1
|
Xia L, Stoika R, Li Y, Zheng Y, Liu Y, Li D, Liu K, Zhang X, Shang X, Jin M. 2,3,4-Trihydroxybenzophenone-induced cardiac and neurological toxicity: Heart-brain interaction mediated by regulation of pgam1a and pgk1 involved in glycolysis and gluconeogenesis in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 974:179212. [PMID: 40157088 DOI: 10.1016/j.scitotenv.2025.179212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
2,3,4-trihydroxybenzophenone (2,3,4-THBP) is a benzophenone-type UV filter commonly used in sunscreens. However, the widespread application of BP-UV filters has led to an appearance of this chemical in the environment and living organisms. Despite of this, there is poor understanding of the bio-toxicity of 2,3,4-THBP. Here, we investigated the adverse effects of 2,3,4-THBP in varying doses (115, 230, 460, 920, and 1840 μg/L) in zebrafish experimental model. Specifically, we assessed its impact on the cardio- and neuro-development, including pericardiac area, heart rate, as well as brain vessels and differentiation of dopaminergic and central nervous system (CNS) neurons. The expression of genes whose products are involved in cardio- and neuro-development was also monitored. It was found that 2,3,4-THBP caused heart failure (HF)-like symptoms in zebrafish embryos including pericardial edema, reduced heart rate, and yolk sac malformation. It also induced dramatic neurotoxicity, namely defective neuron differentiation, cerebrovascular loss, cognition and behavior defects. It disrupted the vascular system, leading to potentially toxic interactions between the heart and brain, further worsening the state of both organs. Notably, RNA-seq findings indicated that 2,3,4-THBP damaged the energy metabolic function via upregulating the expression of phosphoglycerate mutase 1a (pgam1a) and phosphoglycerate kinase 1 (pgk1) whose protein products are involved in regulation of glycolysis and gluconeogenesis, highlighting their role in the interplay between heart and brain. Summarizing, 2,3,4-THBP triggered cardiac and neurological toxicity, which is possibly associated with heart-brain interaction mediated by regulation of pgam1a and pgk1 involved in glycolysis and gluconeogenesis.
Collapse
Affiliation(s)
- Lijie Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, Tangshan 063210, Hebei Province, People's Republic of China
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
| | - Yuqing Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Jinan, 250103, Shandong Province, People's Republic of China
| | - Yuanteng Zheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, Tangshan 063210, Hebei Province, People's Republic of China
| | - Yanao Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Jinan, 250103, Shandong Province, People's Republic of China
| | - Dong Li
- R&D Department, Jinan Perfect Biological Technology Co., Ltd., Jinan 250101, Shandong Province, People's Republic of China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Jinan, 250103, Shandong Province, People's Republic of China
| | - Xiujun Zhang
- School of Psychology and Mental Health, North China University of Science and Technology, Tangshan 063210, Hebei Province, People's Republic of China
| | - Xueliang Shang
- School of Psychology and Mental Health, North China University of Science and Technology, Tangshan 063210, Hebei Province, People's Republic of China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Jinan, 250103, Shandong Province, People's Republic of China.
| |
Collapse
|
2
|
Micovic K, Canuel A, Remtulla A, Chuyen A, Byrsan M, McGarry DJ, Olson MF. Mical1 deletion in tyrosinase expressing cells affects mouse running gaits. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70004. [PMID: 39344934 PMCID: PMC11440367 DOI: 10.1111/gbb.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
Neuronal development is a highly regulated process that is dependent on the correct coordination of cellular responses to extracellular cues. In response to semaphorin axon guidance proteins, the MICAL1 protein is stimulated to produce reactive oxygen species that oxidize actin on specific methionine residues, leading to filamentous actin depolymerization and consequent changes in neuronal growth cone dynamics. Crossing genetically modified mice homozygous for floxed Mical1 (Mical1fl/fl) alleles with transgenic mice expressing Cre recombinase under the control of a tyrosinase gene enhancer/promoter (Tyr::Cre) enabled conditional Mical1 deletion. Immunohistochemical analysis showed Mical1 expression in the cerebellum, which plays a prominent role in the coordination of motor movements, with reduced Mical1 expression in Mical1fl/fl mice co-expressing Tyr::Cre. Analysis of the gaits of mice running on a treadmill showed that both male and female Mical1fl/fl, Tyr::Cre mutant mice had significant alterations to their striding patterns relative to wild-type mice, although the specific aspects of their altered gaits differed between the sexes. Additional motor tests that involved movement on a rotating rod, descending a vertical pole, or crossing a balance beam did not show significant differences between the genotypes, suggesting that the effect of the Mical1fl/fl, Tyr::Cre genetic modifications was only manifested during specific highly coordinated movements that contribute to running. These findings indicate that there is a behavioral consequence in Mical1fl/fl, Tyr::Cre mutant mice that affects motor control as manifested by alterations in their gait.
Collapse
Affiliation(s)
- Katarina Micovic
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Alicia Canuel
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Aasiya Remtulla
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Alexandre Chuyen
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Margarita Byrsan
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| | - David J. McGarry
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Michael F. Olson
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
3
|
Powers RM, Hevner RF, Halpain S. The Neuron Navigators: Structure, function, and evolutionary history. Front Mol Neurosci 2023; 15:1099554. [PMID: 36710926 PMCID: PMC9877351 DOI: 10.3389/fnmol.2022.1099554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,Department of Pathology, UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,*Correspondence: Shelley Halpain, ✉
| |
Collapse
|
4
|
Xue W, Shi W, Kong Y, Kuss M, Duan B. Anisotropic scaffolds for peripheral nerve and spinal cord regeneration. Bioact Mater 2021; 6:4141-4160. [PMID: 33997498 PMCID: PMC8099454 DOI: 10.1016/j.bioactmat.2021.04.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/05/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
The treatment of long-gap (>10 mm) peripheral nerve injury (PNI) and spinal cord injury (SCI) remains a continuous challenge due to limited native tissue regeneration capabilities. The current clinical strategy of using autografts for PNI suffers from a source shortage, while the pharmacological treatment for SCI presents dissatisfactory results. Tissue engineering, as an alternative, is a promising approach for regenerating peripheral nerves and spinal cords. Through providing a beneficial environment, a scaffold is the primary element in tissue engineering. In particular, scaffolds with anisotropic structures resembling the native extracellular matrix (ECM) can effectively guide neural outgrowth and reconnection. In this review, the anatomy of peripheral nerves and spinal cords, as well as current clinical treatments for PNI and SCI, is first summarized. An overview of the critical components in peripheral nerve and spinal cord tissue engineering and the current status of regeneration approaches are also discussed. Recent advances in the fabrication of anisotropic surface patterns, aligned fibrous substrates, and 3D hydrogel scaffolds, as well as their in vitro and in vivo effects are highlighted. Finally, we summarize potential mechanisms underlying the anisotropic architectures in orienting axonal and glial cell growth, along with their challenges and prospects.
Collapse
Affiliation(s)
- Wen Xue
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Mechanical Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
5
|
Doblado LR, Martínez-Ramos C, García-Verdugo JM, Moreno-Manzano V, Pradas MM. Engineered axon tracts within tubular biohybrid scaffolds. J Neural Eng 2021; 18. [PMID: 34311448 DOI: 10.1088/1741-2552/ac17d8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022]
Abstract
Injuries to the nervous system that involve the disruption of axonal pathways are devastating to the individual and require specific tissue engineering strategies. Here we analyse a cells-biomaterials strategy to overcome the obstacles limiting axon regenerationin vivo, based on the combination of a hyaluronic acid (HA) single-channel tubular conduit filled with poly-L-lactide acid (PLA) fibres in its lumen, with pre-cultured Schwann cells (SCs) as cells supportive of axon extension. The HA conduit and PLA fibres sustain the proliferation of SC, which enhance axon growth acting as a feeder layer and growth factor pumps. The parallel unidirectional ensemble formed by PLA fibres and SC tries to recapitulate the directional features of axonal pathways in the nervous system. A dorsal root ganglion (DRG) explant is planted on one of the conduit's ends to follow axon outgrowth from the DRG. After a 21 d co-culture of the DRG + SC-seeded conduit ensemble, we analyse the axonal extension throughout the conduit by scanning, transmission electronic and confocal microscopy, in order to study the features of SC and the grown axons and their association. The separate effects of SC and PLA fibres on the axon growth are also experimentally addressed. The biohybrid thus produced may be considered a synthetic axonal pathway, and the results could be of use in strategies for the regeneration of axonal tracts.
Collapse
Affiliation(s)
- Laura Rodríguez Doblado
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain.,Department of Medicine, Universitat Jaume I, Av. Vicent-Sos Baynat s/n, Castellón 12071, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universitat de València, CIBERNED, Valencia, Spain
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, Valencia, Spain.,Universidad Católica de Valencia, Valencia, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| |
Collapse
|
6
|
Agrawal M, Welshhans K. Local Translation Across Neural Development: A Focus on Radial Glial Cells, Axons, and Synaptogenesis. Front Mol Neurosci 2021; 14:717170. [PMID: 34434089 PMCID: PMC8380849 DOI: 10.3389/fnmol.2021.717170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
In the past two decades, significant progress has been made in our understanding of mRNA localization and translation at distal sites in axons and dendrites. The existing literature shows that local translation is regulated in a temporally and spatially restricted manner and is critical throughout embryonic and post-embryonic life. Here, recent key findings about mRNA localization and local translation across the various stages of neural development, including neurogenesis, axon development, and synaptogenesis, are reviewed. In the early stages of development, mRNAs are localized and locally translated in the endfeet of radial glial cells, but much is still unexplored about their functional significance. Recent in vitro and in vivo studies have provided new information about the specific mechanisms regulating local translation during axon development, including growth cone guidance and axon branching. Later in development, localization and translation of mRNAs help mediate the major structural and functional changes that occur in the axon during synaptogenesis. Clinically, changes in local translation across all stages of neural development have important implications for understanding the etiology of several neurological disorders. Herein, local translation and mechanisms regulating this process across developmental stages are compared and discussed in the context of function and dysfunction.
