1
|
Huang B, Li X. Mechanisms of GPM6A in Malignant Tumors. Cancer Rep (Hoboken) 2025; 8:e70137. [PMID: 39957375 PMCID: PMC11831008 DOI: 10.1002/cnr2.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2024] [Revised: 09/20/2024] [Accepted: 01/25/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Glycoprotein M6A (GPM6A) encodes a transmembrane protein, expressing in large quantities on the cell surface of central nervous system (CNS) neurons. GPM6A acts importantly in neurodevelopment by modulating neuronal differentiation, migration, axon growth, synaptogenesis, and spine formation, but its role in malignancy remains controversial and requires further research. This article reviewed the mechanisms of GPM6A in colorectal cancer, liver cancer, lung cancer, glioblastoma, and other malignant tumors, and made a "one-stop" summary of the relevant mechanisms. RECENT FINDINGS Researches have indicated that GPM6A is related to malignant tumors. It affects epithelial-mesenchymal transition and induces the formation of filopodia, participating in the adhesion, migration, and metastasis of cancer cells. Its role in malignant tumors remains controversial, however. On the one hand, GPM6A may have carcinogenic properties and is related to poor prognosis of malignant tumors. It is highly expressed in lymphoblastic leukemia and is a potential oncogene. It also shows carcinogenic properties in colorectal cancer, glioblastoma, gonadotroph adenomas and so on. On the other hand, the expression of GPM6A decreases in lung adenocarcinoma, liver cancer, thyroid cancer, and so forth as the tumor progresses, and it can inhibit the progression of malignant tumors by inhibiting some signaling pathways, suggesting that it may be a tumor suppressor gene. CONCLUSION Carcinogenic or tumor suppressive? Although the biological function of GPM6A in the development of malignant tumors is still unclear, according to the current research progress, it is still expected to become an effective molecular marker for predicting tumor occurrence, metastasis and prognosis, as well as a new target for diagnosis and treatment.
Collapse
Affiliation(s)
- Bei Huang
- Operation Management and Evaluation Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
- Emergency Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
2
|
Gosain H, Busch KB. TC10 differently controls the dynamics of Exo70 in growth cones of cortical and hippocampal neurons. BIOPHYSICAL REPORTS 2024; 4:100186. [PMID: 39521348 PMCID: PMC11617994 DOI: 10.1016/j.bpr.2024.100186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/03/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The exocyst is an octameric protein complex that acts as a tether for GOLGI-derived vesicles at the plasma membrane during exocytosis. It is involved in membrane expansion during axonal outgrowth. Exo70 is a major subunit of the exocyst complex and is controlled by TC10, a Rho family GTPase. How TC10 affects the dynamics of Exo70 at the plasma membrane is not well understood. There is also evidence that TC10 controls Exo70 dynamics differently in nonpolar cells and axons. To address this, we used super-resolution microscopy to study the spatially resolved effects of TC10 on Exo70 dynamics in HeLa cells and the growth cone of cortical and hippocampal neurons. We generated single-particle localization and trajectory maps and extracted mean square displacements, diffusion coefficients, and alpha coefficients to characterize Exo70 diffusion. We found that the diffusivity of Exo70 was different in nonpolar cells and the growth cone of neurons. TC10 stimulated the mobility of Exo70 in HeLa cells but decreased the diffusion of Exo70 in the growth cone of cortical neurons. In contrast to cortical neurons, TC10 overexpression did not affect the mobility of Exo70 in the axonal growth cone of hippocampal neurons. These data suggest that mainly exocyst tethering in cortical neurons was under the control of TC10.
Collapse
Affiliation(s)
- Hiteshika Gosain
- Institute of Integrative Cell Biology and Physiology, Department of Biology, University of Muenster, Münster, North-Rhine-Westphalia, Germany
| | - Karin B Busch
- Institute of Integrative Cell Biology and Physiology, Department of Biology, University of Muenster, Münster, North-Rhine-Westphalia, Germany.
| |
Collapse
|
3
|
Nozumi M, Sato Y, Nishiyama-Usuda M, Igarashi M. Identification of z-axis filopodia in growth cones using super-resolution microscopy. J Neurochem 2024; 168:2974-2988. [PMID: 38946488 DOI: 10.1111/jnc.16162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2023] [Revised: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
A growth cone is a highly motile tip of an extending axon that is crucial for neural network formation. Three-dimensional-structured illumination microscopy, a type of super-resolution light microscopy with a resolution that overcomes the optical diffraction limitation (ca. 200 nm) of conventional light microscopy, is well suited for studying the molecular dynamics of intracellular events. Using this technique, we discovered a novel type of filopodia distributed along the z-axis ("z-filopodia") within the growth cone. Z-filopodia were typically oriented in the direction of axon growth, not attached to the substratum, protruded spontaneously without microtubule invasion, and had a lifetime that was considerably shorter than that of conventional filopodia. Z-filopodia formation and dynamics were regulated by actin-regulatory proteins, such as vasodilator-stimulated phosphoprotein, fascin, and cofilin. Chromophore-assisted laser inactivation of cofilin induced the rapid turnover of z-filopodia. An axon guidance receptor, neuropilin-1, was concentrated in z-filopodia and was transported together with them, whereas its ligand, semaphorin-3A, was selectively bound to them. Membrane domains associated with z-filopodia were also specialized and resembled those of lipid rafts, and their behaviors were closely related to those of neuropilin-1. The results suggest that z-filopodia have unique turnover properties, and unlike xy-filopodia, do not function as force-generating structures for axon extension.
Collapse
Affiliation(s)
- Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Yuta Sato
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Miyako Nishiyama-Usuda
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
4
|
Liberali P, Schier AF. The evolution of developmental biology through conceptual and technological revolutions. Cell 2024; 187:3461-3495. [PMID: 38906136 DOI: 10.1016/j.cell.2024.05.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Developmental biology-the study of the processes by which cells, tissues, and organisms develop and change over time-has entered a new golden age. After the molecular genetics revolution in the 80s and 90s and the diversification of the field in the early 21st century, we have entered a phase when powerful technologies provide new approaches and open unexplored avenues. Progress in the field has been accelerated by advances in genomics, imaging, engineering, and computational biology and by emerging model systems ranging from tardigrades to organoids. We summarize how revolutionary technologies have led to remarkable progress in understanding animal development. We describe how classic questions in gene regulation, pattern formation, morphogenesis, organogenesis, and stem cell biology are being revisited. We discuss the connections of development with evolution, self-organization, metabolism, time, and ecology. We speculate how developmental biology might evolve in an era of synthetic biology, artificial intelligence, and human engineering.
Collapse
Affiliation(s)
- Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | | |
Collapse
|
5
|
Blake TCA, Fox HM, Urbančič V, Ravishankar R, Wolowczyk A, Allgeyer ES, Mason J, Danuser G, Gallop JL. Filopodial protrusion driven by density-dependent Ena-TOCA-1 interactions. J Cell Sci 2024; 137:jcs261057. [PMID: 38323924 PMCID: PMC11006392 DOI: 10.1242/jcs.261057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
Filopodia are narrow actin-rich protrusions with important roles in neuronal development where membrane-binding adaptor proteins, such as I-BAR- and F-BAR-domain-containing proteins, have emerged as upstream regulators that link membrane interactions to actin regulators such as formins and proteins of the Ena/VASP family. Both the adaptors and their binding partners are part of diverse and redundant protein networks that can functionally compensate for each other. To explore the significance of the F-BAR domain-containing neuronal membrane adaptor TOCA-1 (also known as FNBP1L) in filopodia we performed a quantitative analysis of TOCA-1 and filopodial dynamics in Xenopus retinal ganglion cells, where Ena/VASP proteins have a native role in filopodial extension. Increasing the density of TOCA-1 enhances Ena/VASP protein binding in vitro, and an accumulation of TOCA-1, as well as its coincidence with Ena, correlates with filopodial protrusion in vivo. Two-colour single-molecule localisation microscopy of TOCA-1 and Ena supports their nanoscale association. TOCA-1 clusters promote filopodial protrusion and this depends on a functional TOCA-1 SH3 domain and activation of Cdc42, which we perturbed using the small-molecule inhibitor CASIN. We propose that TOCA-1 clusters act independently of membrane curvature to recruit and promote Ena activity for filopodial protrusion.
