1
|
Kos P, Baumann O. Spatial arrangement, polarity, and posttranslational modifications of the microtubule system in the Drosophila eye. Cell Tissue Res 2024; 398:123-137. [PMID: 39152365 PMCID: PMC11525301 DOI: 10.1007/s00441-024-03914-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
We have analyzed the organization of the microtubule system in photoreceptor cells and pigment cells within the adult Drosophila compound eye. Immunofluorescence localization of tubulin and of Short stop, a spectraplakin that has been reported to be involved in the anchorage of microtubule minus ends at the membrane, suggests the presence of non-centrosomal microtubule-organizing centers at the distal tip of the visual cells. Ultrastructural analyses confirm that microtubules emanate from membrane-associated plaques at the site of contact with cone cells and that all microtubules are aligned in distal-proximal direction within the photoreceptor cells. Determination of microtubule polarities demonstrated that about 95% of the microtubules in photoreceptor cells are oriented with their plus end in the direction of the synapse. Pigment cells in the eye contain only microtubules aligned in distal-proximal direction, with their plus end pointing towards the retinal floor. There, two populations of microtubules can be distinguished, single microtubules and bundled microtubules, the latter associated with actin filaments. Whereas microtubules in both photoreceptor cells and pigment cells are acetylated and mono/bi-glutamylated on α-tubulin, bundled microtubules in pigment cells are apparently also mono/bi-glutamylated on β-tubulin, providing the possibility of binding different microtubule-associated proteins.
Collapse
Affiliation(s)
- Piotr Kos
- Unit of Animal Physiology, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany
| | - Otto Baumann
- Unit of Animal Physiology, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany.
| |
Collapse
|
2
|
Dargan R, Mikheenko A, Johnson NL, Packer B, Li Z, Craig EJ, Sarbanes SL, Bereda C, Mehta PR, Keuss M, Nalls MA, Qi YA, Weller CA, Fratta P, Ryan VH. Altered mRNA transport and local translation in iNeurons with RNA binding protein knockdown. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615153. [PMID: 39386562 PMCID: PMC11463369 DOI: 10.1101/2024.09.26.615153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Neurons rely on mRNA transport and local translation to facilitate rapid protein synthesis in processes far from the cell body. These processes allow precise spatial and temporal control of translation and are mediated by RNA binding proteins (RBPs), including those known to be associated with neurodegenerative diseases. Here, we use proteomics, transcriptomics, and microscopy to investigate the impact of RBP knockdown on mRNA transport and local translation in iPSC-derived neurons. We find thousands of transcripts enriched in neurites and that many of these transcripts are locally translated, possibly due to the shorter length of transcripts in neurites. Loss of frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS)-associated RBPs TDP-43 and hnRNPA1 lead to distinct alterations in the neuritic proteome and transcriptome. TDP-43 knockdown (KD) leads to increased neuritic mRNA and translation. In contrast, hnRNPA1 leads to increased neuritic mRNA, but not translation, and more moderate effects on local mRNA profiles, possibly due to compensation by hnRNPA3. These results highlight the crucial role of FTD/ALS-associated RBPs in mRNA transport and local translation in neurons and the importance of these processes in neuron health and disease.
Collapse
Affiliation(s)
- Rachael Dargan
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Alla Mikheenko
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Nicholas L Johnson
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Benjamin Packer
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Ziyi Li
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Emma J Craig
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie L Sarbanes
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Colleen Bereda
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Puja R Mehta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Matthew Keuss
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Cory A Weller
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- DataTecnica, Washington, DC, USA
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
- Francis Crick Institute, London, UK
| | - Veronica H Ryan
- Center for Alzheimer's and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Louis ED, Kuo SH, Faust PL. Purkinje Cell Dendritic Swellings: A Postmortem Study of Essential Tremor and Other Cerebellar Degenerative Disorders. CEREBELLUM (LONDON, ENGLAND) 2024:10.1007/s12311-024-01739-1. [PMID: 39230844 DOI: 10.1007/s12311-024-01739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Under stress, Purkinje cells (PCs) undergo a variety of reactive morphological changes. These can include swellings of neuronal processes. While axonal swellings, "torpedoes", have been well-studied, dendritic swellings (DS) have not been the centerpiece of study. Surprisingly little is known about their frequency or relationship to other morphological changes in degenerating PCs. Leveraging a large brain bank, we (1) examined the morphology of DS, (2) quantified DS, and (2) examined correlations between counts of DS versus 16 other PC morphological changes in a broad range of cerebellar degenerative disorders. There were 159 brains - 100 essential tremor (ET), 13 Friedreich's ataxia, and 46 spinocerebellar ataxia (SCA) (14 SCA1, 7 SCA2, 13 SCA3, 5 SCA6, 5 SCA7, and 2 SCA8). DS were a feature of PCs across all these disorders, with varying morphologies and changes elsewhere in the dendritic arbor. On Luxol fast blue/hematoxylin and eosin-stained sections, the median number of DS per PC ranged from 0.001 in ET to 0.025 in SCA8. Bielschowsky-stained sections yielded higher counts, from 0.003 in ET to 0.042 in SCA6. Torpedo counts exceeded DS counts by one order of magnitude. DS counts were more robustly correlated with torpedo counts than with counts for any of the other PC morphological changes. In summary, DS ranged in prevalence across cerebellar degenerative disorders, from 1/1,000 to 42/1,000 PCs. Across disorders of cerebellar degeneration, these swellings of the dendritic compartment were most robustly correlated with swellings of the axonal compartment, suggesting a similar type of cellular response to duress.
Collapse
Affiliation(s)
- Elan D Louis
- Department of Neurology, University of Texas Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Sheng-Han Kuo
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Initiative for Columbia Ataxia and Tremor, Columbia University, New York, NY, USA
| | - Phyllis L Faust
- Initiative for Columbia Ataxia and Tremor, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
4
|
Richardson B, Goedert T, Quraishe S, Deinhardt K, Mudher A. How do neurons age? A focused review on the aging of the microtubular cytoskeleton. Neural Regen Res 2024; 19:1899-1907. [PMID: 38227514 DOI: 10.4103/1673-5374.390974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024] Open
Abstract
Aging is the leading risk factor for Alzheimer's disease and other neurodegenerative diseases. We now understand that a breakdown in the neuronal cytoskeleton, mainly underpinned by protein modifications leading to the destabilization of microtubules, is central to the pathogenesis of Alzheimer's disease. This is accompanied by morphological defects across the somatodendritic compartment, axon, and synapse. However, knowledge of what occurs to the microtubule cytoskeleton and morphology of the neuron during physiological aging is comparatively poor. Several recent studies have suggested that there is an age-related increase in the phosphorylation of the key microtubule stabilizing protein tau, a modification, which is known to destabilize the cytoskeleton in Alzheimer's disease. This indicates that the cytoskeleton and potentially other neuronal structures reliant on the cytoskeleton become functionally compromised during normal physiological aging. The current literature shows age-related reductions in synaptic spine density and shifts in synaptic spine conformation which might explain age-related synaptic functional deficits. However, knowledge of what occurs to the microtubular and actin cytoskeleton, with increasing age is extremely limited. When considering the somatodendritic compartment, a regression in dendrites and loss of dendritic length and volume is reported whilst a reduction in soma volume/size is often seen. However, research into cytoskeletal change is limited to a handful of studies demonstrating reductions in and mislocalizations of microtubule-associated proteins with just one study directly exploring the integrity of the microtubules. In the axon, an increase in axonal diameter and age-related appearance of swellings is reported but like the dendrites, just one study investigates the microtubules directly with others reporting loss or mislocalization of microtubule-associated proteins. Though these are the general trends reported, there are clear disparities between model organisms and brain regions that are worthy of further investigation. Additionally, longitudinal studies of neuronal/cytoskeletal aging should also investigate whether these age-related changes contribute not just to vulnerability to disease but also to the decline in nervous system function and behavioral output that all organisms experience. This will highlight the utility, if any, of cytoskeletal fortification for the promotion of healthy neuronal aging and potential protection against age-related neurodegenerative disease. This review seeks to summarize what is currently known about the physiological aging of the neuron and microtubular cytoskeleton in the hope of uncovering mechanisms underpinning age-related risk to disease.
Collapse
Affiliation(s)
- Brad Richardson
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Thomas Goedert
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Shmma Quraishe
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Katrin Deinhardt
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Amritpal Mudher
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
5
|
Gavrilova A, Boström A, Korabel N, Fedotov S, Poulin GB, Allan VJ. The role of kinesin-1 in neuronal dense core vesicle transport, locomotion and lifespan regulation in C. elegans. J Cell Sci 2024; 137:jcs262148. [PMID: 39171448 PMCID: PMC11423817 DOI: 10.1242/jcs.262148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Fast axonal transport is crucial for neuronal function and is driven by kinesins and cytoplasmic dynein. Here, we investigated the role of kinesin-1 in dense core vesicle (DCV) transport in C. elegans, using mutants in the kinesin light chains (klc-1 and klc-2) and the motor subunit (unc-116) expressing an ida-1::gfp transgene that labels DCVs. DCV transport in both directions was greatly impaired in an unc-116 mutant and had reduced velocity in a klc-2 mutant. In contrast, the speed of retrograde DCV transport was increased in a klc-1 mutant whereas anterograde transport was unaffected. We identified striking differences between the klc mutants in their effects on worm locomotion and responses to drugs affecting neuromuscular junction activity. We also determined lifespan, finding that unc-116 mutant was short-lived whereas the klc single mutant lifespan was wild type. The ida-1::gfp transgenic strain was also short-lived, but surprisingly, klc-1 and klc-2 extended the ida-1::gfp lifespan beyond that of wild type. Our findings suggest that kinesin-1 not only influences anterograde and retrograde DCV transport but is also involved in regulating lifespan and locomotion, with the two kinesin light chains playing distinct roles.