Collapse
Affiliation(s)
- Manasi Agrawal
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Kristy Welshhans
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
7
|
Chen S, Liu A, Wu C, Chen Y, Liu C, Zhang Y, Wu K, Wei D, Sun J, Zhou L, Fan H. Static-Dynamic Profited Viscoelastic Hydrogels for Motor-Clutch-Regulated Neurogenesis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:24463-24476. [PMID: 34024102 DOI: 10.1021/acsami.1c03821] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Viscoelasticity, a time-scale mechanical feature of the native extracellular matrix (ECM), is reported to play crucial roles in plentiful cellular behaviors, whereas its effects on neuronal behavior and the underlying molecular mechanism still remain obscure. Challenges are faced in the biocompatible synthesis of neural ECM-mimicked scaffolds solely controlled with viscoelasticity and due to the lack of suitable models for neurons-viscoelastic matrix interaction. Herein, we report difunctional hyaluronan-collagen hydrogels prepared by a static-dynamic strategy. The hydrogels show aldehyde concentration-dependent viscoelasticity and similar initial elastic modulus, fibrillar morphology, swelling as well as degradability. Utilizing the resulting hydrogels, for the first time, we demonstrate matrix viscoelasticity-dependent neuronal responses, including neurite elongation and expression of neurogenic proteins. Then, a motor-clutch model modified with a tension dissipation component is developed to account for the molecular mechanism for viscoelasticity-sensitive neuronal responses. Moreover, we prove enhanced recovery of rat spinal cord injury by implanting cell-free viscoelastic grafts. As a pioneer finding on neurons-viscoelastic matrix interaction both in vitro and in vivo, this work provides intriguing insights not only into nerve repair but also into neuroscience and tissue engineering.
Collapse
Affiliation(s)
- Suping Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Amin Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Yaxing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Kai Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| |
Collapse
|
8
|
Braïni C, Bugnicourt G, Villard C. Neuronal growth from a volume perspective. Phys Biol 2021; 18:016007. [PMID: 33147573 DOI: 10.1088/1478-3975/abc79c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Microfluidic-based fluorescent exclusion method allows to tackle the issue of neuronal growth from a volume perspective. Based on this technology, we studied the two main actin-rich structures accompanying the early stages of neuron development, i.e. growth cones, located at the tip of growing neuronal processes, and propagative actin waves. Our work reveals that growth cones tend to loose volume during their forward motion, as do actin waves during their journey from the cell body to the tip of neuronal processes, before the total transfer of their remaining volume to the growth cone. Actin waves seem thus to supply material to increasingly distant growth cones as neurons develop. In addition, our work may suggest the existence of a membrane recycling phenomena associated to actin waves as a pulsatile anterograde source of material and by a continuous retrograde transport.
Collapse
Affiliation(s)
- Céline Braïni
- Physico-Chimie Curie, CNRS UMR 168, Université PSL, Sorbonne Université, Paris, France
| | | | | |
Collapse
|
9
|
Pollitt SL, Myers KR, Yoo J, Zheng JQ. LIM and SH3 protein 1 localizes to the leading edge of protruding lamellipodia and regulates axon development. Mol Biol Cell 2020; 31:2718-2732. [PMID: 32997597 PMCID: PMC7927181 DOI: 10.1091/mbc.e20-06-0366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The actin cytoskeleton drives cell motility and is essential for neuronal development and function. LIM and SH3 protein 1 (LASP1) is a unique actin-binding protein that is expressed in a wide range of cells including neurons, but its roles in cellular motility and neuronal development are not well understood. We report that LASP1 is expressed in rat hippocampus early in development, and this expression is maintained through adulthood. High-resolution imaging reveals that LASP1 is selectively concentrated at the leading edge of lamellipodia in migrating cells and axonal growth cones. This local enrichment of LASP1 is dynamically associated with the protrusive activity of lamellipodia, depends on the barbed ends of actin filaments, and requires both the LIM domain and the nebulin repeats of LASP1. Knockdown of LASP1 in cultured rat hippocampal neurons results in a substantial reduction in axonal outgrowth and arborization. Finally, loss of the Drosophila homologue Lasp from a subset of commissural neurons in the developing ventral nerve cord produces defasciculated axon bundles that do not reach their targets. Together, our data support a novel role for LASP1 in actin-based lamellipodial protrusion and establish LASP1 as a positive regulator of both in vitro and in vivo axon development.
Collapse
Affiliation(s)
| | | | - Jin Yoo
- Emory College, Emory University, Atlanta, GA 30322
| | - James Q Zheng
- Department of Cell Biology and.,Department of Neurology and Center for Neurodegenerative Diseases, Emory University School of Medicine, and
| |
Collapse
|
10
|
Glasgow SD, Ruthazer ES, Kennedy TE. Guiding synaptic plasticity: Novel roles for netrin-1 in synaptic plasticity and memory formation in the adult brain. J Physiol 2020; 599:493-505. [PMID: 32017127 DOI: 10.1113/jp278704] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Adult neural plasticity engages mechanisms that change synapse structure and function, yet many of the underlying events bear a striking similarity to processes that occur during the initial establishment of neural circuits during development. It is a long-standing hypothesis that the molecular mechanisms critical for neural development may also regulate synaptic plasticity related to learning and memory in adults. Netrins were initially described as chemoattractant guidance cues that direct cell and axon migration during embryonic development, yet they continue to be expressed by neurons in the adult brain. Recent findings have identified roles for netrin-1 in synaptogenesis during postnatal maturation, and in synaptic plasticity in the adult mammalian brain, regulating AMPA glutamate receptor trafficking at excitatory synapses. These findings provide an example of a conserved developmental guidance cue that is expressed by neurons in the adult brain and functions as a key regulator of activity-dependent synaptic plasticity. Notably, in humans, genetic polymorphisms in netrin-1 and its receptors have been linked to neurodevelopmental and neurodegenerative disorders. The molecular mechanisms associated with the synaptic function of netrin-1 therefore present new therapeutic targets for neuropathologies associated with memory dysfunction. Here, we summarize recent findings that link netrin-1 signalling to synaptic plasticity, and discuss the implications of these discoveries for the neurobiological basis of memory consolidation.
Collapse
Affiliation(s)
- Stephen D Glasgow
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Edward S Ruthazer
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, H3A 0C7, Canada
| |
Collapse
|
11
|
Tao T, Sun J, Peng Y, Li Y, Wang P, Chen X, Zhao W, Zheng YY, Wei L, Wang W, Zhou Y, Liu J, Shi YS, Zhu MS. Golgi-resident TRIO regulates membrane trafficking during neurite outgrowth. J Biol Chem 2019; 294:10954-10968. [PMID: 31152060 PMCID: PMC6635450 DOI: 10.1074/jbc.ra118.007318] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/21/2019] [Indexed: 11/06/2022] Open
Abstract
Neurite outgrowth requires coordinated cytoskeletal rearrangements in the growth cone and directional membrane delivery from the neuronal soma. As an essential Rho guanine nucleotide exchange factor (GEF), TRIO is necessary for cytoskeletal dynamics during neurite outgrowth, but its participation in the membrane delivery is unclear. Using co-localization studies, live-cell imaging, and fluorescence recovery after photobleaching analysis, along with neurite outgrowth assay and various biochemical approaches, we here report that in mouse cerebellar granule neurons, TRIO protein pools at the Golgi and regulates membrane trafficking by controlling the directional maintenance of both RAB8 (member RAS oncogene family 8)- and RAB10-positive membrane vesicles. We found that the spectrin repeats in Golgi-resident TRIO confer RAB8 and RAB10 activation by interacting with and activating the RAB GEF RABIN8. Constitutively active RAB8 or RAB10 could partially restore the neurite outgrowth of TRIO-deficient cerebellar granule neurons, suggesting that TRIO-regulated membrane trafficking has an important functional role in neurite outgrowth. Our results also suggest cross-talk between Rho GEF and Rab GEF in controlling both cytoskeletal dynamics and membrane trafficking during neuronal development. They further highlight how protein pools localized to specific organelles regulate crucial cellular activities and functions. In conclusion, our findings indicate that TRIO regulates membrane trafficking during neurite outgrowth in coordination with its GEF-dependent function in controlling cytoskeletal dynamics via Rho GTPases.
Collapse
Affiliation(s)
- Tao Tao
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Jie Sun
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yajing Peng
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Yeqiong Li
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Pei Wang
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Xin Chen
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Wei Zhao
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yan-Yan Zheng
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Lisha Wei
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Wei Wang
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yuwei Zhou
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Jianghuai Liu
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yun Stone Shi
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Min-Sheng Zhu
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and.
| |
Collapse
|
12
|
Shao Q, Yang T, Huang H, Majumder T, Khot BA, Khouzani MM, Alarmanazi F, Gore YK, Liu G. Disease-associated mutations in human TUBB3 disturb netrin repulsive signaling. PLoS One 2019; 14:e0218811. [PMID: 31226147 PMCID: PMC6588280 DOI: 10.1371/journal.pone.0218811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/10/2019] [Indexed: 11/28/2022] Open
Abstract
Missense mutations in the human TUBB3 gene cause a variety of neurological disorders associated with defects in axon guidance and neuronal migration, but the underlying molecular mechanisms are not well understood. Recent studies have shown that direct coupling of dynamic TUBB3 in microtubules with netrin receptors is required for netrin-1-mediated axon guidance, and the interaction of netrin-1 repulsive receptor UNC5C with TUBB3 is involved in netrin-1 mediated axonal repulsion. Here, we report that TUBB3 mutations perturb netrin-1/UNC5C repulsive signaling in the developing nervous system. Among twelve mutants reported in previous studies, five of them show significantly reduced interaction with UNC5C in comparison to the wild-type TUBB3. TUBB3 mutants R262C and R62Q exhibit decreased subcellular colocalization with UNC5C in the peripheral area of the growth cone of primary mouse neurons. Netrin-1 reduces the colocalization of UNC5C with wild-type TUBB3, but not TUBB3 mutants R262C or R62Q, in the growth cone. Results from the in vitro cosedimentation assay indicate that netrin-1 inhibits cosedimentation of UNC5C with polymerized microtubules in primary mouse neurons expressing the wild-type TUBB3, but not R262C or R62Q. Expression of either R262C or R62Q not only blocks netrin-1-induced growth cone collapse and axonal repulsion of primary EGL cells in vitro, but also results in axon projections defects of chicken dorsal root ganglion neurons in ovo. Our study reveals that human TUBB3 mutations specifically perturb netrin-1/UNC5C-mediated repulsion.
