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Guoqiang Xu
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Long Li
- Department of Nephrology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, China
| | - Zhu Zeng
- Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
29
|
Masuda T, Baba K, Nomura T, Tsujita K, Murayama T, Itoh T, Takatani-Nakase T, Sokabe M, Inagaki N, Futaki S. An influenza-derived membrane tension-modulating peptide regulates cell movement and morphology via actin remodeling. Commun Biol 2019; 2:243. [PMID: 31263787 PMCID: PMC6594980 DOI: 10.1038/s42003-019-0486-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/23/2019] [Indexed: 01/01/2023] Open
Abstract
Tension in cell membranes is closely related to various cellular events, including cell movement and morphogenesis. Therefore, modulation of membrane tension can be a new approach for manipulating cellular events. Here, we show that an amphipathic peptide derived from the influenza M2 protein (M2[45-62]) yields lamellipodia at multiple sites in the cell. Effect of M2[45-62] on cell membrane tension was evaluated by optical tweezer. The membrane tension sensor protein FBP17 was involved in M2[45-62]-driven lamellipodium formation. Lysine-to-arginine substitution in M2[45-62] further enhanced its activity of lamellipodium formation. M2[45-62] had an ability to reduce cell motility, evaluated by scratch wound migration and transwell migration assays. An increase in neurite outgrowth was also observed after treatment with M2[45-62]. The above results suggest the potential of M2[45-62] to modulate cell movement and morphology by modulating cell membrane tension.
Collapse
Affiliation(s)
- Toshihiro Masuda
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011 Japan
| | - Kentarou Baba
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192 Japan
| | - Takeshi Nomura
- Department of Agro-environmental Sciences, Kyushu University, Fukuoka, 819-0395 Japan
| | - Kazuya Tsujita
- Division of Membrane Biology, Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501 Japan
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Tomo Murayama
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011 Japan
| | - Toshiki Itoh
- Division of Membrane Biology, Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501 Japan
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Tomoka Takatani-Nakase
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, 65 Tsurumai, Nagoya, 466-8550 Japan
| | - Masahiro Sokabe
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya, Hyogo 663-8179 Japan
| | - Naoyuki Inagaki
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192 Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011 Japan
| |
Collapse
|
30
|
The Architecture of Traveling Actin Waves Revealed by Cryo-Electron Tomography. Structure 2019; 27:1211-1223.e5. [PMID: 31230946 DOI: 10.1016/j.str.2019.05.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Actin waves are dynamic supramolecular structures involved in cell migration, cytokinesis, adhesion, and neurogenesis. Although wave-like propagation of actin networks is a widespread phenomenon, the actin architecture underlying wave propagation remained unknown. In situ cryo-electron tomography of Dictyostelium cells unveils the wave architecture and provides evidence for wave progression by de novo actin nucleation. Subtomogram averaging reveals the structure of Arp2/3 complex-mediated branch junctions in their native state, and enables quantitative analysis of the 3D organization of branching within the waves. We find an excess of branches directed toward the substrate-attached membrane, and tent-like structures at sites of branch clustering. Fluorescence imaging shows that Arp2/3 clusters follow accumulation of the elongation factor VASP. We propose that filament growth toward the membrane lifts up the actin network as the wave propagates, until depolymerization of oblique filaments at the back causes the collapse of horizontal filaments into a compact layer.
Collapse
|
31
|
Collenburg L, Schneider-Schaulies S, Avota E. The neutral sphingomyelinase 2 in T cell receptor signaling and polarity. Biol Chem 2019; 399:1147-1155. [PMID: 29337691 DOI: 10.1515/hsz-2017-0280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/31/2017] [Indexed: 01/13/2023]
Abstract
By hydrolyzing its substrate sphingomyelin at the cytosolic leaflet of cellular membranes, the neutral sphingomyelinase 2 (NSM2) generates microdomains which serve as docking sites for signaling proteins and thereby, functions to regulate signal relay. This has been particularly studied in cellular stress responses while the regulatory role of this enzyme in the immune cell compartment has only recently emerged. In T cells, phenotypic polarization by co-ordinated cytoskeletal remodeling is central to motility and interaction with endothelial or antigen-presenting cells during tissue recruitment or immune synapse formation, respectively. This review highlights studies adressing the role of NSM2 in T cell polarity in which the enzyme plays a major role in regulating cytoskeletal dynamics.
