1
|
Kaushik K, Chapman G, Prakasam R, Batool F, Saleh M, Determan J, Huettner JE, Kroll KL. Requirements for the neurodevelopmental disorder-associated gene ZNF292 in human cortical interneuron development and function. Cell Rep 2025; 44:115597. [PMID: 40257863 DOI: 10.1016/j.celrep.2025.115597] [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: 06/14/2024] [Revised: 12/27/2024] [Accepted: 03/31/2025] [Indexed: 04/23/2025] Open
Abstract
Pathogenic mutation of the zinc-finger transcription factor ZNF292 is a recently defined contributor to human neurodevelopmental disorders (NDDs). However, the gene's roles in cortical development and regulatory networks under its control were previously undefined. Here, human stem cell models of ZNF292 deficiency, resembling pathogenic haploinsufficiency, are used to derive cortical inhibitory neuron progenitors and neurons. ZNF292-deficient progenitors undergo precocious differentiation but subsequently exhibit compromised interneuron maturation and function. In progenitors, genome-wide occupancy and transcriptomic analyses identify direct target genes controlling neuronal differentiation and synapse formation that are upregulated upon ZNF292 deficiency. By contrast, deficiency in interneurons compromises ZNF292 genome-wide association with and causes downregulation of direct target genes promoting interneuron maturation and function, including other NDD genes. ZNF292-deficient interneurons also exhibit altered channel activities, elevated GABA responsiveness, and hallmarks of neuronal hyperactivity. Together, the results of this work define neurodevelopmental requirements for ZNF292, some of which may contribute to pathogenic ZNF292 mutation-related NDDs.
Collapse
Affiliation(s)
- Komal Kaushik
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gareth Chapman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramachandran Prakasam
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Faiza Batool
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maamoon Saleh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Julianna Determan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James E Huettner
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kristen L Kroll
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Jenkins PM, Bender KJ. Axon initial segment structure and function in health and disease. Physiol Rev 2025; 105:765-801. [PMID: 39480263 DOI: 10.1152/physrev.00030.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
At the simplest level, neurons are structured to integrate synaptic input and perform computational transforms on that input, converting it into an action potential (AP) code. This process, converting synaptic input into AP output, typically occurs in a specialized region of the axon termed the axon initial segment (AIS). The AIS, as its name implies, is often contained to the first section of axon abutted to the soma and is home to a dizzying array of ion channels, attendant scaffolding proteins, intracellular organelles, extracellular proteins, and, in some cases, synapses. The AIS serves multiple roles as the final arbiter for determining if inputs are sufficient to evoke APs, as a gatekeeper that physically separates the somatodendritic domain from the axon proper, and as a regulator of overall neuronal excitability, dynamically tuning its size to best suit the needs of parent neurons. These complex roles have received considerable attention from experimentalists and theoreticians alike. Here, we review recent advances in our understanding of the AIS and its role in neuronal integration and polarity in health and disease.
Collapse
Affiliation(s)
- Paul M Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Kevin J Bender
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, United States
| |
Collapse
|
3
|
Fréal A, Hoogenraad CC. The dynamic axon initial segment: From neuronal polarity to network homeostasis. Neuron 2025; 113:649-669. [PMID: 39947181 DOI: 10.1016/j.neuron.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/15/2024] [Accepted: 01/07/2025] [Indexed: 03/08/2025]
Abstract
The axon initial segment (AIS) is a highly specialized compartment in neurons that resides in between axonal and somatodendritic domains. The localization of the AIS in the proximal part of the axon is essential for its two major functions: generating and modulating action potentials and maintaining neuron polarity. Recent findings revealed that the incredibly stable AIS is generated from highly dynamic components and can undergo extensive structural and functional changes in response to alterations in activity levels. These activity-dependent alterations of AIS structure and function have profound consequences for neuronal functioning, and AIS plasticity has emerged as a key regulator of network homeostasis. This review highlights the functions of the AIS, its architecture, and how its organization and remodeling are influenced by developmental plasticity and both acute and chronic adaptations. It also discusses the mechanisms underlying these processes and explores how dysregulated AIS plasticity may contribute to brain disorders.
Collapse
Affiliation(s)
- Amélie Fréal
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam, the Netherlands
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA.
