1
|
Baidoe-Ansah D, Mirzapourdelavar H, Aleshin S, Schott BH, Seidenbecher C, Kaushik R, Dityatev A. Neurocan regulates axon initial segment organization and neuronal activity. Matrix Biol 2025:S0945-053X(25)00001-0. [PMID: 39788215 DOI: 10.1016/j.matbio.2025.01.001] [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: 11/17/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The neural extracellular matrix (ECM) accumulates in the form of perineuronal nets (PNNs), particularly around fast-spiking GABAergic interneurons in the cortex and hippocampus, but also around synapses and in association with the axon initial segments (AIS) and nodes of Ranvier. Increasing evidence highlights the role of Neurocan (Ncan), a brain-specific component of ECM, in the pathophysiology of neuropsychiatric disorders like bipolar disorder and schizophrenia. Ncan localizes at PNNs, perisynaptically, and at the nodes of Ranvier and the AIS, highlighting its potential role in regulating axonal excitability. Here, we used knockdown and knockout approaches in mouse primary cortical neurons in combination with immunocytochemistry, western blotting and electrophysiological techniques to characterize the role of Ncan in the organization of PNNs and AISs and the upregulation of neuronal activity. We found that reduced Ncan levels led to remodeling of PNNs around neurons via upregulation of Aggrecan mRNA and protein levels, increased expression of activity-dependent c-Fos and FosB genes and elevated spontaneous synaptic activity. The latter correlated with increased levels of Ankyrin-G in the AIS, particularly in excitatory neurons, and with the elevated expression of Nav1.6 channels. Our results suggest that Ncan regulates the expression of key proteins in PNNs and AISs and provide new insights into its role in fine-tuning neuronal functions.
Collapse
Affiliation(s)
- David Baidoe-Ansah
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Stepan Aleshin
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Björn Hendrik Schott
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena-Magdeburg-Halle
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
2
|
Thome C, Janssen JM, Karabulut S, Acuna C, D’Este E, Soyka SJ, Baum K, Bock M, Lehmann N, Roos J, Stevens NA, Hasegawa M, Ganea DA, Benoit CM, Gründemann J, Min L, Bird KM, Schultz C, Bennett V, Jenkins PM, Engelhardt M. Live imaging of excitable axonal microdomains in ankyrin-G-GFP mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.01.525891. [PMID: 38948770 PMCID: PMC11212890 DOI: 10.1101/2023.02.01.525891] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The axon initial segment (AIS) constitutes not only the site of action potential initiation, but also a hub for activity-dependent modulation of output generation. Recent studies shedding light on AIS function used predominantly post-hoc approaches since no robust murine in vivo live reporters exist. Here, we introduce a reporter line in which the AIS is intrinsically labeled by an ankyrin-G-GFP fusion protein activated by Cre recombinase, tagging the native Ank3 gene. Using confocal, superresolution, and two-photon microscopy as well as whole-cell patch-clamp recordings in vitro, ex vivo, and in vivo, we confirm that the subcellular scaffold of the AIS and electrophysiological parameters of labeled cells remain unchanged. We further uncover rapid AIS remodeling following increased network activity in this model system, as well as highly reproducible in vivo labeling of AIS over weeks. This novel reporter line allows longitudinal studies of AIS modulation and plasticity in vivo in real-time and thus provides a unique approach to study subcellular plasticity in a broad range of applications.
Collapse
Affiliation(s)
- Christian Thome
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Jan Maximilian Janssen
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Seda Karabulut
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Elisa D’Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Stella J. Soyka
- Institute of Anatomy and Cell Biology, Dept. of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Konrad Baum
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
| | - Michael Bock
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nadja Lehmann
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Johannes Roos
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nikolas A. Stevens
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Masashi Hasegawa
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
| | - Dan A. Ganea
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Chloé M. Benoit
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Jan Gründemann
- German Center for Neurodegenerative Disease (DZNE), Neural Circuit Computations, 53127 Bonn, Germany
- University of Basel, Department of Biomedicine, 4031 Basel, Switzerland
| | - Lia Min
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kalynn M. Bird
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christian Schultz
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Vann Bennett
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Paul M. Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maren Engelhardt
- Institute of Anatomy and Cell Biology, Johannes Kepler University, 4020 Linz, Austria
- Clinical Research Institute of Neuroscience, Johannes Kepler University, 4020 Linz, Austria
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
3
|
Rentsch J, Bandstra S, Sezen B, Sigrist P, Bottanelli F, Schmerl B, Shoichet S, Noé F, Sadeghi M, Ewers H. Sub-membrane actin rings compartmentalize the plasma membrane. J Cell Biol 2024; 223:e202310138. [PMID: 38252080 PMCID: PMC10807028 DOI: 10.1083/jcb.202310138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
The compartmentalization of the plasma membrane (PM) is a fundamental feature of cells. The diffusivity of membrane proteins is significantly lower in biological than in artificial membranes. This is likely due to actin filaments, but assays to prove a direct dependence remain elusive. We recently showed that periodic actin rings in the neuronal axon initial segment (AIS) confine membrane protein motion between them. Still, the local enrichment of ion channels offers an alternative explanation. Here we show, using computational modeling, that in contrast to actin rings, ion channels in the AIS cannot mediate confinement. Furthermore, we show, employing a combinatorial approach of single particle tracking and super-resolution microscopy, that actin rings are close to the PM and that they confine membrane proteins in several neuronal cell types. Finally, we show that actin disruption leads to loss of compartmentalization. Taken together, we here develop a system for the investigation of membrane compartmentalization and show that actin rings compartmentalize the PM.
Collapse
Affiliation(s)
- Jakob Rentsch
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Selle Bandstra
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Batuhan Sezen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Philipp Sigrist
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Francesca Bottanelli
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Bettina Schmerl
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Frank Noé
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Mohsen Sadeghi
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Helge Ewers
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
4
|
Xu Z, Angstmann CN, Wu Y, Stefen H, Parić E, Fath T, Curmi PM. Location of the axon initial segment assembly can be predicted from neuronal shape. iScience 2024; 27:109264. [PMID: 38450155 PMCID: PMC10915628 DOI: 10.1016/j.isci.2024.109264] [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: 09/28/2023] [Revised: 12/21/2023] [Accepted: 02/14/2024] [Indexed: 03/08/2024] Open
Abstract
The axon initial segment (AIS) is located at the proximal axon demarcating the boundary between axonal and somatodendritic compartments. The AIS facilitates the generation of action potentials and maintenance of neuronal polarity. In this study, we show that the location of AIS assembly, as marked by Ankyrin G, corresponds to the nodal plane of the lowest-order harmonic of the Laplace-Beltrami operator solved over the neuronal shape. This correlation establishes a coupling between location of AIS assembly and neuronal cell morphology. We validate this correlation for neurons with atypical morphology and neurons containing multiple AnkG clusters on distinct neurites, where the nodal plane selects the appropriate axon showing enriched Tau. Based on our findings, we propose that Turing patterning systems are candidates for dynamically governing AIS location. Overall, this study highlights the importance of neuronal cell morphology in determining the precise localization of the AIS within the proximal axon.
Collapse
Affiliation(s)
- Zhuang Xu
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
- School of Mathematics and Statistics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Christopher N. Angstmann
- School of Mathematics and Statistics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Yuhuang Wu
- Infection Analytics Program, Kirby Institute for Infection and Immunity, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Holly Stefen
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Esmeralda Parić
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Thomas Fath
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Paul M.G. Curmi
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Fréal A, Jamann N, Ten Bos J, Jansen J, Petersen N, Ligthart T, Hoogenraad CC, Kole MH. Sodium channel endocytosis drives axon initial segment plasticity. SCIENCE ADVANCES 2023; 9:eadf3885. [PMID: 37713493 PMCID: PMC10881073 DOI: 10.1126/sciadv.adf3885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
Activity-dependent plasticity of the axon initial segment (AIS) endows neurons with the ability to adapt action potential output to changes in network activity. Action potential initiation at the AIS highly depends on the clustering of voltage-gated sodium channels, but the molecular mechanisms regulating their plasticity remain largely unknown. Here, we developed genetic tools to label endogenous sodium channels and their scaffolding protein, to reveal their nanoscale organization and longitudinally image AIS plasticity in hippocampal neurons in slices and primary cultures. We find that N-methyl-d-aspartate receptor activation causes both long-term synaptic depression and rapid internalization of AIS sodium channels within minutes. The clathrin-mediated endocytosis of sodium channels at the distal AIS increases the threshold for action potential generation. These data reveal a fundamental mechanism for rapid activity-dependent AIS reorganization and suggests that plasticity of intrinsic excitability shares conserved features with synaptic plasticity.