Collapse
Affiliation(s)
- Lena Collenburg
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | - Sibylle Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| |
Collapse
|
32
|
Cruz-Ortega JS, Boucard AA. Actin cytoskeleton remodeling defines a distinct cellular function for adhesion G protein-coupled receptors ADGRL/latrophilins 1, 2 and 3. Biol Open 2019; 8:bio.039826. [PMID: 30926595 PMCID: PMC6503996 DOI: 10.1242/bio.039826] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Latrophilins represent a subgroup of the adhesion G protein-coupled receptor family, which bind to actin-associated scaffolding proteins. They are expressed in various tissues, suggesting that they might participate in biological processes that are ubiquitous. Here we focus on actin cytoskeleton dynamics to explore the role of latrophilins in mammalian cells. Individual overexpression of each latrophilin isoform comparably increased cell volume while modifying the net profile of F-actin-dependent cell extensions, as evaluated by confocal microscopy analysis. Latrophilin deletion mutants evidenced that direct coupling to the intracellular machinery was a requirement for modulating cell extensions. The association between latrophilins and the actin cytoskeleton was detected by co-immunoprecipitation assays and corroborated with immunocytochemistry analysis. Consistent with the destabilization of F-actin structures, latrophilin isoforms constitutively induced a prominent increase in the activity of actin-depolymerizing factor, cofilin. Intercellular adhesion events stabilized by heterophilic Teneurin-4 trans-interactions disrupted latrophilin colocalization with F-actin and led to an isoform-specific rescue of cell extensions. Thus, we find that the actin cytoskeleton machinery constitutes an important component of constitutive as well as ligand-induced signaling for latrophilins. This article has an associated First Person interview with the first author of the paper. Summary: Synapses involve the adhesion function of latrophilins within existing neuronal extensions. We show that latrophilins engage the actin cytoskeleton, both constitutively and upon ligand stimulation, to dictate cell extension patterns.
Collapse
Affiliation(s)
- Judith S Cruz-Ortega
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City 07360, México
| | - Antony A Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City 07360, México
| |
Collapse
|
33
|
Saha S, Nagy TL, Weiner OD. Joining forces: crosstalk between biochemical signalling and physical forces orchestrates cellular polarity and dynamics. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0145. [PMID: 29632270 DOI: 10.1098/rstb.2017.0145] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Dynamic processes like cell migration and morphogenesis emerge from the self-organized interaction between signalling and cytoskeletal rearrangements. How are these molecular to sub-cellular scale processes integrated to enable cell-wide responses? A growing body of recent studies suggest that forces generated by cytoskeletal dynamics and motor activity at the cellular or tissue scale can organize processes ranging from cell movement, polarity and division to the coordination of responses across fields of cells. To do so, forces not only act mechanically but also engage with biochemical signalling. Here, we review recent advances in our understanding of this dynamic crosstalk between biochemical signalling, self-organized cortical actomyosin dynamics and physical forces with a special focus on the role of membrane tension in integrating cellular motility.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Suvrajit Saha
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Tamas L Nagy
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA.,Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Orion D Weiner
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA .,Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
34
|
Yang Y, Wu M. Rhythmicity and waves in the cortex of single cells. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0116. [PMID: 29632268 PMCID: PMC5904302 DOI: 10.1098/rstb.2017.0116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/15/2022] Open
Abstract