Collapse
Affiliation(s)
- Thomas C. A. Blake
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Helen M. Fox
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Vasja Urbančič
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Roshan Ravishankar
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam Wolowczyk
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Edward S. Allgeyer
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Julia Mason
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jennifer L. Gallop
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
6
|
Honda A, Nozumi M, Ito Y, Natsume R, Kawasaki A, Nakatsu F, Abe M, Uchino H, Matsushita N, Ikeda K, Arita M, Sakimura K, Igarashi M. Very-long-chain fatty acids are crucial to neuronal polarity by providing sphingolipids to lipid rafts. Cell Rep 2023; 42:113195. [PMID: 37816355 DOI: 10.1016/j.celrep.2023.113195] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2023] [Revised: 08/19/2023] [Accepted: 09/14/2023] [Indexed: 10/12/2023] Open
Abstract
Fatty acids have long been considered essential to brain development; however, the involvement of their synthesis in nervous system formation is unclear. We generate mice with knockout of GPSN2, an enzyme for synthesis of very-long-chain fatty acids (VLCFAs) and investigate the effects. Both GPSN2-/- and GPSN2+/- mice show abnormal neuronal networks as a result of impaired neuronal polarity determination. Lipidomics of GPSN2-/- embryos reveal that ceramide synthesis is specifically inhibited depending on FA length; namely, VLCFA-containing ceramide is reduced. We demonstrate that lipid rafts are highly enriched in growth cones and that GPSN2+/- neurons lose gangliosides in their membranes. Application of C24:0 ceramide, but not C16:0 ceramide or C24:0 phosphatidylcholine, to GPSN2+/- neurons rescues both neuronal polarity determination and lipid-raft density in the growth cone. Taken together, our results indicate that VLCFA synthesis contributes to physiological neuronal development in brain network formation, in particular neuronal polarity determination through the formation of lipid rafts.
Collapse
Affiliation(s)
- Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Center for Research Promotion, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Rie Natsume
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Haruki Uchino
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Natsuki Matsushita
- Division of Laboratory Animal Research, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Kazutaka Ikeda
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan; Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
7
|
Rajan S, Yoon J, Wu H, Srapyan S, Baskar R, Ahmed G, Yang T, Grintsevich EE, Reisler E, Terman JR. Disassembly of bundled F-actin and cellular remodeling via an interplay of Mical, cofilin, and F-actin crosslinkers. Proc Natl Acad Sci U S A 2023; 120:e2309955120. [PMID: 37725655 PMCID: PMC10523612 DOI: 10.1073/pnas.2309955120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/16/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
Cellular form and function are controlled by the assembly and stability of actin cytoskeletal structures-but disassembling/pruning these structures is equally essential for the plasticity and remodeling that underlie behavioral adaptations. Importantly, the mechanisms of actin assembly have been well-defined-including that it is driven by actin's polymerization into filaments (F-actin) and then often bundling by crosslinking proteins into stable higher-order structures. In contrast, it remains less clear how these stable bundled F-actin structures are rapidly disassembled. We now uncover mechanisms that rapidly and extensively disassemble bundled F-actin. Using biochemical, structural, and imaging assays with purified proteins, we show that F-actin bundled with one of the most prominent crosslinkers, fascin, is extensively disassembled by Mical, the F-actin disassembly enzyme. Furthermore, the product of this Mical effect, Mical-oxidized actin, is poorly bundled by fascin, thereby further amplifying Mical's disassembly effects on bundled F-actin. Moreover, another critical F-actin regulator, cofilin, also affects fascin-bundled filaments, but we find herein that it synergizes with Mical to dramatically amplify its disassembly of bundled F-actin compared to the sum of their individual effects. Genetic and high-resolution cellular assays reveal that Mical also counteracts crosslinking proteins/bundled F-actin in vivo to control cellular extension, axon guidance, and Semaphorin/Plexin cell-cell repulsion. Yet, our results also support the idea that fascin-bundling serves to dampen Mical's F-actin disassembly in vitro and in vivo-and that physiologically relevant cellular remodeling requires a fine-tuned interplay between the factors that build bundled F-actin networks and those that disassemble them.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
| | - Jimok Yoon
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Heng Wu
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Sargis Srapyan
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA90840
| | - Raju Baskar
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Giasuddin Ahmed
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Taehong Yang
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| | - Elena E. Grintsevich
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA90840
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - Jonathan R. Terman
- Department of Neuroscience, The University of Texas of Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
8
|
Gong J, Jin Z, Chen H, He J, Zhang Y, Yang X. Super-resolution fluorescence microscopic imaging in pathogenesis and drug treatment of neurological disease. Adv Drug Deliv Rev 2023; 196:114791. [PMID: 37004939 DOI: 10.1016/j.addr.2023.114791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2022] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 04/03/2023]
Abstract
Since super-resolution fluorescence microscopic technology breaks the diffraction limit that has existed for a long time in optical imaging, it can observe the process of synapses formed between nerve cells and the protein aggregation related to neurological disease. Thus, super-resolution fluorescence microscopic imaging has significantly impacted several industries, including drug development and pathogenesis research, and it is anticipated that it will significantly alter the future of life science research. Here, we focus on several typical super-resolution fluorescence microscopic technologies, introducing their benefits and drawbacks, as well as applications in several common neurological diseases, in the hope that their services will be expanded and improved in the pathogenesis and drug treatment of neurological diseases.
Collapse
|
9
|
Hasanzadeh A, Hamblin MR, Kiani J, Noori H, Hardie JM, Karimi M, Shafiee H. Could artificial intelligence revolutionize the development of nanovectors for gene therapy and mRNA vaccines? NANO TODAY 2022; 47:101665. [PMID: 37034382 PMCID: PMC10081506 DOI: 10.1016/j.nantod.2022.101665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/19/2023]
Abstract
Gene therapy enables the introduction of nucleic acids like DNA and RNA into host cells, and is expected to revolutionize the treatment of a wide range of diseases. This growth has been further accelerated by the discovery of CRISPR/Cas technology, which allows accurate genomic editing in a broad range of cells and organisms in vitro and in vivo. Despite many advances in gene delivery and the development of various viral and non-viral gene delivery vectors, the lack of highly efficient non-viral systems with low cellular toxicity remains a challenge. The application of cutting-edge technologies such as artificial intelligence (AI) has great potential to find new paradigms to solve this issue. Herein, we review AI and its major subfields including machine learning (ML), neural networks (NNs), expert systems, deep learning (DL), computer vision and robotics. We discuss the potential of AI-based models and algorithms in the design of targeted gene delivery vehicles capable of crossing extracellular and intracellular barriers by viral mimicry strategies. We finally discuss the role of AI in improving the function of CRISPR/Cas systems, developing novel nanobots, and mRNA vaccine carriers.
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Noori
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Joseph M. Hardie
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 141556559, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 1584743311, Iran
| | - Hadi Shafiee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| |
Collapse
|
10
|
Tagliatti E, Cortese K. Imaging Endocytosis Dynamics in Health and Disease. MEMBRANES 2022; 12:membranes12040393. [PMID: 35448364 PMCID: PMC9028293 DOI: 10.3390/membranes12040393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
Abstract
Endocytosis is a critical process for cell growth and viability. It mediates nutrient uptake, guarantees plasma membrane homeostasis, and generates intracellular signaling cascades. Moreover, it plays an important role in dead cell clearance and defense against external microbes. Finally, endocytosis is an important cellular route for the delivery of nanomedicines for therapeutic treatments. Thus, it is not surprising that both environmental and genetic perturbation of endocytosis have been associated with several human conditions such as cancer, neurological disorders, and virus infections, among others. Over the last decades, a lot of research has been focused on developing advanced imaging methods to monitor endocytosis events with high resolution in living cells and tissues. These include fluorescence imaging, electron microscopy, and correlative and super-resolution microscopy. In this review, we outline the major endocytic pathways and briefly discuss how defects in the molecular machinery of these pathways lead to disease. We then discuss the current imaging methodologies used to study endocytosis in different contexts, highlighting strengths and weaknesses.