Collapse
Affiliation(s)
- Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Huai Huang
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Bhakti Ajit Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | | | - Farrah Alarmanazi
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Yasmin K. Gore
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|
13
|
Rink S, Bendella H, Akkin SM, Manthou M, Grosheva M, Angelov DN. Experimental Studies on Facial Nerve Regeneration. Anat Rec (Hoboken) 2019; 302:1287-1303. [DOI: 10.1002/ar.24123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/09/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral MedicineUniversity of Cologne Cologne Germany
| | - Habib Bendella
- Department of NeurosurgeryUniversity of Witten/Herdecke, Cologne Merheim Medical Center (CMMC) Cologne Germany
| | - Salih Murat Akkin
- Department of Anatomy, School of MedicineSANKO University Gaziantep Turkey
| | - Marilena Manthou
- Department of Histology and EmbryologyAristotle University Thessaloniki Thessaloniki Greece
| | - Maria Grosheva
- Department of Oto‐Rhino‐LaryngologyUniversity of Cologne Cologne Germany
| | | |
Collapse
|
14
|
Ye X, Qiu Y, Gao Y, Wan D, Zhu H. A Subtle Network Mediating Axon Guidance: Intrinsic Dynamic Structure of Growth Cone, Attractive and Repulsive Molecular Cues, and the Intermediate Role of Signaling Pathways. Neural Plast 2019; 2019:1719829. [PMID: 31097955 PMCID: PMC6487106 DOI: 10.1155/2019/1719829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023] Open
Abstract
A fundamental feature of both early nervous system development and axon regeneration is the guidance of axonal projections to their targets in order to assemble neural circuits that control behavior. In the navigation process where the nerves grow toward their targets, the growth cones, which locate at the tips of axons, sense the environment surrounding them, including varies of attractive or repulsive molecular cues, then make directional decisions to adjust their navigation journey. The turning ability of a growth cone largely depends on its highly dynamic skeleton, where actin filaments and microtubules play a very important role in its motility. In this review, we summarize some possible mechanisms underlying growth cone motility, relevant molecular cues, and signaling pathways in axon guidance of previous studies and discuss some questions regarding directions for further studies.
Collapse
Affiliation(s)
- Xiyue Ye
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yan Qiu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yuqing Gao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
15
|
Kiss A, Fischer I, Kleele T, Misgeld T, Propst F. Neuronal Growth Cone Size-Dependent and -Independent Parameters of Microtubule Polymerization. Front Cell Neurosci 2018; 12:195. [PMID: 30065631 PMCID: PMC6056669 DOI: 10.3389/fncel.2018.00195] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/17/2018] [Indexed: 01/16/2023] Open
Abstract
Migration and pathfinding of neuronal growth cones during neurite extension is critically dependent on dynamic microtubules. In this study we sought to determine, which aspects of microtubule polymerization relate to growth cone morphology and migratory characteristics. We conducted a multiscale quantitative microscopy analysis using automated tracking of microtubule plus ends in migrating growth cones of cultured murine dorsal root ganglion (DRG) neurons. Notably, this comprehensive analysis failed to identify any changes in microtubule polymerization parameters that were specifically associated with spontaneous extension vs. retraction of growth cones. This suggests that microtubule dynamicity is a basic mechanism that does not determine the polarity of growth cone response but can be exploited to accommodate diverse growth cone behaviors. At the same time, we found a correlation between growth cone size and basic parameters of microtubule polymerization including the density of growing microtubule plus ends and rate and duration of microtubule growth. A similar correlation was observed in growth cones of neurons lacking the microtubule-associated protein MAP1B. However, MAP1B-null growth cones, which are deficient in growth cone migration and steering, displayed an overall reduction in microtubule dynamicity. Our results highlight the importance of taking growth cone size into account when evaluating the influence on growth cone microtubule dynamics of different substrata, guidance factors or genetic manipulations which all can change growth cone morphology and size. The type of large scale multiparametric analysis performed here can help to separate direct effects that these perturbations might have on microtubule dynamics from indirect effects resulting from perturbation-induced changes in growth cone size.
Collapse
Affiliation(s)
- Alexa Kiss
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Irmgard Fischer
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Tatjana Kleele
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich Cluster for Systems Neurology (SyNergy) and German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich Cluster for Systems Neurology (SyNergy) and German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Friedrich Propst
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
16
|
Kim SM, Long DW, Tsang MWK, Wang Y. Zebrafish extracellular matrix improves neuronal viability and network formation in a 3-dimensional culture. Biomaterials 2018; 170:137-146. [PMID: 29665503 DOI: 10.1016/j.biomaterials.2018.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
Mammalian central nervous system (CNS) has limited capacity for regeneration. CNS injuries cause life-long debilitation and lead to $50 billion in healthcare costs in U.S. alone each year. Despite numerous efforts in the last few decades, CNS-related injuries remain as detrimental as they were 50 years ago. Some functional recovery can occur, but most regeneration are limited by an extracellular matrix (ECM) that actively inhibits axonal repair and promotes glial scarring. In most tissues, the ECM is an architectural foundation that plays an active role in supporting cellular development and regenerative response after injury. In mammalian CNS, however, this is not the case - its composition is not conducive for regeneration, with various molecules restricting plasticity and neuronal growth. In fact, the CNS ECM alters its composition dramatically following injury to restrict regeneration and to prioritize containment of injury as well as preservation of intact neural circuitry. This leads us to hypothesize that the inhibitory extracellular environment needs be modified or supplemented to be more regeneration-permissive for significant CNS regeneration. Mammalian nervous tissue cannot provide such ECM, and synthesizing it in a laboratory is beyond current technology. Evolutionarily lower species possess remarkably regenerative neural tissue. For example, small fresh-water dwelling zebrafish (Danio rerio) can regenerate severed spinal cord, re-gaining full motor function in a week. We believe their ECM contributes to its regenerative capability and that it can be harnessed to induce more regeneration in mammalian CNS. This study shows that ECM derived from zebrafish brains promotes more neuronal survival and axonal network formation than the widely studied and available ECM derived from mammalian tissues such as porcine brains, porcine urinary bladder, and rat brains. We believe its regenerative potential, combined with its affordability, easy handling, and fast reproduction, will make zebrafish an excellent candidate as a novel ECM source.
Collapse
Affiliation(s)
- Sung-Min Kim
- Department of Bioengineering, University of Pittsburgh, USA
| | | | | | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, USA.
| |
Collapse
|
17
|
Pearn ML, Schilling JM, Jian M, Egawa J, Wu C, Mandyam CD, Fannon-Pavlich MJ, Nguyen U, Bertoglio J, Kodama M, Mahata SK, DerMardirossian C, Lemkuil BP, Han R, Mobley WC, Patel HH, Patel PM, Head BP. Inhibition of RhoA reduces propofol-mediated growth cone collapse, axonal transport impairment, loss of synaptic connectivity, and behavioural deficits. Br J Anaesth 2018; 120:745-760. [PMID: 29576115 PMCID: PMC6200100 DOI: 10.1016/j.bja.2017.12.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/28/2017] [Accepted: 12/26/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Exposure of the developing brain to propofol results in cognitive deficits. Recent data suggest that inhibition of neuronal apoptosis does not prevent cognitive defects, suggesting mechanisms other than neuronal apoptosis play a role in anaesthetic neurotoxicity. Proper neuronal growth during development is dependent upon growth cone morphology and axonal transport. Propofol modulates actin dynamics in developing neurones, causes RhoA-dependent depolymerisation of actin, and reduces dendritic spines and synapses. We hypothesised that RhoA inhibition prevents synaptic loss and subsequent cognitive deficits. The present study tested whether RhoA inhibition with the botulinum toxin C3 (TAT-C3) prevents propofol-induced synapse and neurite loss, and preserves cognitive function. METHODS RhoA activation, growth cone morphology, and axonal transport were measured in neonatal rat neurones (5-7 days in vitro) exposed to propofol. Synapse counts (electron microscopy), dendritic arborisation (Golgi-Cox), and network connectivity were measured in mice (age 28 days) previously exposed to propofol at postnatal day 5-7. Memory was assessed in adult mice (age 3 months) previously exposed to propofol at postnatal day 5-7. RESULTS Propofol increased RhoA activation, collapsed growth cones, and impaired retrograde axonal transport of quantum dot-labelled brain-derived neurotrophic factor, all of which were prevented with TAT-C3. Adult mice previously treated with propofol had decreased numbers of total hippocampal synapses and presynaptic vesicles, reduced hippocampal dendritic arborisation, and infrapyramidal mossy fibres. These mice also exhibited decreased hippocampal-dependent contextual fear memory recall. All anatomical and behavioural changes were prevented with TAT-C3 pre-treatment. CONCLUSION Inhibition of RhoA prevents propofol-mediated hippocampal neurotoxicity and associated cognitive deficits.