Emergence of dynamic patterns in the form of oscillations and waves on the cortex of single cells is a fascinating and enigmatic phenomenon. Here we outline various theoretical frameworks used to model pattern formation with the goal of reducing complex, heterogeneous patterns into key parameters that are biologically tractable. We also review progress made in recent years on the quantitative and molecular definitions of these terms, which we believe have begun to transform single-cell dynamic patterns from a purely observational and descriptive subject to more mechanistic studies. Specifically, we focus on the nature of local excitable and oscillation events, their spatial couplings leading to propagating waves and the role of active membrane. Instead of arguing for their functional importance, we prefer to consider such patterns as basic properties of dynamic systems. We discuss how knowledge of these patterns could be used to dissect the structure of cellular organization and how the network-centric view could help define cellular functions as transitions between different dynamical states. Last, we speculate on how these patterns could encode temporal and spatial information. This article is part of the theme issue ‘Self-organization in cell biology’.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biological Sciences, Centre for Bioimaging Sciences, Mechanobiology Institute, National University of Singapore, Singapore
| | - Min Wu
- Department of Biological Sciences, Centre for Bioimaging Sciences, Mechanobiology Institute, National University of Singapore, Singapore
| |
Collapse
|
35
|
Zhang B, Xie F, Aziz AUR, Shao S, Li W, Deng S, Liao X, Liu B. Heat Shock Protein 27 Phosphorylation Regulates Tumor Cell Migration under Shear Stress. Biomolecules 2019; 9:biom9020050. [PMID: 30704117 PMCID: PMC6406706 DOI: 10.3390/biom9020050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 01/02/2023] Open
Abstract
Heat shock protein 27 (HSP27) is a multifunctional protein that undergoes significant changes in its expression and phosphorylation in response to shear stress stimuli, suggesting that it may be involved in mechanotransduction. However, the mechanism of HSP27 affecting tumor cell migration under shear stress is still not clear. In this study, HSP27-enhanced cyan fluorescent protein (ECFP) and HSP27-Ypet plasmids are constructed to visualize the self-polymerization of HSP27 in living cells based on fluorescence resonance energy transfer technology. The results show that shear stress induces polar distribution of HSP27 to regulate the dynamic structure at the cell leading edge. Shear stress also promotes HSP27 depolymerization to small molecules and then regulates polar actin accumulation and focal adhesion kinase (FAK) polar activation, which further promotes tumor cell migration. This study suggests that HSP27 plays an important role in the regulation of shear stress-induced HeLa cell migration, and it also provides a theoretical basis for HSP27 as a potential drug target for metastasis.
Collapse
Affiliation(s)
- Baohong Zhang
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Fei Xie
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Shuai Shao
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Wang Li
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Sha Deng
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Xiaoling Liao
- Institute of Biomedical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Bo Liu
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| |
Collapse
|
36
|
Chen CH, Hsu HW, Chang YH, Pan CL. Adhesive L1CAM-Robo Signaling Aligns Growth Cone F-Actin Dynamics to Promote Axon-Dendrite Fasciculation in C. elegans. Dev Cell 2019; 48:215-228.e5. [PMID: 30555000 DOI: 10.1016/j.devcel.2018.10.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/18/2018] [Accepted: 10/26/2018] [Indexed: 11/28/2022]
Abstract
Neurite fasciculation through contact-dependent signaling is important for the wiring and function of the neuronal circuits. Here, we describe a type of axon-dendrite fasciculation in C. elegans, where proximal dendrites of the nociceptor PVD adhere to the axon of the ALA interneuron. This axon-dendrite fasciculation is mediated by a previously uncharacterized adhesive signaling by the ALA membrane signal SAX-7/L1CAM and the PVD receptor SAX-3/Robo but independent of Slit. L1CAM physically interacts with Robo and instructs dendrite adhesion in a Robo-dependent manner. Fasciculation mediated by L1CAM-Robo signaling aligns F-actin dynamics in the dendrite growth cone and facilitates dynamic growth cone behaviors for efficient dendrite guidance. Disruption of PVD dendrite fasciculation impairs nociceptive mechanosensation and rhythmicity in body curvature, suggesting that dendrite fasciculation governs the functions of mechanosensory circuits. Our work elucidates the molecular mechanisms by which adhesive axon-dendrite signaling shapes the construction and function of sensory neuronal circuits.
