1
|
Sebastian A, Shanmuganathan MAA, Tripathy C, Chakravarty S, Ghosh S. Understanding Neurogenesis and Neuritogenesis via Molecular Insights, Gender Influence, and Therapeutic Implications: Intervention of Nanomaterials. ACS APPLIED BIO MATERIALS 2024. [PMID: 39718903 DOI: 10.1021/acsabm.4c01079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Neurological disorders impact global health by affecting both central and peripheral nervous systems. Understanding the neurogenic processes, i.e., neurogenesis and neuritogenesis, is of paramount importance in the context of nervous system development and regeneration as they hold promising therapeutic implications. Neurogenesis forms functional neurons from precursor cells, while neuritogenesis involves extending neurites for neuron connections. This review discusses how these processes are influenced by genetics, epigenetics, neurotrophic factors, environment, neuroinflammation, and neurotransmitters. It also covers gender-specific aspects of neurogenesis and neuritogenesis, their impact on brain plasticity, and susceptibility to neurological disorders. Alterations in these processes, under the influence of cytokines, growth factors, neurotransmitters, and aging, are linked to neurological disorders and potential therapeutic targets. Gender-specific effects of pharmacological interventions, like SSRIs, TCAs, atypical antipsychotics, and lithium, are explored in this review. Hormone-mediated effects of BDNF and PPAR-γ agonists, as well as variations in efficacy and tolerability of MAOIs, AEDs, NMDA receptor modulators, and ampakines, are detailed for accurate therapeutic design. The review also discusses nanotechnology's significant contribution to neural tissue regeneration for mending neurodegenerative disorders, enhancing neuronal connectivity, and stem cell differentiation. Gold nanoparticles support hippocampal neurogenesis, while other nanoparticles aid neuron growth and neurite outgrowth. Quantum dots and nanolayered double hydroxides assist neuroregeneration, which improves brain drug delivery. Gender-specific responses to nanomedicines designed to enhance neuroregeneration have not been extensively investigated. However, we have specified certain gender-related variables that should be taken into account during the development of nanomedicines in an aim to improve therapeutic efficacy. Further research on gender-specific responses to nanomedicines in neural processes could enhance personalized treatments for neurological disorders, paving the way for novel therapeutic approaches in neuroscience.
Collapse
Affiliation(s)
- Aishwarya Sebastian
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mohanraj Alias Ayyappan Shanmuganathan
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chinmayee Tripathy
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sutapa Ghosh
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Sollazzo R, Li Puma DD, Aceto G, Paciello F, Colussi C, Vita MG, Giuffrè GM, Pastore F, Casamassa A, Rosati J, Novelli A, Maietta S, Tiziano FD, Marra C, Ripoli C, Grassi C. Structural and functional alterations of neurons derived from sporadic Alzheimer's disease hiPSCs are associated with downregulation of the LIMK1-cofilin axis. Alzheimers Res Ther 2024; 16:267. [PMID: 39702316 DOI: 10.1186/s13195-024-01632-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by the accumulation of pathological proteins and synaptic dysfunction. This study aims to investigate the molecular and functional differences between human induced pluripotent stem cells (hiPSCs) derived from patients with sporadic AD (sAD) and age-matched controls (healthy subjects, HS), focusing on their neuronal differentiation and synaptic properties in order to better understand the cellular and molecular mechanisms underlying AD pathology. METHODS Skin fibroblasts from sAD patients (n = 5) and HS subjects (n = 5) were reprogrammed into hiPSCs using non-integrating Sendai virus vectors. Through karyotyping, we assessed pluripotency markers (OCT4, SOX2, TRA-1-60) and genomic integrity. Neuronal differentiation was evaluated by immunostaining for MAP2 and NEUN. Electrophysiological properties were measured using whole-cell patch-clamp, while protein expression of Aβ, phosphorylated tau, Synapsin-1, Synaptophysin, PSD95, and GluA1 was quantified by western blot. We then focused on PAK1-LIMK1-Cofilin signaling, which plays a key role in regulating synaptic structure and function, both of which are disrupted in neurodegenerative diseases such as AD. RESULTS sAD and HS hiPSCs displayed similar stemness features and genomic stability. However, they differed in neuronal differentiation and function. sAD-derived neurons (sAD-hNs) displayed increased levels of AD-related proteins, including Aβ and phosphorylated tau. Electrophysiological analyses revealed that while both sAD- and HS-hNs generated action potentials, sAD-hNs exhibited decreased spontaneous synaptic activity. Significant reductions in the expression of synaptic proteins such as Synapsin-1, Synaptophysin, PSD95, and GluA1 were found in sAD-hNs, which are also characterized by reduced neurite length, indicating impaired differentiation. Notably, sAD-hNs demonstrated a marked reduction in LIMK1 phosphorylation, which could be the underlying cause for the changes in cytoskeletal dynamics that we found, leading to the morphological and functional modifications observed in sAD-hNs. To further investigate the involvement of the LIMK1 pathway in the morphological and functional changes observed in sAD neurons, we conducted perturbation experiments using the specific LIMK1 inhibitor, BMS-5. Neurons obtained from healthy subjects treated with the inhibitor showed similar morphological changes to those observed in sAD neurons, confirming that LIMK1 activity is crucial for maintaining normal neuronal structure. Furthermore, administration of the inhibitor to sAD neurons did not exacerbate the morphological alterations, suggesting that LIMK1 activity is already compromised in these cells. CONCLUSION Our findings demonstrate that although sAD- and HS-hiPSCs are similar in their stemness and genomic stability, sAD-hNs exhibit distinct functional and structural anomalies mirroring AD pathology. These anomalies include synaptic dysfunction, altered cytoskeletal organization, and accumulation of AD-related proteins. Our study underscores the usefulness of hiPSCs in modeling AD and provides insights into the disease's molecular underpinnings, thus highlighting potential therapeutic targets.
Collapse
Affiliation(s)
- Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Claudia Colussi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Engineering, Istituto Di Analisi Dei Sistemi Ed Informatica "Antonio Ruberti", National Research Council, 00185, Rome, Italy
| | | | | | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
- Saint Camillus International, University of Health Sciences, 00131, Rome, Italy
| | - Agnese Novelli
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sabrina Maietta
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesco Danilo Tiziano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Camillo Marra
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
3
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Organization of a cytoskeletal superstructure in the apical domain of intestinal tuft cells. J Cell Biol 2024; 223:e202404070. [PMID: 39352498 PMCID: PMC11457492 DOI: 10.1083/jcb.202404070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Tuft cells are a rare epithelial cell type that play important roles in sensing and responding to luminal antigens. A defining morphological feature of this lineage is the actin-rich apical "tuft," which contains large fingerlike protrusions. However, details of the cytoskeletal ultrastructure underpinning the tuft, the molecules involved in building this structure, or how it supports tuft cell biology remain unclear. In the context of the small intestine, we found that tuft cell protrusions are supported by long-core bundles that consist of F-actin crosslinked in a parallel and polarized configuration; they also contain a tuft cell-specific complement of actin-binding proteins that exhibit regionalized localization along the bundle axis. Remarkably, in the sub-apical cytoplasm, the array of core actin bundles interdigitates and co-aligns with a highly ordered network of microtubules. The resulting cytoskeletal superstructure is well positioned to support subcellular transport and, in turn, the dynamic sensing functions of the tuft cell that are critical for intestinal homeostasis.
Collapse
Affiliation(s)
- Jennifer B. Silverman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Evan E. Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ken S. Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
4
|
Falconieri A, Folino P, Da Palmata L, Raffa V. Nano-pulling stimulates axon regeneration in dorsal root ganglia by inducing stabilization of axonal microtubules and activation of local translation. Front Mol Neurosci 2024; 17:1340958. [PMID: 38633213 PMCID: PMC11022966 DOI: 10.3389/fnmol.2024.1340958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Axonal plasticity is strongly related to neuronal development as well as regeneration. It was recently demonstrated that active mechanical tension, intended as an extrinsic factor, is a valid contribution to the modulation of axonal plasticity. Methods In previous publications, our team validated a the "nano-pulling" method used to apply mechanical forces to developing axons of isolated primary neurons using magnetic nanoparticles (MNP) actuated by static magnetic fields. This method was found to promote axon growth and synaptic maturation. Here, we explore the use of nano-pulling as an extrinsic factor to promote axon regeneration in a neuronal tissue explant. Results Whole dorsal root ganglia (DRG) were thus dissected from a mouse spinal cord, incubated with MNPs, and then stretched. We found that particles were able to penetrate the ganglion and thus become localised both in the somas and in sprouting axons. Our results highlight that nano-pulling doubles the regeneration rate, and this is accompanied by an increase in the arborizing capacity of axons, an accumulation of cellular organelles related to mass addition (endoplasmic reticulum and mitochondria) and pre-synaptic proteins with respect to spontaneous regeneration. In line with the previous results on isolated hippocampal neurons, we observed that this process is coupled to an increase in the density of stable microtubules and activation of local translation. Discussion Our data demonstrate that nano-pulling enhances axon regeneration in whole spinal ganglia exposed to MNPs and external magnetic fields. These preliminary data represent an encouraging starting point for proposing nano-pulling as a biophysical tool for the design of novel therapies based on the use of force as an extrinsic factor for promoting nerve regeneration.