Collapse
Affiliation(s)
- Amélie Fréal
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Nora Jamann
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jolijn Ten Bos
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jacqueline Jansen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Naomi Petersen
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Thijmen Ligthart
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Casper C. Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Department of Neuroscience, Genentech Inc, South San Francisco, CA, USA
| | - Maarten H. P. Kole
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
6
|
Chai Z, Gu S, Lykotrafitis G. Dynamics of the axon plasma membrane skeleton. SOFT MATTER 2023; 19:2514-2528. [PMID: 36939651 DOI: 10.1039/d2sm01602h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
It was recently revealed via super-resolution microscopy experiments that the axon plasma membrane skeleton (APMS) comprises a series of periodically arranged azimuthal actin rings connected via longitudinal spectrin filaments forming an orthotropic network. The common perception is that APMS enhances structural stability of the axon but its impact on axon deformation is unknown. To investigate the response of the APMS to extension, we introduce a coarse-grain molecular dynamics model consisting of actin particles forming rings and chains of particles representing spectrin tetramers with repeats than can unfold. We observe that the shape of force-extension curve is initially linear and the force level depends on the extension rate. Even during the initial deformation stage, unfolding of spectrin repeats occurs, but the saw-tooth shape of the corresponding force-extension curve observed in the case of one spectrin tetramer does not appear in the case of the entire APMS. The reason is that spectrin unfolding is not synchronized across filaments during extension. If actin-spectrin associations remain intact, the force-extension response reaches a perfectly plastic region because of increased spectrin unfolding frequency. However, when actin-spectrin links dissociate, which can happen at moderate and high extension rates, APMS softens and the resistance force decreases linearly as the axon elongates until it reaches a point where the APMS is completely severed. Furthermore, when the ring-to-ring distance is maintained fixed under stretch, the resistance force relaxes exponentially as a function of time due to additional unfolding of spectrin tetramers following the Kelvin-Voigt representation of the Zener model.
Collapse
Affiliation(s)
- Zhaojie Chai
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA.
| | - Shiju Gu
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - George Lykotrafitis
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
7
|
Impaired OTUD7A-dependent Ankyrin regulation mediates neuronal dysfunction in mouse and human models of the 15q13.3 microdeletion syndrome. Mol Psychiatry 2023; 28:1747-1769. [PMID: 36604605 DOI: 10.1038/s41380-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.
Collapse
|
8
|
Ferretti G, Romano A, Sirabella R, Serafini S, Maier TJ, Matrone C. An increase in Semaphorin 3A biases the axonal direction and induces an aberrant dendritic arborization in an in vitro model of human neural progenitor differentiation. Cell Biosci 2022; 12:182. [DOI: 10.1186/s13578-022-00916-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Semaphorins (Sema) belong to a large family of repellent guidance cues instrumental in guiding axons during development. In particular, Class 3 Sema (Sema 3) is among the best characterized Sema family members and the only produced as secreted proteins in mammals, thereby exerting both autocrine and paracrine functions. Intriguingly, an increasing number of studies supports the crucial role of the Sema 3A in hippocampal and cortical neurodevelopment. This means that alterations in Sema 3A signaling might compromise hippocampal and cortical circuits and predispose to disorders such as autism and schizophrenia. Consistently, increased Sema 3A levels have been detected in brain of patients with schizophrenia and many polymorphisms in Sema 3A or in the Sema 3A receptors, Neuropilins (Npn 1 and 2) and Plexin As (Plxn As), have been associated to autism.
Results
Here we present data indicating that when overexpressed, Sema 3A causes human neural progenitors (NP) axonal retraction and an aberrant dendritic arborization. Similarly, Sema 3A, when overexpressed in human microglia, triggers proinflammatory processes that are highly detrimental to themselves as well as NP. Indeed, NP incubated in microglia overexpressing Sema 3A media retract axons within an hour and then start suffering and finally die. Sema 3A mediated retraction appears to be related to its binding to Npn 1 and Plxn A2 receptors, thus activating the downstream Fyn tyrosine kinase pathway that promotes the threonine-serine kinase cyclin-dependent kinase 5, CDK5, phosphorylation at the Tyr15 residue and the CDK5 processing to generate the active fragment p35.
Conclusions
All together this study identifies Sema 3A as a critical regulator of human NP differentiation. This may imply that an insult due to Sema 3A overexpression during the early phases of neuronal development might compromise neuronal organization and connectivity and make neurons perhaps more vulnerable to other insults across their lifespan.
Collapse
|
9
|
He L, Jiang W, Li J, Wang C. Crystal structure of Ankyrin-G in complex with a fragment of Neurofascin reveals binding mechanisms required for integrity of the axon initial segment. J Biol Chem 2022; 298:102272. [PMID: 35850303 PMCID: PMC9396398 DOI: 10.1016/j.jbc.2022.102272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
The axon initial segment (AIS) has characteristically dense clustering of voltage-gated sodium channels (Nav), cell adhesion molecule Neurofascin 186 (Nfasc), and neuronal scaffold protein Ankyrin-G (AnkG) in neurons, which facilitates generation of an action potential and maintenance of axonal polarity. However, the mechanisms underlying AIS assembly, maintenance, and plasticity remain poorly understood. Here, we report the high-resolution crystal structure of the AnkG ankyrin repeat (ANK repeat) domain in complex with its binding site in the Nfasc cytoplasmic tail that shows, in conjunction with binding affinity assays with serial truncation variants, the molecular basis of AnkG–Nfasc binding. We confirm AnkG interacts with the FIGQY motif in Nfasc, and we identify another region required for their high affinity binding. Our structural analysis revealed that ANK repeats form 4 hydrophobic or hydrophilic layers in the AnkG inner groove that coordinate interactions with essential Nfasc residues, including F1202, E1204, and Y1212. Moreover, we show disruption of the AnkG–Nfasc complex abolishes Nfasc enrichment at the AIS in cultured mouse hippocampal neurons. Finally, our structural and biochemical analysis indicated that L1 syndrome-associated mutations in L1CAM, a member of the L1 immunoglobulin family proteins including Nfasc, L1CAM, NrCAM, and CHL1, compromise binding with ankyrins. Taken together, these results define the mechanisms underlying AnkG–Nfasc complex formation and show that AnkG-dependent clustering of Nfasc is required for AIS integrity.
Collapse
Affiliation(s)
- Liping He
- Department of Neurology, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Wenli Jiang
- Department of Neurology, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Jianchao Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, P. R. China.
| | - Chao Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China.
| |
Collapse
|
10
|
Eichel K, Shen K. The function of the axon initial segment in neuronal polarity. Dev Biol 2022; 489:47-54. [DOI: 10.1016/j.ydbio.2022.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
|
11
|
Fujitani M, Otani Y, Miyajima H. Pathophysiological Roles of Abnormal Axon Initial Segments in Neurodevelopmental Disorders. Cells 2021; 10:2110. [PMID: 34440880 PMCID: PMC8392614 DOI: 10.3390/cells10082110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 11/17/2022] Open
Abstract
The 20-60 μm axon initial segment (AIS) is proximally located at the interface between the axon and cell body. AIS has characteristic molecular and structural properties regulated by the crucial protein, ankyrin-G. The AIS contains a high density of Na+ channels relative to the cell body, which allows low thresholds for the initiation of action potential (AP). Molecular and physiological studies have shown that the AIS is also a key domain for the control of neuronal excitability by homeostatic mechanisms. The AIS has high plasticity in normal developmental processes and pathological activities, such as injury, neurodegeneration, and neurodevelopmental disorders (NDDs). In the first half of this review, we provide an overview of the molecular, structural, and ion-channel characteristics of AIS, AIS regulation through axo-axonic synapses, and axo-glial interactions. In the second half, to understand the relationship between NDDs and AIS, we discuss the activity-dependent plasticity of AIS, the human mutation of AIS regulatory genes, and the pathophysiological role of an abnormal AIS in NDD model animals and patients. We propose that the AIS may provide a potentially valuable structural biomarker in response to abnormal network activity in vivo as well as a new treatment concept at the neural circuit level.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; (Y.O.); (H.M.)