Collapse
Affiliation(s)
- Chun-Hao Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No.7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Hao-Wei Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No.7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Yun-Hsuan Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No.7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chun-Liang Pan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No.7 Chung-Shan South Road, Taipei 10002, Taiwan.
| |
Collapse
|
37
|
IGARASHI M. Molecular basis of the functions of the mammalian neuronal growth cone revealed using new methods. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:358-377. [PMID: 31406059 PMCID: PMC6766448 DOI: 10.2183/pjab.95.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/26/2019] [Indexed: 05/25/2023]
Abstract
The neuronal growth cone is a highly motile, specialized structure for extending neuronal processes. This structure is essential for nerve growth, axon pathfinding, and accurate synaptogenesis. Growth cones are important not only during development but also for plasticity-dependent synaptogenesis and neuronal circuit rearrangement following neural injury in the mature brain. However, the molecular details of mammalian growth cone function are poorly understood. This review examines molecular findings on the function of the growth cone as a result of the introduction of novel methods such superresolution microscopy and (phospho)proteomics. These results increase the scope of our understating of the molecular mechanisms of growth cone behavior in the mammalian brain.
Collapse
Affiliation(s)
- Michihiro IGARASHI
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
38
|
Minegishi T, Uesugi Y, Kaneko N, Yoshida W, Sawamoto K, Inagaki N. Shootin1b Mediates a Mechanical Clutch to Produce Force for Neuronal Migration. Cell Rep 2018; 25:624-639.e6. [DOI: 10.1016/j.celrep.2018.09.068] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/31/2018] [Accepted: 09/21/2018] [Indexed: 11/26/2022] Open
|
39
|
Baba K, Yoshida W, Toriyama M, Shimada T, Manning CF, Saito M, Kohno K, Trimmer JS, Watanabe R, Inagaki N. Gradient-reading and mechano-effector machinery for netrin-1-induced axon guidance. eLife 2018; 7:34593. [PMID: 30082022 PMCID: PMC6080949 DOI: 10.7554/elife.34593] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/05/2018] [Indexed: 12/28/2022] Open
Abstract
Growth cones navigate axonal projection in response to guidance cues. However, it is unclear how they can decide the migratory direction by transducing the local spatial cues into protrusive forces. Here we show that knockout mice of Shootin1 display abnormal projection of the forebrain commissural axons, a phenotype similar to that of the axon guidance molecule netrin-1. Shallow gradients of netrin-1 elicited highly polarized Pak1-mediated phosphorylation of shootin1 within growth cones. We demonstrate that netrin-1–elicited shootin1 phosphorylation increases shootin1 interaction with the cell adhesion molecule L1-CAM; this, in turn, promotes F-actin–adhesion coupling and concomitant generation of forces for growth cone migration. Moreover, the spatially regulated shootin1 phosphorylation within growth cones is required for axon turning induced by netrin-1 gradients. Our study defines a mechano-effector for netrin-1 signaling and demonstrates that shootin1 phosphorylation is a critical readout for netrin-1 gradients that results in a directional mechanoresponse for axon guidance. Neurons communicate with each other by forming intricate webs that link cells together according to a precise pattern. A neuron can connect to another by growing a branch-like structure known as the axon. To contact the correct neuron, the axon must develop and thread its way to exactly the right place in the brain. Scientists know that the tip of the axon is extraordinarily sensitive to gradients of certain molecules in its surroundings, which guide the budding structure towards its final destination. In particular, two molecules seem to play an important part in this process: netrin-1, which is a protein found outside cells that attracts a growing axon, and shootin1a, which is present inside neurons. Previous studies have shown that netrin-1 can trigger a cascade of reactions that activates shootin1a. In turn, activated shootin1a molecules join the internal skeleton of the cell with L1-CAM, a molecule that attaches the neuron to its surroundings. If the internal skeleton is the engine of the axon, L1-CAMs are the wheels, and shootin1a the clutch. However, it is not clear whether shootin1a is involved in guiding growing axons, and how it could help neurons ‘understand’ and react to gradients of netrin-1. Here, Baba et al. discover that when shootin1a is absent in mice, the axons do not develop properly. Further experiments in rat neurons show that if there is a little more netrin-1 on one side of the tip of an axon, this switches on the shootin1a molecules on that edge. Activated shootin1a promote interactions between the internal skeleton and L1-CAM, helping the axon curve towards the area that has more netrin-1. In fact, if the activated shootin1a is present everywhere on the axon, and not just on one side, the structure can develop, but not turn. Taken together, the results suggest that shootin1a can read the gradients of netrin-1 and then coordinate the turning of a growing axon in response. Wound healing, immune responses or formation of organs are just a few examples of processes that rely on cells moving in an orderly manner through the body. Dissecting how axons are guided through their development may shed light on the migration of cells in general. Ultimately, this could help scientists to understand disorders such as birth abnormalities or neurological disabilities, which arise when this process goes awry.