| |
Collapse
|
4
|
Djukic S, Zhao Z, Jørgensen LMH, Bak AN, Jensen DB, Meehan CF. TDP-43 pathology is sufficient to drive axon initial segment plasticity and hyperexcitability of spinal motoneurones in vivo in the TDP43-ΔNLS model of Amyotrophic Lateral Sclerosis. Acta Neuropathol Commun 2025; 13:42. [PMID: 39994742 PMCID: PMC11849383 DOI: 10.1186/s40478-025-01934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
A hyperexcitability of the motor system is consistently observed in Amyotrophic Lateral Sclerosis (ALS) and has been implicated in the disease pathogenesis. What drives this hyperexcitability in the vast majority of patients is unknown. This is important to know as existing treatments simply reduce all neuronal excitability and fail to distinguish between pathological changes and important homeostatic changes. Understanding what drives the initial pathological changes could therefore provide better treatments. One challenge is that patients represent a heterogeneous population and the vast majority of cases are sporadic. One pathological feature that almost all (~97%) cases (familial and sporadic) have in common are cytoplasmic aggregates of the protein TDP-43 which is normally located in the nucleus. In our experiments we investigated whether this pathology was sufficient to increase neuronal excitability and the mechanisms by which this occurs. We used the TDP-43(ΔNLS) mouse model which successfully recapitulates this pathology in a controllable way. We used in vivo intracellular recordings in this model to demonstrate that TDP-43 pathology is sufficient to drive a severe hyper-excitability of spinal motoneurones. Reductions in soma size and a lengthening and constriction of axon initial segments were observed, which would contribute to enhanced excitability. Resuppression of the transgene resulted in a return to normal excitability parameters by 6-8 weeks. We therefore conclude that TDP-43 pathology itself is sufficient to drive a severe but reversible hyperexcitability of spinal motoneurones.
Collapse
Affiliation(s)
- Svetlana Djukic
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Zhenxiang Zhao
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | - Anna Normann Bak
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Dennis Bo Jensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Claire Francesca Meehan
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
5
|
Di Re J, Marini M, Hussain SI, Singh AK, Venkatesh A, Alshammari MA, Alshammari TK, Hamoud ARA, Imami AS, Haghighijoo Z, Fularcyzk N, Stertz L, Hawes D, Mosebarger A, Jernigan J, Chaljub C, Nehme R, Walss-Bass C, Schulmann A, Vawter MP, McCullumsmith R, Damoiseaux RD, Limon A, Labate D, Wells MF, Laezza F. βIV spectrin abundancy, cellular distribution and sensitivity to AKT/GSK3 regulation in schizophrenia. Mol Psychiatry 2025:10.1038/s41380-025-02917-1. [PMID: 39920295 DOI: 10.1038/s41380-025-02917-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/06/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder with unclear biological mechanisms. Spectrins, cytoskeletal proteins linked to neurodevelopmental disorders, are regulated by the AKT/GSK3 pathway, which is implicated in SCZ. However, the impact of SCZ-related dysregulation of this pathway on spectrin expression and distribution remains unexplored. Here, we show that βIV spectrin protein levels were reduced in neurons of the dorsolateral prefrontal cortex in SCZ postmortem samples compared to healthy control (HC) from the Human Brain Collection Core (HBCC). To investigate potential links between βIV spectrin and the AKT/GSK3 pathway, we analyzed the PsychEncode dataset, revealing elevated SPTBN4 and AKT2 mRNA levels with correlated gene transcription in both HCs and individuals with SCZ. Next, computational tools were employed to identify potential AKT and GSK3 phosphorylation sites on βIV spectrin, and two GSK3 sites were validated through in vitro assays. To assess whether βIV spectrin distribution and sensitivity to AKT/GSK3 are altered in SCZ, we used iPSC-derived neurons from two independent cohorts of patients with significantly increased familial genetic risk for the disorder. Alteration in βIV spectrin levels and sensitivity to AKT/GSK3 inhibitors were consistently observed across both cohorts. Importantly, a Random Forest classifier applied to βIV spectrin imaging achieved up to 98% accuracy in classifying cells by diagnosis in postmortem samples, and by diagnosis or diagnosis × perturbation in iPSC samples. These findings reveal altered βIV spectrin levels and AKT/GSK3 sensitivity in SCZ, identifying βIV spectrin image-based endophenotypes as robust, generalizable predictive biomarkers of SCZ, with the potential for scalable clinical applications.
Collapse
Affiliation(s)
- Jessica Di Re
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Michela Marini
- Department of Mathematics, University of Houston, Houston, TX, USA
| | | | - Aditya K Singh
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Akshaya Venkatesh
- MD-PhD Combined Program, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Musaad A Alshammari
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdul-Rizaq Ali Hamoud
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ali Sajid Imami
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Zahra Haghighijoo
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | | | - Laura Stertz
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Derek Hawes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Angela Mosebarger
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jordan Jernigan
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Claire Chaljub
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Consuelo Walss-Bass
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Anton Schulmann
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Robert McCullumsmith
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Neurosciences Institute, Promedica, Toledo, OH, USA
| | - Robert D Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Agenor Limon
- Department of Neurology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Demetrio Labate
- Department of Mathematics, University of Houston, Houston, TX, USA
| | - Michael F Wells
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
6
|
Eichel K. Endocytosis in the axon initial segment: Roles in neuronal polarity and plasticity. Curr Opin Neurobiol 2025; 90:102949. [PMID: 39689414 DOI: 10.1016/j.conb.2024.102949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/15/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
The axon initial segment (AIS) is a specialized domain that maintains neuronal polarity and is the site of action potential generation, both of which underlie the neuron's ability to send and receive signals. Disruption of the AIS leads to a loss of neuronal polarity, altered neuronal signaling, and an array of neurological disorders. Therefore, understanding how the AIS forms and functions is a central question in cellular neuroscience that is essential to understanding neuronal physiology. Decades of study have identified many molecular components and mechanisms at the AIS. Recently, endocytosis at the AIS has been identified to function in both maintaining neuronal polarity and in mediating AIS plasticity through its ability to dynamically remodel the plasma membrane composition. This review discusses the emerging evidence for the roles of endocytosis in regulating AIS function and structural insights into how endocytosis can occur at the AIS.