Collapse
Affiliation(s)
- Anna Gavrilova
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Rumford St, Manchester M13 9PT, UK
- Department of Mathematics, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Astrid Boström
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Rumford St, Manchester M13 9PT, UK
| | - Nickolay Korabel
- Department of Mathematics, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Sergei Fedotov
- Department of Mathematics, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Gino B Poulin
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Rumford St, Manchester M13 9PT, UK
| | - Victoria J Allan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Rumford St, Manchester M13 9PT, UK
| |
Collapse
|
6
|
Cada AK, Mizuno N. Molecular cartography within axons. Curr Opin Cell Biol 2024; 88:102358. [PMID: 38608424 DOI: 10.1016/j.ceb.2024.102358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
Recent advances in imaging methods begin to further illuminate the inner workings of neurons. Views of the axonal landscape through the lens of in situ cryo-electron tomography (cryo-ET) provide a high-resolution atlas of the macromolecular organization in near-native conditions, leading to our growing understanding of the vital roles of compositional and structural organization in maintaining neuronal homeostasis. In this review, we discuss the latest observations concerning the fundamental components found within neuronal compartments, with special emphasis on the axon, branch points, and growth cone. We describe the similarity and difference in organization of organelles and molecules in varying compartments. Finally, we highlight outstanding questions on the dynamics and localization of various components along the axon that may be answered using orthogonal approaches.
Collapse
Affiliation(s)
- A King Cada
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Bethesda, MD, 20892, USA
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Bethesda, MD, 20892, USA; National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Nelson AC, Rolls MM, Ciocanel MV, McKinley SA. Minimal Mechanisms of Microtubule Length Regulation in Living Cells. Bull Math Biol 2024; 86:58. [PMID: 38627264 PMCID: PMC11413797 DOI: 10.1007/s11538-024-01279-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/05/2024] [Indexed: 04/19/2024]
Abstract
The microtubule cytoskeleton is responsible for sustained, long-range intracellular transport of mRNAs, proteins, and organelles in neurons. Neuronal microtubules must be stable enough to ensure reliable transport, but they also undergo dynamic instability, as their plus and minus ends continuously switch between growth and shrinking. This process allows for continuous rebuilding of the cytoskeleton and for flexibility in injury settings. Motivated by in vivo experimental data on microtubule behavior in Drosophila neurons, we propose a mathematical model of dendritic microtubule dynamics, with a focus on understanding microtubule length, velocity, and state-duration distributions. We find that limitations on microtubule growth phases are needed for realistic dynamics, but the type of limiting mechanism leads to qualitatively different responses to plausible experimental perturbations. We therefore propose and investigate two minimally-complex length-limiting factors: limitation due to resource (tubulin) constraints and limitation due to catastrophe of large-length microtubules. We combine simulations of a detailed stochastic model with steady-state analysis of a mean-field ordinary differential equations model to map out qualitatively distinct parameter regimes. This provides a basis for predicting changes in microtubule dynamics, tubulin allocation, and the turnover rate of tubulin within microtubules in different experimental environments.
Collapse
Affiliation(s)
- Anna C Nelson
- Department of Mathematics, Duke University, Durham, NC, 27710, USA.
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA, 16801, USA
| | - Maria-Veronica Ciocanel
- Department of Mathematics, Duke University, Durham, NC, 27710, USA
- Department of Biology, Duke University, Durham, NC, 27710, USA
| | - Scott A McKinley
- Department of Mathematics, Tulane University, New Orleans, LA, 70118, USA
| |
Collapse
|
8
|
Schijven D, Soheili-Nezhad S, Fisher SE, Francks C. Exome-wide analysis implicates rare protein-altering variants in human handedness. Nat Commun 2024; 15:2632. [PMID: 38565598 PMCID: PMC10987538 DOI: 10.1038/s41467-024-46277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Handedness is a manifestation of brain hemispheric specialization. Left-handedness occurs at increased rates in neurodevelopmental disorders. Genome-wide association studies have identified common genetic effects on handedness or brain asymmetry, which mostly involve variants outside protein-coding regions and may affect gene expression. Implicated genes include several that encode tubulins (microtubule components) or microtubule-associated proteins. Here we examine whether left-handedness is also influenced by rare coding variants (frequencies ≤ 1%), using exome data from 38,043 left-handed and 313,271 right-handed individuals from the UK Biobank. The beta-tubulin gene TUBB4B shows exome-wide significant association, with a rate of rare coding variants 2.7 times higher in left-handers than right-handers. The TUBB4B variants are mostly heterozygous missense changes, but include two frameshifts found only in left-handers. Other TUBB4B variants have been linked to sensorineural and/or ciliopathic disorders, but not the variants found here. Among genes previously implicated in autism or schizophrenia by exome screening, DSCAM and FOXP1 show evidence for rare coding variant association with left-handedness. The exome-wide heritability of left-handedness due to rare coding variants was 0.91%. This study reveals a role for rare, protein-altering variants in left-handedness, providing further evidence for the involvement of microtubules and disorder-relevant genes.
Collapse
Affiliation(s)
- Dick Schijven
- Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Sourena Soheili-Nezhad
- Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Simon E Fisher
- Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Clyde Francks
- Language & Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
9
|
Nelson AC, Rolls MM, Ciocanel MV, McKinley SA. Minimal Mechanisms of Microtubule Length Regulation in Living Cells. ARXIV 2024:arXiv:2310.13666v3. [PMID: 37904745 PMCID: PMC10614985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The microtubule cytoskeleton is responsible for sustained, long-range intracellular transport of mRNAs, proteins, and organelles in neurons. Neuronal microtubules must be stable enough to ensure reliable transport, but they also undergo dynamic instability, as their plus and minus ends continuously switch between growth and shrinking. This process allows for continuous rebuilding of the cytoskeleton and for flexibility in injury settings. Motivated by in vivo experimental data on microtubule behavior in Drosophila neurons, we propose a mathematical model of dendritic microtubule dynamics, with a focus on understanding microtubule length, velocity, and state-duration distributions. We find that limitations on microtubule growth phases are needed for realistic dynamics, but the type of limiting mechanism leads to qualitatively different responses to plausible experimental perturbations. We therefore propose and investigate two minimally-complex length-limiting factors: limitation due to resource (tubulin) constraints and limitation due to catastrophe of large-length microtubules. We combine simulations of a detailed stochastic model with steady-state analysis of a mean-field ordinary differential equations model to map out qualitatively distinct parameter regimes. This provides a basis for predicting changes in microtubule dynamics, tubulin allocation, and the turnover rate of tubulin within microtubules in different experimental environments.
Collapse
Affiliation(s)
- Anna C Nelson
- Department of Mathematics, Duke University, Durham, 27710, NC, USA
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, 16801, PA, USA
| | - Maria-Veronica Ciocanel
- Department of Mathematics, Duke University, Durham, 27710, NC, USA
- Department of Biology, Duke University, Durham, 27710, NC, USA
| | - Scott A McKinley
- Department of Mathematics, Tulane University, New Orleans, 70118, LA, USA
| |
Collapse
|
10
|
Gao Z, Huang E, Wang W, Xu L, Xu W, Zheng T, Rui M. Patronin regulates presynaptic microtubule organization and neuromuscular junction development in Drosophila. iScience 2024; 27:108944. [PMID: 38318379 PMCID: PMC10839449 DOI: 10.1016/j.isci.2024.108944] [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: 08/10/2023] [Revised: 11/20/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Synapses are fundamental components of the animal nervous system. Synaptic cytoskeleton is essential for maintaining proper neuronal development and wiring. Perturbations in neuronal microtubules (MTs) are correlated with numerous neuropsychiatric disorders. Despite discovering multiple synaptic MT regulators, the importance of MT stability, and particularly the polarity of MT in synaptic function, is still under investigation. Here, we identify Patronin, an MT minus-end-binding protein, for its essential role in presynaptic regulation of MT organization and neuromuscular junction (NMJ) development. Analyses indicate that Patronin regulates synaptic development independent of Klp10A. Subsequent research elucidates that it is short stop (Shot), a member of the Spectraplakin family of large cytoskeletal linker molecules, works synergistically with Patronin to govern NMJ development. We further raise the possibility that normal synaptic MT polarity contributes to proper NMJ morphology. Overall, this study demonstrates an unprecedented role of Patronin, and a potential involvement of MT polarity in synaptic development.
Collapse
Affiliation(s)
- Ziyang Gao
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Erqian Huang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Wanting Wang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Lizhong Xu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Wanyue Xu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Ting Zheng
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Menglong Rui
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| |
Collapse
|
11
|
Ugalde MV, Alecki C, Rizwan J, Le P, Jacob-Tomas S, Xu JM, Minotti S, Wu T, Durham H, Yeo G. Localized molecular chaperone synthesis maintains neuronal dendrite proteostasis. RESEARCH SQUARE 2023:rs.3.rs-3673702. [PMID: 38168440 PMCID: PMC10760236 DOI: 10.21203/rs.3.rs-3673702/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Proteostasis is maintained through regulated protein synthesis and degradation and chaperone-assisted protein folding. However, this is challenging in neuronal projections because of their polarized morphology and constant synaptic proteome remodeling. Using high-resolution fluorescence microscopy, we discovered that neurons localize a subset of chaperone mRNAs to their dendrites and use microtubule-based transport to increase this asymmetric localization following proteotoxic stress. The most abundant dendritic chaperone mRNA encodes a constitutive heat shock protein 70 family member (HSPA8). Proteotoxic stress also enhanced HSPA8 mRNA translation efficiency in dendrites. Stress-mediated HSPA8 mRNA localization to the dendrites was impaired by depleting fused in sarcoma-an amyotrophic lateral sclerosis-related protein-in cultured mouse motor neurons and expressing a pathogenic variant of heterogenous nuclear ribonucleoprotein A2/B1 in neurons derived from human induced pluripotent stem cells. These results reveal a crucial and unexpected neuronal stress response in which RNA-binding proteins increase the dendritic localization of HSPA8 mRNA to maintain proteostasis and prevent neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Gene Yeo
- University of California, San Diego
| |
Collapse
|
12
|
Parkes M, Landers NL, Gramlich MW. Recently recycled synaptic vesicles use multi-cytoskeletal transport and differential presynaptic capture probability to establish a retrograde net flux during ISVE in central neurons. Front Cell Dev Biol 2023; 11:1286915. [PMID: 38020880 PMCID: PMC10657820 DOI: 10.3389/fcell.2023.1286915] [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: 09/01/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Presynapses locally recycle synaptic vesicles to efficiently communicate information. During use and recycling, proteins on the surface of synaptic vesicles break down and become less efficient. In order to maintain efficient presynaptic function and accommodate protein breakdown, new proteins are regularly produced in the soma and trafficked to presynaptic locations where they replace older protein-carrying vesicles. Maintaining a balance of new proteins and older proteins is thus essential for presynaptic maintenance and plasticity. While protein production and turnover have been extensively studied, it is still unclear how older synaptic vesicles are trafficked back to the soma for recycling in order to maintain balance. In the present study, we use a combination of fluorescence microscopy, hippocampal cell cultures, and computational analyses to determine the mechanisms that mediate older synaptic vesicle trafficking back to the soma. We show that synaptic vesicles, which have recently undergone exocytosis, can differentially utilize either the microtubule or the actin cytoskeleton networks. We show that axonally trafficked vesicles traveling with higher speeds utilize the microtubule network and are less likely to be captured by presynapses, while slower vesicles utilize the actin network and are more likely to be captured by presynapses. We also show that retrograde-driven vesicles are less likely to be captured by a neighboring presynapse than anterograde-driven vesicles. We show that the loss of synaptic vesicle with bound molecular motor myosin V is the mechanism that differentiates whether vesicles will utilize the microtubule or actin networks. Finally, we present a theoretical framework of how our experimentally observed retrograde vesicle trafficking bias maintains the balance with previously observed rates of new vesicle trafficking from the soma.