Collapse
|
5
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Intestinal tuft cells assemble a cytoskeletal superstructure composed of co-aligned actin bundles and microtubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585757. [PMID: 38562898 PMCID: PMC10983963 DOI: 10.1101/2024.03.19.585757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background & Aims All tissues consist of a distinct set of cell types, which collectively support organ function and homeostasis. Tuft cells are a rare epithelial cell type found in diverse epithelia, where they play important roles in sensing antigens and stimulating downstream immune responses. Exhibiting a unique polarized morphology, tuft cells are defined by an array of giant actin filament bundles that support ∼2 μm of apical membrane protrusion and extend over 7 μm towards the cell's perinuclear region. Despite their established roles in maintaining intestinal epithelial homeostasis, tuft cells remain understudied due to their rarity (e.g. ∼ 1% in the small intestinal epithelium). Details regarding the ultrastructural organization of the tuft cell cytoskeleton, the molecular components involved in building the array of giant actin bundles, and how these cytoskeletal structures support tuft cell biology remain unclear. Methods To begin to answer these questions, we used advanced light and electron microscopy to perform quantitative morphometry of the small intestinal tuft cell cytoskeleton. Results We found that tuft cell core bundles consist of actin filaments that are crosslinked in a parallel "barbed-end out" configuration. These polarized structures are also supported by a unique group of tuft cell enriched actin-binding proteins that are differentially localized along the giant core bundles. Furthermore, we found that tuft cell actin bundles are co-aligned with a highly ordered network of microtubules. Conclusions Tuft cells assemble a cytoskeletal superstructure that is well positioned to serve as a track for subcellular transport along the apical-basolateral axis and in turn, support the dynamic sensing functions that are critical for intestinal epithelial homeostasis. SYNOPSIS This research leveraged advanced light and electron microscopy to perform quantitative morphometry of the intestinal tuft cell cytoskeleton. Three-dimensional reconstructions of segmented image data revealed a co-aligned actin-microtubule superstructure that may play a fundamental role in tuft cell function.
Collapse
|
6
|
Gélin M, Schaeffer A, Gaillard J, Guérin C, Vianay B, Orhant-Prioux M, Braun M, Leterrier C, Blanchoin L, Théry M. Microtubules under mechanical pressure can breach dense actin networks. J Cell Sci 2023; 136:jcs261667. [PMID: 37870087 DOI: 10.1242/jcs.261667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The crosstalk between the actin network and microtubules is essential for cell polarity. It orchestrates microtubule organization within the cell, driven by the asymmetry of actin architecture along the cell periphery. The physical intertwining of these networks regulates spatial organization and force distribution in the microtubule network. Although their biochemical interactions are becoming clearer, the mechanical aspects remain less understood. To explore this mechanical interplay, we developed an in vitro reconstitution assay to investigate how dynamic microtubules interact with various actin filament structures. Our findings revealed that microtubules can align and move along linear actin filament bundles through polymerization force. However, they are unable to pass through when encountering dense branched actin meshworks, similar to those present in the lamellipodium along the periphery of the cell. Interestingly, immobilizing microtubules through crosslinking with actin or other means allow the buildup of pressure, enabling them to breach these dense actin barriers. This mechanism offers insights into microtubule progression towards the cell periphery, with them overcoming obstacles within the denser parts of the actin network and ultimately contributing to cell polarity establishment.
Collapse
Affiliation(s)
- Matthieu Gélin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Alexandre Schaeffer
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Jérémie Gaillard
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Benoit Vianay
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 25250 Vestec, Prague West, Czech Republic
| | - Christophe Leterrier
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto, 13385, Marseille, France
| | - Laurent Blanchoin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Manuel Théry
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| |
Collapse
|
7
|
Villard C. Spatial confinement: A spur for axonal growth. Semin Cell Dev Biol 2023; 140:54-62. [PMID: 35927121 DOI: 10.1016/j.semcdb.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/16/2022] [Accepted: 07/16/2022] [Indexed: 01/28/2023]
Abstract
The concept of spatial confinement is the basis of cell positioning and guidance in in vitro studies. In vivo, it reflects many situations faced during embryonic development. In vitro, spatial confinement of neurons is achieved using different technological approaches: adhesive patterning, topographical structuring, microfluidics and the use of hydrogels. The notion of chemical or physical frontiers is particularly central to the behaviors of growth cones and neuronal processes under confinement. They encompass phenomena of cell spreading, boundary crossing, and path finding on surfaces with different adhesive properties. However, the most universal phenomenon related to confinement, regardless of how it is implemented, is the acceleration of neuronal growth. Overall, a bi-directional causal link emerges between the shape of the growth cone and neuronal elongation dynamics, both in vivo and in vitro. The sensing of adhesion discontinuities by filopodia and the subsequent spatial redistribution and size adaptation of these actin-rich filaments seem critical for the growth rate in conditions in which adhesive contacts and actin-associated clutching forces dominate. On the other hand, the involvement of microtubules, specifically demonstrated in 3D hydrogel environments and leading to ameboid-like locomotion, could be relevant in a wider range of growth situations. This review brings together a literature collected in distinct scientific fields such as development, mechanobiology and bioengineering that highlight the consequences of confinement and raise new questions at different cellular scales. Its ambition is to stimulate new research that could lead to a better understanding of what gives neurons their ability to establish and regulate their exceptional size.
Collapse
Affiliation(s)
- Catherine Villard
- Laboratoire Interdisciplinaire des Energies de Demain (LIED), Université Paris Cité, UMR 8236 CNRS, F-75013 Paris, France.
| |
Collapse
|
8
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
9
|
Raffa V. Force: A messenger of axon outgrowth. Semin Cell Dev Biol 2023; 140:3-12. [PMID: 35817654 DOI: 10.1016/j.semcdb.2022.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 01/28/2023]
Abstract
The axon is a sophisticated macromolecular machine composed of interrelated parts that transmit signals like spur gears transfer motion between parallel shafts. The growth cone is a fine sensor that integrates mechanical and chemical cues and transduces these signals through the generation of a traction force that pushes the tip and pulls the axon shaft forward. The axon shaft, in turn, senses this pulling force and transduces this signal in an orchestrated response, coordinating cytoskeleton remodeling and intercalated mass addition to sustain and support the advancing of the tip. Extensive research suggests that the direct application of active force is per se a powerful inducer of axon growth, potentially bypassing the contribution of the growth cone. This review provides a critical perspective on current knowledge of how the force is a messenger of axon growth and its mode of action for controlling navigation, including aspects that remain unclear. It also focuses on novel approaches and tools designed to mechanically manipulate axons, and discusses their implications in terms of potential novel therapies for re-wiring the nervous system.
Collapse
Affiliation(s)
- Vittoria Raffa
- Department of Biology, University of Pisa, SS12 Abetone e Brennero, 4, 56127 Pisa, Italy.
| |
Collapse
|
10
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
12
|
Ros O, Nicol X. Axon pathfinding and targeting: (R)evolution of insights from in vitro assays. Neuroscience 2023; 508:110-122. [PMID: 36096337 DOI: 10.1016/j.neuroscience.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 01/17/2023]
Abstract
Investigating axonal behaviors while neurons are connecting with each other has been a challenge since the early studies on nervous system development. While molecule-driven axon pathfinding has been theorized by observing neurons at different developmental stages in vivo, direct observation and measurements of axon guidance behaviors required the invention of in vitro systems enabling to test the impact of molecules or cellular extracts on axons growing in vitro. With time, the development of novel in vivo approaches has confirmed the mechanisms highlighted in culture and has led in vitro systems to be adapted for cellular processes that are still inaccessible in intact organisms. We here review the evolution of these in vitro assays, which started with crucial contributions from the Bonhoeffer lab.
Collapse
Affiliation(s)
- Oriol Ros
- Universitat de Barcelona, Department of Cell Biology, Physiology and Immunology, Avinguda Diagonal 643, 08028 Barcelona, Catalonia, Spain
| | - Xavier Nicol
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
13
|
Burute M, Jansen KI, Mihajlovic M, Vermonden T, Kapitein LC. Local changes in microtubule network mobility instruct neuronal polarization and axon specification. SCIENCE ADVANCES 2022; 8:eabo2343. [PMID: 36332030 PMCID: PMC9635826 DOI: 10.1126/sciadv.abo2343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The polarization of neurons into axons and dendrites depends on extracellular cues, intracellular signaling, cytoskeletal rearrangements, and polarized transport, but the interplay between these processes during polarization remains unresolved. Here, we show that axon specification is determined by differences in microtubule network mobility between neurites, regulated by Rho guanosine triphosphatases (GTPases) and extracellular cues. In developing neurons, retrograde microtubule flow prevents the entry of the axon-selective motor protein Kinesin-1 into most neurites. Using inducible assays to control microtubule network flow, we demonstrate that local inhibition of microtubule mobility is sufficient to guide Kinesin-1 into a specific neurite, whereas long-term global inhibition induces the formation of multiple axons. We furthermore show that extracellular mechanical cues and intracellular Rho GTPase signaling control the local differences in microtubule network flow. These results reveal a novel cytoskeletal mechanism for neuronal polarization.