Collapse
Affiliation(s)
- Kelsie Eichel
- Howard Hughes Medical Institute, University of Colorado Boulder, USA.
| |
Collapse
|
7
|
Han Y, Hacker D, Donders BC, Parperis C, Thuenauer R, Leterrier C, Grünewald K, Mikhaylova M. Unveiling the cell biology of hippocampal neurons with dendritic axon origin. J Cell Biol 2025; 224:e202403141. [PMID: 39495320 PMCID: PMC11536041 DOI: 10.1083/jcb.202403141] [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: 03/26/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
In mammalian axon-carrying-dendrite (AcD) neurons, the axon emanates from a basal dendrite, instead of the soma, to create a privileged route for action potential generation at the axon initial segment (AIS). However, it is unclear how such unusual morphology is established and whether the structure and function of the AIS in AcD neurons are preserved. By using dissociated hippocampal cultures as a model, we show that the development of AcD morphology can occur prior to synaptogenesis and independently of the in vivo environment. A single precursor neurite first gives rise to the axon and then to the AcD. The AIS possesses a similar cytoskeletal architecture as the soma-derived AIS and similarly functions as a trafficking barrier to retain axon-specific molecular composition. However, it does not undergo homeostatic plasticity, contains lesser cisternal organelles, and receives fewer inhibitory inputs. Our findings reveal insights into AcD neuron biology and underscore AIS structural differences based on axon onset.
Collapse
Affiliation(s)
- Yuhao Han
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Daniela Hacker
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | - Roland Thuenauer
- Advanced Light and Fluorescence Microscopy (ALFM) Facility, Centre for Structural Systems Biology, Hamburg, Germany
- Technology Platform Light Microscopy, University of Hamburg, Hamburg, Germany
- Technology Platform Microscopy and Image Analysis (TP MIA), Leibniz Institute of Virology (LIV), Hamburg, Germany
| | | | - Kay Grünewald
- Centre for Structural Systems Biology, Hamburg, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany
- AG “Neuronal Protein Transport”, Centre for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Qi Y, Zhao R, Tian J, Lu J, He M, Tai Y. Specific and Plastic: Chandelier Cell-to-Axon Initial Segment Connections in Shaping Functional Cortical Network. Neurosci Bull 2024; 40:1774-1788. [PMID: 39080101 PMCID: PMC11607270 DOI: 10.1007/s12264-024-01266-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 11/30/2024] Open
Abstract
Axon initial segment (AIS) is the most excitable subcellular domain of a neuron for action potential initiation. AISs of cortical projection neurons (PNs) receive GABAergic synaptic inputs primarily from chandelier cells (ChCs), which are believed to regulate action potential generation and modulate neuronal excitability. As individual ChCs often innervate hundreds of PNs, they may alter the activity of PN ensembles and even impact the entire neural network. During postnatal development or in response to changes in network activity, the AISs and axo-axonic synapses undergo dynamic structural and functional changes that underlie the wiring, refinement, and adaptation of cortical microcircuits. Here we briefly introduce the history of ChCs and review recent research advances employing modern genetic and molecular tools. Special attention will be attributed to the plasticity of the AIS and the ChC-PN connections, which play a pivotal role in shaping the dynamic network under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Yanqing Qi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rui Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jifeng Tian
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiangteng Lu
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yilin Tai
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Li Y, Wang H, Wang Y, Chen Z, Liu Y, Tian W, Kang X, Pashang A, Kulasiri D, Yang X, Li HW, Zhang Y. Alterations in the axon initial segment plasticity is involved in early pathogenesis in Alzheimer's disease. MedComm (Beijing) 2024; 5:e768. [PMID: 39415847 PMCID: PMC11473794 DOI: 10.1002/mco2.768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, characterized by the early presence of amyloid-β (Aβ) and hyperphosphorylated tau. Identifying the neuropathological changes preceding cognitive decline is crucial for early intervention. Axon initial segment (AIS) maintains the orderly structure of the axon and is responsible for initiating action potentials (APs). To investigate the role of AIS in early stages of AD pathogenesis, we focused on alterations in the AIS of neurons from APP/PS1 mouse models harboring familial AD mutations. AIS length and electrophysiological properties were assessed in neurons using immunostaining and patch-clamp techniques. The expression and function of ankyrin G (AnkG) isoforms were evaluated by western blot and rescue experiments. We observed a significant shortening of AIS in APP/PS1 mice, which correlated with impaired action potential propagation. Furthermore, a decrease in the 480 kDa isoform of AnkG was observed. Rescue of this isoform restored AIS plasticity and improved long-term potentiation in APP/PS1 neurons. Our study implicates AIS plasticity alterations and AnkG dysregulation as early events in AD. The restoration of AIS integrity by the 480 kDa AnkG isoform presents a potential therapeutic strategy for AD, underscoring the importance of targeting AIS stability in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Li