Collapse
Affiliation(s)
- Kentarou Baba
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Wataru Yoshida
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Tadayuki Shimada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Colleen F Manning
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, United States
| | - Michiko Saito
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kenji Kohno
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, United States
| | - Rikiya Watanabe
- Department of Applied Chemistry, Graduate School of Engineering, University of Tokyo, Tokyo, Japan
| | - Naoyuki Inagaki
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
40
|
Nithianandam V, Chien CT. Actin blobs prefigure dendrite branching sites. J Cell Biol 2018; 217:3731-3746. [PMID: 30042190 PMCID: PMC6168249 DOI: 10.1083/jcb.201711136] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 07/09/2018] [Indexed: 02/08/2023] Open
Abstract
Nithianandam and Chien show via in vivo imaging that a dynamic population of F-actin termed actin blobs propagates bidirectionally in dendrites and stalls at future branching sites. The F-actin–severing protein Tsr/cofilin is a regulator of actin blob dynamics and dendrite branching. The actin cytoskeleton provides structural stability and adaptability to the cell. Neuronal dendrites frequently undergo morphological changes by emanating, elongating, and withdrawing branches. However, the knowledge about actin dynamics in dendrites during these processes is limited. By performing in vivo imaging of F-actin markers, we found that F-actin was highly dynamic and heterogeneously distributed in dendritic shafts with enrichment at terminal dendrites. A dynamic F-actin population that we named actin blobs propagated bidirectionally at an average velocity of 1 µm/min. Interestingly, these actin blobs stalled at sites where new dendrites would branch out in minutes. Overstabilization of F-actin by the G15S mutant abolished actin blobs and dendrite branching. We identified the F-actin–severing protein Tsr/cofilin as a regulator of dynamic actin blobs and branching activity. Hence, actin blob localization at future branching sites represents a dendrite-branching mechanism to account for highly diversified dendritic morphology.
Collapse
Affiliation(s)
- Vanitha Nithianandam
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Cheng-Ting Chien
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan .,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
41
|
Gangatharan G, Schneider-Maunoury S, Breau MA. Role of mechanical cues in shaping neuronal morphology and connectivity. Biol Cell 2018; 110:125-136. [PMID: 29698566 DOI: 10.1111/boc.201800003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
Neuronal circuits, the functional building blocks of the nervous system, assemble during development through a series of dynamic processes including the migration of neurons to their final position, the growth and navigation of axons and their synaptic connection with target cells. While the role of chemical cues in guiding neuronal migration and axonal development has been extensively analysed, the contribution of mechanical inputs, such as forces and stiffness, has received far less attention. In this article, we review the in vitro and more recent in vivo studies supporting the notion that mechanical signals are critical for multiple aspects of neuronal circuit assembly, from the emergence of axons to the formation of functional synapses. By combining live imaging approaches with tools designed to measure and manipulate the mechanical environment of neurons, the emerging field of neuromechanics will add a new paradigm in our understanding of neuronal development and potentially inspire novel regenerative therapies.