| | | | | |
Collapse
|
12
|
Tamada H, Kiryu-Seo S, Sawada S, Kiyama H. Axonal injury alters the extracellular glial environment of the axon initial segment and allows substantial mitochondrial influx into axon initial segment. J Comp Neurol 2021; 529:3621-3632. [PMID: 34235750 DOI: 10.1002/cne.25212] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022]
Abstract
The axon initial segment (AIS) is structurally and functionally distinct from other regions of the axon, yet alterations in the milieu of the AIS after brain injury have not been well characterized. In this study, we have examined extracellular and intracellular changes in the AIS after hypoglossal nerve injury. Microglial adhesions to the AIS were rarely observed in healthy controls, whereas microglial adhesions to the AIS became apparent in the axonal injury model. Regarding intra-AIS morphology, we focused on mitochondria because mitochondrial flow into the injured axon appears critical for axonal regeneration. To visualize mitochondria specifically in injured axons, we used Atf3:BAC transgenic mice whose mitochondria were labeled with GFP in response to nerve injury. These mice clearly showed mitochondrial localization in the AIS after nerve injury. To precisely confirm the light microscopic observations, we performed three-dimensional ultrastructural analysis using focused ion beam/scanning electron microscopy (FIB/SEM). Although the healthy AIS was not surrounded by microglia, tight microglial adhesions with thick processes adhering to the AIS were observed after injury. FIB/SEM simultaneously allowed the observation of mitochondrial localization in the AIS. In the AIS of non-injured neurons, few mitochondria were observed, whereas mitochondria were abundantly localized in the cell body, axon hillock, and axon. Intriguingly, in the injured AIS, numerous mitochondria were observed throughout the AIS. Taken together, axonal injury changes the extracellular glial environment surrounding the AIS and intracellular mitochondrial localization in the AIS. These changes would be crucial responses, perhaps for injured neurons to regenerate after axonal injury.
Collapse
Affiliation(s)
- Hiromi Tamada
- Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sumiko Kiryu-Seo
- Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sohgo Sawada
- Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kiyama
- Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
Liu W, Luque M, Li H, Schrott-Fischer A, Glueckert R, Tylstedt S, Rajan G, Ladak H, Agrawal S, Rask-Andersen H. Spike Generators and Cell Signaling in the Human Auditory Nerve: An Ultrastructural, Super-Resolution, and Gene Hybridization Study. Front Cell Neurosci 2021; 15:642211. [PMID: 33796009 PMCID: PMC8008129 DOI: 10.3389/fncel.2021.642211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background: The human auditory nerve contains 30,000 nerve fibers (NFs) that relay complex speech information to the brain with spectacular acuity. How speech is coded and influenced by various conditions is not known. It is also uncertain whether human nerve signaling involves exclusive proteins and gene manifestations compared with that of other species. Such information is difficult to determine due to the vulnerable, "esoteric," and encapsulated human ear surrounded by the hardest bone in the body. We collected human inner ear material for nanoscale visualization combining transmission electron microscopy (TEM), super-resolution structured illumination microscopy (SR-SIM), and RNA-scope analysis for the first time. Our aim was to gain information about the molecular instruments in human auditory nerve processing and deviations, and ways to perform electric modeling of prosthetic devices. Material and Methods: Human tissue was collected during trans-cochlear procedures to remove petro-clival meningioma after ethical permission. Cochlear neurons were processed for electron microscopy, confocal microscopy (CM), SR-SIM, and high-sensitive in situ hybridization for labeling single mRNA transcripts to detect ion channel and transporter proteins associated with nerve signal initiation and conductance. Results: Transport proteins and RNA transcripts were localized at the subcellular level. Hemi-nodal proteins were identified beneath the inner hair cells (IHCs). Voltage-gated ion channels (VGICs) were expressed in the spiral ganglion (SG) and axonal initial segments (AISs). Nodes of Ranvier (NR) expressed Nav1.6 proteins, and encoding genes critical for inter-cellular coupling were disclosed. Discussion: Our results suggest that initial spike generators are located beneath the IHCs in humans. The first NRs appear at different places. Additional spike generators and transcellular communication may boost, sharpen, and synchronize afferent signals by cell clusters at different frequency bands. These instruments may be essential for the filtering of complex sounds and may be challenged by various pathological conditions.
Collapse
Affiliation(s)
- Wei Liu
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Maria Luque
- Department of Otorhinolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hao Li
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| | | | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sven Tylstedt
- Department of Olaryngology, Västerviks Hospital, Västervik, Sweden
| | - Gunesh Rajan
- Department of Otolaryngology, Head & Neck Surgery, Luzerner Kantonsspital, Luzern, Switzerland
- Department of Otolaryngology, Head & Neck Surgery, Division of Surgery, Medical School, University of Western Australia, Perth, WA, Australia
| | - Hanif Ladak
- Department of Otolaryngology-Head and Neck Surgery, Department of Medical Biophysics and Department of Electrical and Computer Engineering, Western University, London, ON, Canada
| | - Sumit Agrawal
- Department of Otolaryngology-Head and Neck Surgery, Western University, London, ON, Canada
| | - Helge Rask-Andersen
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
14
|
Deardorff AS, Romer SH, Fyffe RE. Location, location, location: the organization and roles of potassium channels in mammalian motoneurons. J Physiol 2021; 599:1391-1420. [DOI: 10.1113/jp278675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Adam S. Deardorff
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
- Department of Neurology and Internal Medicine, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| | - Shannon H. Romer
- Odyssey Systems Environmental Health Effects Laboratory, Navy Medical Research Unit‐Dayton Wright‐Patterson Air Force Base OH 45433 USA
| | - Robert E.W. Fyffe
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| |
Collapse
|
15
|
Torii T, Ogawa Y, Liu CH, Ho TSY, Hamdan H, Wang CC, Oses-Prieto JA, Burlingame AL, Rasband MN. NuMA1 promotes axon initial segment assembly through inhibition of endocytosis. J Cell Biol 2020; 219:jcb.201907048. [PMID: 31727776 PMCID: PMC7041696 DOI: 10.1083/jcb.201907048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/17/2019] [Accepted: 11/01/2019] [Indexed: 11/22/2022] Open
Abstract
Axon initial segments (AISs) initiate action potentials and regulate the trafficking of vesicles between somatodendritic and axonal compartments. Torii et al. show that NuMA1 is transiently located at the AIS and promotes rapid AIS assembly by inhibiting the endocytosis of neurofascin-186. Axon initial segments (AISs) initiate action potentials and regulate the trafficking of vesicles between somatodendritic and axonal compartments. However, the mechanisms controlling AIS assembly remain poorly defined. We performed differential proteomics and found nuclear mitotic apparatus protein 1 (NuMA1) is downregulated in AIS-deficient neonatal mouse brains and neurons. NuMA1 is transiently located at the AIS during development where it interacts with the scaffolding protein 4.1B and the dynein regulator lissencephaly 1 (Lis1). Silencing NuMA1 or protein 4.1B by shRNA disrupts AIS assembly, but not maintenance. Silencing Lis1 or overexpressing NuMA1 during AIS assembly increased the density of AIS proteins, including ankyrinG and neurofascin-186 (NF186). NuMA1 inhibits the endocytosis of AIS NF186 by impeding Lis1’s interaction with doublecortin, a potent facilitator of NF186 endocytosis. Our results indicate the transient expression and AIS localization of NuMA1 stabilizes the developing AIS by inhibiting endocytosis and removal of AIS proteins.