Collapse
Affiliation(s)
- M L Pearn
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J M Schilling
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M Jian
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - J Egawa
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - C Wu
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - C D Mandyam
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M J Fannon-Pavlich
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - U Nguyen
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J Bertoglio
- INSERM U749, Institut Gustave Roussy, Universite Paris-sud, Paris, France
| | - M Kodama
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA; Department of Anesthesiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - S K Mahata
- Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA
| | - C DerMardirossian
- Department of Immunology and Microbial Sciences, TSRI, La Jolla, CA, USA; Department of Cell and Molecular Biology, TSRI, La Jolla, CA, USA
| | - B P Lemkuil
- Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - R Han
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - W C Mobley
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - H H Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - P M Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - B P Head
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA.
| |
Collapse
|
18
|
Huang H, Yang T, Shao Q, Majumder T, Mell K, Liu G. Human TUBB3 Mutations Disrupt Netrin Attractive Signaling. Neuroscience 2018; 374:155-171. [PMID: 29382549 DOI: 10.1016/j.neuroscience.2018.01.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 01/16/2018] [Accepted: 01/21/2018] [Indexed: 02/07/2023]
Abstract
Heterozygous missense mutations in human TUBB3 gene result in a spectrum of brain malformations associated with defects in axon guidance, neuronal migration and differentiation. However, the molecular mechanisms underlying mutation-related axon guidance abnormalities are unclear. Recent studies have shown that netrin-1, a canonical guidance cue, induced the interaction of TUBB3 with the netrin receptor deleted in colorectal cancer (DCC). Furthermore, TUBB3 is required for netrin-1-induced axon outgrowth, branching and pathfinding. Here, we provide evidence that TUBB3 mutations impair netrin/DCC signaling in the developing nervous system. The interaction of DCC with most TUBB3 mutants (eight out of twelve) is significantly reduced compared to the wild-type TUBB3. TUBB3 mutants R262C and A302V exhibit decreased subcellular colocalization with DCC in the growth cones of primary neurons. Netrin-1 increases the interaction of endogenous DCC with wild-type human TUBB3, but not R262C or A302V, in primary neurons. Netrin-1 also increases co-sedimentation of DCC with polymerized microtubules (MTs) in primary neurons expressing the wild-type TUBB3, but not R262C or A302V. Expression of either R262C or A302V not only suppresses netrin-1-induced neurite outgrowth, branching and attraction in vitro, but also causes defects in spinal cord commissural axon (CA) projection and pathfinding in ovo. Our study reveals that missense TUBB3 mutations specifically disrupt netrin/DCC-mediated attractive signaling.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Kristopher Mell
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft St., Toledo, OH 43606, USA.
| |
Collapse
|
19
|
Tanabe Y, Kamimura Y, Ueda M. Parallel signaling pathways regulate excitable dynamics differently for pseudopod formation in eukaryotic chemotaxis. J Cell Sci 2018; 131:jcs.214775. [DOI: 10.1242/jcs.214775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
In eukaryotic chemotaxis, parallel signaling pathways regulate the spatiotemporal pseudopod dynamics at the leading edge of a motile cell through characteristic dynamics of an excitable system; however, differences in the excitability and the physiological roles of individual pathways remain to be elucidated. Here we found that two different pathways, soluble guanylyl cyclase (sGC) and phosphatidylinositol 3-kinase (PI3K), exhibited similar all-or-none responses but different refractory periods by simultaneous observations of their excitable properties. Due to the shorter refractory period, sGC signaling responded more frequently to chemoattractants, leading to pseudopod formation with higher frequency. sGC excitability was regulated negatively by its product, cGMP, and cGMP-binding protein C (GbpC) through the suppression of F-actin polymerization, providing the underlying delayed negative feedback mechanism for the cyclical pseudopod formation. These results suggest that parallel pathways respond on different time-scales to environmental cues for chemotactic motility based on their intrinsic excitability. Key words: cGMP signaling, chemotaxis, excitability, pseudopod formation
Collapse
Affiliation(s)
- Yuki Tanabe
- Laboratory of Single Molecular Biology, Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, 560-0043, Japan
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research (BDR), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Yoichiro Kamimura
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research (BDR), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Masahiro Ueda
- Laboratory of Single Molecular Biology, Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, 560-0043, Japan
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research (BDR), RIKEN, Suita, Osaka, 565-0874, Japan
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
20
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
21
|
Spurlin JW, Nelson CM. Building branched tissue structures: from single cell guidance to coordinated construction. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2015.0527. [PMID: 28348257 DOI: 10.1098/rstb.2015.0527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
Branched networks are ubiquitous throughout nature, particularly found in tissues that require large surface area within a restricted volume. Many tissues with a branched architecture, such as the vasculature, kidney, mammary gland, lung and nervous system, function to exchange fluids, gases and information throughout the body of an organism. The generation of branched tissues requires regulation of branch site specification, initiation and elongation. Branching events often require the coordination of many cells to build a tissue network for material exchange. Recent evidence has emerged suggesting that cell cooperativity scales with the number of cells actively contributing to branching events. Here, we compare mechanisms that regulate branching, focusing on how cell cohorts behave in a coordinated manner to build branched tissues.This article is part of the themed issue 'Systems morphodynamics: understanding the development of tissue hardware'.
Collapse
Affiliation(s)
- James W Spurlin
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA .,Molecular Biology, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| |
Collapse
|
22
|
Marcus M, Baranes K, Park M, Choi IS, Kang K, Shefi O. Interactions of Neurons with Physical Environments. Adv Healthc Mater 2017. [PMID: 28640544 DOI: 10.1002/adhm.201700267] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nerve growth strongly relies on multiple chemical and physical signals throughout development and regeneration. Currently, a cure for injured neuronal tissue is an unmet need. Recent advances in fabrication technologies and materials led to the development of synthetic interfaces for neurons. Such engineered platforms that come in 2D and 3D forms can mimic the native extracellular environment and create a deeper understanding of neuronal growth mechanisms, and ultimately advance the development of potential therapies for neuronal regeneration. This progress report aims to present a comprehensive discussion of this field, focusing on physical feature design and fabrication with additional information about considerations of chemical modifications. We review studies of platforms generated with a range of topographies, from micro-scale features down to topographical elements at the nanoscale that demonstrate effective interactions with neuronal cells. Fabrication methods are discussed as well as their biological outcomes. This report highlights the interplay between neuronal systems and the important roles played by topography on neuronal differentiation, outgrowth, and development. The influence of substrate structures on different neuronal cells and parameters including cell fate, outgrowth, intracellular remodeling, gene expression and activity is discussed. Matching these effects to specific needs may lead to the emergence of clinical solutions for patients suffering from neuronal injuries or brain-machine interface (BMI) applications.
Collapse
Affiliation(s)
- Michal Marcus
- Faculty of Engineering and Bar-Ilan Institute for Nanotechnology and Advanced Materials; Bar-Ilan University; Ramat-Gan 5290002 Israel
| | - Koby Baranes
- Faculty of Engineering and Bar-Ilan Institute for Nanotechnology and Advanced Materials; Bar-Ilan University; Ramat-Gan 5290002 Israel
| | - Matthew Park
- Center for Cell-Encapsulation Research; Department of Chemistry; KAIST; Daejeon 34141 Korea
| | - Insung S. Choi
- Center for Cell-Encapsulation Research; Department of Chemistry; KAIST; Daejeon 34141 Korea
| | - Kyungtae Kang
- Department of Applied Chemistry; Kyung Hee University; Yongin Gyeonggi 17104 Korea
| | - Orit Shefi
- Faculty of Engineering and Bar-Ilan Institute for Nanotechnology and Advanced Materials; Bar-Ilan University; Ramat-Gan 5290002 Israel
| |
Collapse
|
23
|
Kumar A, Pareek V, Faiq MA, Kumar P, Raza K, Prasoon P, Dantham S, Mochan S. Regulatory role of NGFs in neurocognitive functions. Rev Neurosci 2017; 28:649-673. [DOI: 10.1515/revneuro-2016-0031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
AbstractNerve growth factors (NGFs), especially the prototype NGF and brain-derived neurotrophic factor (BDNF), have a diverse array of functions in the central nervous system through their peculiar set of receptors and intricate signaling. They are implicated not only in the development of the nervous system but also in regulation of neurocognitive functions like learning, memory, synaptic transmission, and plasticity. Evidence even suggests their role in continued neurogenesis and experience-dependent neural network remodeling in adult brain. They have also been associated extensively with brain disorders characterized by neurocognitive dysfunction. In the present article, we aimed to make an exhaustive review of literature to get a comprehensive view on the role of NGFs in neurocognitive functions in health and disease. Starting with historical perspective, distribution in adult brain, implied molecular mechanisms, and developmental basis, this article further provides a detailed account of NGFs’ role in specified neurocognitive functions. Furthermore, it discusses plausible NGF-based homeostatic and adaptation mechanisms operating in the pathogenesis of neurocognitive disorders and has presents a survey of such disorders. Finally, it elaborates on current evidence and future possibilities in therapeutic applications of NGFs with an emphasis on recent research updates in drug delivery mechanisms. Conclusive remarks of the article make a strong case for plausible role of NGFs in comprehensive regulation of the neurocognitive functions and pathogenesis of related disorders and advocate that future research should be directed to explore use of NGF-based mechanisms in the prevention of implicated diseases as well as to target these molecules pharmacologically.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Karaikal, Puducherry 609602, India
| | - Vikas Pareek
- Computational Neuroscience and Neuroimaging Division, National Brain Research Centre (NBRC), Manesar, Haryana 122051, India
| | - Muneeb A. Faiq
- Department of Ophthalmology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pavan Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Khursheed Raza
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pranav Prasoon
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Subrahamanyam Dantham
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sankat Mochan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
24
|
Shen Y, Cheng Y, Uyeda TQP, Plaza GR. Cell Mechanosensors and the Possibilities of Using Magnetic Nanoparticles to Study Them and to Modify Cell Fate. Ann Biomed Eng 2017; 45:2475-2486. [PMID: 28744841 DOI: 10.1007/s10439-017-1884-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 07/07/2017] [Indexed: 12/13/2022]
Abstract
The use of magnetic nanoparticles (MNPs) is a promising technique for future advances in biomedical applications. This idea is supported by the availability of MNPs that can target specific cell components, the variety of shapes of MNPs and the possibility of finely controlling the applied magnetic forces. To examine this opportunity, here we review the current developments in the use of MNPs to mechanically stimulate cells and, specifically, the cell mechanotransduction systems. We analyze the cell components that may act as mechanosensors and their effect on cell fate and we focus on the promising possibilities of controlling stem-cell differentiation, inducing cancer-cell death and treating nervous-system diseases.