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Han Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiming Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Zhiya Chen
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiqiong Liu
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Wu Tian
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Xinrui Kang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Xiaoli Yang
- Division of Life Sciences and MedicineDepartment of NeurologyInstitute on Aging and Brain DisordersThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Neurodegenerative Disorder Research CenterAnhui Province Key Laboratory of Biomedical Aging ResearchDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hung Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Yan Zhang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| |
Collapse
|
10
|
Heider J, González EP, Hartmann SM, Kannaiyan N, Vogel S, Wüst R, Fallgatter AJ, Rossner MJ, Kraushaar U, Volkmer H. Aberrant neuronal connectivity and network activity of neurons derived from patients with idiopathic schizophrenia. Neurobiol Dis 2024; 201:106678. [PMID: 39307399 DOI: 10.1016/j.nbd.2024.106678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder with a strong genetic determinant. A major hypothesis to explain disease aetiology comprises synaptic dysfunction associated with excitatory-inhibitory imbalance of synaptic transmission, ultimately contributing to impaired network oscillation and cognitive deficits associated with the disease. Here, we studied the morphological and functional properties of a highly defined co-culture of GABAergic and glutamatergic neurons derived from induced pluripotent stem cells (iPSC) from patients with idiopathic SCZ. Our results indicate upregulation of synaptic genes and increased excitatory synapse formation on GABAergic neurons in co-cultures. In parallel, we observed decreased lengths of axon initial segments, concordant with data from postmortem brains from patients with SCZ. In line with increased synapse density, patch-clamp analyses revealed markedly increased spontaneous excitatory postsynaptic currents (EPSC) recorded from GABAergic SCZ neurons. Finally, MEA recordings from neuronal networks indicate increased strength of network activity, potentially in response to altered synaptic transmission and E-I balance in the co-cultures. In conclusion, our results suggest selective deregulation of neuronal activity in SCZ samples, providing evidence for altered synapse formation and synaptic transmission as a potential base for aberrant network synchronization.
Collapse
Affiliation(s)
- Johanna Heider
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Emilio Pardo González
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Sophia-Marie Hartmann
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Sabrina Vogel
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Andreas J Fallgatter
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Hansjürgen Volkmer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
11
|
Zhao R, Ren B, Xiao Y, Tian J, Zou Y, Wei J, Qi Y, Hu A, Xie X, Huang ZJ, Shu Y, He M, Lu J, Tai Y. Axo-axonic synaptic input drives homeostatic plasticity by tuning the axon initial segment structurally and functionally. SCIENCE ADVANCES 2024; 10:eadk4331. [PMID: 39093969 PMCID: PMC11296346 DOI: 10.1126/sciadv.adk4331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
Homeostatic plasticity maintains the stability of functional brain networks. The axon initial segment (AIS), where action potentials start, undergoes dynamic adjustment to exert powerful control over neuronal firing properties in response to network activity changes. However, it is poorly understood whether this plasticity involves direct synaptic input to the AIS. Here, we show that changes of GABAergic synaptic input from chandelier cells (ChCs) drive homeostatic tuning of the AIS of principal neurons (PNs) in the prelimbic (PL) region, while those from parvalbumin-positive basket cells do not. This tuning is evident in AIS morphology, voltage-gated sodium channel expression, and PN excitability. Moreover, the impact of this homeostatic plasticity can be reflected in animal behavior. Social behavior, inversely linked to PL PN activity, shows time-dependent alterations tightly coupled to changes in AIS plasticity and PN excitability. Thus, AIS-originated homeostatic plasticity in PNs may counteract deficits elicited by imbalanced ChC presynaptic input at cellular and behavioral levels.