Collapse
|
13
|
de Keijzer J, van Spoordonk R, van der Meer-Verweij JE, Janson M, Ketelaar T. Kinesin-4 optimizes microtubule orientations for responsive tip growth guidance in moss. J Cell Biol 2023; 222:e202202018. [PMID: 37389658 PMCID: PMC10316633 DOI: 10.1083/jcb.202202018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/18/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023] Open
Abstract
Tip-growing cells of, amongst others, plants and fungi secrete wall materials in a highly polarized fashion for fast and efficient colonization of the environment. A polarized microtubule cytoskeleton, in which most microtubule ends are directed toward the growing apex, has been implicated in directing growth. Its organizing principles, in particular regarding maintenance of network unipolarity, have remained elusive. We show that a kinesin-4 protein, hitherto best known for a role in cytokinesis, suppresses encounters between antiparallel microtubules. Without this activity, microtubules hyper-aligned along the growth axis and increasingly grew away from the apex. Cells themselves displayed an overly straight growth path and a delayed gravitropic response. This result revealed conflicting systemic needs for a stable growth direction and an ability to change course in response to extracellular cues. Thus, the use of selective inhibition of microtubule growth at antiparallel overlaps constitutes a new organizing principle within a unipolar microtubule array.
Collapse
Affiliation(s)
- Jeroen de Keijzer
- Laboratory of Cell Biology, Wageningen University, Wageningen, Netherlands
| | | | | | - Marcel Janson
- Laboratory of Cell Biology, Wageningen University, Wageningen, Netherlands
| | - Tijs Ketelaar
- Laboratory of Cell Biology, Wageningen University, Wageningen, Netherlands
| |
Collapse
|
14
|
Smith G, Sweeney ST, O’Kane CJ, Prokop A. How neurons maintain their axons long-term: an integrated view of axon biology and pathology. Front Neurosci 2023; 17:1236815. [PMID: 37564364 PMCID: PMC10410161 DOI: 10.3389/fnins.2023.1236815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
Axons are processes of neurons, up to a metre long, that form the essential biological cables wiring nervous systems. They must survive, often far away from their cell bodies and up to a century in humans. This requires self-sufficient cell biology including structural proteins, organelles, and membrane trafficking, metabolic, signalling, translational, chaperone, and degradation machinery-all maintaining the homeostasis of energy, lipids, proteins, and signalling networks including reactive oxygen species and calcium. Axon maintenance also involves specialised cytoskeleton including the cortical actin-spectrin corset, and bundles of microtubules that provide the highways for motor-driven transport of components and organelles for virtually all the above-mentioned processes. Here, we aim to provide a conceptual overview of key aspects of axon biology and physiology, and the homeostatic networks they form. This homeostasis can be derailed, causing axonopathies through processes of ageing, trauma, poisoning, inflammation or genetic mutations. To illustrate which malfunctions of organelles or cell biological processes can lead to axonopathies, we focus on axonopathy-linked subcellular defects caused by genetic mutations. Based on these descriptions and backed up by our comprehensive data mining of genes linked to neural disorders, we describe the 'dependency cycle of local axon homeostasis' as an integrative model to explain why very different causes can trigger very similar axonopathies, providing new ideas that can drive the quest for strategies able to battle these devastating diseases.
Collapse
Affiliation(s)
- Gaynor Smith
- Cardiff University, School of Medicine, College of Biomedical and Life Sciences, Cardiff, United Kingdom
| | - Sean T. Sweeney
- Department of Biology, University of York and York Biomedical Research Institute, York, United Kingdom
| | - Cahir J. O’Kane
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Stone MC, Mauger AS, Rolls MM. Ciliated sensory neurons can regenerate axons after complete axon removal. J Exp Biol 2023; 226:jeb245717. [PMID: 37212026 PMCID: PMC10323231 DOI: 10.1242/jeb.245717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
Axon regeneration helps maintain lifelong function of neurons in many animals. Depending on the site of injury, new axons can grow either from the axon stump (after distal injury) or from the tip of a dendrite (after proximal injury). However, some neuron types do not have dendrites to be converted to a regenerating axon after proximal injury. For example, many sensory neurons receive information from a specialized sensory cilium rather than a branched dendrite arbor. We hypothesized that the lack of traditional dendrites would limit the ability of ciliated sensory neurons to respond to proximal axon injury. We tested this hypothesis by performing laser microsurgery on ciliated lch1 neurons in Drosophila larvae and tracking cells over time. These cells survived proximal axon injury as well as distal axon injury, and, like many other neurons, initiated growth from the axon stump after distal injury. After proximal injury, neurites regrew in a surprisingly flexible manner. Most cells initiated outgrowth directly from the cell body, but neurite growth could also emerge from the short axon stump or base of the cilium. New neurites were often branched. Although outgrowth after proximal axotomy was variable, it depended on the core DLK axon injury signaling pathway. Moreover, each cell had at least one new neurite specified as an axon based on microtubule polarity and accumulation of the endoplasmic reticulum. We conclude that ciliated sensory neurons are not intrinsically limited in their ability to grow a new axon after proximal axon removal.
Collapse
Affiliation(s)
- Michelle C. Stone
- Department of Biochemistry and Molecular Biology, and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Abigail S. Mauger
- Department of Biochemistry and Molecular Biology, and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Melissa M. Rolls
- Department of Biochemistry and Molecular Biology, and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
16
|
Zocchi R, Compagnucci C, Bertini E, Sferra A. Deciphering the Tubulin Language: Molecular Determinants and Readout Mechanisms of the Tubulin Code in Neurons. Int J Mol Sci 2023; 24:ijms24032781. [PMID: 36769099 PMCID: PMC9917122 DOI: 10.3390/ijms24032781] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Microtubules (MTs) are dynamic components of the cell cytoskeleton involved in several cellular functions, such as structural support, migration and intracellular trafficking. Despite their high similarity, MTs have functional heterogeneity that is generated by the incorporation into the MT lattice of different tubulin gene products and by their post-translational modifications (PTMs). Such regulations, besides modulating the tubulin composition of MTs, create on their surface a "biochemical code" that is translated, through the action of protein effectors, into specific MT-based functions. This code, known as "tubulin code", plays an important role in neuronal cells, whose highly specialized morphologies and activities depend on the correct functioning of the MT cytoskeleton and on its interplay with a myriad of MT-interacting proteins. In recent years, a growing number of mutations in genes encoding for tubulins, MT-interacting proteins and enzymes that post-translationally modify MTs, which are the main players of the tubulin code, have been linked to neurodegenerative processes or abnormalities in neural migration, differentiation and connectivity. Nevertheless, the exact molecular mechanisms through which the cell writes and, downstream, MT-interacting proteins decipher the tubulin code are still largely uncharted. The purpose of this review is to describe the molecular determinants and the readout mechanisms of the tubulin code, and briefly elucidate how they coordinate MT behavior during critical neuronal events, such as neuron migration, maturation and axonal transport.
Collapse
Affiliation(s)
- Riccardo Zocchi
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Claudia Compagnucci
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Research Hospital, IRCCS, 00146 Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| | - Antonella Sferra
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| |
Collapse
|
17
|
Aghigh A, Bancelin S, Rivard M, Pinsard M, Ibrahim H, Légaré F. Second harmonic generation microscopy: a powerful tool for bio-imaging. Biophys Rev 2023; 15:43-70. [PMID: 36909955 PMCID: PMC9995455 DOI: 10.1007/s12551-022-01041-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023] Open
Abstract
Second harmonic generation (SHG) microscopy is an important optical imaging technique in a variety of applications. This article describes the history and physical principles of SHG microscopy and its more advanced variants, as well as their strengths and weaknesses in biomedical applications. It also provides an overview of SHG and advanced SHG imaging in neuroscience and microtubule imaging and how these methods can aid in understanding microtubule formation, structuration, and involvement in neuronal function. Finally, we offer a perspective on the future of these methods and how technological advancements can help make SHG microscopy a more widely adopted imaging technique.
Collapse
Affiliation(s)
- Arash Aghigh
- Centre Énergie Matériaux Télécommunications, Institut National de La Recherche Scientifique, Varennes, QC Canada
| | | | - Maxime Rivard
- National Research Council Canada, Boucherville, QC Canada
| | - Maxime Pinsard
- Institut National de Recherche en Sciences Et Technologies Pour L’environnement Et L’agriculture, Paris, France
| | - Heide Ibrahim
- Centre Énergie Matériaux Télécommunications, Institut National de La Recherche Scientifique, Varennes, QC Canada
| | - François Légaré
- Centre Énergie Matériaux Télécommunications, Institut National de La Recherche Scientifique, Varennes, QC Canada
| |
Collapse
|
18
|
Eckel BD, Cruz R, Craig EM, Baas PW. Microtubule polarity flaws as a treatable driver of neurodegeneration. Brain Res Bull 2023; 192:208-215. [PMID: 36442694 DOI: 10.1016/j.brainresbull.2022.11.013] [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: 07/08/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Microtubule disruption is a common downstream mechanism leading to axonal degeneration in a number of neurological diseases. To date, most studies on this topic have focused on the loss of microtubule mass from the axon, as well as changes in the stability properties of the microtubules and/or their tubulin composition. Here we posit corruption of the normal pattern of microtubule polarity orientation as an underappreciated and yet treatable contributor to axonal degeneration. We include computational modeling to fortify the rigor of our considerations. Our simulations demonstrate that even a small deviation from the usual polarity pattern of axonal microtubules is detrimental to motor-based trafficking of organelles and other intracellular cargo. Additional modeling predicts that axons with such deviations will exhibit significantly reduced speed and reliability of organelle transport, and that localized clusters of wrongly oriented microtubules will result in traffic jams of accumulated organelles.