Collapse
Affiliation(s)
- Yajing Shen
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Cheng
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Taro Q P Uyeda
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Gustavo R Plaza
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China. .,Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223, Pozuelo de Alarcón, Spain.
| |
Collapse
|
25
|
Torisawa T, Taniguchi D, Ishihara S, Oiwa K. Spontaneous Formation of a Globally Connected Contractile Network in a Microtubule-Motor System. Biophys J 2017; 111:373-385. [PMID: 27463139 PMCID: PMC4968425 DOI: 10.1016/j.bpj.2016.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 04/19/2016] [Accepted: 06/08/2016] [Indexed: 11/01/2022] Open
Abstract
Microtubule (MT) networks play key roles in cell division, intracellular transport, and cell motility. These functions of MT networks occur through interactions between MTs and various associated proteins, notably motor proteins that bundle and slide MTs. Our objective in this study was to address the question of how motors determine the nature of MT networks. We conducted in vitro assays using homotetrameric kinesin Eg5, a motor protein involved in the formation and maintenance of the mitotic spindle. The mixing of Eg5 and MTs produced a range of spatiotemporal dynamics depending on the motor/filament ratio. Low motor/filament ratios produced globally connected static MT networks with sparsely distributed contractile active nodes (motor-accumulating points with radially extending MTs). Increasing the motor/filament ratio facilitated the linking of contractile active nodes and led to a global contraction of the network. When the motor/filament ratio was further increased, densely distributed active nodes formed local clusters and segmented the network into pieces with their strong contractile forces. Altering the properties of the motor through the use of chimeric Eg5, which has kinesin-1 heads, resulted in the generation of many isolated asters. These results suggest that the spatial distribution of contractile active nodes determines the dynamics of MT-motor networks. We then developed a coarse-grained model of MT-motor networks and identified two essential features for reproducing the experimentally observed patterns: an accumulation of motors that form the active nodes necessary to generate contractile forces, and a nonlinear dependency of contractile force on motor densities. Our model also enabled us to characterize the mechanical properties of the contractile network. Our study provides insight into how local motor-MT interactions generate the spatiotemporal dynamics of macroscopic network structures.
Collapse
Affiliation(s)
- Takayuki Torisawa
- National Institute of Information and Communications Technology, Advanced ICT Research Institute, Kobe, Hyogo, Japan
| | | | - Shuji Ishihara
- Department of Physics, Meiji University, Kawasaki, Kanagawa, Japan
| | - Kazuhiro Oiwa
- National Institute of Information and Communications Technology, Advanced ICT Research Institute, Kobe, Hyogo, Japan.
| |
Collapse
|
26
|
Uncoupling of UNC5C with Polymerized TUBB3 in Microtubules Mediates Netrin-1 Repulsion. J Neurosci 2017; 37:5620-5633. [PMID: 28483977 DOI: 10.1523/jneurosci.2617-16.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 04/22/2017] [Accepted: 04/28/2017] [Indexed: 11/21/2022] Open
Abstract
Modulation of microtubule (MT) dynamics is a key event of cytoskeleton remodeling in the growth cone (GC) during axon outgrowth and pathfinding. Our previous studies have shown that the direct interaction of netrin receptor DCC and DSCAM with polymerized TUBB3, a neuron-specific MT subunit in the brain, is required for netrin-1-mediated axon outgrowth, branching, and attraction. Here, we show that uncoupling of polymerized TUBB3 with netrin-1-repulsive receptor UNC5C is involved in netrin-1-mediated axonal repulsion. TUBB3 directly interacted with UNC5C and partially colocalized with UNC5C in the peripheral area of the GC of primary neurons from the cerebellar external granule layer of P2 mouse pups of both sexes. Netrin-1 reduced this interaction as well as the colocalization of UNC5C and TUBB3 in the GC. Results from the in vitro cosedimentation assay indicated that UNC5C interacted with polymerized TUBB3 in MTs and netrin-1 decreased this interaction. Knockdown of either TUBB3 or UNC5C blocked netrin-1-promoted axon repulsion in vitro and caused defects in axon projection of DRG toward the spinal cord in vivo Furthermore, live-cell imaging of end-binding protein 3 tagged with EGFP (EB3-GFP) in primary external granule layer cells showed that netrin-1 differentially increased MT dynamics in the GC with more MT growth in the distal than the proximal region of the GC during repulsion, and knockdown of either UNC5C or TUBB3 abolished the netrin-1 effect. Together, these data indicate that the disengagement of UNC5C with polymerized TUBB3 plays an essential role in netrin-1/UNC5C-mediated axon repulsion.SIGNIFICANCE STATEMENT Proper regulation of microtubule (MT) dynamics in the growth cone plays an important role in axon guidance. However, whether guidance cues modulate MT dynamics directly or indirectly is unclear. Here, we report that dissociation of UNC5C and polymerized TUBB3, the highly dynamic β-tubulin isoform in neurons, is essential for netrin-1/UNC5C-promoted axon repulsion. These results not only provide a working model of direct modulation of MTs by guidance cues in growth cone navigation but also help us to understand molecular mechanisms underlying developmental brain disorders associated with TUBB3 mutations.
Collapse
|
27
|
Riveiro AR, Mariani L, Malmberg E, Amendola PG, Peltonen J, Wong G, Salcini AE. JMJD-1.2/PHF8 controls axon guidance by regulating Hedgehog-like signaling. Development 2017; 144:856-865. [PMID: 28126843 DOI: 10.1242/dev.142695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/09/2017] [Indexed: 01/10/2023]
Abstract
Components of the KDM7 family of histone demethylases are implicated in neuronal development and one member, PHF8, is often found to be mutated in cases of X-linked mental retardation. However, how PHF8 regulates neurodevelopmental processes and contributes to the disease is still largely unknown. Here, we show that the catalytic activity of a PHF8 homolog in Caenorhabditis elegans, JMJD-1.2, is required non-cell-autonomously for proper axon guidance. Loss of JMJD-1.2 dysregulates transcription of the Hedgehog-related genes wrt-8 and grl-16, the overexpression of which is sufficient to induce the axonal defects. Deficiency of either wrt-8 or grl-16, or reduced expression of homologs of genes promoting Hedgehog signaling, restores correct axon guidance in jmjd-1.2 mutants. Genetic and overexpression data indicate that Hedgehog-related genes act on axon guidance through actin remodelers. Thus, our study highlights a novel function of jmjd-1.2 in axon guidance that might be relevant for the onset of X-linked mental retardation and provides compelling evidence of a conserved function of the Hedgehog pathway in C. elegans axon migration.
Collapse
Affiliation(s)
- Alba Redo Riveiro
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.,Centre for Epigenetics, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Luca Mariani
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.,Centre for Epigenetics, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Emily Malmberg
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.,Centre for Epigenetics, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Pier Giorgio Amendola
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.,Centre for Epigenetics, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Juhani Peltonen
- A. I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, 70211, Kuopio, Finland
| | - Garry Wong
- Faculty of Health Sciences, University of Macau, 999078, Macau, China
| | - Anna Elisabetta Salcini
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark .,Centre for Epigenetics, University of Copenhagen, 2200, Copenhagen, Denmark
| |
Collapse
|
28
|
Law V, Dong S, Rosales JL, Jeong MY, Zochodne D, Lee KY. Enhancement of Peripheral Nerve Regrowth by the Purine Nucleoside Analog and Cell Cycle Inhibitor, Roscovitine. Front Cell Neurosci 2016; 10:238. [PMID: 27799897 PMCID: PMC5066473 DOI: 10.3389/fncel.2016.00238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/30/2016] [Indexed: 02/03/2023] Open
Abstract
Peripheral nerve regeneration is a slow process that can be associated with limited outcomes and thus a search for novel and effective therapy for peripheral nerve injury and disease is crucial. Here, we found that roscovitine, a synthetic purine nucleoside analog, enhances neurite outgrowth in neuronal-like PC12 cells. Furthermore, ex vivo analysis of pre-injured adult rat dorsal root ganglion (DRG) neurons showed that roscovitine enhances neurite regrowth in these cells. Likewise, in vivo transected sciatic nerves in rats locally perfused with roscovitine had augmented repopulation of new myelinated axons beyond the transection zone. By mass spectrometry, we found that roscovitine interacts with tubulin and actin. It interacts directly with tubulin and causes a dose-dependent induction of tubulin polymerization as well as enhances Guanosine-5′-triphosphate (GTP)-dependent tubulin polymerization. Conversely, roscovitine interacts indirectly with actin and counteracts the inhibitory effect of cyclin-dependent kinases 5 (Cdk5) on Actin-Related Proteins 2/3 (Arp2/3)-dependent actin polymerization, and thus, causes actin polymerization. Moreover, in the presence of neurotrophic factors such as nerve growth factor (NGF), roscovitine-enhanced neurite outgrowth is mediated by increased activation of the extracellular signal-regulated kinases 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK) pathways. Since microtubule and F-actin dynamics are critical for axonal regrowth, the ability of roscovitine to activate the ERK1/2 and p38 MAPK pathways and support polymerization of tubulin and actin indicate a major role for this purine nucleoside analog in the promotion of axonal regeneration. Together, our findings demonstrate a therapeutic potential for the purine nucleoside analog, roscovitine, in peripheral nerve injury.
Collapse
Affiliation(s)
- Vincent Law
- Department of Cell Biology and Anatomy, Arnie Charbonneau Cancer Institute, Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| | - Sophie Dong
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| | - Jesusa L Rosales
- Department of Biochemistry and Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary Calgary, AB, Canada
| | - Myung-Yung Jeong
- Department of Cogno-Mechatronics Engineering, Pusan National University Pusan, South Korea
| | - Douglas Zochodne
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| | - Ki-Young Lee
- Department of Cell Biology and Anatomy, Arnie Charbonneau Cancer Institute, Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| |
Collapse
|
29
|
Stanic K, Saldivia N, Förstera B, Torrejón M, Montecinos H, Caprile T. Expression Patterns of Extracellular Matrix Proteins during Posterior Commissure Development. Front Neuroanat 2016; 10:89. [PMID: 27733818 PMCID: PMC5039192 DOI: 10.3389/fnana.2016.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022] Open
Abstract
Extracellular matrix (ECM) molecules are pivotal for central nervous system (CNS) development, facilitating cell migration, axonal growth, myelination, dendritic spine formation, and synaptic plasticity, among other processes. During axon guidance, the ECM not only acts as a permissive or non-permissive substrate for navigating axons, but also modulates the effects of classical guidance cues, such as netrin or Eph/ephrin family members. Despite being highly important, little is known about the expression of ECM molecules during CNS development. Therefore, this study assessed the molecular expression patterns of tenascin, HNK-1, laminin, fibronectin, perlecan, decorin, and osteopontin along chick embryo prosomere 1 during posterior commissure development. The posterior commissure is the first transversal axonal tract of the embryonic vertebrate brain. Located in the dorso-caudal portion of prosomere 1, posterior commissure axons primarily arise from the neurons of basal pretectal nuclei that run dorsally to the roof plate midline, where some turn toward the ipsilateral side. Expressional analysis of ECM molecules in this area these revealed to be highly arranged, and molecule interactions with axon fascicles suggested involvement in processes other than structural support. In particular, tenascin and the HNK-1 epitope extended in ventro-dorsal columns and enclosed axons during navigation to the roof plate. Laminin and osteopontin were expressed in the midline, very close to axons that at this point must decide between extending to the contralateral side or turning to the ipsilateral side. Finally, fibronectin, decorin, and perlecan appeared unrelated to axonal pathfinding in this region and were instead restricted to the external limiting membrane. In summary, the present report provides evidence for an intricate expression of different extracellular molecules that may cooperate in guiding posterior commissure axons.