Collapse
Affiliation(s)
- Rui Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Baihui Ren
- Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yujie Xiao
- Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Jifeng Tian
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Zou
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiafan Wei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanqing Qi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoying Xie
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Z. Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC 27708, USA
| | - Yousheng Shu
- Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiangteng Lu
- Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yilin Tai
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Zhao R, Ren B, Xiao Y, Tian J, Zou Y, Wei J, Qi Y, Hu A, Xie X, Huang ZJ, Shu Y, He M, Lu J, Tai Y. Axo-axonic synaptic input drives homeostatic plasticity by tuning the axon initial segment structurally and functionally. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589005. [PMID: 38659885 PMCID: PMC11042219 DOI: 10.1101/2024.04.11.589005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The stability of functional brain network is maintained by homeostatic plasticity, which restores equilibrium following perturbation. As the initiation site of action potentials, the axon initial segment (AIS) of glutamatergic projection neurons (PyNs) undergoes dynamic adjustment that exerts powerful control over neuronal firing properties in response to changes in network states. Although AIS plasticity has been reported to be coupled with the changes of network activity, it is poorly understood whether it involves direct synaptic input to the AIS. Here we show that changes of GABAergic synaptic input to the AIS of cortical PyNs, specifically from chandelier cells (ChCs), are sufficient to drive homeostatic tuning of the AIS within 1-2 weeks, while those from parvalbumin-positive basket cells do not. This tuning is reflected in the morphology of the AIS, the expression level of voltage-gated sodium channels, and the intrinsic neuronal excitability of PyNs. Interestingly, the timing of AIS tuning in PyNs of the prefrontal cortex corresponds to the recovery of changes in social behavior caused by alterations of ChC synaptic transmission. Thus, homeostatic plasticity of the AIS at postsynaptic PyNs may counteract deficits elicited by imbalanced ChC presynaptic input. Teaser Axon initial segment dynamically responds to changes in local input from chandelier cells to prevent abnormal neuronal functions.
Collapse
|
13
|
Micinski D, Hotulainen P. Actin polymerization and longitudinal actin fibers in axon initial segment plasticity. Front Mol Neurosci 2024; 17:1376997. [PMID: 38799616 PMCID: PMC11120970 DOI: 10.3389/fnmol.2024.1376997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
The location of the axon initial segment (AIS) at the junction between the soma and axon of neurons makes it instrumental in maintaining neural polarity and as the site for action potential generation. The AIS is also capable of large-scale relocation in an activity-dependent manner. This represents a form of homeostatic plasticity in which neurons regulate their own excitability by changing the size and/or position of the AIS. While AIS plasticity is important for proper functionality of AIS-containing neurons, the cellular and molecular mechanisms of AIS plasticity are poorly understood. Here, we analyzed changes in the AIS actin cytoskeleton during AIS plasticity using 3D structured illumination microscopy (3D-SIM). We showed that the number of longitudinal actin fibers increased transiently 3 h after plasticity induction. We further showed that actin polymerization, especially formin mediated actin polymerization, is required for AIS plasticity and formation of longitudinal actin fibers. From the formin family of proteins, Daam1 localized to the ends of longitudinal actin fibers. These results indicate that active re-organization of the actin cytoskeleton is required for proper AIS plasticity.
Collapse
Affiliation(s)
- David Micinski
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- HiLIFE-Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Wang Y, Chen Y, Chen L, Herron BJ, Chen XY, Wolpaw JR. Motor learning changes the axon initial segment of the spinal motoneuron. J Physiol 2024; 602:2107-2126. [PMID: 38568869 PMCID: PMC11196014 DOI: 10.1113/jp283875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
We are studying the mechanisms of H-reflex operant conditioning, a simple form of learning. Modelling studies in the literature and our previous data suggested that changes in the axon initial segment (AIS) might contribute. To explore this, we used blinded quantitative histological and immunohistochemical methods to study in adult rats the impact of H-reflex conditioning on the AIS of the spinal motoneuron that produces the reflex. Successful, but not unsuccessful, H-reflex up-conditioning was associated with greater AIS length and distance from soma; greater length correlated with greater H-reflex increase. Modelling studies in the literature suggest that these increases may increase motoneuron excitability, supporting the hypothesis that they may contribute to H-reflex increase. Up-conditioning did not affect AIS ankyrin G (AnkG) immunoreactivity (IR), p-p38 protein kinase IR, or GABAergic terminals. Successful, but not unsuccessful, H-reflex down-conditioning was associated with more GABAergic terminals on the AIS, weaker AnkG-IR, and stronger p-p38-IR. More GABAergic terminals and weaker AnkG-IR correlated with greater H-reflex decrease. These changes might potentially contribute to the positive shift in motoneuron firing threshold underlying H-reflex decrease; they are consistent with modelling suggesting that sodium channel change may be responsible. H-reflex down-conditioning did not affect AIS dimensions. This evidence that AIS plasticity is associated with and might contribute to H-reflex conditioning adds to evidence that motor learning involves both spinal and brain plasticity, and both neuronal and synaptic plasticity. AIS properties of spinal motoneurons are likely to reflect the combined influence of all the motor skills that share these motoneurons. KEY POINTS: Neuronal action potentials normally begin in the axon initial segment (AIS). AIS plasticity affects neuronal excitability in development and disease. Whether it does so in learning is unknown. Operant conditioning of a spinal reflex, a simple learning model, changes the rat spinal motoneuron AIS. Successful, but not unsuccessful, H-reflex up-conditioning is associated with greater AIS length and distance from soma. Successful, but not unsuccessful, down-conditioning is associated with more AIS GABAergic terminals, less ankyrin G, and more p-p38 protein kinase. The associations between AIS plasticity and successful H-reflex conditioning are consistent with those between AIS plasticity and functional changes in development and disease, and with those predicted by modelling studies in the literature. Motor learning changes neurons and synapses in spinal cord and brain. Because spinal motoneurons are the final common pathway for behaviour, their AIS properties probably reflect the combined impact of all the behaviours that use these motoneurons.