Collapse
Affiliation(s)
- Erica Tagliatti
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Milano, Italy
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Correspondence: (E.T.); (K.C.)
| | - Katia Cortese
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine (DIMES), Human Anatomy, Università di Genova, Via Antonio de Toni 14, 16132 Genova, Italy
- Correspondence: (E.T.); (K.C.)
| |
Collapse
|
11
|
Okada M, Kawagoe Y, Takasugi T, Nozumi M, Ito Y, Fukusumi H, Kanemura Y, Fujii Y, Igarashi M. JNK1-Dependent Phosphorylation of GAP-43 Serine 142 is a Novel Molecular Marker for Axonal Growth. Neurochem Res 2022; 47:2668-2682. [PMID: 35347634 DOI: 10.1007/s11064-022-03580-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
Abstract
Mammalian axon growth has mechanistic similarities with axon regeneration. The growth cone is an important structure that is involved in both processes, and GAP-43 (growth associated protein-43 kDa) is believed to be the classical molecular marker. Previously, we used growth cone phosphoproteomics to demonstrate that S96 and T172 of GAP-43 in rodents are highly phosphorylated sites that are phosphorylated by c-jun N-terminal protein kinase (JNK). We also revealed that phosphorylated (p)S96 and pT172 antibodies recognize growing axons in the developing brain and regenerating axons in adult peripheral nerves. In rodents, S142 is another putative JNK-dependent phosphorylation site that is modified at a lower frequency than S96 and T172. Here, we characterized this site using a pS142-specific antibody. We confirmed that pS142 was detected by co-expressing mouse GAP-43 and JNK1. pS142 antibody labeled growth cones and growing axons in developing mouse neurons. pS142 was sustained until at least nine weeks after birth in mouse brains. The pS142 antibody could detect regenerating axons following sciatic nerve injury in adult mice. Comparison of amino acid sequences indicated that rodent S142 corresponds to human T151, which is predicted to be a substrate of the MAPK family, which includes JNK. Thus, we confirmed that the pS142 antibody recognized human phospho-GAP-43 using activated JNK1, and also that its immunostaining pattern in neurons differentiated from human induced pluripotent cells was similar to those observed in mice. These results indicate that the S142 residue is phosphorylated by JNK1 and that the pS142 antibody is a new candidate molecular marker for axonal growth in both rodents and human.
Collapse
Affiliation(s)
- Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Neurosurgery, Medical and Dental Hospital, Niigata University, Niigata, Japan
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Yosuke Kawagoe
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Toshiyuki Takasugi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Hayato Fukusumi
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan.
| |
Collapse
|
12
|
Tsai CL, Huang CY, Lu YC, Pai LM, Horák D, Ma YH. Cyclic Strain Mitigates Nanoparticle Internalization by Vascular Smooth Muscle Cells. Int J Nanomedicine 2022; 17:969-981. [PMID: 35280334 PMCID: PMC8909538 DOI: 10.2147/ijn.s337942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2021] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Chia-Liang Tsai
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Ching-Yun Huang
- Institute of Biomedical Sciences, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Yi-Ching Lu
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
| | - Li-Mei Pai
- Department of Biochemistry & Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan, Republic of China
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague 6, 162 06, Czech Republic
| | - Yunn-Hwa Ma
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, 33302, Taiwan, Republic of China
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan, Republic of China
- Correspondence: Yunn-Hwa Ma, Department of Physiology and Pharmacology, Chang Gung University, Guishan, Taoyuan, 33302, Taiwan, Republic of China, Email
| |
Collapse
|
13
|
Gagliano G, Nelson T, Saliba N, Vargas-Hernández S, Gustavsson AK. Light Sheet Illumination for 3D Single-Molecule Super-Resolution Imaging of Neuronal Synapses. Front Synaptic Neurosci 2021; 13:761530. [PMID: 34899261 PMCID: PMC8651567 DOI: 10.3389/fnsyn.2021.761530] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/20/2021] [Accepted: 10/27/2021] [Indexed: 01/02/2023] Open
Abstract
The function of the neuronal synapse depends on the dynamics and interactions of individual molecules at the nanoscale. With the development of single-molecule super-resolution microscopy over the last decades, researchers now have a powerful and versatile imaging tool for mapping the molecular mechanisms behind the biological function. However, imaging of thicker samples, such as mammalian cells and tissue, in all three dimensions is still challenging due to increased fluorescence background and imaging volumes. The combination of single-molecule imaging with light sheet illumination is an emerging approach that allows for imaging of biological samples with reduced fluorescence background, photobleaching, and photodamage. In this review, we first present a brief overview of light sheet illumination and previous super-resolution techniques used for imaging of neurons and synapses. We then provide an in-depth technical review of the fundamental concepts and the current state of the art in the fields of three-dimensional single-molecule tracking and super-resolution imaging with light sheet illumination. We review how light sheet illumination can improve single-molecule tracking and super-resolution imaging in individual neurons and synapses, and we discuss emerging perspectives and new innovations that have the potential to enable and improve single-molecule imaging in brain tissue.
Collapse
Affiliation(s)
- Gabriella Gagliano
- Department of Chemistry, Rice University, Houston, TX, United States
- Applied Physics Program, Rice University, Houston, TX, United States
- Smalley-Curl Institute, Rice University, Houston, TX, United States
| | - Tyler Nelson
- Department of Chemistry, Rice University, Houston, TX, United States
- Applied Physics Program, Rice University, Houston, TX, United States
- Smalley-Curl Institute, Rice University, Houston, TX, United States
| | - Nahima Saliba
- Department of Chemistry, Rice University, Houston, TX, United States
| | - Sofía Vargas-Hernández
- Department of Chemistry, Rice University, Houston, TX, United States
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, United States
- Institute of Biosciences & Bioengineering, Rice University, Houston, TX, United States
| | - Anna-Karin Gustavsson
- Department of Chemistry, Rice University, Houston, TX, United States
- Smalley-Curl Institute, Rice University, Houston, TX, United States
- Institute of Biosciences & Bioengineering, Rice University, Houston, TX, United States
- Department of Biosciences, Rice University, Houston, TX, United States
- Laboratory for Nanophotonics, Rice University, Houston, TX, United States
| |
Collapse
|
14
|
Weaver CJ, Poulain FE. From whole organism to ultrastructure: progress in axonal imaging for decoding circuit development. Development 2021; 148:271122. [PMID: 34328171 DOI: 10.1242/dev.199717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023]
Abstract
Since the pioneering work of Ramón y Cajal, scientists have sought to unravel the complexities of axon development underlying neural circuit formation. Micrometer-scale axonal growth cones navigate to targets that are often centimeters away. To reach their targets, growth cones react to dynamic environmental cues that change in the order of seconds to days. Proper axon growth and guidance are essential to circuit formation, and progress in imaging has been integral to studying these processes. In particular, advances in high- and super-resolution microscopy provide the spatial and temporal resolution required for studying developing axons. In this Review, we describe how improved microscopy has revolutionized our understanding of axonal development. We discuss how novel technologies, specifically light-sheet and super-resolution microscopy, led to new discoveries at the cellular scale by imaging axon outgrowth and circuit wiring with extreme precision. We next examine how advanced microscopy broadened our understanding of the subcellular dynamics driving axon growth and guidance. We finally assess the current challenges that the field of axonal biology still faces for imaging axons, and examine how future technology could meet these needs.