Collapse
Affiliation(s)
- Mithila Burute
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Klara I. Jansen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Marko Mihajlovic
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, Netherlands
| | - Tina Vermonden
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, Netherlands
| | - Lukas C. Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| |
Collapse
|
14
|
Ovejero-Sánchez M, Asensio-Juárez G, González M, Puebla P, Vicente-Manzanares M, Pélaez R, González-Sarmiento R, Herrero AB. Panobinostat Synergistically Enhances the Cytotoxicity of Microtubule Destabilizing Drugs in Ovarian Cancer Cells. Int J Mol Sci 2022; 23:13019. [PMID: 36361809 PMCID: PMC9657298 DOI: 10.3390/ijms232113019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 12/02/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecologic neoplasia and has the highest mortality rate, which is mainly due to late-stage diagnosis and chemotherapy resistance. There is an urgent need to explore new and better therapeutic strategies. We have previously described a family of Microtubule Destabilizing Sulfonamides (MDS) that does not trigger multidrug-mediated resistance in OC cell lines. MDS bind to the colchicine site of tubulin, disrupting the microtubule network and causing antiproliferative and cytotoxic effects. In this work, a novel microtubule-destabilizing agent (PILA9) was synthetized and characterized. This compound also inhibited OC cell proliferation and induced G2/M cell cycle arrest and apoptosis. Interestingly, PILA9 was significantly more cytotoxic than MDS. Here, we also analyzed the effect of these microtubule-destabilizing agents (MDA) in combination with Panobinostat, a pan-histone deacetylase inhibitor. We found that Panobinostat synergistically enhanced MDA-cytotoxicity. Mechanistically, we observed that Panobinostat and MDA induced α-tubulin acetylation and that the combination of both agents enhanced this effect, which could be related to the observed synergy. Altogether, our results suggest that MDA/Panobinostat combinations could represent new therapeutic strategies against OC.
Collapse
Affiliation(s)
- María Ovejero-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Gloria Asensio-Juárez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Myriam González
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Pilar Puebla
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Rafael Pélaez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Rogelio González-Sarmiento
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| | - Ana Belén Herrero
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007 Salamanca, Spain
| |
Collapse
|
15
|
Lin CH, Chen YC, Chan SP, Ou CY. TIAM-1 differentially regulates dendritic and axonal microtubule organization in patterning neuronal development through its multiple domains. PLoS Genet 2022; 18:e1010454. [PMID: 36223408 PMCID: PMC9612824 DOI: 10.1371/journal.pgen.1010454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 10/27/2022] [Accepted: 09/29/2022] [Indexed: 12/02/2022] Open
Abstract
Axon and dendrite development require the cooperation of actin and microtubule cytoskeletons. Microtubules form a well-organized network to direct polarized trafficking and support neuronal processes formation with distinct actin structures. However, it is largely unknown how cytoskeleton regulators differentially regulate microtubule organization in axon and dendrite development. Here, we characterize the role of actin regulators in axon and dendrite development and show that the RacGEF TIAM-1 regulates dendritic patterns through its N-terminal domains and suppresses axon growth through its C-terminal domains. TIAM-1 maintains plus-end-out microtubule orientation in posterior dendrites and prevents the accumulation of microtubules in the axon. In somatodendritic regions, TIAM-1 interacts with UNC-119 and stabilizes the organization between actin filaments and microtubules. UNC-119 is required for TIAM-1 to control axon growth, and its expression levels determine axon length. Taken together, TIAM-1 regulates neuronal microtubule organization and patterns axon and dendrite development respectively through its different domains.
Collapse
Affiliation(s)
- Chih-Hsien Lin
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Chun Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Peng Chan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chan-Yen Ou
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
16
|
Vela-Alcantara AM, Rios-Ramirez A, Santiago-Garcia J, Rodriguez-Alba JC, Tamariz Domínguez E. Modulation of DRG neurons response to semaphorin 3A via substrate stiffness. Cells Dev 2022; 171:203800. [PMID: 35717026 DOI: 10.1016/j.cdev.2022.203800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/25/2023]
Abstract
Semaphorin 3A (Sema3a) is a chemotropic protein that acts as a neuronal guidance cue and plays a major role in dorsal root ganglion (DRG) sensory neurons projection during embryo development. The present study evaluated the impact of stiffness in the repulsive response of DRG neurons to Sema3a when cultured over substrates of variable stiffness. Stiffness modified DRG neurons morphology and regulated their response to Sema3a, reducing the collapse of growth cones when they were cultured on softer substrates. Sema3a receptors expression was also regulated by stiffness, neuropilin-1 was overexpressed and plexin A4 mRNA was downregulated in stiffer substrates. Cytoskeleton distribution was also modified by stiffness. In softer substrates, βIII-tubulin and actin co-localized up to the leading edge of the growth cones, and as the substrate became stiffer, βIII-tubulin was confined to the transition and peripheral domains of the growth cone. Moreover, a decrease in the α-actinin adaptor protein was also observed in softer substrates. Our results show that substrate stiffness plays an important role in regulating the collapse response to Sema3a and that the modulation of cytoskeleton distribution and Sema3a receptors expression are related to the differential collapse responses of the growth cones.
Collapse
Affiliation(s)
- Ana Monserrat Vela-Alcantara
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico; Maestría y Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Mexico.
| | - Ariadna Rios-Ramirez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Blvd. Juriquilla #3001, 76230 Juriquilla, Querétaro, Mexico.
| | - Juan Santiago-Garcia
- Instituto de Investigaciones Biológicas, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| | - Juan Carlos Rodriguez-Alba
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| | - Elisa Tamariz Domínguez
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Av. Luis Castelazo Ayala s/n, 91190 Xalapa, Veracruz, Mexico.
| |
Collapse
|
17
|
Higgs VE, Das RM. Establishing neuronal polarity: microtubule regulation during neurite initiation. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac007. [PMID: 38596701 PMCID: PMC10913830 DOI: 10.1093/oons/kvac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 04/11/2024]
Abstract
The initiation of nascent projections, or neurites, from the neuronal cell body is the first stage in the formation of axons and dendrites, and thus a critical step in the establishment of neuronal architecture and nervous system development. Neurite formation relies on the polarized remodelling of microtubules, which dynamically direct and reinforce cell shape, and provide tracks for cargo transport and force generation. Within neurons, microtubule behaviour and structure are tightly controlled by an array of regulatory factors. Although microtubule regulation in the later stages of axon development is relatively well understood, how microtubules are regulated during neurite initiation is rarely examined. Here, we discuss how factors that direct microtubule growth, remodelling, stability and positioning influence neurite formation. In addition, we consider microtubule organization by the centrosome and modulation by the actin and intermediate filament networks to provide an up-to-date picture of this vital stage in neuronal development.
Collapse
Affiliation(s)
- Victoria E Higgs
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Raman M Das
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
18
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
19
|
Atherton J, Stouffer M, Francis F, Moores CA. Visualising the cytoskeletal machinery in neuronal growth cones using cryo-electron tomography. J Cell Sci 2022; 135:274968. [PMID: 35383828 PMCID: PMC9016625 DOI: 10.1242/jcs.259234] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Neurons extend axons to form the complex circuitry of the mature brain. This depends on the coordinated response and continuous remodelling of the microtubule and F-actin networks in the axonal growth cone. Growth cone architecture remains poorly understood at nanoscales. We therefore investigated mouse hippocampal neuron growth cones using cryo-electron tomography to directly visualise their three-dimensional subcellular architecture with molecular detail. Our data showed that the hexagonal arrays of actin bundles that form filopodia penetrate and terminate deep within the growth cone interior. We directly observed the modulation of these and other growth cone actin bundles by alteration of individual F-actin helical structures. Microtubules with blunt, slightly flared or gently curved ends predominated in the growth cone, frequently contained lumenal particles and exhibited lattice defects. Investigation of the effect of absence of doublecortin, a neurodevelopmental cytoskeleton regulator, on growth cone cytoskeleton showed no major anomalies in overall growth cone organisation or in F-actin subpopulations. However, our data suggested that microtubules sustained more structural defects, highlighting the importance of microtubule integrity during growth cone migration. Summary: Cryo-electron tomographic reconstruction of neuronal growth cone subdomains reveals distinctive F-actin and microtubule cytoskeleton architectures and modulation at molecular detail.