Collapse
Affiliation(s)
- Tomohiro Torii
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Cheng-Hsin Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Tammy Szu-Yu Ho
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Hamdan Hamdan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Chih-Chuan Wang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
| | | |
Collapse
|
16
|
Ghosh A, Malavasi EL, Sherman DL, Brophy PJ. Neurofascin and Kv7.3 are delivered to somatic and axon terminal surface membranes en route to the axon initial segment. eLife 2020; 9:60619. [PMID: 32903174 PMCID: PMC7511229 DOI: 10.7554/elife.60619] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Ion channel complexes promote action potential initiation at the mammalian axon initial segment (AIS), and modulation of AIS size by recruitment or loss of proteins can influence neuron excitability. Although endocytosis contributes to AIS turnover, how membrane proteins traffic to this proximal axonal domain is incompletely understood. Neurofascin186 (Nfasc186) has an essential role in stabilising the AIS complex to the proximal axon, and the AIS channel protein Kv7.3 regulates neuron excitability. Therefore, we have studied how these proteins reach the AIS. Vesicles transport Nfasc186 to the soma and axon terminal where they fuse with the neuronal plasma membrane. Nfasc186 is highly mobile after insertion in the axonal membrane and diffuses bidirectionally until immobilised at the AIS through its interaction with AnkyrinG. Kv7.3 is similarly recruited to the AIS. This study reveals how key proteins are delivered to the AIS and thereby how they may contribute to its functional plasticity.
Collapse
Affiliation(s)
- Aniket Ghosh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Elise Lv Malavasi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Diane L Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter J Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
17
|
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: 6.0] [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
|
18
|
Abouelezz A, Stefen H, Segerstråle M, Micinski D, Minkeviciene R, Lahti L, Hardeman EC, Gunning PW, Hoogenraad CC, Taira T, Fath T, Hotulainen P. Tropomyosin Tpm3.1 Is Required to Maintain the Structure and Function of the Axon Initial Segment. iScience 2020; 23:101053. [PMID: 32344377 PMCID: PMC7186529 DOI: 10.1016/j.isci.2020.101053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 03/05/2020] [Accepted: 04/06/2020] [Indexed: 12/26/2022] Open
Abstract
The axon initial segment (AIS) is the site of action potential initiation and serves as a cargo transport filter and diffusion barrier that helps maintain neuronal polarity. The AIS actin cytoskeleton comprises actin patches and periodic sub-membranous actin rings. We demonstrate that tropomyosin isoform Tpm3.1 co-localizes with actin patches and that the inhibition of Tpm3.1 led to a reduction in the density of actin patches. Furthermore, Tpm3.1 showed a periodic distribution similar to sub-membranous actin rings but Tpm3.1 was only partially congruent with sub-membranous actin rings. Nevertheless, the inhibition of Tpm3.1 affected the uniformity of the periodicity of actin rings. Furthermore, Tpm3.1 inhibition led to reduced accumulation of AIS structural and functional proteins, disruption in sorting somatodendritic and axonal proteins, and a reduction in firing frequency. These results show that Tpm3.1 is necessary for the structural and functional maintenance of the AIS. Tropomyosin isoform Tpm3.1 co-localizes with the actin cytoskeleton in the AIS Tpm3.1 inhibition led to a less organized AIS actin cytoskeleton Perturbation of Tpm3.1 function reduced the accumulation of AIS scaffolding proteins Tpm3.1 inhibition compromised cargo sorting and rapidly reduced firing frequency
Collapse
Affiliation(s)
- Amr Abouelezz
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland; HiLIFE - Neuroscience Center, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Holly Stefen
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Mikael Segerstråle
- Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 1, 00790 Helsinki, Finland
| | - David Micinski
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Rimante Minkeviciene
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Lauri Lahti
- Department of Computer Science, Aalto University School of Science, Espoo, Finland
| | - Edna C Hardeman
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Peter W Gunning
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584CH Utrecht, the Netherlands
| | - Tomi Taira
- Faculty of Veterinary Medicine, University of Helsinki, Agnes Sjöbergin katu 2, 00790 Helsinki, Finland
| | - Thomas Fath
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; Dementia Research Centre, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
19
|
Goethals S, Brette R. Theoretical relation between axon initial segment geometry and excitability. eLife 2020; 9:53432. [PMID: 32223890 PMCID: PMC7170651 DOI: 10.7554/elife.53432] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/30/2020] [Indexed: 12/29/2022] Open
Abstract
In most vertebrate neurons, action potentials are triggered at the distal end of the axon initial segment (AIS). Both position and length of the AIS vary across and within neuron types, with activity, development and pathology. What is the impact of AIS geometry on excitability? Direct empirical assessment has proven difficult because of the many potential confounding factors. Here, we carried a principled theoretical analysis to answer this question. We provide a simple formula relating AIS geometry and sodium conductance density to the somatic voltage threshold. A distal shift of the AIS normally produces a (modest) increase in excitability, but we explain how this pattern can reverse if a hyperpolarizing current is present at the AIS, due to resistive coupling with the soma. This work provides a theoretical tool to assess the significance of structural AIS plasticity for electrical function.
Collapse
Affiliation(s)
- Sarah Goethals
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Romain Brette
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
20
|
Lorenzo DN. Cargo hold and delivery: Ankyrins, spectrins, and their functional patterning of neurons. Cytoskeleton (Hoboken) 2020; 77:129-148. [PMID: 32034889 DOI: 10.1002/cm.21602] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 01/12/2023]
Abstract
The highly polarized, typically very long, and nonmitotic nature of neurons present them with unique challenges in the maintenance of their homeostasis. This architectural complexity serves a rich and tightly controlled set of functions that enables their fast communication with neighboring cells and endows them with exquisite plasticity. The submembrane neuronal cytoskeleton occupies a pivotal position in orchestrating the structural patterning that determines local and long-range subcellular specialization, membrane dynamics, and a wide range of signaling events. At its center is the partnership between ankyrins and spectrins, which self-assemble with both remarkable long-range regularity and micro- and nanoscale specificity to precisely position and stabilize cell adhesion molecules, membrane transporters, ion channels, and other cytoskeletal proteins. To accomplish these generally conserved, but often functionally divergent and spatially diverse, roles these partners use a combinatorial program of a couple of dozens interacting family members, whose code is not fully unraveled. In a departure from their scaffolding roles, ankyrins and spectrins also enable the delivery of material to the plasma membrane by facilitating intracellular transport. Thus, it is unsurprising that deficits in ankyrins and spectrins underlie several neurodevelopmental, neurodegenerative, and psychiatric disorders. Here, I summarize key aspects of the biology of spectrins and ankyrins in the mammalian neuron and provide a snapshot of the latest advances in decoding their roles in the nervous system.
Collapse
Affiliation(s)
- Damaris N Lorenzo
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
21
|
Hefting LL, D'Este E, Arvedsen E, Benned-Jensen T, Rasmussen HB. Multiple Domains in the Kv7.3 C-Terminus Can Regulate Localization to the Axon Initial Segment. Front Cell Neurosci 2020; 14:10. [PMID: 32116557 PMCID: PMC7010958 DOI: 10.3389/fncel.2020.00010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/14/2020] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated Kv7.2/Kv7.3 potassium channel is a critical regulator of neuronal excitability. It is strategically positioned at the axon initial segment (AIS) of neurons, where it effectively inhibits repetitive action potential firing. While the selective accumulation of Kv7.2/Kv7.3 channels at the AIS requires binding to the adaptor protein ankyrin G, it is currently unknown if additional molecular mechanisms contribute to the localization and fine-tuning of channel numbers at the AIS. Here, we utilized a chimeric approach to pinpoint regions within the Kv7.3 C-terminal tail with an impact upon AIS localization. This strategy identified two domains with opposing effects upon the AIS localization of Kv7.3 chimeras expressed in cultured hippocampal neurons. While a membrane proximal domain reduced AIS localization of Kv7.3 chimeras, helix D increased and stabilized chimera AIS localization. None of the identified domains were required for AIS localization. However, the domains modulated the relative efficiency of the localization raising the possibility that the two domains contribute to the regulation of Kv7 channel numbers and nanoscale organization at the AIS.