Collapse
Affiliation(s)
- Cory J Weaver
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Fabienne E Poulain
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
15
|
Hou X, Nozumi M, Nakamura H, Igarashi M, Sugiyama S. Coactosin Promotes F-Actin Protrusion in Growth Cones Under Cofilin-Related Signaling Pathway. Front Cell Dev Biol 2021; 9:660349. [PMID: 34235144 PMCID: PMC8256272 DOI: 10.3389/fcell.2021.660349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
During brain development, axon outgrowth and its subsequent pathfinding are reliant on a highly motile growth cone located at the tip of the axon. Actin polymerization that is regulated by actin-depolymerizing factors homology (ADF-H) domain-containing family drives the formation of lamellipodia and filopodia at the leading edge of growth cones for axon guidance. However, the precise localization and function of ADF-H domain-containing proteins involved in axon extension and retraction remain unclear. We have previously shown that transcripts and proteins of coactosin-like protein 1 (COTL1), an ADF-H domain-containing protein, are observed in neurites and axons in chick embryos. Coactosin overexpression analysis revealed that this protein was localized to axonal growth cones and involved in axon extension in the midbrain. We further examined the specific distribution of coactosin and cofilin within the growth cone using superresolution microscopy, structured illumination microscopy, which overcomes the optical diffraction limitation and is suitable to the analysis of cellular dynamic movements. We found that coactosin was tightly associated with F-actin bundles at the growth cones and that coactosin overexpression promoted the expansion of lamellipodia and extension of growth cones. Coactosin knockdown in oculomotor neurons resulted in an increase in the levels of the inactive, phosphorylated form of cofilin and dysregulation of actin polymerization and axonal elongation, which suggests that coactosin promoted axonal growth in a cofilin-dependent manner. Indeed, the application of a dominant-negative form of LIMK1, a downstream effector of GTPases, reversed the effect of coactosin knockdown on axonal growth by enhancing cofilin activity. Combined, our results indicate that coactosin functions promote the assembly of protrusive actin filament arrays at the leading edge for growth cone motility.
Collapse
Affiliation(s)
- Xubin Hou
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Harukazu Nakamura
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sayaka Sugiyama
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
16
|
Prakash K, Diederich B, Reichelt S, Heintzmann R, Schermelleh L. Super-resolution structured illumination microscopy: past, present and future. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200143. [PMID: 33896205 PMCID: PMC8366908 DOI: 10.1098/rsta.2020.0143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/03/2023]
Abstract
Structured illumination microscopy (SIM) has emerged as an essential technique for three-dimensional (3D) and live-cell super-resolution imaging. However, to date, there has not been a dedicated workshop or journal issue covering the various aspects of SIM, from bespoke hardware and software development and the use of commercial instruments to biological applications. This special issue aims to recap recent developments as well as outline future trends. In addition to SIM, we cover related topics such as complementary super-resolution microscopy techniques, computational imaging, visualization and image processing methods. This article is part of the Theo Murphy meeting issue 'Super-resolution structured illumination microscopy (part 1)'.
Collapse
Affiliation(s)
- Kirti Prakash
- National Physical Laboratory, TW11 0LW Teddington, UK
- Department of Paediatrics, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Benedict Diederich
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University, Helmholtzweg 4, Jena, Germany
| | - Stefanie Reichelt
- CRUK Cambridge Research Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Rainer Heintzmann
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University, Helmholtzweg 4, Jena, Germany
- Faculty of Physics and Astronomy, Friedrich-Schiller-University, Jena, Germany
| | - Lothar Schermelleh
- Micron Advanced Bioimaging Unit, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| |
Collapse
|
17
|
Okada M, Kawagoe Y, Sato Y, Nozumi M, Ishikawa Y, Tamada A, Yamazaki H, Sekino Y, Kanemura Y, Shinmyo Y, Kawasaki H, Kaneko N, Sawamoto K, Fujii Y, Igarashi M. Phosphorylation of GAP-43 T172 is a molecular marker of growing axons in a wide range of mammals including primates. Mol Brain 2021; 14:66. [PMID: 33832520 PMCID: PMC8034164 DOI: 10.1186/s13041-021-00755-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
GAP-43 is a vertebrate neuron-specific protein and that is strongly related to axon growth and regeneration; thus, this protein has been utilized as a classical molecular marker of these events and growth cones. Although GAP-43 was biochemically characterized more than a quarter century ago, how this protein is related to these events is still not clear. Recently, we identified many phosphorylation sites in the growth cone membrane proteins of rodent brains. Two phosphorylation sites of GAP-43, S96 and T172, were found within the top 10 hit sites among all proteins. S96 has already been characterized (Kawasaki et al., 2018), and here, phosphorylation of T172 was characterized. In vitro (cultured neurons) and in vivo, an antibody specific to phosphorylated T172 (pT172 antibody) specifically recognized cultured growth cones and growing axons in developing mouse neurons, respectively. Immunoblotting showed that pT172 antigens were more rapidly downregulated throughout development than those of pS96 antibody. From the primary structure, this phosphorylation site was predicted to be conserved in a wide range of animals including primates. In the developing marmoset brainstem and in differentiated neurons derived from human induced pluripotent stem cells, immunoreactivity with pT172 antibody revealed patterns similar to those in mice. pT172 antibody also labeled regenerating axons following sciatic nerve injury. Taken together, the T172 residue is widely conserved in a wide range of mammals including primates, and pT172 is a new candidate molecular marker for growing axons.
Collapse
Affiliation(s)
- Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Medical and Dental Hospital, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yosuke Kawagoe
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yuta Sato
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Motohiro Nozumi
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yuya Ishikawa
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of Orthopedic Surgery, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Atsushi Tamada
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yuko Sekino
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yonehiro Kanemura
- Division of Regenerative Medicine, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Medical and Dental Hospital, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan.
| |
Collapse
|
18
|
Choquet D, Sainlos M, Sibarita JB. Advanced imaging and labelling methods to decipher brain cell organization and function. Nat Rev Neurosci 2021; 22:237-255. [PMID: 33712727 DOI: 10.1038/s41583-021-00441-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 02/05/2021] [Indexed: 01/31/2023]
Abstract
The brain is arguably the most complex organ. The branched and extended morphology of nerve cells, their subcellular complexity, the multiplicity of brain cell types as well as their intricate connectivity and the scattering properties of brain tissue present formidable challenges to the understanding of brain function. Neuroscientists have often been at the forefront of technological and methodological developments to overcome these hurdles to visualize, quantify and modify cell and network properties. Over the last few decades, the development of advanced imaging methods has revolutionized our approach to explore the brain. Super-resolution microscopy and tissue imaging approaches have recently exploded. These instrumentation-based innovations have occurred in parallel with the development of new molecular approaches to label protein targets, to evolve new biosensors and to target them to appropriate cell types or subcellular compartments. We review the latest developments for labelling and functionalizing proteins with small localization and functionalized reporters. We present how these molecular tools are combined with the development of a wide variety of imaging methods that break either the diffraction barrier or the tissue penetration depth limits. We put these developments in perspective to emphasize how they will enable step changes in our understanding of the brain.
Collapse
Affiliation(s)
- Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France. .,University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, Bordeaux, France.
| | - Matthieu Sainlos
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| | - Jean-Baptiste Sibarita
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
19
|
Doki C, Nishida K, Saito S, Shiga M, Ogara H, Kuramoto A, Kuragano M, Nozumi M, Igarashi M, Nakagawa H, Kotani S, Tokuraku K. Microtubule elongation along actin filaments induced by microtubule-associated protein 4 contributes to the formation of cellular protrusions. J Biochem 2021; 168:295-303. [PMID: 32289170 DOI: 10.1093/jb/mvaa046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2020] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Actin-microtubule crosstalk is implicated in the formation of cellular protrusions, but the mechanism remains unclear. In this study, we examined the regulation of cell protrusion involving a ubiquitously expressed microtubule-associated protein (MAP) 4, and its superfamily proteins, neuronal MAP2 and tau. Fluorescence microscopy revealed that these MAPs bound to F-actin and microtubules simultaneously, and formed F-actin/microtubule hybrid bundles. The hybrid bundle-forming activity was in the order of MAP2 > MAP4 ≫ tau. Interestingly, the microtubule assembly-promoting activity of MAP4 and MAP2, but not of tau, was upregulated by their interaction with F-actin. When MAP4 was overexpressed in NG108-15 cells, the number of cell processes and maximum process length of each cell increased significantly by 28% and 30%, respectively. Super-resolution microscopy revealed that 95% of microtubules in cell processes colocalized with F-actin, and MAP4 was always found in their vicinity. These results suggest that microtubule elongation along F-actin induced by MAP4 contributes to the formation of cellular protrusions. Since MAP4, MAP2 and tau had different crosstalk activity between F-actin and microtubules, it is likely that the functional differentiation of these MAPs is a driving force for neural evolution, causing significant changes in cell morphology.