Collapse
Affiliation(s)
- Yu Wang
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Yi Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Lu Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Bruce J. Herron
- Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| | - Xiang Yang Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| | - Jonathan R. Wolpaw
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| |
Collapse
|
15
|
Konietzny A, Han Y, Popp Y, van Bommel B, Sharma A, Delagrange P, Arbez N, Moutin MJ, Peris L, Mikhaylova M. Efficient axonal transport of endolysosomes relies on the balanced ratio of microtubule tyrosination and detyrosination. J Cell Sci 2024; 137:jcs261737. [PMID: 38525600 PMCID: PMC11112122 DOI: 10.1242/jcs.261737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
In neurons, the microtubule (MT) cytoskeleton forms the basis for long-distance protein transport from the cell body into and out of dendrites and axons. To maintain neuronal polarity, the axon initial segment (AIS) serves as a physical barrier, separating the axon from the somatodendritic compartment and acting as a filter for axonal cargo. Selective trafficking is further instructed by axonal enrichment of MT post-translational modifications, which affect MT dynamics and the activity of motor proteins. Here, we compared two knockout mouse lines lacking the respective enzymes for MT tyrosination and detyrosination, and found that both knockouts led to a shortening of the AIS. Neurons from both lines also showed an increased immobile fraction of endolysosomes present in the axon, whereas mobile organelles displayed shortened run distances in the retrograde direction. Overall, our results highlight the importance of maintaining the balance of tyrosinated and detyrosinated MTs for proper AIS length and axonal transport processes.
Collapse
Affiliation(s)
- Anja Konietzny
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan
| | - Yuhao Han
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Centre for Structural Systems Biology, Hamburg 22607, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg 20251, Germany
| | - Yannes Popp
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Charité – Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany
| | - Bas van Bommel
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Aditi Sharma
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | | | - Nicolas Arbez
- Institut de Recherche Servier, Croissy 78290, France
| | - Marie-Jo Moutin
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marina Mikhaylova
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| |
Collapse
|
16
|
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has become an increasingly popular tool to modulate neural excitability and induce neural plasticity in clinical and preclinical models; however, the physiological mechanisms in which it exerts these effects remain largely unknown. To date, studies have primarily focused on characterizing rTMS-induced changes occurring at the synapse, with little attention given to changes in intrinsic membrane properties. However, accumulating evidence suggests that rTMS may induce its effects, in part, via intrinsic plasticity mechanisms, suggesting a new and potentially complementary understanding of how rTMS alters neural excitability and neural plasticity. In this review, we provide an overview of several intrinsic plasticity mechanisms before reviewing the evidence for rTMS-induced intrinsic plasticity. In addition, we discuss a select number of neurological conditions where rTMS-induced intrinsic plasticity has therapeutic potential before speculating on the temporal relationship between rTMS-induced intrinsic and synaptic plasticity.
Collapse
Affiliation(s)
- Emily S King
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
17
|
Beros JL, King ES, Clarke D, Jaeschke-Angi L, Rodger J, Tang AD. Static magnetic stimulation induces structural plasticity at the axon initial segment of inhibitory cortical neurons. Sci Rep 2024; 14:1479. [PMID: 38233493 PMCID: PMC10794225 DOI: 10.1038/s41598-024-51845-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/10/2024] [Indexed: 01/19/2024] Open
Abstract
Static magnetic stimulation (SMS) is a form of non-invasive brain stimulation that alters neural activity and induces neural plasticity that outlasts the period of stimulation. This can modify corticospinal excitability or motor behaviours, suggesting that SMS may alter the intrinsic excitability of neurons. In mammalian neurons, the axon initial segment (AIS) is the site of action potential initiation and undergoes structural plasticity (changes in length and position from the soma) as a homeostatic mechanism to counteract chronic changes in neuronal activity. We investigated whether the chronic application of SMS (6 and 48 h, 0.5 T) induces structural AIS plasticity in postnatally derived primary cortical neurons. Following 6 h of SMS, we observed a shortening in mean AIS length compared to control, that persisted 24 h post stimulation. In contrast, 48 h of SMS induced an immediate distal shift that persisted 24 h post-stimulation. Pharmacological blockade of voltage gated L/T-type calcium channels during stimulation did not prevent SMS-induced AIS structural plasticity. Our findings provide the foundation to expand the use of chronic SMS as a non-invasive method to promote AIS plasticity.
Collapse
Affiliation(s)
- J L Beros
- School of Biological Sciences, The University of Western Australia, Crawley, 6009, Australia.