Collapse
Affiliation(s)
- Joseph Atherton
- Randall Centre for Cell and Molecular Biophysics, King's College, London SE1 1YR, UK.,Institute of Structural and Molecular Biology, Birkbeck, University of London, London WC1E 7HX, UK
| | - Melissa Stouffer
- INSERM UMR-S 1270, 17 Rue du Fer à Moulin, 75005 Paris, France.,Sorbonne University UMR-S 1270, 4 Place Jussieu, 75005 Paris, France.,Institut du Fer à Moulin, 17 Rue du Fer à Moulin, 75005 Paris, France.,Institute of Science and Technology Austria, Am campus 1, 3400 Klosterneuberg, Austria
| | - Fiona Francis
- INSERM UMR-S 1270, 17 Rue du Fer à Moulin, 75005 Paris, France.,Sorbonne University UMR-S 1270, 4 Place Jussieu, 75005 Paris, France.,Institut du Fer à Moulin, 17 Rue du Fer à Moulin, 75005 Paris, France
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck, University of London, London WC1E 7HX, UK
| |
Collapse
|
20
|
Del Castillo U, Norkett R, Lu W, Serpinskaya A, Gelfand VI. Ataxin-2 is essential for cytoskeletal dynamics and neurodevelopment in Drosophila. iScience 2022; 25:103536. [PMID: 34977501 PMCID: PMC8689088 DOI: 10.1016/j.isci.2021.103536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/19/2021] [Accepted: 11/25/2021] [Indexed: 12/03/2022] Open
Abstract
Ataxin-2 (Atx2) is a highly conserved RNA binding protein. Atx2 undergoes polyglutamine expansion leading to amyotrophic lateral sclerosis (ALS) or spinocerebellar ataxia type 2 (SCA2). However, the physiological functions of Atx2 in neurons remain unknown. Here, using the powerful genetics of Drosophila, we show that Atx2 is essential for normal neuronal cytoskeletal dynamics and organelle trafficking. Upon neuron-specific Atx2 loss, the microtubule and actin networks were abnormally stabilized and cargo transport was drastically inhibited. Depletion of Atx2 caused multiple morphological defects in the nervous system of third instar larvae. These include reduced brain size, impaired axon development, and decreased dendrite outgrowth. Defects in the nervous system caused loss of the ability to crawl and lethality at the pupal stage. Taken together, these data mark Atx2 as a major regulator of cytoskeletal dynamics and denote Atx2 as an essential gene in neurodevelopment, as well as a neurodegenerative factor. Atx2 is a major regulator of the cytoskeleton in neurons Atx2 is responsible for maintaining dynamic cytoskeletal networks Atx2 depletion in the Drosophila larval CNS severely impairs organelle transport Atx2 is necessary for correct neurite outgrowth and CNS development in Drosophila
Collapse
Affiliation(s)
- Urko Del Castillo
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rosalind Norkett
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Wen Lu
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anna Serpinskaya
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Vladimir I Gelfand
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
21
|
Kano T, Tsumagari R, Nakashima A, Kikkawa U, Ueda S, Yamanoue M, Takei N, Shirai Y. RalA, PLD and mTORC1 Are Required for Kinase-Independent Pathways in DGKβ-Induced Neurite Outgrowth. Biomolecules 2021; 11:1814. [PMID: 34944458 PMCID: PMC8699322 DOI: 10.3390/biom11121814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Diacylglycerol kinase β (DGKβ) is an enzyme that converts diacylglycerol to phosphatidic acid and is mainly expressed in the cerebral cortex, hippocampus and striatum. We previously reported that DGKβ induces neurite outgrowth and spinogenesis, contributing to higher brain functions, including emotion and memory. To elucidate the mechanisms involved in neuronal development by DGKβ, we investigated the importance of DGKβ activity in the induction of neurite outgrowth using human neuroblastoma SH-SY5Y cells. Interestingly, both wild-type DGKβ and the kinase-negative (KN) mutant partially induced neurite outgrowth, and these functions shared a common pathway via the activation of mammalian target of rapamycin complex 1 (mTORC1). In addition, we found that DGKβ interacted with the small GTPase RalA and that siRNA against RalA and phospholipase D (PLD) inhibitor treatments abolished DGKβKN-induced neurite outgrowth. These results indicate that binding of RalA and activation of PLD and mTORC1 are involved in DGKβKN-induced neurite outgrowth. Taken together with our previous reports, mTORC1 is a key molecule in both kinase-dependent and kinase-independent pathways of DGKβ-mediated neurite outgrowth, which is important for higher brain functions.
Collapse
Affiliation(s)
- Takuya Kano
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Ryosuke Tsumagari
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Akio Nakashima
- Division of Signal Functions, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan; (A.N.); (U.K.)
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Ushio Kikkawa
- Division of Signal Functions, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan; (A.N.); (U.K.)
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Shuji Ueda
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Minoru Yamanoue
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan;
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences, Kobe University, Kobe 657-8501, Japan; (T.K.); (R.T.); (S.U.); (M.Y.)
| |
Collapse
|
22
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
23
|
Ming Y, Abedin MJ, Tatic-Lucic S, Berdichevsky Y. Microdevice for directional axodendritic connectivity between micro 3D neuronal cultures. MICROSYSTEMS & NANOENGINEERING 2021; 7:67. [PMID: 34567779 PMCID: PMC8433170 DOI: 10.1038/s41378-021-00292-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/27/2021] [Accepted: 06/20/2021] [Indexed: 06/13/2023]
Abstract
Neuronal cultures are widely used in neuroscience research. However, the randomness of circuits in conventional cultures prevents accurate in vitro modeling of cortical development and of the pathogenesis of neurological and psychiatric disorders. A basic feature of cortical circuits that is not captured in standard cultures of dissociated cortical cells is directional connectivity. In this work, a polydimethylsiloxane (PDMS)-based device that achieves directional connectivity between micro 3D cultures is demonstrated. The device consists of through-holes for micro three-dimensional (μ3D) clusters of cortical cells connected by microtrenches for axon and dendrite guidance. The design of the trenches relies in part on the concept of axonal edge guidance, as well as on the novel concept of specific dendrite targeting. This replicates dominant excitatory connectivity in the cortex, enables the guidance of the axon after it forms a synapse in passing (an "en passant" synapse), and ensures that directional selectivity is preserved over the lifetime of the culture. The directionality of connections was verified morphologically and functionally. Connections were dependent on glutamatergic synapses. The design of this device has the potential to serve as a building block for the reconstruction of more complex cortical circuits in vitro.
Collapse
Affiliation(s)
- Yixuan Ming
- Department of Electrical & Computer Engineering, Lehigh University, Bethlehem, PA USA
| | - Md Joynal Abedin
- Department of Bioengineering, Lehigh University, Bethlehem, PA USA
| | - Svetlana Tatic-Lucic
- Department of Electrical & Computer Engineering, Lehigh University, Bethlehem, PA USA
- Department of Bioengineering, Lehigh University, Bethlehem, PA USA
| | - Yevgeny Berdichevsky
- Department of Electrical & Computer Engineering, Lehigh University, Bethlehem, PA USA
- Department of Bioengineering, Lehigh University, Bethlehem, PA USA
| |
Collapse
|
24
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
25
|
Kundu T, Dutta P, Nagar D, Maiti S, Ghose A. Coupling of dynamic microtubules to F-actin by Fmn2 regulates chemotaxis of neuronal growth cones. J Cell Sci 2021; 134:jcs252916. [PMID: 34313311 DOI: 10.1242/jcs.252916] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Dynamic co-regulation of the actin and microtubule subsystems enables the highly precise and adaptive remodelling of the cytoskeleton necessary for critical cellular processes, such as axonal pathfinding. The modes and mediators of this interpolymer crosstalk, however, are inadequately understood. We identify Fmn2, a non-diaphanous-related formin associated with cognitive disabilities, as a novel regulator of cooperative actin-microtubule remodelling in growth cones of both chick and zebrafish neurons. We show that Fmn2 stabilizes microtubules in the growth cones of cultured spinal neurons and in vivo. Super-resolution imaging revealed that Fmn2 facilitates guidance of exploratory microtubules along actin bundles into the chemosensory filopodia. Using live imaging, biochemistry and single-molecule assays, we show that a C-terminal domain in Fmn2 is necessary for the dynamic association between microtubules and actin filaments. In the absence of the cross-bridging function of Fmn2, filopodial capture of microtubules is compromised, resulting in destabilized filopodial protrusions and deficits in growth cone chemotaxis. Our results uncover a critical function for Fmn2 in actin-microtubule crosstalk in neurons and demonstrate that the modulation of microtubule dynamics via associations with F-actin is central to directional motility.