Collapse
Affiliation(s)
- Chihiro Doki
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Kohei Nishida
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Shoma Saito
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Miyuki Shiga
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Hikari Ogara
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Ayumu Kuramoto
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Masahiro Kuragano
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Hiroyuki Nakagawa
- Division of Biology, Faculty of Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Susumu Kotani
- Department Biological Science, Faculty of Science, Kanagawa University, Kanagawa 259-1293, Japan
| | - Kiyotaka Tokuraku
- Department of Applied Sciences, Muroran Institute of Technology, Muroran, Hokkaido 050-8585, Japan
| |
Collapse
|
20
|
Wei R, Sugiyama A, Sato Y, Nozumi M, Nishino H, Takahashi M, Saito T, Ando K, Fukuda M, Tomomura M, Igarashi M, Hisanaga SI. Isoform-dependent subcellular localization of LMTK1A and LMTK1B and their roles in axon outgrowth and spine formation. J Biochem 2021; 168:23-32. [PMID: 32044995 DOI: 10.1093/jb/mvaa019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/25/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Lemur kinase 1 (LMTK1) is a membrane-bound Ser/Thr kinase that is expressed in neurons. There are two splicing variants of LMTK1 with different membrane binding modes, viz., cytosolic LMTK1A that binds to membranes through palmitoylation at the N-terminal cysteines and LMTK1B, an integral membrane protein with transmembrane sequences. We recently reported that LMTK1A regulates axon outgrowth and spine formation in neurons. However, data about LMTK1B are scarce. We analysed the expression and cellular localization of LMTK1B along with its role in axon and spine formation. We found that both LMTK1B and LMTK1A were expressed equally in the cerebral cortex and cerebellum of the mouse brain. Similar to LMTK1A, the wild type of LMTK1B was localized to Rab11-positive pericentrosomal compartment. The kinase negative (kn) mutant of LMTK1B was found to be associated with an increase in the tubular form of endoplasmic reticulum (ER), which was not the case with LMTK1A kn. Furthermore, unlike LMTK1A kn, LMTK1B kn did not stimulate the axon outgrowth and spine formation. These results suggest that while LMTK1A and LMTK1B share a common function in recycling endosomal trafficking at the pericentrosomal compartment, LMTK1B has an additional unique function in vesicle transport in the ER region.
Collapse
Affiliation(s)
- Ran Wei
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Arika Sugiyama
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Yuta Sato
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Hironori Nishino
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Miyuki Takahashi
- Miyuki Takahashi, Department of Life Science and Medical Bioscience, Laboratory for Molecular Brain Science, Waseda University, Tokyo 162-8480
| | - Taro Saito
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Kanae Ando
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mineko Tomomura
- Department of Oral Health Sciences, Meikai University School of Health Sciences, Urayasu, Chiba 279-9950, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Faculty of Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397, Japan
| |
Collapse
|
21
|
Remsburg C, Testa M, Song JL. Rab35 regulates skeletogenesis and gastrulation by facilitating actin remodeling and vesicular trafficking. Cells Dev 2021; 165:203660. [PMID: 34024337 DOI: 10.1016/j.cdev.2021.203660] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2020] [Revised: 11/11/2020] [Accepted: 12/21/2020] [Indexed: 11/15/2022]
Abstract
Rab35 is a small GTPase that regulates plasma membrane to early endosome vesicular trafficking and mediates actin remodeling to form actin-rich cellular structures. While the function of Rab35 in the cellular context has been examined, its role during development has not been well studied. In this study, we take advantage of the sea urchin's high fecundity, external fertilization, and transparent embryos to determine the function of Rab35 during development. We found that loss of function of Rab35 results in defects in skeletogenesis and gastrulation, which were rescued by co-injection of sea urchin Rab35. The loss of Rab35's function results in decreased endocytosis and impaired exocytosis, which may be important for skeletogenesis and gastrulation. Skeletal spicules of Rab35 knockdown embryos have reduced organized actin compared to the control, supporting the notion that Rab35 regulates actin dynamics. In addition, the skeletal and gastrulation defects induced by Rab35 knockdown were rescued by co-injection with Fascin, an actin-bundling protein, indicating that proper actin dynamics play a critical role for both skeletogenesis and gastrulation. Overall, results indicate that through its role in mediating vesicular trafficking and actin remodeling, Rab35 is an important regulator of embryonic structure formation in early development.
Collapse
Affiliation(s)
- Carolyn Remsburg
- University of Delaware, Department of Biological Sciences, Newark, DE, USA
| | - Michael Testa
- University of Delaware, Department of Biological Sciences, Newark, DE, USA
| | - Jia L Song
- University of Delaware, Department of Biological Sciences, Newark, DE, USA.
| |
Collapse
|
22
|
Lee S, Kim H, Higuchi H, Ishikawa M. Visualization Method for the Cell-Level Vesicle Transport Using Optical Flow and a Diverging Colormap. SENSORS (BASEL, SWITZERLAND) 2021; 21:E522. [PMID: 33450927 PMCID: PMC7828387 DOI: 10.3390/s21020522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 12/01/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 11/25/2022]
Abstract
Elucidation of cell-level transport mediated by vesicles within a living cell provides key information regarding viral infection processes and also drug delivery mechanisms. Although the single-particle tracking method has enabled clear analysis of individual vesicle trajectories, information regarding the entire cell-level intracellular transport is hardly obtainable, due to the difficulty in collecting a large dataset with current methods. In this paper, we propose a visualization method of vesicle transport using optical flow, based on geometric cell center estimation and vector analysis, for measuring the trafficking directions. As a quantitative visualization method for determining the intracellular transport status, the proposed method is expected to be universally exploited in various biomedical cell image analyses.
Collapse
Affiliation(s)
- Seohyun Lee
- Information Technology Center, Data Science Research Division, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan; (H.K.); (M.I.)
| | - Hyuno Kim
- Information Technology Center, Data Science Research Division, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan; (H.K.); (M.I.)
| | - Hideo Higuchi
- Department of Physics, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Masatoshi Ishikawa
- Information Technology Center, Data Science Research Division, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8656, Japan; (H.K.); (M.I.)
| |
Collapse
|
23
|
Leterrier C. A Pictorial History of the Neuronal Cytoskeleton. J Neurosci 2021; 41:11-27. [PMID: 33408133 PMCID: PMC7786211 DOI: 10.1523/jneurosci.2872-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Christophe Leterrier
- Aix Marseille Université, Centre National de la Recherche Scientifique, INP Unité Mixte de Recherche 7051, NeuroCyto, Marseille 13005, France
| |
Collapse
|
24
|
Jarsch IK, Gadsby JR, Nuccitelli A, Mason J, Shimo H, Pilloux L, Marzook B, Mulvey CM, Dobramysl U, Bradshaw CR, Lilley KS, Hayward RD, Vaughan TJ, Dobson CL, Gallop JL. A direct role for SNX9 in the biogenesis of filopodia. J Cell Biol 2020; 219:151579. [PMID: 32328641 PMCID: PMC7147113 DOI: 10.1083/jcb.201909178] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/08/2019] [Revised: 01/24/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
Filopodia are finger-like actin-rich protrusions that extend from the cell surface and are important for cell-cell communication and pathogen internalization. The small size and transient nature of filopodia combined with shared usage of actin regulators within cells confounds attempts to identify filopodial proteins. Here, we used phage display phenotypic screening to isolate antibodies that alter the actin morphology of filopodia-like structures (FLS) in vitro. We found that all of the antibodies that cause shorter FLS interact with SNX9, an actin regulator that binds phosphoinositides during endocytosis and at invadopodia. In cells, we discover SNX9 at specialized filopodia in Xenopus development and that SNX9 is an endogenous component of filopodia that are hijacked by Chlamydia entry. We show the use of antibody technology to identify proteins used in filopodia-like structures, and a role for SNX9 in filopodia.