Collapse
Affiliation(s)
- Louise Leth Hefting
- Membrane Trafficking Group, Department of Biomedical Sciences, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Emil Arvedsen
- Membrane Trafficking Group, Department of Biomedical Sciences, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tau Benned-Jensen
- Membrane Trafficking Group, Department of Biomedical Sciences, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Borger Rasmussen
- Membrane Trafficking Group, Department of Biomedical Sciences, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Mechanistic insights into the interactions of dynein regulator Ndel1 with neuronal ankyrins and implications in polarity maintenance. Proc Natl Acad Sci U S A 2019; 117:1207-1215. [PMID: 31889000 DOI: 10.1073/pnas.1916987117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ankyrin-G (AnkG), a highly enriched scaffold protein in the axon initial segment (AIS) of neurons, functions to maintain axonal polarity and the integrity of the AIS. At the AIS, AnkG regulates selective intracellular cargo trafficking between soma and axons via interaction with the dynein regulator protein Ndel1, but the molecular mechanism underlying this binding remains elusive. Here we report that Ndel1's C-terminal coiled-coil region (CT-CC) binds to giant neuron-specific insertion regions present in both AnkG and AnkB with 2:1 stoichiometry. The high-resolution crystal structure of AnkB in complex with Ndel1 CT-CC revealed the detailed molecular basis governing the AnkB/Ndel1 complex formation. Mechanistically, AnkB binds with Ndel1 by forming a stable 5-helix bundle dominated by hydrophobic interactions spread across 6 distinct interaction layers. Moreover, we found that AnkG is essential for Ndel1 accumulation at the AIS. Finally, we found that cargo sorting at the AIS can be disrupted by blocking the AnkG/Ndel1 complex formation using a peptide designed based on our structural data. Collectively, the atomic structure of the AnkB/Ndel1 complex together with studies of cargo sorting through the AIS establish the mechanistic basis for AnkG/Ndel1 complex formation and for the maintenance of axonal polarity. Our study will also be valuable for future studies of the interaction between AnkB and Ndel1 perhaps at distal axonal cargo transport.
Collapse
|
23
|
Abstract
Neuronal activity can be modulated by mechanical stimuli. To study this phenomenon quantitatively, we mechanically stimulated rat cortical neurons by shear stress and local indentation. Neurons show 2 distinct responses, classified as transient and sustained. Transient responses display fast kinetics, similar to spontaneous neuronal activity, whereas sustained responses last several minutes before returning to baseline. Local soma stimulations with micrometer-sized beads evoke transient responses at low forces of ∼220 nN and pressures of ∼5.6 kPa and sustained responses at higher forces of ∼360 nN and pressures of ∼9.2 kPa. Among the neuronal compartments, axons are highly susceptible to mechanical stimulation and predominantly show sustained responses, whereas the less susceptible dendrites predominantly respond transiently. Chemical perturbation experiments suggest that mechanically evoked responses require the influx of extracellular calcium through ion channels. We propose that subtraumatic forces/pressures applied to neurons evoke neuronal responses via nonspecific gating of ion channels.
Collapse
|
24
|
Beets I, Zhang G, Fenk LA, Chen C, Nelson GM, Félix MA, de Bono M. Natural Variation in a Dendritic Scaffold Protein Remodels Experience-Dependent Plasticity by Altering Neuropeptide Expression. Neuron 2019; 105:106-121.e10. [PMID: 31757604 PMCID: PMC6953435 DOI: 10.1016/j.neuron.2019.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 08/18/2019] [Accepted: 09/28/2019] [Indexed: 12/13/2022]
Abstract
The extent to which behavior is shaped by experience varies between individuals. Genetic differences contribute to this variation, but the neural mechanisms are not understood. Here, we dissect natural variation in the behavioral flexibility of two Caenorhabditis elegans wild strains. In one strain, a memory of exposure to 21% O2 suppresses CO2-evoked locomotory arousal; in the other, CO2 evokes arousal regardless of previous O2 experience. We map that variation to a polymorphic dendritic scaffold protein, ARCP-1, expressed in sensory neurons. ARCP-1 binds the Ca2+-dependent phosphodiesterase PDE-1 and co-localizes PDE-1 with molecular sensors for CO2 at dendritic ends. Reducing ARCP-1 or PDE-1 activity promotes CO2 escape by altering neuropeptide expression in the BAG CO2 sensors. Variation in ARCP-1 alters behavioral plasticity in multiple paradigms. Our findings are reminiscent of genetic accommodation, an evolutionary process by which phenotypic flexibility in response to environmental variation is reset by genetic change. Behavioral flexibility varies across Caenorhabditis and C. elegans wild isolates A natural polymorphism in ARCP-1 underpins inter-individual variation in plasticity ARCP-1 is a dendritic scaffold protein localizing cGMP signaling machinery to cilia Disrupting ARCP-1 alters behavioral plasticity by changing neuropeptide expression
Collapse
Affiliation(s)
- Isabel Beets
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Gaotian Zhang
- Institut de Biologie de l'École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris 75005, France
| | - Lorenz A Fenk
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Changchun Chen
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Geoffrey M Nelson
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris 75005, France.
| | - Mario de Bono
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
25
|
Khalaf B, Roncador A, Pischedda F, Casini A, Thomas S, Piccoli G, Kiebler M, Macchi P. Ankyrin-G induces nucleoporin Nup358 to associate with the axon initial segment of neurons. J Cell Sci 2019; 132:jcs.222802. [PMID: 31427429 DOI: 10.1242/jcs.222802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Nup358 (also known as RanBP2) is a member of the large nucleoporin family that constitutes the nuclear pore complex. Depending on the cell type and the physiological state, Nup358 interacts with specific partner proteins and influences distinct mechanisms independent of its role in nucleocytoplasmic transport. Here, we provide evidence that Nup358 associates selectively with the axon initial segment (AIS) of mature neurons, mediated by the AIS scaffold protein ankyrin-G (AnkG, also known as Ank3). The N-terminus of Nup358 is demonstrated to be sufficient for its localization at the AIS. Further, we show that Nup358 is expressed as two isoforms, one full-length and another shorter form of Nup358. These isoforms differ in their subcellular distribution in neurons and expression level during neuronal development. Overall, the present study highlights an unprecedented localization of Nup358 within the AIS and suggests its involvement in neuronal function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Bouchra Khalaf
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Alessandro Roncador
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Francesca Pischedda
- Dulbecco Telethon Laboratory of Biology of Synapses, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Antonio Casini
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Sabine Thomas
- Department for Cell Biology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University of Munich, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Giovanni Piccoli
- Dulbecco Telethon Laboratory of Biology of Synapses, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Michael Kiebler
- Department for Cell Biology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University of Munich, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Paolo Macchi
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| |
Collapse
|
26
|
Kvarnung M, Shahsavani M, Taylan F, Moslem M, Breeuwsma N, Laan L, Schuster J, Jin Z, Nilsson D, Lieden A, Anderlid BM, Nordenskjöld M, Syk Lundberg E, Birnir B, Dahl N, Nordgren A, Lindstrand A, Falk A. Ataxia in Patients With Bi-Allelic NFASC Mutations and Absence of Full-Length NF186. Front Genet 2019; 10:896. [PMID: 31608123 PMCID: PMC6769111 DOI: 10.3389/fgene.2019.00896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/23/2019] [Indexed: 01/29/2023] Open
Abstract
The etiology of hereditary ataxia syndromes is heterogeneous, and the mechanisms underlying these disorders are often unknown. Here, we utilized exome sequencing in two siblings with progressive ataxia and muscular weakness and identified a novel homozygous splice mutation (c.3020-1G > A) in neurofascin (NFASC). In RNA extracted from fibroblasts, we showed that the mutation resulted in inframe skipping of exon 26, with a deprived expression of the full-length transcript that corresponds to NFASC isoform NF186. To further investigate the disease mechanisms, we reprogrammed fibroblasts from one affected sibling to induced pluripotent stem cells, directed them to neuroepithelial stem cells and finally differentiated to neurons. In early neurogenesis, differentiating cells with selective depletion of the NF186 isoform showed significantly reduced neurite outgrowth as well as fewer emerging neurites. Furthermore, whole-cell patch-clamp recordings of patient-derived neuronal cells revealed a lower threshold for openings, indicating altered Na+ channel kinetics, suggesting a lower threshold for openings as compared to neuronal cells without the NFASC mutation. Taken together, our results suggest that loss of the full-length NFASC isoform NF186 causes perturbed neurogenesis and impaired neuronal biophysical properties resulting in a novel early-onset autosomal recessive ataxia syndrome.