Collapse
Affiliation(s)
- Karen Stanic
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Natalia Saldivia
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Benjamín Förstera
- Department of Physiology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Marcela Torrejón
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Hernán Montecinos
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Teresa Caprile
- Axon Guidance Laboratory, Department of Cell Biology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| |
Collapse
|
30
|
Back to the tubule: microtubule dynamics in Parkinson's disease. Cell Mol Life Sci 2016; 74:409-434. [PMID: 27600680 PMCID: PMC5241350 DOI: 10.1007/s00018-016-2351-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
Cytoskeletal homeostasis is essential for the development, survival and maintenance of an efficient nervous system. Microtubules are highly dynamic polymers important for neuronal growth, morphology, migration and polarity. In cooperation with several classes of binding proteins, microtubules regulate long-distance intracellular cargo trafficking along axons and dendrites. The importance of a delicate interplay between cytoskeletal components is reflected in several human neurodegenerative disorders linked to abnormal microtubule dynamics, including Parkinson’s disease (PD). Mounting evidence now suggests PD pathogenesis might be underlined by early cytoskeletal dysfunction. Advances in genetics have identified PD-associated mutations and variants in genes encoding various proteins affecting microtubule function including the microtubule-associated protein tau. In this review, we highlight the role of microtubules, their major posttranslational modifications and microtubule associated proteins in neuronal function. We then present key evidence on the contribution of microtubule dysfunction to PD. Finally, we discuss how regulation of microtubule dynamics with microtubule-targeting agents and deacetylase inhibitors represents a promising strategy for innovative therapeutic development.
Collapse
|
31
|
Kilinc D, Dennis CL, Lee GU. Bio-Nano-Magnetic Materials for Localized Mechanochemical Stimulation of Cell Growth and Death. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5672-80. [PMID: 26780501 PMCID: PMC5536250 DOI: 10.1002/adma.201504845] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/12/2015] [Indexed: 05/16/2023]
Abstract
Magnetic nanoparticles are promising new tools for therapeutic applications, such as magnetic nanoparticle hyperthermia therapy and targeted drug delivery. Recent in vitro studies have demonstrated that a force application with magnetic tweezers can also affect cell fate, suggesting a therapeutic potential for magnetically modulated mechanical stimulation. The magnetic properties of nanoparticles that induce physical responses and the subtle responses that result from mechanically induced membrane damage and/or intracellular signaling are evaluated. Magnetic particles with various physical, geometric, and magnetic properties and specific functionalization can now be used to apply mechanical force to specific regions of cells, which permit the modulation of cellular behavior through the use of spatially and time controlled magnetic fields. On one hand, mechanochemical stimulation has been used to direct the outgrowth on neuronal growth cones, indicating a therapeutic potential for neural repair. On the other hand, it has been used to kill cancer cells that preferentially express specific receptors. Advances made in the synthesis and characterization of magnetic nanomaterials and a better understanding of cellular mechanotransduction mechanisms may support the translation of mechanochemical stimulation into the clinic as an emerging therapeutic approach.
Collapse
Affiliation(s)
- Devrim Kilinc
- Bionanosciences Lab, School of Chemistry and Chemical Biology, UCD
Conway Institute of Biomolecular and Biomedical Research, University College Dublin,
Belfield, Dublin 4, Ireland
| | - Cindi L. Dennis
- Material Measurement Laboratory, National Institute of Standards and
Technology, 100 Bureau Drive, Gaithersburg, MD 20899–8552, USA
| | - Gil U. Lee
- Bionanosciences Lab, School of Chemistry and Chemical Biology, UCD
Conway Institute of Biomolecular and Biomedical Research, University College Dublin,
Belfield, Dublin 4, Ireland
| |
Collapse
|
32
|
Mariani L, Lussi YC, Vandamme J, Riveiro A, Salcini AE. The H3K4me3/2 histone demethylase RBR-2 controls axon guidance by repressing the actin-remodeling gene wsp-1. Development 2016; 143:851-63. [PMID: 26811384 DOI: 10.1242/dev.132985] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/16/2016] [Indexed: 12/25/2022]
Abstract
The dynamic regulation of histone modifications is important for modulating transcriptional programs during development. Aberrant H3K4 methylation is associated with neurological disorders, but how the levels and the recognition of this modification affect specific neuronal processes is unclear. Here, we show that RBR-2, the sole homolog of the KDM5 family of H3K4me3/2 demethylases in Caenorhabditis elegans, ensures correct axon guidance by controlling the expression of the actin regulator wsp-1. Loss of rbr-2 results in increased levels of H3K4me3 at the transcriptional start site of wsp-1, with concomitant higher wsp-1 expression responsible for defective axon guidance. In agreement, overexpression of WSP-1 mimics rbr-2 loss, and its depletion restores normal axon guidance in rbr-2 mutants. NURF-1, an H3K4me3-binding protein and member of the chromatin-remodeling complex NURF, is required for promoting aberrant wsp-1 transcription in rbr-2 mutants and its ablation restores wild-type expression of wsp-1 and axon guidance. Thus, our results establish a precise role for epigenetic regulation in neuronal development by demonstrating a functional link between RBR-2 activity, H3K4me3 levels, the NURF complex and the expression of WSP-1.
Collapse
Affiliation(s)
- Luca Mariani
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yvonne C Lussi
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Julien Vandamme
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alba Riveiro
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anna Elisabetta Salcini
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark Centre for Epigenetics, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
33
|
Blasiak A, Lee GU, Kilinc D. Neuron Subpopulations with Different Elongation Rates and DCC Dynamics Exhibit Distinct Responses to Isolated Netrin-1 Treatment. ACS Chem Neurosci 2015; 6:1578-90. [PMID: 26190161 DOI: 10.1021/acschemneuro.5b00142] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Correct wiring of the nervous system requires guidance cues, diffusible or substrate-bound proteins that steer elongating axons to their target tissues. Netrin-1, the best characterized member of the Netrins family of guidance molecules, is known to induce axon turning and modulate axon elongation rate; however, the factors regulating the axonal response to Netrin-1 are not fully understood. Using microfluidics, we treated fluidically isolated axons of mouse primary cortical neurons with Netrin-1 and characterized axon elongation rates, as well as the membrane localization of deleted in colorectal cancer (DCC), a well-established receptor of Netrin-1. The capacity to stimulate and observe a large number of individual axons allowed us to conduct distribution analyses, through which we identified two distinct neuron subpopulations based on different elongation behavior and different DCC membrane dynamics. Netrin-1 reduced the elongation rates in both subpopulations, where the effect was more pronounced in the slow growing subpopulation. Both the source of Ca(2+) influx and the basal cytosolic Ca(2+) levels regulated the effect of Netrin-1, for example, Ca(2+) efflux from the endoplasmic reticulum due to the activation of Ryanodine channels blocked Netrin-1-induced axon slowdown. Netrin-1 treatment resulted in a rapid membrane insertion of DCC, followed by a gradual internalization. DCC membrane dynamics were different in the central regions of the growth cones compared to filopodia and axon shafts, highlighting the temporal and spatial heterogeneity in the signaling events downstream of Netrin-1. Cumulatively, these results demonstrate the power of microfluidic compartmentalization and distribution analysis in describing the complex axonal Netrin-1 response.
Collapse
Affiliation(s)
- Agata Blasiak
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| | - Gil U. Lee
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| | - Devrim Kilinc
- Bionanoscience Group, School
of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin
4, Ireland
| |
Collapse
|
34
|
Stamatakou E, Hoyos-Flight M, Salinas PC. Wnt Signalling Promotes Actin Dynamics during Axon Remodelling through the Actin-Binding Protein Eps8. PLoS One 2015; 10:e0134976. [PMID: 26252776 PMCID: PMC4529215 DOI: 10.1371/journal.pone.0134976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/07/2015] [Indexed: 11/26/2022] Open
Abstract
Upon arrival at their synaptic targets, axons slow down their growth and extensively remodel before the assembly of presynaptic boutons. Wnt proteins are target-derived secreted factors that promote axonal remodelling and synaptic assembly. In the developing spinal cord, Wnts secreted by motor neurons promote axonal remodelling of NT-3 responsive dorsal root ganglia neurons. Axon remodelling induced by Wnts is characterised by growth cone pausing and enlargement, processes that depend on the re-organisation of microtubules. However, the contribution of the actin cytoskeleton has remained unexplored. Here, we demonstrate that Wnt3a regulates the actin cytoskeleton by rapidly inducing F-actin accumulation in growth cones from rodent DRG neurons through the scaffold protein Dishevelled-1 (Dvl1) and the serine-threonine kinase Gsk3β. Importantly, these changes in actin cytoskeleton occurs before enlargement of the growth cones is evident. Time-lapse imaging shows that Wnt3a increases lamellar protrusion and filopodia velocity. In addition, pharmacological inhibition of actin assembly demonstrates that Wnt3a increases actin dynamics. Through a yeast-two hybrid screen, we identified the actin-binding protein Eps8 as a direct interactor of Dvl1, a scaffold protein crucial for the Wnt signalling pathway. Gain of function of Eps8 mimics Wnt-mediated axon remodelling, whereas Eps8 silencing blocks the axon remodelling activity of Wnt3a. Importantly, blockade of the Dvl1-Eps8 interaction completely abolishes Wnt3a-mediated axonal remodelling. These findings demonstrate a novel role for Wnt-Dvl1 signalling through Eps8 in the regulation of axonal remodeling.