Collapse
Affiliation(s)
- Iris K Jarsch
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Jonathan R Gadsby
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Annalisa Nuccitelli
- Antibody Discovery and Protein Engineering, AstraZeneca, Granta Park, Cambridge, UK
| | - Julia Mason
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Hanae Shimo
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Ludovic Pilloux
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Bishara Marzook
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Claire M Mulvey
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Ulrich Dobramysl
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Charles R Bradshaw
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Kathryn S Lilley
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Tristan J Vaughan
- Antibody Discovery and Protein Engineering, AstraZeneca, Granta Park, Cambridge, UK
| | - Claire L Dobson
- Antibody Discovery and Protein Engineering, AstraZeneca, Granta Park, Cambridge, UK
| | - Jennifer L Gallop
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Igarashi M, Honda A, Kawasaki A, Nozumi M. Neuronal Signaling Involved in Neuronal Polarization and Growth: Lipid Rafts and Phosphorylation. Front Mol Neurosci 2020; 13:150. [PMID: 32922262 PMCID: PMC7456915 DOI: 10.3389/fnmol.2020.00150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/25/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Neuronal polarization and growth are developmental processes that occur during neuronal cell differentiation. The molecular signaling mechanisms involved in these events in in vivo mammalian brain remain unclear. Also, cellular events of the neuronal polarization process within a given neuron are thought to be constituted of many independent intracellular signal transduction pathways (the "tug-of-war" model). However, in vivo results suggest that such pathways should be cooperative with one another among a given group of neurons in a region of the brain. Lipid rafts, specific membrane domains with low fluidity, are candidates for the hotspots of such intracellular signaling. Among the signals reported to be involved in polarization, a number are thought to be present or translocated to the lipid rafts in response to extracellular signals. As part of our analysis, we discuss how such novel molecular mechanisms are combined for effective regulation of neuronal polarization and growth, focusing on the significance of the lipid rafts, including results based on recently introduced methods.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| |
Collapse
|
26
|
Casamento A, Boucrot E. Molecular mechanism of Fast Endophilin-Mediated Endocytosis. Biochem J 2020; 477:2327-2345. [PMID: 32589750 PMCID: PMC7319585 DOI: 10.1042/bcj20190342] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/10/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Endocytosis mediates the cellular uptake of micronutrients and cell surface proteins. Clathrin-mediated endocytosis (CME) is the housekeeping pathway in resting cells but additional Clathrin-independent endocytic (CIE) routes, including Fast Endophilin-Mediated Endocytosis (FEME), internalize specific cargoes and support diverse cellular functions. FEME is part of the Dynamin-dependent subgroup of CIE pathways. Here, we review our current understanding of the molecular mechanism of FEME. Key steps are: (i) priming, (ii) cargo selection, (iii) membrane curvature and carrier formation, (iv) membrane scission and (v) cytosolic transport. All steps are controlled by regulatory mechanisms mediated by phosphoinositides and by kinases such as Src, LRRK2, Cdk5 and GSK3β. A key feature of FEME is that it is not constitutively active but triggered upon the stimulation of selected cell surface receptors by their ligands. In resting cells, there is a priming cycle that concentrates Endophilin into clusters on discrete locations of the plasma membrane. In the absence of receptor activation, the patches quickly abort and new cycles are initiated nearby, constantly priming the plasma membrane for FEME. Upon activation, receptors are swiftly sorted into pre-existing Endophilin clusters, which then bud to form FEME carriers within 10 s. We summarize the hallmarks of FEME and the techniques and assays required to identify it. Next, we review similarities and differences with other CIE pathways and proposed cargoes that may use FEME to enter cells. Finally, we submit pending questions and future milestones and discuss the exciting perspectives that targeting FEME may boost treatments against cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandra Casamento
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
- Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, U.K
| |
Collapse
|
27
|
Gallop J. Filopodia and their links with membrane traffic and cell adhesion. Semin Cell Dev Biol 2020; 102:81-89. [DOI: 10.1016/j.semcdb.2019.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/13/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 01/24/2023]
|
28
|
Phosphoproteomic and bioinformatic methods for analyzing signaling in vertebrate axon growth and regeneration. J Neurosci Methods 2020; 339:108723. [DOI: 10.1016/j.jneumeth.2020.108723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
|
29
|
Durso W, Martins M, Marchetti L, Cremisi F, Luin S, Cardarelli F. Lysosome Dynamic Properties during Neuronal Stem Cell Differentiation Studied by Spatiotemporal Fluctuation Spectroscopy and Organelle Tracking. Int J Mol Sci 2020; 21:ijms21093397. [PMID: 32403391 PMCID: PMC7247004 DOI: 10.3390/ijms21093397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 01/20/2023] Open
Abstract
We investigated lysosome dynamics during neuronal stem cell (NSC) differentiation by two quantitative and complementary biophysical methods based on fluorescence: imaging-derived mean square displacement (iMSD) and single-particle tracking (SPT). The former extracts the average dynamics and size of the whole population of moving lysosomes directly from imaging, with no need to calculate single trajectories; the latter resolves the finest heterogeneities and dynamic features at the single-lysosome level, which are lost in the iMSD analysis. In brief, iMSD analysis reveals that, from a structural point of view, lysosomes decrement in size during NSC differentiation, from 1 μm average diameter in the embryonic cells to approximately 500 nm diameter in the fully differentiated cells. Concomitantly, iMSD analysis highlights modification of key dynamic parameters, such as the average local organelle diffusivity and anomalous coefficient, which may parallel cytoskeleton remodeling during the differentiation process. From average to local, SPT allows mapping heterogeneous dynamic responses of single lysosomes in different districts of the cells. For instance, a dramatic decrease of lysosomal transport in the soma is followed by a rapid increase of transport in the projections at specific time points during neuronal differentiation, an observation compatible with the hypothesis that lysosomal active mobilization shifts from the soma to the newborn projections. Our combined results provide new insight into the lysosome size and dynamics regulation throughout NSC differentiation, supporting new functions proposed for this organelle.
Collapse
Affiliation(s)
- William Durso
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Manuella Martins
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Laura Marchetti
- Center for Nanotechnology Innovation@NEST (CNI@NEST), Piazza San Silvestro 12, 56126 Pisa, Italy;
| | - Federico Cremisi
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Stefano Luin
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- NEST, Istituto Nanoscienze, CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
- Correspondence: (S.L.); (F.C.)
| | - Francesco Cardarelli
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- Correspondence: (S.L.); (F.C.)
| |
Collapse
|
30
|
Badawi Y, Nishimune H. Super-resolution microscopy for analyzing neuromuscular junctions and synapses. Neurosci Lett 2020; 715:134644. [PMID: 31765730 PMCID: PMC6937598 DOI: 10.1016/j.neulet.2019.134644] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/27/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
Super-resolution microscopy techniques offer subdiffraction limited resolution that is two- to ten-fold improved compared to that offered by conventional confocal microscopy. This breakthrough in resolution for light microscopy has contributed to new findings in neuroscience and synapse biology. This review will focus on the Structured Illumination Microscopy (SIM), Stimulated emission depletion (STED) microscopy, and Stochastic optical reconstruction microscopy (STORM) / Single molecule localization microscopy (SMLM) techniques and compare them for the better understanding of their differences and their suitability for the analysis of synapse biology. In addition, we will discuss a few practical aspects of these microscopic techniques, including resolution, image acquisition speed, multicolor capability, and other advantages and disadvantages. Tips for the improvement of microscopy will be introduced; for example, information resources for recommended dyes, the limitations of multicolor analysis, and capabilities for live imaging. In addition, we will summarize how super-resolution microscopy has been used for analyses of neuromuscular junctions and synapses.