Collapse
Affiliation(s)
- Malin Kvarnung
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mansoureh Shahsavani
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Fulya Taylan
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mohsen Moslem
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Nicole Breeuwsma
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Loora Laan
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Jens Schuster
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Zhe Jin
- Department of Neuroscience, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Daniel Nilsson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Agne Lieden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Nordenskjöld
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisabeth Syk Lundberg
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bryndis Birnir
- Department of Neuroscience, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Niklas Dahl
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Biomedical Centre, Uppsala University, Uppsala, Sweden
| | - Ann Nordgren
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
27
|
Fréal A, Rai D, Tas RP, Pan X, Katrukha EA, van de Willige D, Stucchi R, Aher A, Yang C, Altelaar AFM, Vocking K, Post JA, Harterink M, Kapitein LC, Akhmanova A, Hoogenraad CC. Feedback-Driven Assembly of the Axon Initial Segment. Neuron 2019; 104:305-321.e8. [PMID: 31474508 PMCID: PMC6839619 DOI: 10.1016/j.neuron.2019.07.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/14/2019] [Accepted: 07/22/2019] [Indexed: 11/01/2022]
Abstract
The axon initial segment (AIS) is a unique neuronal compartment that plays a crucial role in the generation of action potential and neuronal polarity. The assembly of the AIS requires membrane, scaffolding, and cytoskeletal proteins, including Ankyrin-G and TRIM46. How these components cooperate in AIS formation is currently poorly understood. Here, we show that Ankyrin-G acts as a scaffold interacting with End-Binding (EB) proteins and membrane proteins such as Neurofascin-186 to recruit TRIM46-positive microtubules to the plasma membrane. Using in vitro reconstitution and cellular assays, we demonstrate that TRIM46 forms parallel microtubule bundles and stabilizes them by acting as a rescue factor. TRIM46-labeled microtubules drive retrograde transport of Neurofascin-186 to the proximal axon, where Ankyrin-G prevents its endocytosis, resulting in stable accumulation of Neurofascin-186 at the AIS. Neurofascin-186 enrichment in turn reinforces membrane anchoring of Ankyrin-G and subsequent recruitment of TRIM46-decorated microtubules. Our study reveals feedback-based mechanisms driving AIS assembly.
Collapse
Affiliation(s)
- Amélie Fréal
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands; Department of Axonal Signaling, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Dipti Rai
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Roderick P Tas
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Xingxiu Pan
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Dieudonnée van de Willige
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Riccardo Stucchi
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences and the Netherlands Proteomics Center, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Amol Aher
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Chao Yang
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - A F Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences and the Netherlands Proteomics Center, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Karin Vocking
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Jan Andries Post
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Martin Harterink
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands.
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands; Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
28
|
Le Large TYS, El Hassouni B, Funel N, Kok B, Piersma SR, Pham TV, Olive KP, Kazemier G, van Laarhoven HWM, Jimenez CR, Bijlsma MF, Giovannetti E. Proteomic analysis of gemcitabine-resistant pancreatic cancer cells reveals that microtubule-associated protein 2 upregulation associates with taxane treatment. Ther Adv Med Oncol 2019; 11:1758835919841233. [PMID: 31205498 PMCID: PMC6535709 DOI: 10.1177/1758835919841233] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 01/23/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Chemoresistance hampers the treatment of patients suffering from pancreatic ductal adenocarcinoma (PDAC). Here we aimed to evaluate the (phospho)proteome of gemcitabine-sensitive and gemcitabine-resistant PDAC cells to identify novel therapeutic targets and predictive biomarkers. Methods: The oncogenic capabilities of gemcitabine-sensitive and resistant PDAC cells were evaluated in vitro and in vivo. Cultured cells were analyzed by label-free proteomics. Differential proteins and phosphopeptides were evaluated by gene ontology and for their predictive or prognostic biomarker potential with immunohistochemistry of tissue microarrays. Results: Gemcitabine-resistant cells had increased potential to induce xenograft tumours (p value < 0.001). Differential analyses showed that proteins associated with gemcitabine resistance are correlated with microtubule regulation. Indeed, gemcitabine-resistant cells displayed an increased sensitivity for paclitaxel in vitro (p < 0.001) and nab-paclitaxel had a strong anti-tumour efficacy in vivo. Microtubule-associated protein 2 (MAP2) was found to be highly upregulated (p = 0.002, fold change = 10) and phosphorylated in these resistant cells. Expression of MAP2 was correlated with a poorer overall survival in patients treated with gemcitabine in the palliative (p = 0.037) and adjuvant setting (p = 0.014). Conclusions: These data show an explanation as to why the combination of gemcitabine with nab-paclitaxel is effective in PDAC patients. The identified gemcitabine-resistance marker, MAP2, emerged as a novel prognostic marker in PDAC patients treated with gemcitabine and warrants further clinical investigation.
Collapse
Affiliation(s)
- Tessa Ya Sung Le Large
- Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, NetherlandsLEXOR, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, NetherlandsCancer Pharmacology Lab, AIRC-Start-Up, University Hospital of Pisa, Pisa, Italy
| | - Btissame El Hassouni
- Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC-Start-Up, University Hospital of Pisa, Pisa, Italy
| | - Bart Kok
- Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sander R Piersma
- Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Thang V Pham
- Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Kenneth P Olive
- Departments of Medicine and Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York City, NY, USA
| | - Geert Kazemier
- Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Hanneke W M van Laarhoven
- Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Connie R Jimenez
- Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maarten F Bijlsma
- LEXOR, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Elisa Giovannetti
- Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| |
Collapse
|
29
|
Fletcher-Jones A, Hildick KL, Evans AJ, Nakamura Y, Wilkinson KA, Henley JM. The C-terminal helix 9 motif in rat cannabinoid receptor type 1 regulates axonal trafficking and surface expression. eLife 2019; 8:44252. [PMID: 31036155 PMCID: PMC6491034 DOI: 10.7554/elife.44252] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Cannabinoid type one receptor (CB1R) is only stably surface expressed in axons, where it downregulates neurotransmitter release. How this tightly regulated axonal surface polarity is established and maintained is unclear. To address this question, we used time-resolved imaging to determine the trafficking of CB1R from biosynthesis to mature polarised localisation in cultured rat hippocampal neurons. We show that the secretory pathway delivery of CB1R is axonally biased and that surface expressed CB1R is more stable in axons than in dendrites. This dual mechanism is mediated by the CB1R C-terminus and involves the Helix 9 (H9) domain. Removal of the H9 domain increases secretory pathway delivery to dendrites and decreases surface stability. Furthermore, CB1RΔH9 is more sensitive to agonist-induced internalisation and less efficient at downstream signalling than CB1RWT. Together, these results shed new light on how polarity of CB1R is mediated and indicate that the C-terminal H9 domain plays key roles in this process. The brain contains around 100 billion neurons that are in constant communication with one another. Each consists of a cell body, plus two components specialized for exchanging information. These are the axon, which delivers information, and the dendrites, which receive it. This exchange takes place at contact points between neurons called synapses. To send a message, a neuron releases chemicals called neurotransmitters from its axon terminals into the synapse. The neurotransmitters cross the synapse and bind to receptor proteins on the dendrites of another neuron. In doing so, they pass on the message. Cannabinoid type 1 receptors (CB1Rs) help control the flow of information at synapses. They do this by binding neurotransmitters called endocannabinoids, which are unusual among neurotransmitters. Rather than sending messages from axons to dendrites, endocannabinoids send them in the opposite direction. Thus, it is dendrites that release endocannabinoids, which then bind to CB1Rs in axon terminals. This backwards, or 'retrograde', signalling dampens the release of other neurotransmitters. This slows down brain activity, and gives rise to the 'mellow' sensation that recreational cannabis users often describe. Like most other proteins, CB1Rs are built inside the cell body. So, how do these receptors end up in the axon terminals where they are needed? Are they initially sent to both axons and dendrites, with the CB1Rs that travel to dendrites being rerouted back to axons? Or do the receptors travel directly to the axon itself? Fletcher-Jones et al. tracked newly made CB1Rs in rat neurons growing in a dish. The results revealed that the receptors go directly to the axon, before moving on to the axon terminals. A specific region of the CB1R protein is crucial for sending the receptors to the axon, and for ensuring that they do not get diverted to the dendrite surface. This region stabilizes CB1Rs at the axon surface, and helps to make the receptors available to bind endocannabinoids. CB1Rs also respond to medical marijuana, a topic that continues to generate interest as well as controversy. Activating CB1Rs could help treat a wide range of diseases, such as chronic pain, epilepsy and multiple sclerosis. Future studies should build on our understanding of CB1Rs to explore and optimize new therapeutic approaches.