Collapse
Affiliation(s)
- Tanushree Kundu
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Priyanka Dutta
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Dhriti Nagar
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| | - Sankar Maiti
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, Nadia, West Bengal, India
| | - Aurnab Ghose
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhabha Road, Pune 411008, India
| |
Collapse
|
26
|
Hou X, Nozumi M, Nakamura H, Igarashi M, Sugiyama S. Coactosin Promotes F-Actin Protrusion in Growth Cones Under Cofilin-Related Signaling Pathway. Front Cell Dev Biol 2021; 9:660349. [PMID: 34235144 PMCID: PMC8256272 DOI: 10.3389/fcell.2021.660349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/31/2021] [Indexed: 12/03/2022] Open
Abstract
During brain development, axon outgrowth and its subsequent pathfinding are reliant on a highly motile growth cone located at the tip of the axon. Actin polymerization that is regulated by actin-depolymerizing factors homology (ADF-H) domain-containing family drives the formation of lamellipodia and filopodia at the leading edge of growth cones for axon guidance. However, the precise localization and function of ADF-H domain-containing proteins involved in axon extension and retraction remain unclear. We have previously shown that transcripts and proteins of coactosin-like protein 1 (COTL1), an ADF-H domain-containing protein, are observed in neurites and axons in chick embryos. Coactosin overexpression analysis revealed that this protein was localized to axonal growth cones and involved in axon extension in the midbrain. We further examined the specific distribution of coactosin and cofilin within the growth cone using superresolution microscopy, structured illumination microscopy, which overcomes the optical diffraction limitation and is suitable to the analysis of cellular dynamic movements. We found that coactosin was tightly associated with F-actin bundles at the growth cones and that coactosin overexpression promoted the expansion of lamellipodia and extension of growth cones. Coactosin knockdown in oculomotor neurons resulted in an increase in the levels of the inactive, phosphorylated form of cofilin and dysregulation of actin polymerization and axonal elongation, which suggests that coactosin promoted axonal growth in a cofilin-dependent manner. Indeed, the application of a dominant-negative form of LIMK1, a downstream effector of GTPases, reversed the effect of coactosin knockdown on axonal growth by enhancing cofilin activity. Combined, our results indicate that coactosin functions promote the assembly of protrusive actin filament arrays at the leading edge for growth cone motility.
Collapse
Affiliation(s)
- Xubin Hou
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Harukazu Nakamura
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sayaka Sugiyama
- Laboratory of Neuronal Development, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
27
|
Blazejewski SM, Bennison SA, Liu X, Toyo-Oka K. High-throughput kinase inhibitor screening reveals roles for Aurora and Nuak kinases in neurite initiation and dendritic branching. Sci Rep 2021; 11:8156. [PMID: 33854138 PMCID: PMC8047044 DOI: 10.1038/s41598-021-87521-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2021] [Indexed: 12/25/2022] Open
Abstract
Kinases are essential regulators of a variety of cellular signaling processes, including neurite formation—a foundational step in neurodevelopment. Aberrant axonal sprouting and failed regeneration of injured axons are associated with conditions like traumatic injury, neurodegenerative disease, and seizures. Investigating the mechanisms underlying neurite formation will allow for identification of potential therapeutics. We used a kinase inhibitor library to screen 493 kinase inhibitors and observed that 45% impacted neuritogenesis in Neuro2a (N-2a) cells. Based on the screening, we further investigated the roles of Aurora kinases A, B, and C and Nuak kinases 1 and 2. The roles of Aurora and Nuak kinases have not been thoroughly studied in the nervous system. Inhibition or overexpression of Aurora and Nuak kinases in primary cortical neurons resulted in various neuromorphological defects, with Aurora A regulating neurite initiation, Aurora B and C regulating neurite initiation and elongation, all Aurora kinases regulating arborization, and all Nuak kinases regulating neurite initiation and elongation and arborization. Our high-throughput screening and analysis of Aurora and Nuak kinases revealed their functions and may contribute to the identification of therapeutics.
Collapse
Affiliation(s)
- Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
28
|
Mini-review: Microtubule sliding in neurons. Neurosci Lett 2021; 753:135867. [PMID: 33812935 DOI: 10.1016/j.neulet.2021.135867] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022]
Abstract
Microtubule sliding is an underappreciated mechanism that contributes to the establishment, organization, preservation, and plasticity of neuronal microtubule arrays. Powered by molecular motor proteins and regulated in part by static crosslinker proteins, microtubule sliding is the movement of microtubules relative to other microtubules or to non-microtubule structures such as the actin cytoskeleton. In addition to other important functions, microtubule sliding significantly contributes to the establishment and maintenance of microtubule polarity patterns in different regions of the neuron. The purpose of this article is to review the state of knowledge on microtubule sliding in the neuron, with emphasis on its mechanistic underpinnings as well as its functional significance.
Collapse
|
29
|
Liu X, Blazejewski SM, Bennison SA, Toyo-oka K. Glutathione S-transferase Pi (Gstp) proteins regulate neuritogenesis in the developing cerebral cortex. Hum Mol Genet 2021; 30:30-45. [PMID: 33437989 PMCID: PMC8033146 DOI: 10.1093/hmg/ddab003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022] Open
Abstract
GSTP proteins are metabolic enzymes involved in the removal of oxidative stress and intracellular signaling and also have inhibitory effects on JNK activity. However, the functions of Gstp proteins in the developing brain are unknown. In mice, there are three Gstp proteins, Gstp1, 2 and 3, whereas there is only one GSTP in humans. By reverse transcription-polymerase chain reaction (RT-PCR) analysis, we found that Gstp1 was expressed beginning at E15.5 in the cortex, but Gstp2 and 3 started expressing at E18.5. Gstp 1 and 2 knockdown (KD) caused decreased neurite number in cortical neurons, implicating them in neurite initiation. Using in utero electroporation (IUE) to knock down Gstp1 and 2 in layer 2/3 pyramidal neurons in vivo, we found abnormal swelling of the apical dendrite at P3 and reduced neurite number at P15. Using time-lapse live imaging, we found that the apical dendrite orientation was skewed compared with the control. We explored the molecular mechanism and found that JNK inhibition rescued reduced neurite number caused by Gstp knockdown, indicating that Gstp regulates neurite formation through JNK signaling. Thus, we found novel functions of Gstp proteins in neurite initiation during cortical development. These findings not only provide novel functions of Gstp proteins in neuritogenesis during cortical development but also help us to understand the complexity of neurite formation.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| | - Kazuhito Toyo-oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| |
Collapse
|
30
|
Sánchez-Huertas C, Bonhomme M, Falco A, Fagotto-Kaufmann C, van Haren J, Jeanneteau F, Galjart N, Debant A, Boudeau J. The +TIP Navigator-1 is an actin-microtubule crosslinker that regulates axonal growth cone motility. J Cell Biol 2021; 219:151835. [PMID: 32497170 PMCID: PMC7480110 DOI: 10.1083/jcb.201905199] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 04/03/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Microtubule (MT) plus-end tracking proteins (+TIPs) are central players in the coordination between the MT and actin cytoskeletons in growth cones (GCs) during axon guidance. The +TIP Navigator-1 (NAV1) is expressed in the developing nervous system, yet its neuronal functions remain poorly elucidated. Here, we report that NAV1 controls the dynamics and motility of the axonal GCs of cortical neurons in an EB1-dependent manner and is required for axon turning toward a gradient of netrin-1. NAV1 accumulates in F-actin-rich domains of GCs and binds actin filaments in vitro. NAV1 can also bind MTs independently of EB1 in vitro and crosslinks nonpolymerizing MT plus ends to actin filaments in axonal GCs, preventing MT depolymerization in F-actin-rich areas. Together, our findings pinpoint NAV1 as a key player in the actin-MT crosstalk that promotes MT persistence at the GC periphery and regulates GC steering. Additionally, we present data assigning to NAV1 an important role in the radial migration of cortical projection neurons in vivo.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Marion Bonhomme
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Amandine Falco
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Christine Fagotto-Kaufmann
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Jeffrey van Haren
- Department of Cell Biology and Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Niels Galjart
- Department of Cell Biology and Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Debant
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| | - Jérôme Boudeau
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique, Montpellier, France
| |
Collapse
|
31
|
Pinto-Costa R, Sousa MM. Microtubules, actin and cytolinkers: how to connect cytoskeletons in the neuronal growth cone. Neurosci Lett 2021; 747:135693. [PMID: 33529653 DOI: 10.1016/j.neulet.2021.135693] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 01/20/2023]
Abstract
Cytolinkers ensure the integration of the different cytoskeleton components in the neuronal growth cone during development and in the course of axon regeneration. In neurons, an integrated skeleton guarantees appropriate function, and connectivity of high order circuits. Over the past years, several cytoskeleton regulatory proteins with actin-microtubule crosslinking activity have been identified. In neurons, the importance of spectrins, formins and other cytolinkers capable of coupling actin and microtubules has been extensively highlighted during axon outgrowth and guidance. In this Review, we discuss the current knowledge on cytolinkers specifically expressed in the neuronal growth cone of developing and regenerating axons.
Collapse
Affiliation(s)
- Rita Pinto-Costa
- Nerve Regeneration Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
| | - Monica Mendes Sousa
- Nerve Regeneration Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
32
|
Manipulation of Axonal Outgrowth via Exogenous Low Forces. Int J Mol Sci 2020; 21:ijms21218009. [PMID: 33126477 PMCID: PMC7663625 DOI: 10.3390/ijms21218009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Neurons are mechanosensitive cells. The role of mechanical force in the process of neurite initiation, elongation and sprouting; nerve fasciculation; and neuron maturation continues to attract considerable interest among scientists. Force is an endogenous signal that stimulates all these processes in vivo. The axon is able to sense force, generate force and, ultimately, transduce the force in a signal for growth. This opens up fascinating scenarios. How are forces generated and sensed in vivo? Which molecular mechanisms are responsible for this mechanotransduction signal? Can we exploit exogenously applied forces to mimic and control this process? How can these extremely low forces be generated in vivo in a non-invasive manner? Can these methodologies for force generation be used in regenerative therapies? This review addresses these questions, providing a general overview of current knowledge on the applications of exogenous forces to manipulate axonal outgrowth, with a special focus on forces whose magnitude is similar to those generated in vivo. We also review the principal methodologies for applying these forces, providing new inspiration and insights into the potential of this approach for future regenerative therapies.