Collapse
Affiliation(s)
- Yomna Badawi
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, 66160, USA
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, 66160, USA.
| |
Collapse
|
31
|
Ishikawa Y, Okada M, Honda A, Ito Y, Tamada A, Endo N, Igarashi M. Phosphorylation sites of microtubule-associated protein 1B (MAP 1B) are involved in axon growth and regeneration. Mol Brain 2019; 12:93. [PMID: 31711525 PMCID: PMC6849251 DOI: 10.1186/s13041-019-0510-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2019] [Accepted: 10/10/2019] [Indexed: 01/29/2023] Open
Abstract
The growth cone is a specialized structure that forms at the tip of extending axons in developing and regenerating neurons. This structure is essential for accurate synaptogenesis at developmental stages, and is also involved in plasticity-dependent synaptogenesis and axon regeneration in the mature brain. Thus, understanding the molecular mechanisms utilized by growth cones is indispensable to understanding neuronal network formation and rearrangement. Phosphorylation is the most important and commonly utilized protein modification in signal transduction. We previously identified microtubule-associated protein 1B (MAP 1B) as the most frequently phosphorylated protein among ~ 1200 phosphorylated proteins. MAP 1B has more than 10 phosphorylation sites that were present more than 50 times among these 1200 proteins. Here, we produced phospho-specific antibodies against phosphorylated serines at positions 25 and 1201 of MAP 1B that specifically recognize growing axons both in cultured neurons and in vivo in various regions of the embryonic brain. Following sciatic nerve injury, immunoreactivity with each antibody increased compared to the sham operated group. Experiments with transected and sutured nerves revealed that regenerating axons were specifically recognized by these antibodies. These results suggest that these MAP 1B phosphorylation sites are specifically involved in axon growth and that phospho-specific antibodies against MAP 1B are useful markers of growing/regenerating axons.
Collapse
Affiliation(s)
- Yuya Ishikawa
- Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata, Japan.,Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan
| | - Masayasu Okada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Trans-disciplinary Research Programs, Brain Research Institute, Niigata University, Niigata, Japan.,Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan
| | - Atsushi Tamada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.,Trans-disciplinary Research Programs, Brain Research Institute, Niigata University, Niigata, Japan.,Department of iPS Cell Applied Medicine, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Naoto Endo
- Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan. .,Trans-disciplinary Research Programs, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
32
|
New observations in neuroscience using superresolution microscopy. J Neurosci 2019; 38:9459-9467. [PMID: 30381437 DOI: 10.1523/jneurosci.1678-18.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/31/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 12/28/2022] Open
Abstract
Superresolution microscopy (SM) techniques are among the revolutionary methods for molecular and cellular observations in the 21st century. SM techniques overcome optical limitations, and several new observations using SM lead us to expect these techniques to have a large impact on neuroscience in the near future. Several types of SM have been developed, including structured illumination microscopy (SIM), stimulated emission depletion microscopy (STED), and photoactivated localization microscopy (PALM)/stochastic optical reconstruction microscopy (STORM), each with special features. In this Minisymposium, experts in these different types of SM discuss the new structural and functional information about specific important molecules in neuroscience that has been gained with SM. Using these techniques, we have revealed novel mechanisms of endocytosis in nerve growth, fusion pore dynamics, and described quantitative new properties of excitatory and inhibitory synapses. Additional powerful techniques, including single molecule-guided Bayesian localization SM (SIMBA) and expansion microscopy (ExM), alone or combined with super-resolution observation, are also introduced in this session.
Collapse
|
33
|
Chasing Uptake: Super-Resolution Microscopy in Endocytosis and Phagocytosis. Trends Cell Biol 2019; 29:727-739. [PMID: 31227311 DOI: 10.1016/j.tcb.2019.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 11/21/2022]
Abstract
Since their invention about two decades ago, super-resolution microscopes have become a method of choice in cell biology. Owing to a spatial resolution below 50 nm, smaller than the size of most organelles, and an order of magnitude better than the diffraction limit of conventional light microscopes, super-resolution microscopy is a powerful technique for resolving intracellular trafficking. In this review we discuss discoveries in endocytosis and phagocytosis that have been made possible by super-resolution microscopy - from uptake at the plasma membrane, endocytic coat formation, and cytoskeletal rearrangements to endosomal maturation. The detailed visualization of the diverse molecular assemblies that mediate endocytic uptake will provide a better understanding of how cells ingest extracellular material.
Collapse
|
34
|
Tanaka M, Fujii Y, Hirano K, Higaki T, Nagasaki A, Ishikawa R, Okajima T, Katoh K. Fascin in lamellipodia contributes to cell elasticity by controlling the orientation of filamentous actin. Genes Cells 2019; 24:202-213. [PMID: 30664308 DOI: 10.1111/gtc.12671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 11/30/2022]
Abstract
Fascin, an actin-bundling protein, is present in the filopodia and lamellipodia of growth cones. However, few studies have examined lamellipodial fascin because it is difficult to observe. In this study, we evaluated lamellipodial fascin. We visualized the actin meshwork of lamellipodia in live growth cones by super-resolution microscopy. Fascin was colocalized with the actin meshwork in lamellipodia. Ser39 of fascin is a well-known phosphorylation site that controls the binding of fascin to actin filaments. Fluorescence recovery after photobleaching experiments with confocal microscopy showed that binding of fascin was controlled by phosphorylation of Ser39 in lamellipodia. Moreover, TPA, an agonist of protein kinase C, induced phosphorylation of fascin and dissociation from actin filaments in lamellipodia. Time series images showed that dissociation of fascin from the actin meshwork was induced by TPA. As fascin dissociated from actin filaments, the orientation of the actin filaments became parallel to the leading edge. The angle of actin filaments against the leading edge was changed from 73° to 15°. This decreased the elasticity of the lamellipodia by 40%, as measured by atomic force microscopy. These data suggest that actin bundles made by fascin contribute to elasticity of the growth cone.
Collapse
Affiliation(s)
- Minami Tanaka
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yuki Fujii
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Kazumi Hirano
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takumi Higaki
- International Research Organization for Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Akira Nagasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Ryoki Ishikawa
- School of Nursing, Gunma Prefectural College of Health Sciences, Maebashi, Japan
| | - Takaharu Okajima
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
35
|
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] [Academic Contribution 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
|
36
|
A cell surface protein controls endocrine ring gland morphogenesis and steroid production. Dev Biol 2018; 445:16-28. [PMID: 30367846 DOI: 10.1016/j.ydbio.2018.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/10/2018] [Revised: 10/09/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
Identification of signals for systemic adaption of hormonal regulation would help to understand the crosstalk between cells and environmental cues contributing to growth, metabolic homeostasis and development. Physiological states are controlled by precise pulsatile hormonal release, including endocrine steroids in human and ecdysteroids in insects. We show in Drosophila that regulation of genes that control biosynthesis and signaling of the steroid hormone ecdysone, a central regulator of developmental progress, depends on the extracellular matrix protein Obstructor-A (Obst-A). Ecdysone is produced by the prothoracic gland (PG), where sensory neurons projecting axons from the brain integrate stimuli for endocrine control. By defining the extracellular surface, Obst-A promotes morphogenesis and axonal growth in the PG. This process requires Obst-A-matrix reorganization by Clathrin/Wurst-mediated endocytosis. Our data identifies the extracellular matrix as essential for endocrine ring gland function, which coordinates physiology, axon morphogenesis, and developmental programs. As Obst-A and Wurst homologs are found among all arthropods, we propose that this mechanism is evolutionary conserved.
Collapse
|
37
|
Vesicular movements in the growth cone. Neurochem Int 2018; 119:71-76. [DOI: 10.1016/j.neuint.2017.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2017] [Revised: 08/29/2017] [Accepted: 09/24/2017] [Indexed: 01/03/2023]
|
38
|
Lee S, Kim H, Higuchi H. Numerical method for vesicle movement analysis in a complex cytoskeleton network. OPTICS EXPRESS 2018; 26:16236-16249. [PMID: 30119458 DOI: 10.1364/oe.26.016236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/13/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
The detection of the precise movement of a vesicle during transport in a live cell provides key information for the intracellular delivery process. Here we report a novel numerical method for analyzing three-dimensional vesicle movement. Since the vesicle moves along a linear cytoskeleton during the active transport, our method first detects the orientation and position of the cytoskeleton as a linear section based on angle correlation and linear regression, after noise reduction. Then, the precise vesicle movement is calculated using vector analysis, in terms of rotation angle and translational displacement. Using this method, various vesicle trajectories obtained via high spatiotemporal resolution microscopy can be understood..