Collapse
Affiliation(s)
- Alexandra Fletcher-Jones
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Keri L Hildick
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Ashley J Evans
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Yasuko Nakamura
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
30
|
Alpizar SA, Cho IH, Hoppa MB. Subcellular control of membrane excitability in the axon. Curr Opin Neurobiol 2019; 57:117-125. [PMID: 30784979 DOI: 10.1016/j.conb.2019.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Abstract
Ion channels are microscopic pore proteins in the membrane that open and close in response to chemical and electrical stimuli. This simple concept underlies rapid electrical signaling in the brain as well as several important aspects of neural plasticity. Although the soma accounts for less than 1% of many neurons by membrane area, it has been the major site of measuring ion channel function. However, the axon is one of the longest processes found in cellular biology and hosts a multitude of critical signaling functions in the brain. Not only does the axon initiate and rapidly propagate action potentials (APs) across the brain but it also forms the presynaptic terminals that convert these electrical inputs into chemical outputs. Here, we review recent advances in the physiological role of ion channels within the diverse landscape of the axon and presynaptic terminals.
Collapse
Affiliation(s)
- Scott A Alpizar
- Dartmouth College, Department of Biological Sciences, Hanover, NH, United States
| | - In Ha Cho
- Dartmouth College, Department of Biological Sciences, Hanover, NH, United States
| | - Michael B Hoppa
- Dartmouth College, Department of Biological Sciences, Hanover, NH, United States.
| |
Collapse
|
31
|
Alpizar SA, Baker AL, Gulledge AT, Hoppa MB. Loss of Neurofascin-186 Disrupts Alignment of AnkyrinG Relative to Its Binding Partners in the Axon Initial Segment. Front Cell Neurosci 2019; 13:1. [PMID: 30723396 PMCID: PMC6349729 DOI: 10.3389/fncel.2019.00001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/07/2019] [Indexed: 12/14/2022] Open
Abstract
The axon initial segment (AIS) is a specialized region within the proximal portion of the axon that initiates action potentials thanks in large part to an enrichment of sodium channels. The scaffolding protein ankyrinG (AnkG) is essential for the recruitment of sodium channels as well as several other intracellular and extracellular proteins to the AIS. In the present study, we explore the role of the cell adhesion molecule (CAM) neurofascin-186 (NF-186) in arranging the individual molecular components of the AIS in cultured rat hippocampal neurons. Using a CRISPR depletion strategy to ablate NF expression, we found that the loss of NF selectively perturbed AnkG accumulation and its relative proximal distribution within the AIS. We found that the overexpression of sodium channels could restore AnkG accumulation, but not its altered distribution within the AIS without NF present. We go on to show that although the loss of NF altered AnkG distribution, sodium channel function within the AIS remained normal. Taken together, these results demonstrate that the regulation of AnkG and sodium channel accumulation within the AIS can occur independently of one another, potentially mediated by other binding partners such as NF.
Collapse
Affiliation(s)
- Scott A Alpizar
- Department of Biological Sciences, Dartmouth College, Hanover, NH, United States
| | - Arielle L Baker
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States
| | - Allan T Gulledge
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States
| | - Michael B Hoppa
- Department of Biological Sciences, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
32
|
Maturation Dynamics of the Axon Initial Segment (AIS) of Newborn Dentate Granule Cells in Young Adult C57BL/6J Mice. J Neurosci 2019; 39:1605-1620. [PMID: 30651327 DOI: 10.1523/jneurosci.2253-18.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
Newborn dentate granule cells (DGCs) are generated in the hippocampal dentate gyrus (DG) of rodents through a process called adult hippocampal neurogenesis, which is subjected to tight intrinsic and extrinsic regulation. The use of retroviruses encoding fluorescent proteins has allowed the characterization of the maturation dynamics of newborn DGCs, including their morphological development and the establishment and maturation of their afferent and efferent synaptic connections. However, the study of a crucial cellular compartment of these cells, namely, the axon initial segment (AIS), has remained unexplored to date. The AIS is not only the site of action potential initiation, but it also has a unique molecular identity that makes it one of the master regulators of neural plasticity and excitability. Here we examined the dynamics of AIS formation in newborn DGCs of young female adult C57BL/6J mice in vivo Our data reveal notable changes in AIS length and thickness throughout cell maturation under physiological conditions and show that the most remarkable structural changes coincide with periods of intense morphological and functional remodeling. Moreover, we demonstrate that AIS development can be modulated extrinsically by both neuroprotective (environmental enrichment) and detrimental (lipopolysaccharide from Escherichia coli) stimuli.SIGNIFICANCE STATEMENT The hippocampal dentate gyrus (DG) of rodents generates newborn dentate granule cells (DGCs) throughout life. This process, named adult hippocampal neurogenesis, confers a unique degree of plasticity to the hippocampal circuit, and it is crucial for learning and memory. Here we studied, for the first time, the formation of a key cellular compartment of newborn DGCs, namely, the axon initial segment (AIS) in vivo Our data reveal remarkable AIS structural remodeling throughout the maturation of these cells under physiological conditions. Moreover, AIS development can be modulated extrinsically by both neuroprotective (environmental enrichment) and detrimental (lipopolysaccharide from Escherichia coli) stimuli.
Collapse
|
33
|
Disruption of the psychiatric risk gene Ankyrin 3 enhances microtubule dynamics through GSK3/CRMP2 signaling. Transl Psychiatry 2018; 8:135. [PMID: 30046097 PMCID: PMC6060177 DOI: 10.1038/s41398-018-0182-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/11/2018] [Indexed: 12/23/2022] Open
Abstract
The ankyrin 3 gene (ANK3) is a well-established risk gene for psychiatric illness, but the mechanisms underlying its pathophysiology remain elusive. We examined the molecular effects of disrupting brain-specific Ank3 isoforms in mouse and neuronal model systems. RNA sequencing of hippocampus from Ank3+/- and Ank3+/+ mice identified altered expression of 282 genes that were enriched for microtubule-related functions. Results were supported by increased expression of microtubule end-binding protein 3 (EB3), an indicator of microtubule dynamics, in Ank3+/- mouse hippocampus. Live-cell imaging of EB3 movement in primary neurons from Ank3+/- mice revealed impaired elongation of microtubules. Using a CRISPR-dCas9-KRAB transcriptional repressor in mouse neuro-2a cells, we determined that repression of brain-specific Ank3 increased EB3 expression, decreased tubulin acetylation, and increased the soluble:polymerized tubulin ratio, indicating enhanced microtubule dynamics. These changes were rescued by inhibition of glycogen synthase kinase 3 (GSK3) with lithium or CHIR99021, a highly selective GSK3 inhibitor. Brain-specific Ank3 repression in neuro-2a cells increased GSK3 activity (reduced inhibitory phosphorylation) and elevated collapsin response mediator protein 2 (CRMP2) phosphorylation, a known GSK3 substrate and microtubule-binding protein. Pharmacological inhibition of CRMP2 activity attenuated the rescue of EB3 expression and tubulin polymerization in Ank3-repressed cells by lithium or CHIR99021, suggesting microtubule instability induced by Ank3 repression is dependent on CRMP2 activity. Taken together, our data indicate that ANK3 functions in neuronal microtubule dynamics through GSK3 and its downstream substrate CRMP2. These findings reveal cellular and molecular mechanisms underlying brain-specific ANK3 disruption that may be related to its role in psychiatric illness.