Collapse
|
33
|
Ghate K, Mutalik SP, Sthanam LK, Sen S, Ghose A. Fmn2 Regulates Growth Cone Motility by Mediating a Molecular Clutch to Generate Traction Forces. Neuroscience 2020; 448:160-171. [PMID: 33002558 DOI: 10.1016/j.neuroscience.2020.09.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/10/2020] [Accepted: 09/22/2020] [Indexed: 01/06/2023]
Abstract
Growth cone-mediated axonal outgrowth and accurate synaptic targeting are central to brain morphogenesis. Translocation of the growth cone necessitates mechanochemical regulation of cell-extracellular matrix interactions and the generation of propulsive traction forces onto the growth environment. However, the molecular mechanisms subserving force generation by growth cones remain poorly characterized. The formin family member, Fmn2, has been identified earlier as a regulator of growth cone motility. Here, we explore the mechanisms underlying Fmn2 function in the growth cone. Evaluation of multiple components of the adhesion complexes suggests that Fmn2 regulates point contact stability. Analysis of F-actin retrograde flow reveals that Fmn2 functions as a clutch molecule and mediates the coupling of the actin cytoskeleton to the growth substrate, via point contact adhesion complexes. Using traction force microscopy, we show that the Fmn2-mediated clutch function is necessary for the generation of traction stresses by neurons. Our findings suggest that Fmn2, a protein associated with neurodevelopmental and neurodegenerative disorders, is a key regulator of a molecular clutch activity and consequently motility of neuronal growth cones.
Collapse
Affiliation(s)
- Ketakee Ghate
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, India
| | - Sampada P Mutalik
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, India
| | - Lakshmi Kavitha Sthanam
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Mumbai 400076, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Mumbai 400076, India
| | - Aurnab Ghose
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, India.
| |
Collapse
|
34
|
Gallo G. The bioenergetics of neuronal morphogenesis and regeneration: Frontiers beyond the mitochondrion. Dev Neurobiol 2020; 80:263-276. [PMID: 32750228 DOI: 10.1002/dneu.22776] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
The formation of axons and dendrites during development, and their regeneration following injury, are energy intensive processes. The underlying assembly and dynamics of the cytoskeleton, axonal transport mechanisms, and extensive signaling networks all rely on ATP and GTP consumption. Cellular ATP is generated through oxidative phosphorylation (OxP) in mitochondria, glycolysis and "regenerative" kinase systems. Recent investigations have focused on the role of the mitochondrion in axonal development and regeneration emphasizing the importance of this organelle and OxP in axon development and regeneration. In contrast, the understanding of alternative sources of ATP in neuronal morphogenesis and regeneration remains largely unexplored. This review focuses on the current state of the field of neuronal bioenergetics underlying morphogenesis and regeneration and considers the literature on the bioenergetics of non-neuronal cell motility to emphasize the potential contributions of non-mitochondrial energy sources.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, PA, USA
| |
Collapse
|
35
|
Costa AR, Sousa MM. Non-Muscle Myosin II in Axonal Cell Biology: From the Growth Cone to the Axon Initial Segment. Cells 2020; 9:cells9091961. [PMID: 32858875 PMCID: PMC7563147 DOI: 10.3390/cells9091961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
By binding to actin filaments, non-muscle myosin II (NMII) generates actomyosin networks that hold unique contractile properties. Their dynamic nature is essential for neuronal biology including the establishment of polarity, growth cone formation and motility, axon growth during development (and axon regeneration in the adult), radial and longitudinal axonal tension, and synapse formation and function. In this review, we discuss the current knowledge on the spatial distribution and function of the actomyosin cytoskeleton in different axonal compartments. We highlight some of the apparent contradictions and open questions in the field, including the role of NMII in the regulation of axon growth and regeneration, the possibility that NMII structural arrangement along the axon shaft may control both radial and longitudinal contractility, and the mechanism and functional purpose underlying NMII enrichment in the axon initial segment. With the advances in live cell imaging and super resolution microscopy, it is expected that in the near future the spatial distribution of NMII in the axon, and the mechanisms by which it participates in axonal biology will be further untangled.
Collapse
|
36
|
Rodemer W, Gallo G, Selzer ME. Mechanisms of Axon Elongation Following CNS Injury: What Is Happening at the Axon Tip? Front Cell Neurosci 2020; 14:177. [PMID: 32719586 PMCID: PMC7347967 DOI: 10.3389/fncel.2020.00177] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
After an injury to the central nervous system (CNS), functional recovery is limited by the inability of severed axons to regenerate and form functional connections with appropriate target neurons beyond the injury. Despite tremendous advances in our understanding of the mechanisms of axon growth, and of the inhibitory factors in the injured CNS that prevent it, disappointingly little progress has been made in restoring function to human patients with CNS injuries, such as spinal cord injury (SCI), through regenerative therapies. Clearly, the large number of overlapping neuron-intrinsic and -extrinsic growth-inhibitory factors attenuates the benefit of neutralizing any one target. More daunting is the distances human axons would have to regenerate to reach some threshold number of target neurons, e.g., those that occupy one complete spinal segment, compared to the distances required in most experimental models, such as mice and rats. However, the difficulties inherent in studying mechanisms of axon regeneration in the mature CNS in vivo have caused researchers to rely heavily on extrapolation from studies of axon regeneration in peripheral nerve, or of growth cone-mediated axon development in vitro and in vivo. Unfortunately, evidence from several animal models, including the transected lamprey spinal cord, has suggested important differences between regeneration of mature CNS axons and growth of axons in peripheral nerve, or during embryonic development. Specifically, long-distance regeneration of severed axons may not involve the actin-myosin molecular motors that guide embryonic growth cones in developing axons. Rather, non-growth cone-mediated axon elongation may be required to propel injured axons in the mature CNS. If so, it may be necessary to use other experimental models to promote regeneration that is sufficient to contact a critical number of target neurons distal to a CNS lesion. This review examines the cytoskeletal underpinnings of axon growth, focusing on the elongating axon tip, to gain insights into how CNS axons respond to injury, and how this might affect the development of regenerative therapies for SCI and other CNS injuries.
Collapse
Affiliation(s)
- William Rodemer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
37
|
McElmurry K, Stone JE, Ma D, Lamoureux P, Zhang Y, Steidemann M, Fix L, Huang F, Miller KE, Suter DM. Dynein-mediated microtubule translocation powering neurite outgrowth in chick and Aplysia neurons requires microtubule assembly. J Cell Sci 2020; 133:133/8/jcs232983. [PMID: 32332091 DOI: 10.1242/jcs.232983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 03/02/2020] [Indexed: 12/22/2022] Open
Abstract
Previously, we have shown that bulk microtubule (MT) movement correlates with neurite elongation, and blocking either dynein activity or MT assembly inhibits both processes. However, whether the contributions of MT dynamics and dynein activity to neurite elongation are separate or interdependent is unclear. Here, we investigated the underlying mechanism by testing the roles of dynein and MT assembly in neurite elongation of Aplysia and chick neurites using time-lapse imaging, fluorescent speckle microscopy, super-resolution imaging and biophysical analysis. Pharmacologically inhibiting either dynein activity or MT assembly reduced neurite elongation rates as well as bulk and individual MT anterograde translocation. Simultaneously suppressing both processes did not have additive effects, suggesting a shared mechanism of action. Single-molecule switching nanoscopy revealed that inhibition of MT assembly decreased the association of dynein with MTs. Finally, inhibiting MT assembly prevented the rise in tension induced by dynein inhibition. Taken together, our results suggest that MT assembly is required for dynein-driven MT translocation and neurite outgrowth.
Collapse
Affiliation(s)
- Kristi McElmurry
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jessica E Stone
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Donghan Ma
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Phillip Lamoureux
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Yueyun Zhang
- Department of Statistics, Purdue University, West Lafayette, IN 47904, USA
| | - Michelle Steidemann
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Lucas Fix
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Fang Huang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Kyle E Miller
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA .,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
38
|
Zhan J, Li X, Luo D, Hou Y, Hou Y, Chen S, Xiao Z, Luan J, Lin D. Polydatin promotes the neuronal differentiation of bone marrow mesenchymal stem cells in vitro and in vivo: Involvement of Nrf2 signalling pathway. J Cell Mol Med 2020; 24:5317-5329. [PMID: 32299154 PMCID: PMC7205798 DOI: 10.1111/jcmm.15187] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/26/2020] [Accepted: 02/23/2020] [Indexed: 12/11/2022] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC) transplantation represents a promising repair strategy following spinal cord injury (SCI), although the therapeutic effects are minimal due to their limited neural differentiation potential. Polydatin (PD), a key component of the Chinese herb Polygonum cuspidatum, exerts significant neuroprotective effects in various central nervous system disorders and protects BMSCs against oxidative injury. However, the effect of PD on the neuronal differentiation of BMSCs, and the underlying mechanisms remain inadequately understood. In this study, we induced neuronal differentiation of BMSCs in the presence of PD, and analysed the Nrf2 signalling and neuronal differentiation markers using routine molecular assays. We also established an in vivo model of SCI and assessed the locomotor function of the mice through hindlimb movements and electrophysiological measurements. Finally, tissue regeneration was evaluated by H&E staining, Nissl staining and transmission electron microscopy. PD (30 μmol/L) markedly facilitated BMSC differentiation into neuron‐like cells by activating the Nrf2 pathway and increased the expression of neuronal markers in the transplanted BMSCs at the injured spinal cord sites. Furthermore, compared with either monotherapy, the combination of PD and BMSC transplantation promoted axonal rehabilitation, attenuated glial scar formation and promoted axonal generation across the glial scar, thereby enhancing recovery of hindlimb locomotor function. Taken together, PD augments the neuronal differentiation of BMSCs via Nrf2 activation and improves functional recovery, indicating a promising new therapeutic approach against SCI.