Collapse
|
39
|
Kawasaki A, Okada M, Tamada A, Okuda S, Nozumi M, Ito Y, Kobayashi D, Yamasaki T, Yokoyama R, Shibata T, Nishina H, Yoshida Y, Fujii Y, Takeuchi K, Igarashi M. Growth Cone Phosphoproteomics Reveals that GAP-43 Phosphorylated by JNK Is a Marker of Axon Growth and Regeneration. iScience 2018; 4:190-203. [PMID: 30240740 PMCID: PMC6147025 DOI: 10.1016/j.isci.2018.05.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2018] [Revised: 05/05/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022] Open
Abstract
Neuronal growth cones are essential for nerve growth and regeneration, as well as for the formation and rearrangement of the neural network. To elucidate phosphorylation-dependent signaling pathways and establish useful molecular markers for axon growth and regeneration, we performed a phosphoproteomics study of mammalian growth cones, which identified >30,000 phosphopeptides of ∼1,200 proteins. The phosphorylation sites were highly proline directed and primarily MAPK dependent, owing to the activation of JNK, suggesting that proteins that undergo proline-directed phosphorylation mediate nerve growth in the mammalian brain. Bioinformatics analysis revealed that phosphoproteins were enriched in microtubules and the cortical cytoskeleton. The most frequently phosphorylated site was S96 of GAP-43 (growth-associated protein 43-kDa), a vertebrate-specific protein involved in axon growth. This previously uncharacterized phosphorylation site was JNK dependent. S96 phosphorylation was specifically detected in growing and regenerating axons as the most frequent target of JNK signaling; thus it represents a promising new molecular marker for mammalian axonal growth and regeneration. Phosphoproteomics of mammalian growth cone membranes reveals activation of MAPK JNK is the activated MAPK in growth cones and phosphorylates S96 of GAP-43 pS96 of GAP-43, the most frequent site, is observed in growing axons pS96 is biochemically detected in the regenerating axons of the peripheral nerves
Collapse
Affiliation(s)
- Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Masayasu Okada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Department of Neurosurgery, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Atsushi Tamada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Shujiro Okuda
- Laboratory of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Daiki Kobayashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Tokiwa Yamasaki
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryo Yokoyama
- K.K. Sciex Japan, Shinagawa-ku, Tokyo 140-0001, Japan
| | | | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yutaka Yoshida
- Center for Coordination of Research, Institute for Research Promotion, Niigata University, Ikarashi, Niigata 951-2181, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Kosei Takeuchi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Department of Medical Cell Biology, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
40
|
Ito Y, Honda A, Igarashi M. Glycoprotein M6a as a signaling transducer in neuronal lipid rafts. Neurosci Res 2018; 128:19-24. [DOI: 10.1016/j.neures.2017.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
41
|
Chen X, Chen C, Hao J, Zhang J, Zhang F. Effect of CLIP3 Upregulation on Astrocyte Proliferation and Subsequent Glial Scar Formation in the Rat Spinal Cord via STAT3 Pathway After Injury. J Mol Neurosci 2017; 64:117-128. [PMID: 29218499 DOI: 10.1007/s12031-017-0998-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/14/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Spinal cord injury (SCI) is a devastating event resulting in neuron degeneration and permanent paralysis through inflammatory cytokine overproduction and glial scar formation. Presently, the endogenous molecular mechanisms coordinating glial scar formation in the injured spinal cord remain elusive. Signal transducer and activator of transcription 3 (STAT3) is a well-known transcription factor particularly involving in cell proliferation and inflammation in the lesion site following SCI. Meanwhile, CAP-Gly domain containing linker protein 3(CLIP3), a vital cytoplasmic protein, has been confirmed to providing an optimal conduit for intracellular signal transduction and interacting with STAT3 with mass spectrometry analysis. In this study, we aimed to identify the expression of CLIP3 in the spinal cord as well as its role in mediating astrocyte activation and glial scar formation after SCI by establishing an acute traumatic SCI model in male adult rats. Western blot analysis revealed that CLIP3 increased gradually after injury, reached a peak at day 3. The immunohistochemistry staining showed the same result in white matter. With double immunofluorescence staining, we found that CLIP3 was expressed in glial cells and significant changes of CLIP3 expression occurred in astrocytes during the pathological process. Statistical analysis demonstrated there was a correlation between the number of positive cells stained by CLIP3 and STAT3 in the spinal cord after SCI. Co-immunoprecipitation further indicated that CLIP3 interacted with STAT3 in the injured spinal cord. Taken together, our study clearly suggested that CLIP3 played an essential role in astrocyte activation, associating with the STAT3 pathway activation induced by SCI.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China
| | - Cheng Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Medical Colleges of Nantong University, Nantong, Jiangsu, 226001, China
| | - Jie Hao
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China
| | - Jiyun Zhang
- Medical Colleges of Nantong University, Nantong, Jiangsu, 226001, China.,Department of Radiology, Third Municipal People's Hospital, Nantong, Jiangsu, 226001, China
| | - Feng Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China. .,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
42
|
|
43
|
Honda A, Usui H, Sakimura K, Igarashi M. Rufy3 is an adapter protein for small GTPases that activates a Rac guanine nucleotide exchange factor to control neuronal polarity. J Biol Chem 2017; 292:20936-20946. [PMID: 29089386 DOI: 10.1074/jbc.m117.809541] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2017] [Revised: 10/25/2017] [Indexed: 01/01/2023] Open
Abstract
RUN and FYVE domain-containing 3 (Rufy3) is an adapter protein for small GTPase proteins and is bound to activated Rap2, a Ras family protein in the developing neuron. Previously, we reported the presence of a rapid cell polarity determination mechanism involving Rufy3, which is likely required for in vivo neuronal development. However, the molecular details of this mechanism are unclear. To this end, here we produced Rufy3 knock-out (Rufy3-KO) mice to study the role of Rufy3 in more detail. Examining Rufy3-KO neurons, we found that Rufy3 is recruited via glycoprotein M6A to detergent-resistant membrane domains, which are biochemically similar to lipid rafts. We also clarified that Rufy3, as a component of a ternary complex, induces the assembly of Rap2 in the axonal growth cone, whereas in the absence of Rufy3, the accumulation of a Rac guanine nucleotide exchange factor, T-cell lymphoma invasion and metastasis 2 (Tiam2/STEF), is inhibited downstream of Rap2. We also found that Rufy3 regulates the cellular localization of Rap2 and Tiam2/STEF. Taken together, we conclude that Rufy3 is a physiological adapter for Rap2 and activates Tiam2/STEF in glycoprotein M6A-regulated neuronal polarity and axon growth.
Collapse
Affiliation(s)
- Atsuko Honda
- From the Department of Neurochemistry and Molecular Cell Biology.,Trans-disciplinary Research Program, and
| | - Hiroshi Usui
- Department of Cellular Neurobiology, Institute for Brain Research, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Institute for Brain Research, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Michihiro Igarashi
- From the Department of Neurochemistry and Molecular Cell Biology, .,Trans-disciplinary Research Program, and
| |
Collapse
|
44
|
Extracellular Signals Induce Glycoprotein M6a Clustering of Lipid Rafts and Associated Signaling Molecules. J Neurosci 2017; 37:4046-4064. [PMID: 28275160 DOI: 10.1523/jneurosci.3319-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2016] [Revised: 01/25/2017] [Accepted: 02/18/2017] [Indexed: 01/08/2023] Open
Abstract
Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. To examine how signaling protein complexes are clustered in rafts, we focused on the functions of glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing mouse neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a GPM6a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a, such as Rufy3, Rap2, and Tiam2/STEF, accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation in neuronal development. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of neuronal polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.SIGNIFICANCE STATEMENT Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. We focused on glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.
Collapse
|