Collapse
|
34
|
Huang CYM, Rasband MN. Axon initial segments: structure, function, and disease. Ann N Y Acad Sci 2018; 1420:46-61. [PMID: 29749636 DOI: 10.1111/nyas.13718] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 11/28/2022]
Abstract
The axon initial segment (AIS) is located at the proximal axon and is the site of action potential initiation. This reflects the high density of ion channels found at the AIS. Adaptive changes to the location and length of the AIS can fine-tune the excitability of neurons and modulate plasticity in response to activity. The AIS plays an important role in maintaining neuronal polarity by regulating the trafficking and distribution of proteins that function in somatodendritic or axonal compartments of the neuron. In this review, we provide an overview of the AIS cytoarchitecture, mechanism of assembly, and recent studies revealing mechanisms of differential transport at the AIS that maintain axon and dendrite identities. We further discuss how genetic mutations in AIS components (i.e., ankyrins, ion channels, and spectrins) and injuries may cause neurological disorders.
Collapse
Affiliation(s)
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
35
|
Berger SL, Leo-Macias A, Yuen S, Khatri L, Pfennig S, Zhang Y, Agullo-Pascual E, Caillol G, Zhu MS, Rothenberg E, Melendez-Vasquez CV, Delmar M, Leterrier C, Salzer JL. Localized Myosin II Activity Regulates Assembly and Plasticity of the Axon Initial Segment. Neuron 2018; 97:555-570.e6. [PMID: 29395909 PMCID: PMC5805619 DOI: 10.1016/j.neuron.2017.12.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/24/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023]
Abstract
The axon initial segment (AIS) is the site of action potential generation and a locus of activity-dependent homeostatic plasticity. A multimeric complex of sodium channels, linked via a cytoskeletal scaffold of ankyrin G and beta IV spectrin to submembranous actin rings, mediates these functions. The mechanisms that specify the AIS complex to the proximal axon and underlie its plasticity remain poorly understood. Here we show phosphorylated myosin light chain (pMLC), an activator of contractile myosin II, is highly enriched in the assembling and mature AIS, where it associates with actin rings. MLC phosphorylation and myosin II contractile activity are required for AIS assembly, and they regulate the distribution of AIS components along the axon. pMLC is rapidly lost during depolarization, destabilizing actin and thereby providing a mechanism for activity-dependent structural plasticity of the AIS. Together, these results identify pMLC/myosin II activity as a common link between AIS assembly and plasticity.
Collapse
Affiliation(s)
- Stephen L Berger
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Stephanie Yuen
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Latika Khatri
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Sylvia Pfennig
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Yanqing Zhang
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Ghislaine Caillol
- Aix Marseille Université, CNRS, INP UMR7051, 13344 Cedex 15, Marseille, France
| | - Min-Sheng Zhu
- Model Animal Research Center and MOE Key Laboratory of Model Animal and Disease Study, Nanjing University, Nanjing 210061, China
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Carmen V Melendez-Vasquez
- Department of Biological Sciences, Hunter College, New York, NY 10065, USA; Department of Molecular, Cellular, and Developmental Biology, The Graduate Center, The City University of New York, NY 10016, USA
| | - Mario Delmar
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - James L Salzer
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
36
|
The Axon Initial Segment: An Updated Viewpoint. J Neurosci 2018; 38:2135-2145. [PMID: 29378864 DOI: 10.1523/jneurosci.1922-17.2018] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/20/2018] [Accepted: 01/25/2018] [Indexed: 12/20/2022] Open
Abstract
At the base of axons sits a unique compartment called the axon initial segment (AIS). The AIS generates and shapes the action potential before it is propagated along the axon. Neuronal excitability thus depends crucially on the AIS composition and position, and these adapt to developmental and physiological conditions. The AIS also demarcates the boundary between the somatodendritic and axonal compartments. Recent studies have brought insights into the molecular architecture of the AIS and how it regulates protein trafficking. This Viewpoints article summarizes current knowledge about the AIS and highlights future challenges in understanding this key actor of neuronal physiology.
Collapse
|
37
|
Jamann N, Jordan M, Engelhardt M. Activity-dependent axonal plasticity in sensory systems. Neuroscience 2017; 368:268-282. [PMID: 28739523 DOI: 10.1016/j.neuroscience.2017.07.035] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/23/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022]
Abstract
The rodent whisker-to-barrel cortex pathway is a classic model to study the effects of sensory experience and deprivation on neuronal circuit formation, not only during development but also in the adult. Decades of research have produced a vast body of evidence highlighting the fundamental role of neuronal activity (spontaneous and/or sensory-evoked) for circuit formation and function. In this context, it has become clear that neuronal adaptation and plasticity is not just a function of the neonatal brain, but persists into adulthood, especially after experience-driven modulation of network status. Mechanisms for structural remodeling of the somatodendritic or axonal domain include microscale alterations of neurites or synapses. At the same time, functional alterations at the nanoscale such as expression or activation changes of channels and receptors contribute to the modulation of intrinsic excitability or input-output relationships. However, it remains elusive how these forms of structural and functional plasticity come together to shape neuronal network formation and function. While specifically somatodendritic plasticity has been studied in great detail, the role of axonal plasticity, (e.g. at presynaptic boutons, branches or axonal microdomains), is rather poorly understood. Therefore, this review will only briefly highlight somatodendritic plasticity and instead focus on axonal plasticity. We discuss (i) the role of spontaneous and sensory-evoked plasticity during critical periods, (ii) the assembly of axonal presynaptic sites, (iii) axonal plasticity in the mature brain under baseline and sensory manipulation conditions, and finally (iv) plasticity of electrogenic axonal microdomains, namely the axon initial segment, during development and in the mature CNS.
Collapse
Affiliation(s)
- Nora Jamann
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany
| | - Merryn Jordan
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Medical Faculty Mannheim, CBTM, Heidelberg University, Germany.
| |
Collapse
|
38
|
Nelson AD, Jenkins PM. Axonal Membranes and Their Domains: Assembly and Function of the Axon Initial Segment and Node of Ranvier. Front Cell Neurosci 2017; 11:136. [PMID: 28536506 PMCID: PMC5422562 DOI: 10.3389/fncel.2017.00136] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022] Open
Abstract
Neurons are highly specialized cells of the nervous system that receive, process and transmit electrical signals critical for normal brain function. Here, we review the intricate organization of axonal membrane domains that facilitate rapid action potential conduction underlying communication between complex neuronal circuits. Two critical excitable domains of vertebrate axons are the axon initial segment (AIS) and the nodes of Ranvier, which are characterized by the high concentrations of voltage-gated ion channels, cell adhesion molecules and specialized cytoskeletal networks. The AIS is located at the proximal region of the axon and serves as the site of action potential initiation, while nodes of Ranvier, gaps between adjacent myelin sheaths, allow rapid propagation of the action potential through saltatory conduction. The AIS and nodes of Ranvier are assembled by ankyrins, spectrins and their associated binding partners through the clustering of membrane proteins and connection to the underlying cytoskeleton network. Although the AIS and nodes of Ranvier share similar protein composition, their mechanisms of assembly are strikingly different. Here we will cover the mechanisms of formation and maintenance of these axonal excitable membrane domains, specifically highlighting the similarities and differences between them. We will also discuss recent advances in super resolution fluorescence imaging which have elucidated the arrangement of the submembranous axonal cytoskeleton revealing a surprising structural organization necessary to maintain axonal organization and function. Finally, human mutations in axonal domain components have been associated with a growing number of neurological disorders including severe cognitive dysfunction, epilepsy, autism, neurodegenerative diseases and psychiatric disorders. Overall, this review highlights the assembly, maintenance and function of axonal excitable domains, particularly the AIS and nodes of Ranvier, and how abnormalities in these processes may contribute to disease.
Collapse
Affiliation(s)
- Andrew D Nelson
- Department of Pharmacology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Paul M Jenkins
- Department of Pharmacology, University of Michigan Medical SchoolAnn Arbor, MI, USA.,Department of Psychiatry, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|