Collapse
Affiliation(s)
- Bridie D Eckel
- Dept Neurobiol/Anat, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Roy Cruz
- Dept Physics, Central Washington University, Ellensburg, WA 98926, USA
| | - Erin M Craig
- Dept Physics, Central Washington University, Ellensburg, WA 98926, USA
| | - Peter W Baas
- Dept Neurobiol/Anat, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA.
| |
Collapse
|
19
|
Dzaki N, Bu S, Lau SSY, Yong WL, Yu F. Drosophila GSK3β promotes microtubule disassembly and dendrite pruning in sensory neurons. Development 2022; 149:281771. [PMID: 36264221 DOI: 10.1242/dev.200844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022]
Abstract
The evolutionarily conserved Glycogen Synthase Kinase 3β (GSK3β), a negative regulator of microtubules, is crucial for neuronal polarization, growth and migration during animal development. However, it remains unknown whether GSK3β regulates neuronal pruning, which is a regressive process. Here, we report that the Drosophila GSK3β homologue Shaggy (Sgg) is cell-autonomously required for dendrite pruning of ddaC sensory neurons during metamorphosis. Sgg is necessary and sufficient to promote microtubule depolymerization, turnover and disassembly in the dendrites. Although Sgg is not required for the minus-end-out microtubule orientation in dendrites, hyperactivated Sgg can disturb the dendritic microtubule orientation. Moreover, our pharmacological and genetic data suggest that Sgg is required to promote dendrite pruning at least partly via microtubule disassembly. We show that Sgg and Par-1 kinases act synergistically to promote microtubule disassembly and dendrite pruning. Thus, Sgg and Par-1 might converge on and phosphorylate a common downstream microtubule-associated protein(s) to disassemble microtubules and thereby facilitate dendrite pruning.
Collapse
Affiliation(s)
- Najat Dzaki
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Shufeng Bu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604.,Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Samuel Song Yuan Lau
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Wei Lin Yong
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604.,Department of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|
20
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
21
|
Jakobs MAH, Zemel A, Franze K. Unrestrained growth of correctly oriented microtubules instructs axonal microtubule orientation. eLife 2022; 11:77608. [PMID: 36214669 PMCID: PMC9550224 DOI: 10.7554/elife.77608] [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: 02/04/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
In many eukaryotic cells, directed molecular transport occurs along microtubules. Within neuronal axons, transport over vast distances particularly relies on uniformly oriented microtubules, whose plus-ends point towards the distal axon tip (anterogradely polymerizing, or plus-end-out). However, axonal microtubules initially have mixed orientations, and how they orient during development is not yet fully understood. Using live imaging of primary Drosophila melanogaster neurons, we found that, in the distal part of the axon, catastrophe rates of plus-end-out microtubules were significantly reduced compared to those of minus-end-out microtubules. Physical modelling revealed that plus-end-out microtubules should therefore exhibit persistent long-term growth, while growth of minus-end-out microtubules should be limited, leading to a bias in overall axonal microtubule orientation. Using chemical and physical perturbations of microtubule growth and genetic perturbations of the anti -catastrophe factor p150, which was enriched in the distal axon tip, we confirmed that the enhanced growth of plus-end-out microtubules is critical for achieving uniform microtubule orientation. Computer simulations of axon development integrating the enhanced plus-end-out microtubule growth identified here with previously suggested mechanisms, that is, dynein-based microtubule sliding and augmin-mediated templating, correctly predicted the long-term evolution of axonal microtubule orientation as found in our experiments. Our study thus leads to a holistic explanation of how axonal microtubules orient uniformly, a prerequisite for efficient long-range transport essential for neuronal functioning. For humans to be able to wiggle their toes, messages need to travel from the brain to the foot, a distance well over a meter in many adults. This is made possible by neurons, the cells that form the nervous system, which transmit electrical signals along long extensions called ‘axons’. Axons can only transmit signals if all the required molecules, which are produced in a part of the neuron known as the cell body, are ferried to the ends of the axons. This ferrying around of molecules is carried out by long, filamentous molecules called microtubules, which act as a directed carrier system, shuttling molecules along the axon, either towards or away from the cell body. Microtubules can be thought of as asymmetrical rods. One end – known as the plus end – is dynamic and can undergo growth or shrinkage, while the other end – called the minus end – is stable. For transport along the axon to happen efficiently, microtubules in the neuron need to be oriented with their plus end pointing towards the ends of the axon. Microtubules in growing neurons develop this orientation, but how that is achieved is not fully understood. To understand the basis of this cellular phenomenon, Jakobs, Zemel and Franze examined the behaviour of microtubules in developing neurons from fruit fly larvae. A fluorescent protein, which emits light when the microtubules are growing, helped the researchers visualise the plus end of microtubules, the microtubule orientation, and their growth in developing axons. This experiment showed that microtubules that had their plus end pointing towards the axon end shrank more slowly than those with the opposite orientation, leading them to grow longer. This resulted in a higher proportion of the correctly-oriented microtubules in the axon. Treating the neurons with Nocodazole, a chemical that disrupts microtubule growth, or with sodium chloride, which changes the osmotic pressure, caused the microtubules that were oriented with their plus end towards the axon to grow less, and disrupted the uniform orientation of the microtubules in the axon. The next step was to determine whether specific axonal proteins such as p150 – a protein that is enriched at the tip of the axon and decreases microtubule shrinkage rates – are involved in this process. Reducing the levels of p150 in fruit flies using molecular and genetic methods resulted in microtubules with their plus end pointing towards the axon tip shrinking faster, reducing the proportion of microtubules with this orientation in the axon. This role of proteins enriched in the axonal tip, along with previously discovered mechanisms, explains how microtubules align unidirectionally in axons. These findings open new avenues of research into neurodegenerative diseases like Alzheimer’s and Parkinson’s, which might manifest due to a breakdown of transport along microtubules in neurons.
Collapse
Affiliation(s)
- Maximilian A H Jakobs
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom.,DeepMirror, Cambridge, United Kingdom
| | - Assaf Zemel
- Institute of Biomedical and Oral Research, and the Fritz Haber Center for Molecular Dynamics, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom.,Institute for Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| |
Collapse
|
22
|
H-ABC tubulinopathy revealed by label-free second harmonic generation microscopy. Sci Rep 2022; 12:14417. [PMID: 36002546 PMCID: PMC9402540 DOI: 10.1038/s41598-022-18370-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 08/10/2022] [Indexed: 11/08/2022] Open
Abstract
Hypomyelination with atrophy of the basal ganglia and cerebellum is a recently described tubulinopathy caused by a mutation in the tubulin beta 4a isoform, expressed in oligodendrocytes. The taiep rat is the only spontaneous tubulin beta 4a mutant available for the study of this pathology. We aimed to identify the effects of the tubulin mutation on freshly collected, unstained samples of the central white matter of taiep rats using second harmonic generation microscopy. Cytoskeletal differences between the central white matter of taiep rats and control animals were found. Nonlinear emissions from the processes and somata of oligodendrocytes in tubulin beta 4a mutant rats were consistently detected, in the shape of elongated structures and cell-like bodies, which were never detected in the controls. This signal represents the second harmonic trademark of the disease. The tissue was also fluorescently labeled and analyzed to corroborate the origin of the nonlinear signal. Besides enabling the description of structural and molecular aspects of H-ABC, our data open the door to the diagnostic use of nonlinear optics in the study of neurodegenerative diseases, with the additional advantage of a label-free approach that preserves tissue morphology and vitality.
Collapse
|
23
|
Lin G, Lin H, Zhuo R, He W, Ma C, Liu Y, Liu M. GCN5/KAT2A contributes to axon growth and neurogenesis. Neurosci Lett 2022; 784:136742. [PMID: 35716963 DOI: 10.1016/j.neulet.2022.136742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
Posttranslational modification (PTM) of tubulin proteins is involved in microtubule dynamics. Acetylation, an important alpha-tubulin PTM, which is regarded as a hallmark event of stable microtubules, often occurs in neurogenesis and axon outgrowth. GCN5/KAT2A is a well-known histone acetyltransferase and has also been reported to hold the activity of nonhistone acetyltransferases, such as acetylated tubulin (Ace-tubulin). In this study, we investigated the role of GCN5/KAT2A in axon growth and neurogenesis. E18 cortical neurons obtained from day 18 embryos of pregnant Sprague-Dawley (SD) rats were cultured and transfected with GCN5 siRNA or treated with the GCN5 inhibitor MB-3. Neural stem cells (NSCs) derived from the cerebral cortexes of E14 SD rats were cultured and differentiated. During differentiation, MB-3 was applied to investigate the effect of GCN5 dysfunction on neurogenesis. The axonal length and the ratio and distribution of acetylated and tyrosinated tubulin (Tyr-tubulin) were evaluated by immunostaining assay. The expression levels of Nestin, Tuj1, acetylated tubulin, and tyrosinated tubulin proteins were analyzed by Western blotting assays. In primary neurons, both GCN5 siRNA and MB-3 treatment reduced acetylated tubulin protein, changed the ratio of acetylated and tyrosinated tubulin, and decreased axonal length. During NSC differentiation, MB-3 application reduced axon outgrowth, decreased acetylated tubulin and altered the distribution of acetylated tubulin and tyrosinated tubulin. This study revealed for the first time that the acetyltransferase GCN5/KAT2A could contribute to axon outgrowth by altering the ratio and distribution of acetylated tubulin.