- The Perron Institute for Neurological and Translational Science, Nedlands, 6009, Australia.
| | - E S King
- The Perron Institute for Neurological and Translational Science, Nedlands, 6009, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, 6009, Australia
| | - D Clarke
- Département de Neurosciences, Université de Montréal, Quebec, H3C 3J7, Canada
| | - L Jaeschke-Angi
- School of Biological Sciences, The University of Western Australia, Crawley, 6009, Australia
- The Perron Institute for Neurological and Translational Science, Nedlands, 6009, Australia
| | - J Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, 6009, Australia
- The Perron Institute for Neurological and Translational Science, Nedlands, 6009, Australia
| | - A D Tang
- The Perron Institute for Neurological and Translational Science, Nedlands, 6009, Australia.
- School of Biomedical Sciences, The University of Western Australia, Crawley, 6009, Australia.
| |
Collapse
|
18
|
Harley P, Kerins C, Gatt A, Neves G, Riccio F, Machado CB, Cheesbrough A, R'Bibo L, Burrone J, Lieberam I. Aberrant axon initial segment plasticity and intrinsic excitability of ALS hiPSC motor neurons. Cell Rep 2023; 42:113509. [PMID: 38019651 DOI: 10.1016/j.celrep.2023.113509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Dysregulated neuronal excitability is a hallmark of amyotrophic lateral sclerosis (ALS). We sought to investigate how functional changes to the axon initial segment (AIS), the site of action potential generation, could impact neuronal excitability in ALS human induced pluripotent stem cell (hiPSC) motor neurons. We find that early TDP-43 and C9orf72 hiPSC motor neurons show an increase in the length of the AIS and impaired activity-dependent AIS plasticity that is linked to abnormal homeostatic regulation of neuronal activity and intrinsic hyperexcitability. In turn, these hyperactive neurons drive increased spontaneous myofiber contractions of in vitro hiPSC motor units. In contrast, late hiPSC and postmortem ALS motor neurons show AIS shortening, and hiPSC motor neurons progress to hypoexcitability. At a molecular level, aberrant expression of the AIS master scaffolding protein ankyrin-G and AIS-specific voltage-gated sodium channels mirror these dynamic changes in AIS function and excitability. Our results point toward the AIS as an important site of dysfunction in ALS motor neurons.
Collapse
Affiliation(s)
- Peter Harley
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK; UCL Queen Square Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Caoimhe Kerins
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK; Centre for Craniofacial & Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Ariana Gatt
- Queen Square Brain Bank, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Guilherme Neves
- Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK
| | - Federica Riccio
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK
| | - Carolina Barcellos Machado
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK
| | - Aimee Cheesbrough
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK
| | - Lea R'Bibo
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, Kings College London, London SE1 1UL, UK.
| | - Ivo Lieberam
- Centre for Gene Therapy & Regenerative Medicine, Kings College London, London SE1 9RT, UK; Centre for Developmental Neurobiology, Kings College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, Kings College London, London SE1 1UL, UK.
| |
Collapse
|
19
|
Iwahashi M, Yoshimura T, Harigai W, Takuma K, Hashimoto H, Katayama T, Hayata-Takano A. Pituitary adenylate cyclase-activating polypeptide deficient mice show length abnormalities of the axon initial segment. J Pharmacol Sci 2023; 153:175-182. [PMID: 37770159 DOI: 10.1016/j.jphs.2023.08.006] [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: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
We previously found that pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP-/-) mice exhibit dendritic spine morphology impairment and neurodevelopmental disorder (NDD)-like behaviors such as hyperactivity, increased novelty-seeking behavior, and deficient pre-pulse inhibition. Recent studies have indicated that rodent models of NDDs (e.g., attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder) show abnormalities in the axon initial segment (AIS). Here, we revealed that PACAP-/- mice exhibited a longer AIS length in layer 2/3 pyramidal neurons of the primary somatosensory barrel field compared with wild-type control mice. Further, we previously showed that a single injection of atomoxetine, an ADHD drug, improved hyperactivity in PACAP-/- mice. In this study, we found that repeated treatments of atomoxetine significantly improved AIS abnormality along with hyperactivity in PACAP-/- mice. These results suggest that AIS abnormalities are associated with NDDs-like behaviors in PACAP-/- mice. Thus, improvement in AIS abnormalities will be a novel drug therapy for NDDs.