Collapse
Affiliation(s)
- Girisaran Gangatharan
- Sorbonne Université, CNRS UMR 7622, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine (LBD-IBPS), INSERM, Paris, 75005, France
| | - Sylvie Schneider-Maunoury
- Sorbonne Université, CNRS UMR 7622, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine (LBD-IBPS), INSERM, Paris, 75005, France
| | - Marie Anne Breau
- Sorbonne Université, CNRS UMR 7622, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine (LBD-IBPS), INSERM, Paris, 75005, France.,Sorbonne Université, CNRS UMR 8237, Laboratoire Jean Perrin, Paris, 75005, France
| |
Collapse
|
42
|
Papandréou MJ, Leterrier C. The functional architecture of axonal actin. Mol Cell Neurosci 2018; 91:151-159. [PMID: 29758267 DOI: 10.1016/j.mcn.2018.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 11/17/2022] Open
Abstract
The cytoskeleton builds and supports the complex architecture of neurons. It orchestrates the specification, growth, and compartmentation of the axon: axon initial segment, axonal shaft, presynapses. The cytoskeleton must then maintain this intricate architecture for the whole life of its host, but also drive its adaptation to new network demands and changing physiological conditions. Microtubules are readily visible inside axon shafts by electron microscopy, whereas axonal actin study has long been focused on dynamic structures of the axon such as growth cones. Super-resolution microscopy and live-cell imaging have recently revealed new actin-based structures in mature axons: rings, hotspots and trails. This has caused renewed interest for axonal actin, with efforts underway to understand the precise organization and cellular functions of these assemblies. Actin is also present in presynapses, where its arrangement is still poorly defined, and its functions vigorously debated. Here we review the organization of axonal actin, focusing on recent advances and current questions in this rejuvenated field.
Collapse
|
43
|
Segal D, Zaritsky A, Schejter ED, Shilo BZ. Feedback inhibition of actin on Rho mediates content release from large secretory vesicles. J Cell Biol 2018; 217:1815-1826. [PMID: 29496739 PMCID: PMC5940311 DOI: 10.1083/jcb.201711006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/24/2017] [Accepted: 01/30/2018] [Indexed: 12/02/2022] Open
Abstract
Secretion of adhesive glycoproteins to the lumen of Drosophila melanogaster larval salivary glands is performed by contraction of an actomyosin network assembled around large secretory vesicles, after their fusion to the apical membranes. We have identified a cycle of actin coat nucleation and disassembly that is independent of myosin. Recruitment of active Rho1 to the fused vesicle triggers activation of the formin Diaphanous and actin nucleation. This leads to actin-dependent localization of a RhoGAP protein that locally shuts off Rho1, promoting disassembly of the actin coat. When contraction of vesicles is blocked, the strict temporal order of the recruited elements generates repeated oscillations of actin coat formation and disassembly. Interestingly, different blocks to actin coat disassembly arrested vesicle contraction, indicating that actin turnover is an integral part of the actomyosin contraction cycle. The capacity of F-actin to trigger a negative feedback on its own production may be widely used to coordinate a succession of morphogenetic events or maintain homeostasis.
Collapse
Affiliation(s)
- Dagan Segal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Assaf Zaritsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
44
|
Inner Workings: Uncovering the neuron's internal skeleton. Proc Natl Acad Sci U S A 2018; 113:13931-13933. [PMID: 27930346 DOI: 10.1073/pnas.1617651113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
45
|
Mortal S, Iseppon F, Perissinotto A, D'Este E, Cojoc D, Napolitano LMR, Torre V. Actin Waves Do Not Boost Neurite Outgrowth in the Early Stages of Neuron Maturation. Front Cell Neurosci 2017; 11:402. [PMID: 29326552 PMCID: PMC5741660 DOI: 10.3389/fncel.2017.00402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/01/2017] [Indexed: 11/27/2022] Open
Abstract
During neurite development, Actin Waves (AWs) emerge at the neurite base and move up to its tip, causing a transient retraction of the Growth Cone (GC). Many studies have shown that AWs are linked to outbursts of neurite growth and, therefore, contribute to the fast elongation of the nascent axon. Using long term live cell-imaging, we show that AWs do not boost neurite outgrowth and that neurites without AWs can elongate for several hundred microns. Inhibition of Myosin II abolishes the transient GC retraction and strongly modifies the AWs morphology. Super-resolution nanoscopy shows that Myosin IIB shapes the growth cone-like AWs structure and is differently distributed in AWs and GCs. Interestingly, depletion of membrane cholesterol and inhibition of Rho GTPases decrease AWs frequency and velocity. Our results indicate that Myosin IIB, membrane tension, and small Rho GTPases are important players in the regulation of the AW dynamics. Finally, we suggest a role for AWs in maintaining the GCs active during environmental exploration.