Collapse
Affiliation(s)
- Ge Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Haixu Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Run Zhuo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Wei He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Chao Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China.
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China.
| |
Collapse
|
24
|
Damuka N, Orr ME, Bansode AH, Krizan I, Miller M, Lee J, Macauley SL, Whitlow CT, Mintz A, Craft S, Solingapuram Sai KK. Preliminary mechanistic insights of a brain-penetrant microtubule imaging PET ligand in a tau-knockout mouse model. EJNMMI Res 2022; 12:41. [PMID: 35881263 PMCID: PMC9325934 DOI: 10.1186/s13550-022-00912-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/29/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Microtubules (MTs) are critical for cell structure, function, and survival. MT instability may contribute to Alzheimer's disease (AD) pathogenesis as evidenced by persistent negative regulation (phosphorylation) of the neuronal microtubule-associated protein tau. Hyperphosphorylated tau, not bound to MTs, forms intraneuronal pathology that correlates with dementia and can be tracked using positron emission tomography (PET) imaging. The contribution of MT instability in AD remains unknown, though it may be more proximal to neuronal dysfunction than tau accumulation. Our lab reported the first brain-penetrant MT-based PET ligand, [11C]MPC-6827, and its PET imaging with this ligand in normal rodents and non-human primates demonstrated high brain uptake and excellent pharmacokinetics. Target engagement and mechanism of action using in vitro, in vivo, and ex vivo methods were evaluated here. METHODS In vitro cell uptake assay was performed in SH-SY5Y neuronal cells with [11C]MPC-6827, with various MT stabilizing and destabilizing agents. To validate the in vitro results, wild type (WT) mice (n = 4) treated with a brain-penetrant MT stabilizing drug (EpoD) underwent microPET/CT brain imaging with [11C]MPC-6827. To determine the influence of tau protein on radiotracer binding in the absence of protein accumulation, we utilized tau knockout (KO) mice. In vivo microPET imaging, ex vivo biodistribution, and autoradiography studies were performed in tau KO and WT mice (n = 6/group) with [11C]MPC-6827. Additionally, α, β, and acetylated tubulin levels in both brain samples were determined using commercially available cytoskeleton-based MT kit and capillary electrophoresis immunoblotting assays. RESULTS Cell uptake demonstrated higher radioactive uptake with MT destabilizing agents and lower uptake with stabilizing agents compared to untreated cells. Similarly, acute treatment with EpoD in WT mice decreased [11C]MPC-6827 brain uptake, assessed with microPET/CT imaging. Compared to WT mice, tau KO mice expressed significantly lower β tubulin, which contains the MPC-6827 binding domain, and modestly lower levels of acetylated α tubulin, indicative of unstable MTs. In vivo imaging revealed significantly higher [11C]MPC-6827 uptake in tau KOs than WT, particularly in AD-relevant brain regions known to express high levels of tau. Ex vivo post-PET biodistribution and autoradiography confirmed the in vivo results. CONCLUSIONS Collectively, our data indicate that [11C]MPC-6827 uptake inversely correlates with MT stability and may better reflect the absence of tau than total tubulin levels. Given the radiotracer binding does not require the presence of aggregated tau, we hypothesize that [11C]MPC-6827 may be particularly useful in preclinical stages of AD prior to tau deposition. Our study provides immediate clarity on high uptake of the MT-based radiotracer in AD brains, which directly informs clinical utility in MT/tau-based PET imaging studies.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Miranda E. Orr
- Department of Gerontology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Avinash H. Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Jillian Lee
- Department of Gerontology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | - Shannon L. Macauley
- Department of Gerontology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | | | - Akiva Mintz
- Department of Radiology, Columbia Medical Center, New York, NY 10032 USA
| | - Suzanne Craft
- Department of Gerontology, Wake Forest School of Medicine, Winston Salem, NC 27157 USA
| | | |
Collapse
|
25
|
Thyagarajan P, Feng C, Lee D, Shorey M, Rolls MM. Microtubule polarity is instructive for many aspects of neuronal polarity. Dev Biol 2022; 486:56-70. [PMID: 35341730 PMCID: PMC9058238 DOI: 10.1016/j.ydbio.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022]
Abstract
Many neurons in bilaterian animals are polarized with functionally distinct axons and dendrites. Microtubule polarity, microtubule stability, and the axon initial segment (AIS) have all been shown to influence polarized transport in neurons. Each of these cytoskeletal cues could act independently to control axon and dendrite identity, or there could be a hierarchy in which one acts upstream of the others. Here we test the hypothesis that microtubule polarity acts as a master regulator of neuronal polarity by using a Drosophila genetic background in which some dendrites have normal minus-end-out microtubule polarity and others have the axonal plus-end-out polarity. In these mosaic dendrite arbors, we found that ribosomes, which are more abundant in dendrites than axons, were reduced in plus-end-out dendrites, while an axonal cargo was increased. In addition, we determined that microtubule stability was different in plus-end-out and minus-end-out dendrites, with plus-end-out ones having more stable microtubules like axons. Similarly, we found that ectopic diffusion barriers, like those at the AIS, formed at the base of dendrites with plus-end-out regions. Thus, changes in microtubule polarity were sufficient to rearrange other cytoskeletal features associated with neuronal polarization. However, overall neuron shape was maintained with only subtle changes in branching in mosaic arbors. We conclude that microtubule polarity can act upstream of many aspects of intracellular neuronal polarization, but shape is relatively resilient to changes in microtubule polarity in vivo.
Collapse
Affiliation(s)
- Pankajam Thyagarajan
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Chengye Feng
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - David Lee
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Matthew Shorey
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Melissa M Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
26
|
Yap CC, Winckler B. Spatial regulation of endosomes in growing dendrites. Dev Biol 2022; 486:5-14. [PMID: 35306006 PMCID: PMC10646839 DOI: 10.1016/j.ydbio.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/21/2022] [Accepted: 03/13/2022] [Indexed: 01/19/2023]
Abstract
Many membrane proteins are highly enriched in either dendrites or axons. This non-uniform distribution is a critical feature of neuronal polarity and underlies neuronal function. The molecular mechanisms responsible for polarized distribution of membrane proteins has been studied for some time and many answers have emerged. A less well studied feature of neurons is that organelles are also frequently non-uniformly distributed. For instance, EEA1-positive early endosomes are somatodendritic whereas synaptic vesicles are axonal. In addition, some organelles are present in both axons and dendrites, but not distributed uniformly along the processes. One well known example are lysosomes which are abundant in the soma and proximal dendrite, but sparse in the distal dendrite and the distal axon. The mechanisms that determine the spatial distribution of organelles along dendrites are only starting to be studied. In this review, we will discuss the cell biological mechanisms of how the distribution of diverse sets of endosomes along the proximal-distal axis of dendrites might be regulated. In particular, we will focus on the regulation of bulk homeostatic mechanisms as opposed to local regulation. We posit that immature dendrites regulate organelle motility differently from mature dendrites in order to spatially organize dendrite growth, branching and sculpting.
Collapse
|
27
|
Bu S, Tang Q, Wang Y, Lau SSY, Yong WL, Yu F. Drosophila CLASP regulates microtubule orientation and dendrite pruning by suppressing Par-1 kinase. Cell Rep 2022; 39:110887. [PMID: 35649352 DOI: 10.1016/j.celrep.2022.110887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/04/2022] [Accepted: 05/06/2022] [Indexed: 02/06/2023] Open
Abstract
The evolutionarily conserved CLASPs (cytoplasmic linker-associated proteins) are microtubule-associated proteins that inhibit microtubule catastrophe and promote rescue. CLASPs can regulate axonal elongation and dendrite branching in growing neurons. However, their roles in microtubule orientation and neurite pruning in remodeling neurons remain unknown. Here, we identify the Drosophila CLASP homolog Orbit/MAST, which is required for dendrite pruning in ddaC sensory neurons during metamorphosis. Orbit is important for maintenance of the minus-end-out microtubule orientation in ddaC dendrites. Our structural analysis reveals that the microtubule lattice-binding TOG2 domain is required for Orbit to regulate dendritic microtubule orientation and dendrite pruning. In a genetic modifier screen, we further identify the conserved Par-1 kinase as a suppressor of Orbit in dendritic microtubule orientation. Moreover, elevated Par-1 function impairs dendritic microtubule orientation and dendrite pruning, phenocopying orbit mutants. Overall, our study demonstrates that Drosophila CLASP governs dendritic microtubule orientation and dendrite pruning at least partly via suppressing Par-1 kinase.
Collapse
Affiliation(s)
- Shufeng Bu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Quan Tang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | - Yan Wang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | - Samuel Song Yuan Lau
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | - Wei Lin Yong
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Nedozralova H, Basnet N, Ibiricu I, Bodakuntla S, Biertümpfel C, Mizuno N. In situ cryo-electron tomography reveals local cellular machineries for axon branch development. J Biophys Biochem Cytol 2022; 221:213057. [PMID: 35262630 PMCID: PMC8916118 DOI: 10.1083/jcb.202106086] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Neurons are highly polarized cells forming an intricate network of dendrites and axons. They are shaped by the dynamic reorganization of cytoskeleton components and cellular organelles. Axon branching allows the formation of new paths and increases circuit complexity. However, our understanding of branch formation is sparse due to the lack of direct in-depth observations. Using in situ cellular cryo-electron tomography on primary mouse neurons, we directly visualized the remodeling of organelles and cytoskeleton structures at axon branches. Strikingly, branched areas functioned as hotspots concentrating organelles to support dynamic activities. Unaligned actin filaments assembled at the base of premature branches accompanied by filopodia-like protrusions. Microtubules and ER comigrated into preformed branches to support outgrowth together with accumulating compact, ∼500-nm mitochondria and locally clustered ribosomes. We obtained a roadmap of events supporting the hypothesis of local protein synthesis selectively taking place at axon branches, allowing them to serve as unique control hubs for axon development and downstream neural network formation.