Collapse
Affiliation(s)
- Eleanna Stamatakou
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Monica Hoyos-Flight
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Regulators of Actin Dynamics in Gastrointestinal Tract Tumors. Gastroenterol Res Pract 2015; 2015:930157. [PMID: 26345720 PMCID: PMC4539459 DOI: 10.1155/2015/930157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 02/07/2023] Open
Abstract
Reorganization of the actin cytoskeleton underlies cell migration in a wide variety of physiological and pathological processes, such as embryonic development, wound healing, and tumor cell invasion. It has been shown that actin assembly and disassembly are precisely regulated by intracellular signaling cascades that respond to changes in the cell microenvironment, ligand binding to surface receptors, or oncogenic transformation of the cell. Actin-nucleating and actin-depolymerizing (ANFs/ADFs) and nucleation-promoting factors (NPFs) regulate cytoskeletal dynamics at the leading edge of migrating cells, thereby modulating cell shape; these proteins facilitate cellular movement and mediate degradation of the surrounding extracellular matrix by secretion of lytic proteases, thus eliminating barriers for tumor cell invasion. Accordingly, expression and activity of these actin-binding proteins have been linked to enhanced metastasis and poor prognosis in a variety of malignancies. In this review, we will summarize what is known about expression patterns and the functional role of actin regulators in gastrointestinal tumors and evaluate first pharmacological approaches to prevent invasion and metastatic dissemination of malignant cells.
Collapse
|
36
|
Lopez BJ, Valentine MT. Molecular control of stress transmission in the microtubule cytoskeleton. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [PMID: 26225932 DOI: 10.1016/j.bbamcr.2015.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In this article, we will summarize recent progress in understanding the mechanical origins of rigidity, strength, resiliency and stress transmission in the MT cytoskeleton using reconstituted networks formed from purified components. We focus on the role of network architecture, crosslinker compliance and dynamics, and molecular determinants of single filament elasticity, while highlighting open questions and future directions for this work.
Collapse
Affiliation(s)
- Benjamin J Lopez
- Department of Mechanical Engineering and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106-5070, USA
| | - Megan T Valentine
- Department of Mechanical Engineering and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106-5070, USA.
| |
Collapse
|
37
|
Kilinc D, Blasiak A, Lee GU. Microtechnologies for studying the role of mechanics in axon growth and guidance. Front Cell Neurosci 2015; 9:282. [PMID: 26283918 PMCID: PMC4515553 DOI: 10.3389/fncel.2015.00282] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/10/2015] [Indexed: 12/18/2022] Open
Abstract
The guidance of axons to their proper targets is not only a crucial event in neurodevelopment, but also a potential therapeutic target for neural repair. Axon guidance is mediated by various chemo- and haptotactic cues, as well as the mechanical interactions between the cytoskeleton and the extracellular matrix (ECM). Axonal growth cones, dynamic ends of growing axons, convert external stimuli to biochemical signals, which, in turn, are translated into behavior, e.g., turning or retraction, via cytoskeleton-matrix linkages. Despite the inherent mechanical nature of the problem, the role of mechanics in axon guidance is poorly understood. Recent years has witnessed the application of a range of microtechnologies in neurobiology, from microfluidic circuits to single molecule force spectroscopy. In this mini-review, we describe microtechnologies geared towards dissecting the mechanical aspects of axon guidance, divided into three categories: controlling the growth cone microenvironment, stimulating growth cones with externally applied forces, and measuring forces exerted by the growth cones. A particular emphasis is given to those studies that combine multiple techniques, as dictated by the complexity of the problem.
Collapse
Affiliation(s)
- Devrim Kilinc
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| | - Agata Blasiak
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| | - Gil U Lee
- Bionanosciences Group, School of Chemisty and Chemical Biology, University College Dublin Belfield, Dublin, Ireland
| |
Collapse
|
38
|
Kamudzandu M, Yang Y, Roach P, Fricker RA. Efficient alignment of primary CNS neurites using structurally engineered surfaces and biochemical cues. RSC Adv 2015. [DOI: 10.1039/c4ra15739g] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Micro- and nano-structured materials were used to investigate directional alignment of primary CNS neurons (red) co-cultured with astrocytes (green).
Collapse
Affiliation(s)
| | - Ying Yang
- Guy Hilton Research Centre
- Keele University
- UK
| | - Paul Roach
- Guy Hilton Research Centre
- Keele University
- UK
| | | |
Collapse
|
39
|
Ammar MR, Kassas N, Bader MF, Vitale N. Phosphatidic acid in neuronal development: A node for membrane and cytoskeleton rearrangements. Biochimie 2014; 107 Pt A:51-7. [DOI: 10.1016/j.biochi.2014.07.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/30/2014] [Indexed: 12/22/2022]
|
40
|
Kilinc D, Blasiak A, O'Mahony JJ, Lee GU. Low piconewton towing of CNS axons against diffusing and surface-bound repellents requires the inhibition of motor protein-associated pathways. Sci Rep 2014; 4:7128. [PMID: 25417891 PMCID: PMC4241520 DOI: 10.1038/srep07128] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/03/2014] [Indexed: 12/24/2022] Open
Abstract
Growth cones, dynamic structures at axon tips, integrate chemical and physical stimuli and translate them into coordinated axon behaviour, e.g., elongation or turning. External force application to growth cones directs and enhances axon elongation in vitro; however, direct mechanical stimulation is rarely combined with chemotactic stimulation. We describe a microfluidic device that exposes isolated cortical axons to gradients of diffusing and substrate-bound molecules, and permits the simultaneous application of piconewton (pN) forces to multiple individual growth cones via magnetic tweezers. Axons treated with Y-27632, a RhoA kinase inhibitor, were successfully towed against Semaphorin 3A gradients, which repel untreated axons, with less than 12 pN acting on a small number of neural cell adhesion molecules. Treatment with Y-27632 or monastrol, a kinesin-5 inhibitor, promoted axon towing on substrates coated with chondroitin sulfate proteoglycans, potent axon repellents. Thus, modulating key molecular pathways that regulate contractile stress generation in axons counteracts the effects of repellent molecules and promotes tension-induced growth. The demonstration of parallel towing of axons towards inhibitory environments with minute forces suggests that mechanochemical stimulation may be a promising therapeutic approach for the repair of the damaged central nervous system, where regenerating axons face repellent factors over-expressed in the glial scar.
Collapse
Affiliation(s)
- Devrim Kilinc
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Agata Blasiak
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - James J O'Mahony
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gil U Lee
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
41
|
Chen K, Zhang W, Chen J, Li S, Guo G. Rho-associated protein kinase modulates neurite extension by regulating microtubule remodeling and vinculin distribution. Neural Regen Res 2014; 8:3027-35. [PMID: 25206623 PMCID: PMC4146208 DOI: 10.3969/j.issn.1673-5374.2013.32.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/25/2013] [Indexed: 01/11/2023] Open
Abstract
Rho-associated protein kinase is an essential regulator of cytoskeletal dynamics during the process of neurite extension. However, whether Rho kinase regulates microtubule remodeling or the distribution of adhesive proteins to mediate neurite outgrowth remains unclear. By specifically modulating Rho kinase activity with pharmacological agents, we studied the morpho-dynamics of neurite outgrowth. We found that lysophosphatidic acid, an activator of Rho kinase, inhibited neurite outgrowth, which could be reversed by Y-27632, an inhibitor of Rho kinase. Meanwhile, reorganization of microtubules was noticed during these processes, as indicated by their significant changes in the soma and growth cone. In addition, exposure to lysophosphatidic acid led to a decreased membrane distribution of vinculin, a focal adhesion protein in neurons, whereas Y-27632 recruited vinculin to the membrane. Taken together, our data suggest that Rho kinase regulates rat hippocampal neurite growth and microtubule formation via a mechanism associated with the redistribution of vinculin.
Collapse
Affiliation(s)
- Ke'en Chen
- Department of Neurosurgery, First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Wenbin Zhang
- Department of Emergency, First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Jing Chen
- Department of Anatomy, Medical College of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Sumei Li
- Department of Anatomy, Medical College of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Guoqing Guo
- Department of Anatomy, Medical College of Jinan University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
42
|
Liu G, Dwyer T. Microtubule dynamics in axon guidance. Neurosci Bull 2014; 30:569-83. [PMID: 24968808 DOI: 10.1007/s12264-014-1444-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/10/2014] [Indexed: 12/18/2022] Open
Abstract
Precise modulation of the cytoskeleton is involved in a variety of cellular processes including cell division, migration, polarity, and adhesion. In developing post-mitotic neurons, extracellular guidance cues not only trigger signaling cascades that act at a distance to indirectly regulate microtubule distribution, and assembly and disassembly in the growth cone, but also directly modulate microtubule stability and dynamics through coupling of guidance receptors with microtubules to control growth-cone turning. Microtubule-associated proteins including classical microtubule-associated proteins and microtubule plus-end tracking proteins are required for modulating microtubule dynamics to influence growth-cone steering. Multiple key signaling components, such as calcium, small GTPases, glycogen synthase kinase-3β, and c-Jun N-terminal kinase, link upstream signal cascades to microtubule stability and dynamics in the growth cone to control axon outgrowth and projection. Understanding the functions and regulation of microtubule dynamics in the growth cone provides new insights into the molecular mechanisms of axon guidance.
Collapse
Affiliation(s)
- Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, 43606, USA,
| | | |
Collapse
|
43
|
Steketee MB, Oboudiyat C, Daneman R, Trakhtenberg E, Lamoureux P, Weinstein JE, Heidemann S, Barres BA, Goldberg JL. Regulation of intrinsic axon growth ability at retinal ganglion cell growth cones. Invest Ophthalmol Vis Sci 2014; 55:4369-77. [PMID: 24906860 DOI: 10.1167/iovs.14-13882] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Mammalian central nervous system neurons fail to regenerate after injury or disease, in part due to a progressive loss in intrinsic axon growth ability after birth. Whether lost axon growth ability is due to limited growth resources or to changes in the axonal growth cone is unknown. METHODS Static and time-lapse images of purified retinal ganglion cells (RGCs) were analyzed for axon growth rate and growth cone morphology and dynamics without treatment and after manipulating Kruppel-like transcription factor (KLF) expression or applying mechanical tension. RESULTS Retinal ganglion cells undergo a developmental switch in growth cone dynamics that mirrors the decline in postnatal axon growth rates, with increased filopodial adhesion and decreased lamellar protrusion area in postnatal axonal growth cones. Moreover, expressing growth-suppressive KLF4 or growth-enhancing KLF6 transcription factors elicits similar changes in postnatal growth cones that correlate with axon growth rates. Postnatal RGC axon growth rate is not limited by an inability to achieve axon growth rates similar to embryonic RGCs; indeed, postnatal axons support elongation rates up to 100-fold faster than postnatal axonal growth rates. Rather, the intrinsic capacity for rapid axon growth is due to both growth cone pausing and retraction, as well as to a slightly decreased ability to achieve rapid instantaneous rates of forward progression. Finally, we observed that RGC axon and dendrite growth are regulated independently in vitro. CONCLUSIONS Together, these data support the hypothesis that intrinsic axon growth rate is regulated by an axon-specific growth program that differentially regulates growth cone motility.