Collapse
Affiliation(s)
- Simone Mortal
- Neurobiology Department, International School for Advanced Studies, Trieste, Italy
| | - Federico Iseppon
- Neurobiology Department, International School for Advanced Studies, Trieste, Italy
| | - Andrea Perissinotto
- Neurobiology Department, International School for Advanced Studies, Trieste, Italy
| | - Elisa D'Este
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Dan Cojoc
- Optical Manipulation Lab, Istituto Officina dei Materiali (CNR), Trieste, Italy
| | - Luisa M R Napolitano
- Neurobiology Department, International School for Advanced Studies, Trieste, Italy
| | - Vincent Torre
- Neurobiology Department, International School for Advanced Studies, Trieste, Italy
| |
Collapse
|
46
|
|
47
|
Collenburg L, Beyersdorf N, Wiese T, Arenz C, Saied EM, Becker-Flegler KA, Schneider-Schaulies S, Avota E. The Activity of the Neutral Sphingomyelinase Is Important in T Cell Recruitment and Directional Migration. Front Immunol 2017; 8:1007. [PMID: 28871263 PMCID: PMC5566967 DOI: 10.3389/fimmu.2017.01007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/07/2017] [Indexed: 01/13/2023] Open
Abstract
Breakdown of sphingomyelin as catalyzed by the activity of sphingomyelinases profoundly affects biophysical properties of cellular membranes which is particularly important with regard to compartmentalization of surface receptors and their signaling relay. As it is activated both upon TCR ligation and co-stimulation in a spatiotemporally controlled manner, the neutral sphingomyelinase (NSM) has proven to be important in T cell activation, where it appears to play a particularly important role in cytoskeletal reorganization and cell polarization. Because these are important parameters in directional T cell migration and motility in tissues, we analyzed the role of the NSM in these processes. Pharmacological inhibition of NSM interfered with early lymph node homing of T cells in vivo indicating that the enzyme impacts on endothelial adhesion, transendothelial migration, sensing of chemokine gradients or, at a cellular level, acquisition of a polarized phenotype. NSM inhibition reduced adhesion of T cells to TNF-α/IFN-γ activated, but not resting endothelial cells, most likely via inhibiting high-affinity LFA-1 clustering. NSM activity proved to be highly important in directional T cell motility in response to SDF1-α, indicating that their ability to sense and translate chemokine gradients might be NSM dependent. In fact, pharmacological or genetic NSM ablation interfered with T cell polarization both at an overall morphological level and redistribution of CXCR4 and pERM proteins on endothelial cells or fibronectin, as well as with F-actin polymerization in response to SDF1-α stimulation, indicating that efficient directional perception and signaling relay depend on NSM activity. Altogether, these data support a central role of the NSM in T cell recruitment and migration both under homeostatic and inflamed conditions by regulating polarized redistribution of receptors and their coupling to the cytoskeleton.