Collapse
Affiliation(s)
- Hana Nedozralova
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Martinsried, Germany.,Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Nirakar Basnet
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Iosune Ibiricu
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Satish Bodakuntla
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.,National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
29
|
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: 13] [Impact Index Per Article: 6.5] [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
|
30
|
Rolls MM. Principles of microtubule polarity in linear cells. Dev Biol 2022; 483:112-117. [PMID: 35016908 PMCID: PMC10071391 DOI: 10.1016/j.ydbio.2022.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/06/2022] [Indexed: 01/30/2023]
Abstract
The microtubule cytoskeleton is critical for maintenance of long and long-lived neurons. The overlapping array of microtubules extends from the major site of synthesis in the cell body to the far reaches of axons and dendrites. New materials are transported from the cell body along these neuronal roads by motor proteins, and building blocks and information about the state of affairs in other parts of the cell are returned by motors moving in the opposite direction. As motor proteins walk only in one direction along microtubules, the combination of correct motor and correctly oriented microtubules is essential for moving cargoes in the right direction. In this review, we focus on how microtubule polarity is established and maintained in neurons. At first thought, it seems that figuring out how microtubules are organized in neurons should be simple. After all, microtubules are essentially sticks with a slow-growing minus end and faster-growing plus end, and arranging sticks within the constrained narrow tubes of axons and dendrites should be straightforward. It is therefore quite surprising how many mechanisms contribute to making sure they are arranged in the correct polarity. Some of these mechanisms operate to generate plus-end-out polarity of axons, and others control mixed or minus-end-out dendrites.
Collapse
Affiliation(s)
- Melissa M Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
31
|
Pchitskaya E, Rakovskaya A, Chigray M, Bezprozvanny I. Cytoskeleton Protein EB3 Contributes to Dendritic Spines Enlargement and Enhances Their Resilience to Toxic Effects of Beta-Amyloid. Int J Mol Sci 2022; 23:2274. [PMID: 35216391 PMCID: PMC8875759 DOI: 10.3390/ijms23042274] [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: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/16/2022] Open
Abstract
EB3 protein is expressed abundantly in the nervous system and transiently enters the dendritic spines at the tip of the growing microtubule, which leads to spine enlargement. Nevertheless, the role of dynamic microtubules, and particularly EB3 protein, in synapse function is still elusive. By manipulating the EB3 expression level, we have shown that this protein is required for a normal dendritogenesis. Nonetheless, EB3 overexpression also reduces hippocampal neurons dendritic branching and total dendritic length. This effect likely occurs due to the speeding neuronal development cycle from dendrite outgrowth to the step when dendritic spines are forming. Implementing direct morphometric characterization of dendritic spines, we showed that EB3 overexpression leads to a dramatic increase in the dendritic spine head area. EB3 knockout oppositely reduces spine head area and increases spine neck length and spine neck/spine length ratio. The same effect is observed in conditions of amyloid-beta toxicity, modeling Alzheimer`s disease. Neck elongation is supposed to be a common detrimental effect on the spine's shape, which makes them biochemically and electrically less connected to the dendrite. EB3 also potentiates the formation of presynaptic protein Synapsin clusters and CaMKII-alpha preferential localization in spines rather than in dendrites of hippocampal neurons, while its downregulation has an opposite effect and reduces the size of presynaptic protein clusters Synapsin and PSD95. EB3's role in spine development and maturation determines its neuroprotective effect. EB3 overexpression makes dendritic spines resilient to amyloid-beta toxicity, restores altered PSD95 clustering, and reduces CaMKII-alpha localization in spines observed in this pathological state.
Collapse
Affiliation(s)
- Ekaterina Pchitskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.P.); (A.R.); (M.C.)
| | - Anastasiya Rakovskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.P.); (A.R.); (M.C.)
| | - Margarita Chigray
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.P.); (A.R.); (M.C.)
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.P.); (A.R.); (M.C.)
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| |
Collapse
|
32
|
Postnatal development of inner lamina II interneurons of the rat medullary dorsal horn. Pain 2021; 163:984-998. [PMID: 34433770 DOI: 10.1097/j.pain.0000000000002459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/16/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT Pain processing in young mammals is immature. Despite the central role of the medullary dorsal horn (MDH) in processing orofacial sensory information, the maturation of the neurons within the MDH has been largely overlooked. Combining in vitro electrophysiological recordings and 3D morphological analysis over the first postnatal month in rats, we investigated the age-dependent development of the neurons within the inner lamina II (IIi) of the MDH. We show the lamina IIi neuronal population transition into a more hyperpolarized state, with modification of the action potential waveform, and a shift from single spiking, at early postnatal ages, to tonic firing and initial bursting at later stages. These physiological changes are associated with a strong structural remodelling of the neuronal morphology with most of the modifications occurring after the third postnatal week. Among the lamina IIi neuronal population, the subpopulation of interneurons expressing the γ isoform of the protein kinase C (PKCγ+) are key elements for the circuits underlying facial mechanical allodynia. How do they develop from the rest of the lamina IIi constitute an important question that remained to be addressed. Here, we show that PKCγ+ interneurons display electrophysiological changes over time comparable with the PKCγ- population. However, they show a distinctive increase of the soma volume and primary branches length, as opposed to the PKCγ- population. Together, our data demonstrate a novel pattern of late postnatal maturation of lamina IIi interneurons, with a spotlight on PKCγ+ interneurons, that may be relevant for the development of orofacial sensitivity.
Collapse
|
33
|
Yang TC, Yarmishyn AA, Yang YP, Lu PC, Chou SJ, Wang ML, Lin TC, Hwang DK, Chou YB, Chen SJ, Yu WK, Wang AG, Hsu CC, Chiou SH. Mitochondrial transport mediates survival of retinal ganglion cells in affected LHON patients. Hum Mol Genet 2021; 29:1454-1464. [PMID: 32277753 DOI: 10.1093/hmg/ddaa063] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/02/2020] [Accepted: 03/30/2020] [Indexed: 01/17/2023] Open
Abstract
The mutations in the genes encoding the subunits of complex I of the mitochondrial electron transport chain are the most common cause of Leber's hereditary optic neuropathy (LHON), a maternal hereditary disease characterized by retinal ganglion cell (RGC) degeneration. The characteristics of incomplete penetrance indicate that nuclear genetic and environmental factors also determine phenotypic expression of LHON. Therefore, further understanding of the role of mutant mitochondrial nicotinamide adenine dinucleotide dehydrogenase subunit proteins and nuclear genetic factors/environmental effects in the etiology of LHON is needed. In this study, we generated human-induced pluripotent stem cells (hiPSCs) from healthy control, unaffected LHON mutation carrier, and affected LHON patient. hiPSC-derived RGCs were used to study the differences between affected and unaffected carriers of mitochondrial DNA point mutation m.11778G > A in the MT-ND4 gene. We found that both mutated cell lines were characterized by increase in reactive oxygen species production, however, only affected cell line had increased levels of apoptotic cells. We found a significant increase in retrograde mitochondria and a decrease in stationary mitochondria in the affected RGC axons. In addition, the messenger RNA and protein levels of KIF5A in the LHON-affected RGCs were significantly reduced. Antioxidant N-acetyl-L-cysteine could restore the expression of KIF5A and the normal pattern of mitochondrial movement in the affected RGCs. To conclude, we found essential differences in the mutually dependent processes of oxidative stress, mitochondrial transport and apoptosis between two LHON-specific mutation carrier RGC cell lines, asymptomatic carrier and disease-affected, and identified KIF5A as a central modulator of these differences.
Collapse
Affiliation(s)
- Tien-Chun Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | | | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Pin-Chen Lu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Tai-Chi Lin
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Bai Chou
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Jen Chen
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Wei-Kuang Yu
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - An-Guor Wang
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chih-Chien Hsu
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
34
|
Tracking Fungal Growth: Establishment of Arp1 as a Marker for Polarity Establishment and Active Hyphal Growth in Filamentous Ascomycetes. J Fungi (Basel) 2021; 7:jof7070580. [PMID: 34356959 PMCID: PMC8304394 DOI: 10.3390/jof7070580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/20/2022] Open
Abstract
Polar growth is a key characteristic of all filamentous fungi. It allows these eukaryotes to not only effectively explore organic matter but also interact within its own colony, mating partners, and hosts. Therefore, a detailed understanding of the dynamics in polar growth establishment and maintenance is crucial for several fields of fungal research. We developed a new marker protein, the actin-related protein 1 (Arp1) fused to red and green fluorescent proteins, which allows for the tracking of polar axis establishment and active hyphal growth in microscopy approaches. To exclude a probable redundancy with known polarity markers, we compared the localizations of the Spitzenkörper (SPK) and Arp1 using an FM4-64 staining approach. As we show in applications with the coprophilous fungus Sordaria macrospora and the hemibiotrophic plant pathogen Colletotrichum graminicola, the monitoring of Arp1 can be used for detailed studies of hyphal growth dynamics and ascospore germination, the interpretation of chemotropic growth processes, and the tracking of elongating penetration pegs into plant material. Since the Arp1 marker showed the same dynamics in both fungi tested, we believe this marker can be broadly applied in fungal research to study the manifold polar growth processes determining fungal life.
Collapse
|
35
|
Shorey M, Rao K, Stone MC, Mattie FJ, Sagasti A, Rolls MM. Microtubule organization of vertebrate sensory neurons in vivo. Dev Biol 2021; 478:1-12. [PMID: 34147472 DOI: 10.1016/j.ydbio.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/30/2023]
Abstract
Dorsal root ganglion (DRG) neurons are the predominant cell type that innervates the vertebrate skin. They are typically described as pseudounipolar cells that have central and peripheral axons branching from a single root exiting the cell body. The peripheral axon travels within a nerve to the skin, where free sensory endings can emerge and branch into an arbor that receives and integrates information. In some immature vertebrates, DRG neurons are preceded by Rohon-Beard (RB) neurons. While the sensory endings of RB and DRG neurons function like dendrites, we use live imaging in zebrafish to show that they have axonal plus-end-out microtubule polarity at all stages of maturity. Moreover, we show both cell types have central and peripheral axons with plus-end-out polarity. Surprisingly, in DRG neurons these emerge separately from the cell body, and most cells never acquire the signature pseudounipolar morphology. Like another recently characterized cell type that has multiple plus-end-out neurites, ganglion cells in Nematostella, RB and DRG neurons maintain a somatic microtubule organizing center even when mature. In summary, we characterize key cellular and subcellular features of vertebrate sensory neurons as a foundation for understanding their function and maintenance.