Collapse
Affiliation(s)
- Jiheng Zhan
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Luo
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Hou
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonghui Hou
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shudong Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhifeng Xiao
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiyao Luan
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dingkun Lin
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
39
|
Bennison SA, Blazejewski SM, Smith TH, Toyo-Oka K. Protein kinases: master regulators of neuritogenesis and therapeutic targets for axon regeneration. Cell Mol Life Sci 2020; 77:1511-1530. [PMID: 31659414 PMCID: PMC7166181 DOI: 10.1007/s00018-019-03336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Proper neurite formation is essential for appropriate neuronal morphology to develop and defects at this early foundational stage have serious implications for overall neuronal function. Neuritogenesis is tightly regulated by various signaling mechanisms that control the timing and placement of neurite initiation, as well as the various processes necessary for neurite elongation to occur. Kinases are integral components of these regulatory pathways that control the activation and inactivation of their targets. This review provides a comprehensive summary of the kinases that are notably involved in regulating neurite formation, which is a complex process that involves cytoskeletal rearrangements, addition of plasma membrane to increase neuronal surface area, coupling of cytoskeleton/plasma membrane, metabolic regulation, and regulation of neuronal differentiation. Since kinases are key regulators of these functions during neuromorphogenesis, they have high potential for use as therapeutic targets for axon regeneration after injury or disease where neurite formation is disrupted.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
40
|
RhoA-GTPase Modulates Neurite Outgrowth by Regulating the Expression of Spastin and p60-Katanin. Cells 2020; 9:cells9010230. [PMID: 31963385 PMCID: PMC7016723 DOI: 10.3390/cells9010230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/17/2022] Open
Abstract
RhoA-GTPase (RhoA) is widely regarded as a key molecular switch to inhibit neurite outgrowth by rigidifying the actin cytoskeleton. However, during neurite outgrowth, whether and how microtubule dynamics are regulated by RhoA remains to be elucidated. Herein, CT04 and Y27632 were used to inactivate RhoA and its downstream effector Rho-associated coiled coil-forming kinase (ROCK), while the RhoAQ63L lentiviral vector was utilized to overexpress the constitutively activated RhoA in dorsal root ganglion (DRG) neurons or neuronal differentiated PC12 cells. The current data illustrate that the RhoA signaling pathway negatively modulates neurite outgrowth and elevates the expression of Glu-tubulin (a marker for a stabilized microtubule). Meanwhile, the microtubule-severing proteins spastin and p60-katanin were downregulated by the RhoA signaling pathway. When spastin and p60-katanin were knocked down, the effects of RhoA inhibition on neurite outgrowth were significantly reversed. Taken together, this study demonstrates that the RhoA pathway-mediated inhibition of neurite outgrowth is not only related to the modulation of microfilament dynamics but is also attributable to the regulation of the expression of spastin and p60-katanin and thus influences microtubule dynamics.
Collapse
|
41
|
Nakai H, Tsumagari R, Maruo K, Nakashima A, Kikkawa U, Ueda S, Yamanoue M, Saito N, Takei N, Shirai Y. mTORC1 is involved in DGKβ-induced neurite outgrowth and spinogenesis. Neurochem Int 2019; 134:104645. [PMID: 31891737 DOI: 10.1016/j.neuint.2019.104645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/30/2019] [Accepted: 12/12/2019] [Indexed: 02/08/2023]
Abstract
Diacylglycerol kinase β (DGKβ) is an enzyme converting DG to phosphatidic acid (PA) and is specifically expressed in neurons, especially those in the cerebral cortex, hippocampus and striatum. We previously reported that DGKβ induces neurite outgrowth and spinogenesis, contributing to higher brain function including emotion and memory, and plasma membrane localization of DGKβ via the C1 domain and a cluster of basic amino acids at the C-terminus is necessary for its function. To clarify the mechanisms involved in neuronal development by DGKβ, we investigated whether DGKβ activity induces neurite outgrowth using human neuroblastoma SH-SY5Y cells. DGKβ induced neurite outgrowth by activation of mammalian target of rapamycin complex 1 (mTORC1) through a kinase-dependent pathway. In addition, in primary cultured cortical and hippocampal neurons, inhibition of mTORC1 abolished DGKβ induced-neurite outgrowth, branching and spinogenesis. These results indicated that DGKβ induces neurite outgrowth and spinogenesis by activating mTORC1 in a kinase-dependent pathway.
Collapse
Affiliation(s)
- Hiroko Nakai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| | - Ryosuke Tsumagari
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| | - Kenta Maruo
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| | - Akio Nakashima
- Laboratory of Cell Signaling, Biosignal Research Center, Kobe University, Kobe, Japan.
| | - Ushio Kikkawa
- Laboratory of Cell Signaling, Biosignal Research Center, Kobe University, Kobe, Japan.
| | - Shuji Ueda
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| | - Minoru Yamanoue
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Yasuhito Shirai
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Sciences & Faculty of Agriculture, Kobe University, Kobe, Japan.
| |
Collapse
|
42
|
Dupraz S, Hilton BJ, Husch A, Santos TE, Coles CH, Stern S, Brakebusch C, Bradke F. RhoA Controls Axon Extension Independent of Specification in the Developing Brain. Curr Biol 2019; 29:3874-3886.e9. [PMID: 31679934 DOI: 10.1016/j.cub.2019.09.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
The specification of an axon and its subsequent outgrowth are key steps during neuronal polarization, a prerequisite to wire the brain. The Rho-guanosine triphosphatase (GTPase) RhoA is believed to be a central player in these processes. However, its physiological role has remained undefined. Here, genetic loss- and gain-of-function experiments combined with time-lapse microscopy, cell culture, and in vivo analysis show that RhoA is not involved in axon specification but confines the initiation of neuronal polarization and axon outgrowth during development. Biochemical analysis and super-resolution microscopy together with molecular and pharmacological manipulations reveal that RhoA restrains axon growth by activating myosin-II-mediated actin arc formation in the growth cone to prevent microtubules from protruding toward the leading edge. Through this mechanism, RhoA regulates the duration of axon growth and pause phases, thus controlling the tightly timed extension of developing axons. Thereby, this work unravels physiologically relevant players coordinating actin-microtubule interactions during axon growth.
Collapse
Affiliation(s)
- Sebastian Dupraz
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Brett J Hilton
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Andreas Husch
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Telma E Santos
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Charlotte H Coles
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Sina Stern
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Cord Brakebusch
- Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany.
| |
Collapse
|
43
|
Logan CM, Menko AS. Microtubules: Evolving roles and critical cellular interactions. Exp Biol Med (Maywood) 2019; 244:1240-1254. [PMID: 31387376 DOI: 10.1177/1535370219867296] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microtubules are cytoskeletal elements known as drivers of directed cell migration, vesicle and organelle trafficking, and mitosis. In this review, we discuss new research in the lens that has shed light into further roles for stable microtubules in the process of development and morphogenesis. In the lens, as well as other systems, distinct roles for characteristically dynamic microtubules and stabilized populations are coming to light. Understanding the mechanisms of microtubule stabilization and the associated microtubule post-translational modifications is an evolving field of study. Appropriate cellular homeostasis relies on not only one cytoskeletal element, but also rather an interaction between cytoskeletal proteins as well as other cellular regulators. Microtubules are key integrators with actin and intermediate filaments, as well as cell–cell junctional proteins and other cellular regulators including myosin and RhoGTPases to maintain this balance.Impact statementThe role of microtubules in cellular functioning is constantly expanding. In this review, we examine new and exciting fields of discovery for microtubule’s involvement in morphogenesis, highlight our evolving understanding of differential roles for stabilized versus dynamic subpopulations, and further understanding of microtubules as a cellular integrator.