Collapse
Affiliation(s)
- Lena Collenburg
- Institute for Virology and Immunobiology, University of Würzburg, Wuerzburg, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Wuerzburg, Germany
| | - Teresa Wiese
- Institute for Virology and Immunobiology, University of Würzburg, Wuerzburg, Germany
| | - Christoph Arenz
- Institute for Organic and Bioorganic Chemistry, Humboldt University of Berlin, Berlin, Germany
| | - Essa M Saied
- Institute for Organic and Bioorganic Chemistry, Humboldt University of Berlin, Berlin, Germany.,Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | | | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Wuerzburg, Germany
| |
Collapse
|
48
|
Huang DL, Bax NA, Buckley CD, Weis WI, Dunn AR. Vinculin forms a directionally asymmetric catch bond with F-actin. Science 2017; 357:703-706. [PMID: 28818948 PMCID: PMC5821505 DOI: 10.1126/science.aan2556] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/11/2017] [Indexed: 11/02/2022]
Abstract
Vinculin is an actin-binding protein thought to reinforce cell-cell and cell-matrix adhesions. However, how mechanical load affects the vinculin-F-actin bond is unclear. Using a single-molecule optical trap assay, we found that vinculin forms a force-dependent catch bond with F-actin through its tail domain, but with lifetimes that depend strongly on the direction of the applied force. Force toward the pointed (-) end of the actin filament resulted in a bond that was maximally stable at 8 piconewtons, with a mean lifetime (12 seconds) 10 times as long as the mean lifetime when force was applied toward the barbed (+) end. A computational model of lamellipodial actin dynamics suggests that the directionality of the vinculin-F-actin bond could establish long-range order in the actin cytoskeleton. The directional and force-stabilized binding of vinculin to F-actin may be a mechanism by which adhesion complexes maintain front-rear asymmetry in migrating cells.
Collapse
Affiliation(s)
- Derek L Huang
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Nicolas A Bax
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
| | - Craig D Buckley
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - William I Weis
- Biophysics Program, Stanford University, Stanford, CA 94305, USA.
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA
| | - Alexander R Dunn
- Biophysics Program, Stanford University, Stanford, CA 94305, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
49
|
Actin Waves: Origin of Cell Polarization and Migration? Trends Cell Biol 2017; 27:515-526. [DOI: 10.1016/j.tcb.2017.02.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/26/2017] [Accepted: 02/07/2017] [Indexed: 01/22/2023]
|
50
|
Iizuka Y, Mooneyham A, Sieben A, Chen K, Maile M, Hellweg R, Schütz F, Teckle K, Starr T, Thayanithy V, Vogel RI, Lou E, Lee MK, Bazzaro M. UNC-45A is required for neurite extension via controlling NMII activation. Mol Biol Cell 2017; 28:1337-1346. [PMID: 28356421 PMCID: PMC5426848 DOI: 10.1091/mbc.e16-06-0381] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 01/13/2023] Open
Abstract
UNC-45A is a novel regulator of neuronal differentiation. UNC-45A localizes at the growth cone, binds to NMIIA and NMIIB, and is disposable for neuronal survival but is required for neurite initiation and extension via regulating NMII activation. Thus UNC-45A is a potential master regulator of a number of NMII-mediated cellular processes. UNC-45A is a highly conserved member of the UNC-45/CRO1/She4p family of proteins, which act as chaperones for conventional and nonconventional myosins. NMII mediates contractility and actin-based motility, which are fundamental for proper growth cone motility and neurite extension. The presence and role of UNC-45A in neuronal differentiation have been largely unknown. Here we demonstrate that UNC-45A is a novel growth cone–localized, NMII-associated component of the multiprotein complex regulating growth cone dynamics. We show that UNC-45A is dispensable for neuron survival but required for neurite elongation. Mechanistically, loss of UNC-45A results in increased levels of NMII activation. Collectively our results provide novel insights into the molecular mechanisms of neurite growth and define UNC-45A as a novel and master regulator of NMII-mediated cellular processes in neurons.
Collapse
Affiliation(s)
- Yoshie Iizuka
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Ashley Mooneyham
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Andrew Sieben
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Kevin Chen
- Department of Biology, University of Maryland, Baltimore, MD 21250
| | - Makayla Maile
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Raffaele Hellweg
- Breast Unit, University of Heidelberg, 69120 Heidelberg, Germany
| | - Florian Schütz
- Breast Unit, University of Heidelberg, 69120 Heidelberg, Germany
| | - Kebebush Teckle
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Timothy Starr
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Venugopal Thayanithy
- Division of Hematology, Oncology and Transplantation, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Rachel Isaksson Vogel
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women's Heath, University of Minnesota Twin Cities, Minneapolis, MN 55455
| |
Collapse
|