Collapse
Affiliation(s)
- Matthew Shorey
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kavitha Rao
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Michelle C Stone
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Floyd J Mattie
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Alvaro Sagasti
- Molecular, Cell and Developmental Biology Department and Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
36
|
Feng C, Cleary JM, Kothe GO, Stone MC, Weiner AT, Hertzler JI, Hancock WO, Rolls MM. Trim9 and Klp61F promote polymerization of new dendritic microtubules along parallel microtubules. J Cell Sci 2021; 134:jcs258437. [PMID: 34096607 PMCID: PMC8214762 DOI: 10.1242/jcs.258437] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/04/2021] [Indexed: 02/03/2023] Open
Abstract
Axons and dendrites are distinguished by microtubule polarity. In Drosophila, dendrites are dominated by minus-end-out microtubules, whereas axons contain plus-end-out microtubules. Local nucleation in dendrites generates microtubules in both orientations. To understand why dendritic nucleation does not disrupt polarity, we used live imaging to analyze the fate of microtubules generated at branch points. We found that they had different rates of success exiting the branch based on orientation: correctly oriented minus-end-out microtubules succeeded in leaving about twice as often as incorrectly oriented microtubules. Increased success relied on other microtubules in a parallel orientation. From a candidate screen, we identified Trim9 and kinesin-5 (Klp61F) as machinery that promoted growth of new microtubules. In S2 cells, Eb1 recruited Trim9 to microtubules. Klp61F promoted microtubule growth in vitro and in vivo, and could recruit Trim9 in S2 cells. In summary, the data argue that Trim9 and kinesin-5 act together at microtubule plus ends to help polymerizing microtubules parallel to pre-existing ones resist catastrophe.
Collapse
Affiliation(s)
- Chengye Feng
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Joseph M. Cleary
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gregory O. Kothe
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Michelle C. Stone
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Alexis T. Weiner
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - James I. Hertzler
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - William O. Hancock
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Melissa M. Rolls
- Biochemistry and Molecular Biology Department and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
37
|
Coquand L, Victoria GS, Tata A, Carpentieri JA, Brault JB, Guimiot F, Fraisier V, Baffet AD. CAMSAPs organize an acentrosomal microtubule network from basal varicosities in radial glial cells. J Cell Biol 2021; 220:212175. [PMID: 34019079 PMCID: PMC8144914 DOI: 10.1083/jcb.202003151] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 11/30/2020] [Accepted: 04/29/2021] [Indexed: 01/05/2023] Open
Abstract
Neurons of the neocortex are generated by stem cells called radial glial cells. These polarized cells extend a short apical process toward the ventricular surface and a long basal fiber that acts as a scaffold for neuronal migration. How the microtubule cytoskeleton is organized in these cells to support long-range transport is unknown. Using subcellular live imaging within brain tissue, we show that microtubules in the apical process uniformly emanate for the pericentrosomal region, while microtubules in the basal fiber display a mixed polarity, reminiscent of the mammalian dendrite. We identify acentrosomal microtubule organizing centers localized in varicosities of the basal fiber. CAMSAP family members accumulate in these varicosities, where they control microtubule growth. Double knockdown of CAMSAP1 and 2 leads to a destabilization of the entire basal process. Finally, using live imaging of human fetal cortex, we reveal that this organization is conserved in basal radial glial cells, a related progenitor cell population associated with human brain size expansion.
Collapse
Affiliation(s)
- Laure Coquand
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France
| | - Guiliana Soraya Victoria
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France
| | - Alice Tata
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France
| | - Jacopo Amerigo Carpentieri
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France
| | - Jean-Baptiste Brault
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France
| | - Fabien Guimiot
- Unité de Fœtopathologie-Université de Paris et Institut national de la santé et de la recherche médicale UMR1141, Hôpital Robert Debré, Paris, France
| | - Vincent Fraisier
- UMR144-Cell and Tissue Imaging Facility, Centre national de la recherche scientifique-Institut Curie, Paris, France
| | - Alexandre D Baffet
- Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR144, Paris, France.,Institut national de la santé et de la recherche médicale, Paris, France
| |
Collapse
|
38
|
Parato J, Bartolini F. The microtubule cytoskeleton at the synapse. Neurosci Lett 2021; 753:135850. [PMID: 33775740 DOI: 10.1016/j.neulet.2021.135850] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
In neurons, microtubules (MTs) provide routes for transport throughout the cell and structural support for dendrites and axons. Both stable and dynamic MTs are necessary for normal neuronal functions. Research in the last two decades has demonstrated that MTs play additional roles in synaptic structure and function in both pre- and postsynaptic elements. Here, we review current knowledge of the functions that MTs perform in excitatory and inhibitory synapses, as well as in the neuromuscular junction and other specialized synapses, and discuss the implications that this knowledge may have in neurological disease.
Collapse
Affiliation(s)
- Julie Parato
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States; SUNY Empire State College, Department of Natural Sciences, 177 Livingston Street, Brooklyn, NY, 11201, United States
| | - Francesca Bartolini
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States.
| |
Collapse
|
39
|
To nucleate or not, that is the question in neurons. Neurosci Lett 2021; 751:135806. [PMID: 33705928 DOI: 10.1016/j.neulet.2021.135806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 01/19/2023]
Abstract
Microtubules are the structural center of neurons, stretching in overlapping arrays from the cell body to the far reaches of axons and dendrites. They also act as the tracks for long-range transport mediated by dynein and kinesin motors. Transcription and most translation take place in the cell body, and newly made cargoes must be shipped from this site of synthesis to sites of function in axons and dendrites. This constant demand for transport means that the microtubule array must be present without gaps throughout the cell over the lifetime of the animal. This task is made slightly easier in many animals by the relatively long, stable microtubules present in neurons. However, even stable neuronal microtubules have ends that are dynamic, and individual microtubules typically last on the order of hours, while the neurons around them last a lifetime. "Birth" of new microtubules is therefore required to maintain the neuronal microtubule array. In this review we discuss the nucleation of new microtubules in axons and dendrites, including how and where they are nucleated. In addition, it is becoming clear that neuronal microtubule nucleation is highly regulated, with unexpected machinery impinging on the decision of whether nucleation sites are active or inactive through space and time.
Collapse
|
40
|
Chen YC, Huang HR, Hsu CH, Ou CY. CRMP/UNC-33 organizes microtubule bundles for KIF5-mediated mitochondrial distribution to axon. PLoS Genet 2021; 17:e1009360. [PMID: 33571181 PMCID: PMC7904166 DOI: 10.1371/journal.pgen.1009360] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/24/2021] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons are highly specialized cells with polarized cellular processes and subcellular domains. As vital organelles for neuronal functions, mitochondria are distributed by microtubule-based transport systems. Although the essential components of mitochondrial transport including motors and cargo adaptors are identified, it is less clear how mitochondrial distribution among somato-dendritic and axonal compartment is regulated. Here, we systematically study mitochondrial motors, including four kinesins, KIF5, KIF17, KIF1, KLP-6, and dynein, and transport regulators in C. elegans PVD neurons. Among all these motors, we found that mitochondrial export from soma to neurites is mainly mediated by KIF5/UNC-116. Interestingly, UNC-116 is especially important for axonal mitochondria, while dynein removes mitochondria from all plus-end dendrites and the axon. We surprisingly found one mitochondrial transport regulator for minus-end dendritic compartment, TRAK-1, and two mitochondrial transport regulators for axonal compartment, CRMP/UNC-33 and JIP3/UNC-16. While JIP3/UNC-16 suppresses axonal mitochondria, CRMP/UNC-33 is critical for axonal mitochondria; nearly no axonal mitochondria present in unc-33 mutants. We showed that UNC-33 is essential for organizing the population of UNC-116-associated microtubule bundles, which are tracks for mitochondrial trafficking. Disarrangement of these tracks impedes mitochondrial transport to the axon. In summary, we identified a compartment-specific transport regulation of mitochondria by UNC-33 through organizing microtubule tracks for different kinesin motors other than microtubule polarity.
Collapse
Affiliation(s)
- Ying-Chun Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hao-Ru Huang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hao Hsu
- Institute of Biochemistry and Molecular Biology, 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
|
41
|
Li P, Li L, Yu B, Wang X, Wang Q, Lin J, Zheng Y, Zhu J, He M, Xia Z, Tu M, Liu JS, Lin Z, Fu X. Doublecortin facilitates the elongation of the somatic Golgi apparatus into proximal dendrites. Mol Biol Cell 2021; 32:422-434. [PMID: 33405953 PMCID: PMC8098852 DOI: 10.1091/mbc.e19-09-0530] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in the doublecortin (DCX) gene, which encodes a microtubule (MT)-binding protein, cause human cortical malformations, including lissencephaly and subcortical band heterotopia. A deficiency in DCX and DCX-like kinase 1 (DCLK1), a functionally redundant and structurally similar cognate of DCX, decreases neurite length and increases the number of primary neurites directly arising from the soma. The underlying mechanism is not completely understood. In this study, the elongation of the somatic Golgi apparatus into proximal dendrites, which have been implicated in dendrite patterning, was significantly decreased in the absence of DCX/DCLK1. Phosphorylation of DCX at S47 or S327 was involved in this process. DCX deficiency shifted the distribution of CLASP2 proteins to the soma from the dendrites. In addition to CLASP2, dynein and its cofactor JIP3 were abnormally distributed in DCX-deficient neurons. The association between JIP3 and dynein was significantly increased in the absence of DCX. Down-regulation of CLASP2 or JIP3 expression with specific shRNAs rescued the Golgi phenotype observed in DCX-deficient neurons. We conclude that DCX regulates the elongation of the Golgi apparatus into proximal dendrites through MT-associated proteins and motors.