Collapse
Affiliation(s)
- Michael B Steketee
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States Department of Ophthalmology and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Carly Oboudiyat
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Richard Daneman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
| | - Ephraim Trakhtenberg
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Philip Lamoureux
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Jessica E Weinstein
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Steve Heidemann
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
| | - Jeffrey L Goldberg
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States Department of Ophthalmology, Shiley Eye Center, University of California San Diego, San Diego, California, United States
| |
Collapse
|
44
|
Giridharan SSP, Caplan S. MICAL-family proteins: Complex regulators of the actin cytoskeleton. Antioxid Redox Signal 2014; 20:2059-73. [PMID: 23834433 PMCID: PMC3993057 DOI: 10.1089/ars.2013.5487] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
SIGNIFICANCE The molecules interacting with CasL (MICAL) family members participate in a multitude of activities, including axonal growth cone repulsion, membrane trafficking, apoptosis, and bristle development in flies. An interesting feature of MICAL proteins is the presence of an N-terminal flavo-mono-oxygenase domain. This mono-oxygenase domain generates redox potential with which MICALs can either oxidize proteins or produce reactive oxygen species (ROS). Actin is one such protein that is affected by MICAL function, leading to dramatic cytoskeletal rearrangements. This review describes the MICAL-family members, and discusses their mechanisms of actin-binding and regulation of actin cytoskeleton organization. RECENT ADVANCES Recent studies show that MICALs directly induce oxidation of actin molecules, leading to actin depolymerization. ROS production by MICALs also causes oxidation of collapsin response mediator protein-2, a microtubule assembly promoter, which subsequently undergoes phosphorylation. CRITICAL ISSUES MICAL proteins oxidize proteins through two mechanisms: either directly by oxidizing methionine residues or indirectly via the production of ROS. It remains unclear whether MICAL proteins employ both mechanisms or whether the activity of MICAL-family proteins might vary with different substrates. FUTURE DIRECTIONS The identification of additional substrates oxidized by MICAL will shed new light on MICAL protein function. Additional directions include expanding studies toward the MICAL-like homologs that lack flavin adenine dinucleotide domains and oxidation activity.
Collapse
Affiliation(s)
- Sai Srinivas Panapakkam Giridharan
- Department of Biochemistry and Molecular Biology, and the Pamela and Fred Buffett Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska
| | | |
Collapse
|
45
|
Liu W, Xing S, Yuan B, Zheng W, Jiang X. Change of laminin density stimulates axon branching via growth cone myosin II-mediated adhesion. Integr Biol (Camb) 2014; 5:1244-52. [PMID: 23959160 DOI: 10.1039/c3ib40131f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Axon branching enables neurons to contact with multiple targets and respond to their microenvironment. Owing to its importance in neuronal network formation, axon branching has been studied extensively during the past decades. The chemical properties of extracellular matrices have been proposed to regulate axonal development, but the effects of their density changes on axon branching are not well understood. Here, we demonstrate that both the sharp broadening of substrate geometry and the sharp change of laminin density stimulate axon branching by using microcontact printing (μCP) and microfluidic printing (μFP) techniques. We also found that the change of axon branching stimulated by laminin density depends on myosin II activity. The change of laminin density induces asymmetric extensions of filopodia on the growth cone, which is the precondition for axon branching. These previously unknown mechanisms of change of laminin density-stimulated axon branching may explain how the extracellular matrices regulate axon branching in vivo and facilitate the establishment of neuronal networks in vitro.
Collapse
Affiliation(s)
- Wenwen Liu
- CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, P. R. China.
| | | | | | | | | |
Collapse
|
46
|
Prokosch V, Chiwitt C, Rose K, Thanos S. Deciphering proteins and their functions in the regenerating retina. Expert Rev Proteomics 2014; 7:775-95. [DOI: 10.1586/epr.10.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
47
|
Ip JPK, Fu AKY, Ip NY. CRMP2: functional roles in neural development and therapeutic potential in neurological diseases. Neuroscientist 2014; 20:589-98. [PMID: 24402611 DOI: 10.1177/1073858413514278] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cytoskeletal restructuring is essential for nearly all cellular processes in the developing brain. After cell fate determination, newborn cortical neurons must migrate to their final positions while establishing proper axon-dendrite polarity. Significant progress has recently been made towards understanding the cellular and molecular mechanisms underlying neuronal polarization in vivo. Collapsin response mediator protein 2 (CRMP2) has long been identified as a microtubule-binding protein that regulates neuronal polarity in vitro. Recent studies provide new insights into the roles of CRMP2 in neuronal migration and subsequent neuronal differentiation. Both the expression and activity of CRMP2 are tightly regulated during cortex development. CRMP2 is suggested to be important in the multipolar-bipolar transition in radial migration. The increasing number of known interaction partners indicates that CRMP2 has functions beyond cytoskeletal regulation, including axonal transport, vesicle trafficking, and neurotransmitter release. This review discusses the current knowledge about CRMP2 in the context of neuronal development and highlights a recent emerging theme regarding its potential therapeutic applications.
Collapse
Affiliation(s)
- Jacque P K Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
48
|
Long JB, Van Vactor D. Embryonic and larval neural connectivity: progressive changes in synapse form and function at the neuromuscular junction mediated by cytoskeletal regulation. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2013; 2:747-65. [PMID: 24123935 DOI: 10.1002/wdev.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
During development, precise formation of millions of synaptic connections is critical for the formation of a functional nervous system. Synaptogenesis is a complex multistep process in which axons follow gradients of secreted and cell surface guidance cues to reach their target area, at which point they must accurately distinguish their specific target. Upon target recognition, the axonal growth cone undergoes rapid growth and morphological changes, ultimately forming a functional synapse that continues to remodel during activity-dependent plasticity. Significant evidence suggests that the underlying actin and microtubule (MT) cytoskeletons are key effectors throughout synaptogenesis downstream of numerous receptors and signaling pathways. An increasing number of cytoskeletal-associated proteins have been shown to influence actin and MT stability and dynamics and many of these regulators have been implicated during synaptic morphogenesis using both mammalian and invertebrate model systems. In this review, we present an overview of the role cytoskeletal regulators play during the formation of the Drosophila neuromuscular junction.
Collapse
Affiliation(s)
- Jennifer B Long
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
49
|
Li L, Fothergill T, Hutchins BI, Dent EW, Kalil K. Wnt5a evokes cortical axon outgrowth and repulsive guidance by tau mediated reorganization of dynamic microtubules. Dev Neurobiol 2013; 74:797-817. [PMID: 23818454 PMCID: PMC4087151 DOI: 10.1002/dneu.22102] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/24/2013] [Accepted: 06/20/2013] [Indexed: 01/08/2023]
Abstract
Wnt5a guides cortical axons in vivo by repulsion and in vitro evokes cortical axon outgrowth and repulsion by calcium signaling pathways. Here we examined the role of microtubule (MT) reorganization and dynamics in mediating effects of Wnt5a. Inhibiting MT dynamics with nocodazole and taxol abolished Wnt5a evoked axon outgrowth and repulsion of cultured hamster cortical neurons. EGFP-EB3 labeled dynamic MTs visualized in live cell imaging revealed that growth cone MTs align with the nascent axon. Wnt5a increased axon outgrowth by reorganization of dynamic MTs from a splayed to a bundled array oriented in the direction of axon extension, and Wnt5a gradients induced asymmetric redistribution of dynamic MTs toward the far side of the growth cone. Wnt5a gradients also evoked calcium transients that were highest on the far side of the growth cone. Calcium signaling and the reorganization of dynamic MTs could be linked by tau, a MT associated protein that stabilizes MTs. Tau is phosphorylated at the Ser 262 MT binding site by CaMKII, and is required for Wnt5a induced axon outgrowth and repulsive turning. Phosphorylation of tau at Ser262 is known to detach tau from MTs to increase their dynamics. Using transfection with tau constructs mutated at Ser262, we found that this site is required for the growth and guidance effects of Wnt5a by mediating reorganization of dynamic MTs in cortical growth cones. Moreover, CaMKII inhibition also prevents MT reorganization required for Wnt5a induced axon outgrowth, thus linking Wnt/calcium signaling to tau mediated MT reorganization during growth cone behaviors. © 2013 The Authors. Developmental Neurobiology Published by Wiley Periodicals, Inc.Develop Neurobiol 74: 797–817, 2014
Collapse
Affiliation(s)
- Li Li
- Neuroscience Training Program, University of Wisconsin-Madison, Wisconsin, 53706
| | | | | | | | | |
Collapse
|
50
|
Different modes of growth cone collapse in NG 108-15 cells. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2013; 42:591-605. [PMID: 23644679 PMCID: PMC3705140 DOI: 10.1007/s00249-013-0907-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/27/2013] [Accepted: 04/13/2013] [Indexed: 10/27/2022]
Abstract
In the fundamental process of neuronal path-finding, a growth cone at the tip of every neurite detects and follows multiple guidance cues regulating outgrowth and initiating directional changes. While the main focus of research lies on the cytoskeletal dynamics underlying growth cone advancement, we investigated collapse and retraction mechanisms in NG108-15 growth cones transiently transfected with mCherry-LifeAct and pCS2+/EMTB-3XGFP for filamentous actin and microtubules, respectively. Using fluorescence time lapse microscopy we could identify two distinct modes of growth cone collapse leading either to neurite retraction or to a controlled halt of neurite extension. In the latter case, lateral movement and folding of actin bundles (filopodia) confine microtubule extension and limit microtubule-based expansion processes without the necessity of a constantly engaged actin turnover machinery. We term this previously unreported second type fold collapse and suggest that it marks an intermediate-term mode of growth regulation closing the gap between full retraction and small scale fluctuations.
Collapse
|