Collapse
Affiliation(s)
- Caitlin M Logan
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - A Sue Menko
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
44
|
Slater PG, Cammarata GM, Samuelson AG, Magee A, Hu Y, Lowery LA. XMAP215 promotes microtubule-F-actin interactions to regulate growth cone microtubules during axon guidance in Xenopus laevis. J Cell Sci 2019; 132:jcs.224311. [PMID: 30890650 DOI: 10.1242/jcs.224311] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/08/2019] [Indexed: 12/24/2022] Open
Abstract
It has long been established that neuronal growth cone navigation depends on changes in microtubule (MT) and F-actin architecture downstream of guidance cues. However, the mechanisms by which MTs and F-actin are dually coordinated remain a fundamentally unresolved question. Here, we report that the well-characterized MT polymerase, XMAP215 (also known as CKAP5), plays an important role in mediating MT-F-actin interaction within the growth cone. We demonstrate that XMAP215 regulates MT-F-actin alignment through its N-terminal TOG 1-5 domains. Additionally, we show that XMAP215 directly binds to F-actin in vitro and co-localizes with F-actin in the growth cone periphery. We also find that XMAP215 is required for regulation of growth cone morphology and response to the guidance cue, Ephrin A5. Our findings provide the first strong evidence that XMAP215 coordinates MT and F-actin interaction in vivo We suggest a model in which XMAP215 regulates MT extension along F-actin bundles into the growth cone periphery and that these interactions may be important to control cytoskeletal dynamics downstream of guidance cues. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Paula G Slater
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | | | | | - Alexandra Magee
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
45
|
Woo JAA, Liu T, Fang CC, Cazzaro S, Kee T, LePochat P, Yrigoin K, Penn C, Zhao X, Wang X, Liggett SB, Kang DE. Activated cofilin exacerbates tau pathology by impairing tau-mediated microtubule dynamics. Commun Biol 2019; 2:112. [PMID: 30911686 PMCID: PMC6430779 DOI: 10.1038/s42003-019-0359-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. While the accumulation of Aβ is pivotal to the etiology of AD, both the microtubule-associated protein tau (MAPT) and the F-actin severing protein cofilin are necessary for the deleterious effects of Aβ. However, the molecular link between tau and cofilin remains unclear. In this study, we found that cofilin competes with tau for direct microtubule binding in vitro, in cells, and in vivo, which inhibits tau-induced microtubule assembly. Genetic reduction of cofilin mitigates tauopathy and synaptic defects in Tau-P301S mice and movement deficits in tau transgenic C. elegans. The pathogenic effects of cofilin are selectively mediated by activated cofilin, as active but not inactive cofilin selectively interacts with tubulin, destabilizes microtubules, and promotes tauopathy. These results therefore indicate that activated cofilin plays an essential intermediary role in neurotoxic signaling that promotes tauopathy.
Collapse
Affiliation(s)
- Jung-A. A. Woo
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Tian Liu
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Cenxiao C. Fang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Sara Cazzaro
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Teresa Kee
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Patrick LePochat
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Ksenia Yrigoin
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Courtney Penn
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Xingyu Zhao
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Xinming Wang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - Stephen B. Liggett
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
| | - David E. Kang
- USF Health Byrd Alzheimer’s Institute, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- Department of Molecular Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33613 USA
- James A. Haley Veteran’s Administration Hospital, Tampa, FL 33612 USA
| |
Collapse
|
46
|
Miller KE, Suter DM. An Integrated Cytoskeletal Model of Neurite Outgrowth. Front Cell Neurosci 2018; 12:447. [PMID: 30534055 PMCID: PMC6275320 DOI: 10.3389/fncel.2018.00447] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Neurite outgrowth underlies the wiring of the nervous system during development and regeneration. Despite a significant body of research, the underlying cytoskeletal mechanics of growth and guidance are not fully understood, and the relative contributions of individual cytoskeletal processes to neurite growth are controversial. Here, we review the structural organization and biophysical properties of neurons to make a semi-quantitative comparison of the relative contributions of different processes to neurite growth. From this, we develop the idea that neurons are active fluids, which generate strong contractile forces in the growth cone and weaker contractile forces along the axon. As a result of subcellular gradients in forces and material properties, actin flows rapidly rearward in the growth cone periphery, and microtubules flow forward in bulk along the axon. With this framework, an integrated model of neurite outgrowth is proposed that hopefully will guide new approaches to stimulate neuronal growth.
Collapse
Affiliation(s)
- Kyle E Miller
- Department of Integrative Biology, Michigan State University, East Lansing, MI, United States
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
47
|
Craig EM. Model for Coordination of Microtubule and Actin Dynamics in Growth Cone Turning. Front Cell Neurosci 2018; 12:394. [PMID: 30450038 PMCID: PMC6225807 DOI: 10.3389/fncel.2018.00394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/15/2018] [Indexed: 11/16/2022] Open
Abstract
In the developing nervous system, axons are guided to their synaptic targets by motile structures at the axon tip called growth cones, which reorganize their cytoskeleton in order to steer in response to chemotactic cues. Growth cone motility is mediated by an actin-adhesion “clutch” mechanism, in which mechanical attachment to a substrate, coupled with polarized actin growth, produces leading-edge protrusion. Several studies suggest that dynamic microtubules (MTs) in the growth cone periphery play an essential role in growth cone steering. It is not yet well-understood how the MT cytoskeleton and the dynamic actin-adhesion clutch system are coordinated to promote growth cone navigation. I introduce an experimentally motivated stochastic model of the dynamic reorganization of the growth cone cytoskeleton in response to external guidance cues. According to this model, asymmetric decoupling of MTs from actin retrograde flow leads to a local influx of MTs to the growth cone leading edge, and the leading-edge MT accumulation is amplified by positive feedback between MTs and the actin-adhesion clutch system. Local accumulation of MTs at the leading edge is hypothesized to increase actin adhesion to the substrate, which attenuates actin retrograde flow and promotes leading-edge protrusion. Growth cone alignment with the chemotactic gradient is predicted to be most effective for intermediate levels of sensitivity of the adhesion strength to the presence of leading-edge MTs. Quantitative predictions of the MT distribution and the local rate of retrograde actin flow will allow the hypothetical positive feedback mechanism to be experimentally tested.
Collapse
Affiliation(s)
- Erin M Craig
- Department of Physics, Central Washington University, Ellensburg, WA, United States
| |
Collapse
|
48
|
Roles of 5-HT on phase transition of neurite outgrowth in the identified serotoninergic neuron C1, Helisoma trivolvis. INVERTEBRATE NEUROSCIENCE 2018; 18:10. [PMID: 30128715 DOI: 10.1007/s10158-018-0214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/27/2018] [Indexed: 10/28/2022]
Abstract
Neurite outgrowth is a morphological marker of neuronal differentiation and neuroregeneration, and the process includes four essential phases, namely initiation, elongation, guidance and cessation. Intrinsic and extrinsic signaling molecules seem to involve morphological changes of neurite outgrowth via various cellular signaling cascades phase transition. Although mechanisms associated with neurite outgrowth have been studied extensively, little is known about how phase transition is regulated during neurite outgrowth. 5-HT has long been studied with regard to its relationship to neurite outgrowth in invertebrate and vertebrate culture systems, and many studies have suggested 5-HT inhibits neurite elongation and growth cone motility, in particular, at the growing parts of neurite such as growth cones and filopodia. However, the underlying mechanisms need to be investigated. In this study, we investigated roles of 5-HT on neurite outgrowth using single serotonergic neurons C1 isolated from Helisoma trivolvis. We observed that 5-HT delayed phase transitions from initiation to elongation of neurite outgrowth. This study for the first time demonstrated that 5-HT has a critical role in phase-controlling mechanisms of neurite outgrowth in neuronal cell cultures.
Collapse
|
49
|
Schlam D, Grinstein S, Freeman SA. Screening for Rho GTPase Modulators in Actin-Dependent Processes Exemplified by Phagocytosis. Methods Mol Biol 2018; 1821:107-127. [PMID: 30062408 DOI: 10.1007/978-1-4939-8612-5_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Rho GTPases, a family of molecular switches, are essential for the assembly and rearrangement of the cellular actin network. Actin remodeling is a central component of many important biological phenomena including chemotaxis, immunological synapse formation, and phagocytosis. Proper execution of these processes requires careful modulation of Rho GTPase activity in space and time. This is accomplished by delicate coordination of Rho GTPase activation and inactivation by Rho guanine nucleotide exchange factors (RhoGEFs) and Rho GTPase-activating proteins (RhoGAPs), respectively. Elucidating the function of these Rho GTPase modulators is complicated by their diversity, varied expression across different tissues, and multiplicity of substrates. To overcome some of these hurdles, we describe here a systematic and unbiased screening approach consisting of three sequential steps: (1) monitoring the subcellular localization of a library of Rho GTPase modulators; (2) assessing endogenous levels of expression of the suitably localized candidates in the cell type of interest; and (3) validating the functional relevance of the identified candidates by siRNA, followed by determining the effects of gene silencing on Rho GTPase activity and actin polymerization. To this end, we describe the expression and visualization of fluorescent Rho GTPase modulators, and the use of genetically encoded biosensors for active Rac/Cdc42 and of fluorescent probes of polymerized actin. Phagocytosis by macrophages is used in this chapter as an experimental paradigm, but the methods described herein can be easily extended to other cells and actin-dependent processes.
Collapse
Affiliation(s)
- Daniel Schlam
- Division of Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sergio Grinstein
- Division of Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.
| | - Spencer A Freeman
- Division of Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
50
|
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
|