Collapse
Affiliation(s)
- Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Luyao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Binyuan Yu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xinye Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qi Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jingjing Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yihui Zheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jinjin Zhu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Minzhi He
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhaonan Xia
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Mengjing Tu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Judy S Liu
- Department of Neurology, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903
| | - Zhenlang Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaoqin Fu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
42
|
Leterrier C. A Pictorial History of the Neuronal Cytoskeleton. J Neurosci 2021; 41:11-27. [PMID: 33408133 PMCID: PMC7786211 DOI: 10.1523/jneurosci.2872-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Christophe Leterrier
- Aix Marseille Université, Centre National de la Recherche Scientifique, INP Unité Mixte de Recherche 7051, NeuroCyto, Marseille 13005, France
| |
Collapse
|
43
|
Stone MC, Kothe GO, Rolls MM, Jegla T. Cytoskeletal and synaptic polarity of LWamide-like+ ganglion neurons in the sea anemone Nematostella vectensis. J Exp Biol 2020; 223:jeb233197. [PMID: 32968001 PMCID: PMC7673360 DOI: 10.1242/jeb.233197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022]
Abstract
The centralized nervous systems of bilaterian animals rely on directional signaling facilitated by polarized neurons with specialized axons and dendrites. It is not known whether axo-dendritic polarity is exclusive to bilaterians or was already present in early metazoans. We therefore examined neurite polarity in the starlet sea anemone Nematostella vectensis (Cnidaria). Cnidarians form a sister clade to bilaterians and share many neuronal building blocks characteristic of bilaterians, including channels, receptors and synaptic proteins, but their nervous systems comprise a comparatively simple net distributed throughout the body. We developed a tool kit of fluorescent polarity markers for live imaging analysis of polarity in an identified neuron type, large ganglion cells of the body column nerve net that express the LWamide-like neuropeptide. Microtubule polarity differs in bilaterian axons and dendrites, and this in part underlies polarized distribution of cargo to the two types of processes. However, in LWamide-like+ neurons, all neurites had axon-like microtubule polarity suggesting that they may have similar contents. Indeed, presynaptic and postsynaptic markers trafficked to all neurites and accumulated at varicosities where neurites from different neurons often crossed, suggesting the presence of bidirectional synaptic contacts. Furthermore, we could not identify a diffusion barrier in the plasma membrane of any of the neurites like the axon initial segment barrier that separates the axonal and somatodendritic compartments in bilaterian neurons. We conclude that at least one type of neuron in Nematostella vectensis lacks the axo-dendritic polarity characteristic of bilaterian neurons.
Collapse
Affiliation(s)
- Michelle C Stone
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gregory O Kothe
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Timothy Jegla
- Department of Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
44
|
Microtubule Dysfunction: A Common Feature of Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21197354. [PMID: 33027950 PMCID: PMC7582320 DOI: 10.3390/ijms21197354] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons are particularly susceptible to microtubule (MT) defects and deregulation of the MT cytoskeleton is considered to be a common insult during the pathogenesis of neurodegenerative disorders. Evidence that dysfunctions in the MT system have a direct role in neurodegeneration comes from findings that several forms of neurodegenerative diseases are associated with changes in genes encoding tubulins, the structural units of MTs, MT-associated proteins (MAPs), or additional factors such as MT modifying enzymes which modulating tubulin post-translational modifications (PTMs) regulate MT functions and dynamics. Efforts to use MT-targeting therapeutic agents for the treatment of neurodegenerative diseases are underway. Many of these agents have provided several benefits when tested on both in vitro and in vivo neurodegenerative model systems. Currently, the most frequently addressed therapeutic interventions include drugs that modulate MT stability or that target tubulin PTMs, such as tubulin acetylation. The purpose of this review is to provide an update on the relevance of MT dysfunctions to the process of neurodegeneration and briefly discuss advances in the use of MT-targeting drugs for the treatment of neurodegenerative disorders.
Collapse
|
45
|
Zupa E, Liu P, Würtz M, Schiebel E, Pfeffer S. The structure of the γ-TuRC: a 25-years-old molecular puzzle. Curr Opin Struct Biol 2020; 66:15-21. [PMID: 33002806 DOI: 10.1016/j.sbi.2020.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
The nucleation of microtubules from αβ-tubulin dimers is an essential cellular process dependent on γ-tubulin complexes. Mechanistic understanding of the nucleation reaction was hampered by the lack of γ-tubulin complex structures at sufficiently high resolution. The recent technical developments in cryo-electron microscopy have allowed resolving the vertebrate γ-tubulin ring complex (γ-TuRC) structure at near-atomic resolution. These studies clarified the arrangement and stoichiometry of gamma-tubulin complex proteins in the γ-TuRC, characterized the surprisingly versatile integration of the small proteins MZT1/2 into the complex, and identified actin as an integral component of the γ-TuRC. In this review, we summarize the structural insights into the molecular architecture, the assembly pathway, and the regulation of the microtubule nucleation reaction.
Collapse
Affiliation(s)
- Erik Zupa
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Peng Liu
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany.
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany.
| |
Collapse
|
46
|
Cuentas-Condori A, Miller Rd DM. Synaptic remodeling, lessons from C. elegans. J Neurogenet 2020; 34:307-322. [PMID: 32808848 PMCID: PMC7855814 DOI: 10.1080/01677063.2020.1802725] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/07/2020] [Indexed: 02/08/2023]
Abstract
Sydney Brenner's choice of Caenorhabditis elegans as a model organism for understanding the nervous system has accelerated discoveries of gene function in neural circuit development and behavior. In this review, we discuss a striking example of synaptic remodeling in the C. elegans motor circuit in which DD class motor neurons effectively reverse polarity as presynaptic and postsynaptic domains at opposite ends of the DD neurite switch locations. Originally revealed by EM reconstruction conducted over 40 years ago, DD remodeling has since been investigated by live cell imaging methods that exploit the power of C. elegans genetics to reveal key effectors of synaptic plasticity. Although synapses are also extensively rewired in developing mammalian circuits, the underlying remodeling mechanisms are largely unknown. Here, we highlight the possibility that studies in C. elegans can reveal pathways that orchestrate synaptic remodeling in more complex organisms. Specifically, we describe (1) transcription factors that regulate DD remodeling, (2) the cellular and molecular cascades that drive synaptic remodeling and (3) examples of circuit modifications in vertebrate neurons that share some similarities with synaptic remodeling in C. elegans DD neurons.
Collapse
|
47
|
Shorey M, Stone MC, Mandel J, Rolls MM. Neurons survive simultaneous injury to axons and dendrites and regrow both types of processes in vivo. Dev Biol 2020; 465:108-118. [PMID: 32687893 DOI: 10.1016/j.ydbio.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Neurons extend dendrites and axons to receive and send signals. If either type of process is removed, the cell cannot function. Rather than undergoing cell death, some neurons can regrow axons and dendrites. Axon and dendrite regeneration have been examined separately and require sensing the injury and reinitiating the correct growth program. Whether neurons in vivo can sense and respond to simultaneous axon and dendrite injury with polarized regeneration has not been explored. To investigate the outcome of simultaneous axon and dendrite damage, we used a Drosophila model system in which neuronal polarity, axon regeneration, and dendrite regeneration have been characterized. After removal of the axon and all but one dendrite, the remaining dendrite was converted to a process that had a long unbranched region that extended over long distances and a region where shorter branched processes were added. These observations suggested axons and dendrites could regrow at the same time. To further test the capacity of neurons to implement polarized regeneration after axon and dendrite damage, we removed all neurites from mature neurons. In this case a long unbranched neurite and short branched neurites were regrown from the stripped cell body. Moreover, the long neurite had axonal plus-end-out microtubule polarity and the shorter neurites had mixed polarity consistent with dendrite identity. The long process also accumulated endoplasmic reticulum at its tip like regenerating axons. We conclude that neurons in vivo can respond to simultaneous axon and dendrite injury by initiating growth of a new axon and new dendrites.
Collapse
Affiliation(s)
- Matthew Shorey
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Michelle C Stone
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Jenna Mandel
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Melissa M Rolls
- Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
48
|
Hill SE, Colón-Ramos DA. The Journey of the Synaptic Autophagosome: A Cell Biological Perspective. Neuron 2020; 105:961-973. [PMID: 32191859 DOI: 10.1016/j.neuron.2020.01.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 01/13/2023]
Abstract
Autophagy is a key cellular degradative pathway, important for neuronal homeostasis and function. Disruption of autophagy is associated with neuronal dysfunction and neurodegeneration. Autophagy is compartmentalized in neurons, with specific stages of the pathway occurring in distinct subcellular compartments. Coordination of these stages drives progression of autophagy and enables clearance of substrates. Yet, we are only now learning how these distributed processes are integrated across the neuron. In this review, we focus on the cell biological course of autophagy in neurons, from biogenesis at the synapse to degradation in the soma. We describe how the steps of autophagy are distributed across neuronal subcellular compartments, how local machinery regulates autophagy, and the impact of coordinated regulation on neuronal physiology and disease. We also discuss how recent advances in our understanding of neuronal autophagic mechanisms have reframed how we think about the role of local regulation of autophagy in all tissues.
Collapse
Affiliation(s)
- Sarah E Hill
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, PO Box 9812, New Haven, CT 06536-0812, USA; Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Daniel A Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine, PO Box 9812, New Haven, CT 06536-0812, USA; Instituto de Neurobiología José del Castillo, Universidad de Puerto Rico, San Juan, PR, USA.
| |
Collapse
|
49
|
Prokop A. Cytoskeletal organization of axons in vertebrates and invertebrates. J Cell Biol 2020; 219:e201912081. [PMID: 32369543 PMCID: PMC7337489 DOI: 10.1083/jcb.201912081] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The maintenance of axons for the lifetime of an organism requires an axonal cytoskeleton that is robust but also flexible to adapt to mechanical challenges and to support plastic changes of axon morphology. Furthermore, cytoskeletal organization has to adapt to axons of dramatically different dimensions, and to their compartment-specific requirements in the axon initial segment, in the axon shaft, at synapses or in growth cones. To understand how the cytoskeleton caters to these different demands, this review summarizes five decades of electron microscopic studies. It focuses on the organization of microtubules and neurofilaments in axon shafts in both vertebrate and invertebrate neurons, as well as the axon initial segments of vertebrate motor- and interneurons. Findings from these ultrastructural studies are being interpreted here on the basis of our contemporary molecular understanding. They strongly suggest that axon architecture in animals as diverse as arthropods and vertebrates is dependent on loosely cross-linked bundles of microtubules running all along axons, with only minor roles played by neurofilaments.
Collapse
Affiliation(s)
- Andreas Prokop
- School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
50
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|