Collapse
Affiliation(s)
- Misaki Iwahashi
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeshi Yoshimura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Wakana Harigai
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hitoshi Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, 2-8 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsuko Hayata-Takano
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Townsend LN, Clarke H, Maddison D, Jones KM, Amadio L, Jefferson A, Chughtai U, Bis DM, Züchner S, Allen ND, Van der Goes van Naters W, Peters OM, Smith GA. Cdk12 maintains the integrity of adult axons by suppressing actin remodeling. Cell Death Discov 2023; 9:348. [PMID: 37730761 PMCID: PMC10511712 DOI: 10.1038/s41420-023-01642-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
The role of cyclin-dependent kinases (CDKs) that are ubiquitously expressed in the adult nervous system remains unclear. Cdk12 is enriched in terminally differentiated neurons where its conical role in the cell cycle progression is redundant. We find that in adult neurons Cdk12 acts a negative regulator of actin formation, mitochondrial dynamics and neuronal physiology. Cdk12 maintains the size of the axon at sites proximal to the cell body through the transcription of homeostatic enzymes in the 1-carbon by folate pathway which utilize the amino acid homocysteine. Loss of Cdk12 leads to elevated homocysteine and in turn leads to uncontrolled F-actin formation and axonal swelling. Actin remodeling further induces Drp1-dependent fission of mitochondria and the breakdown of axon-soma filtration barrier allowing soma restricted cargos to enter the axon. We demonstrate that Cdk12 is also an essential gene for long-term neuronal survival and loss of this gene causes age-dependent neurodegeneration. Hyperhomocysteinemia, actin changes, and mitochondrial fragmentation are associated with several neurodegenerative conditions such as Alzheimer's disease and we provide a candidate molecular pathway to link together such pathological events.
Collapse
Affiliation(s)
- L N Townsend
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - H Clarke
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - D Maddison
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - K M Jones
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - L Amadio
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - A Jefferson
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - U Chughtai
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - D M Bis
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - S Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - N D Allen
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | | | - O M Peters
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - G A Smith
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK.
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
21
|
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
|
22
|
Teunissen MWA, Lewerissa E, van Hugte EJH, Wang S, Ockeloen CW, Koolen DA, Pfundt R, Marcelis CLM, Brilstra E, Howe JL, Scherer SW, Le Guillou X, Bilan F, Primiano M, Roohi J, Piton A, de Saint Martin A, Baer S, Seiffert S, Platzer K, Jamra RA, Syrbe S, Doering JH, Lakhani S, Nangia S, Gilissen C, Vermeulen RJ, Rouhl RPW, Brunner HG, Willemsen MH, Nadif Kasri N. ANK2 loss-of-function variants are associated with epilepsy, and lead to impaired axon initial segment plasticity and hyperactive network activity in hiPSC-derived neuronal networks. Hum Mol Genet 2023; 32:2373-2385. [PMID: 37195288 PMCID: PMC10321384 DOI: 10.1093/hmg/ddad081] [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: 10/14/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023] Open
Abstract
PURPOSE To characterize a novel neurodevelopmental syndrome due to loss-of-function (LoF) variants in Ankyrin 2 (ANK2), and to explore the effects on neuronal network dynamics and homeostatic plasticity in human-induced pluripotent stem cell-derived neurons. METHODS We collected clinical and molecular data of 12 individuals with heterozygous de novo LoF variants in ANK2. We generated a heterozygous LoF allele of ANK2 using CRISPR/Cas9 in human-induced pluripotent stem cells (hiPSCs). HiPSCs were differentiated into excitatory neurons, and we measured their spontaneous electrophysiological responses using micro-electrode arrays (MEAs). We also characterized their somatodendritic morphology and axon initial segment (AIS) structure and plasticity. RESULTS We found a broad neurodevelopmental disorder (NDD), comprising intellectual disability, autism spectrum disorders and early onset epilepsy. Using MEAs, we found that hiPSC-derived neurons with heterozygous LoF of ANK2 show a hyperactive and desynchronized neuronal network. ANK2-deficient neurons also showed increased somatodendritic structures and altered AIS structure of which its plasticity is impaired upon activity-dependent modulation. CONCLUSIONS Phenotypic characterization of patients with de novo ANK2 LoF variants defines a novel NDD with early onset epilepsy. Our functional in vitro data of ANK2-deficient human neurons show a specific neuronal phenotype in which reduced ANKB expression leads to hyperactive and desynchronized neuronal network activity, increased somatodendritic complexity and AIS structure and impaired activity-dependent plasticity of the AIS.
Collapse
Affiliation(s)
- Maria W A Teunissen
- Department of Neurology, Maastricht University Medical Center, Maastricht, HX 6229, The Netherlands
- Academic Center for Epileptology Kempenhaeghe/Maastricht University Medical Center, Heeze 5591 VE, The Netherlands
| | - Elly Lewerissa
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
| | - Eline J H van Hugte
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
| | - Shan Wang
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
| | - Charlotte W Ockeloen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, GA 6525, the Netherlands
| | - David A Koolen
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, GA 6525, the Netherlands
| | - Carlo L M Marcelis
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, GA 6525, the Netherlands
| | - Eva Brilstra
- Department of Human Genetics, University Medical Center Utrecht, Utrecht, CX 3584, The Netherlands
| | - Jennifer L Howe
- The Centre for Applied Genomics and Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3H7, Canada
| | - Xavier Le Guillou
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers 86000, France
| | - Frédéric Bilan
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers 86000, France
- Laboratory of Experimental and Clinical Neurosciences University of Poitiers, INSERM U1084, Poitiers 86000, France
| | - Michelle Primiano
- Department of Clinical Genetics, Morgan Stanley Children’s Hospital of New York-Presbytarian, New York, NY, 10032, USA
| | - Jasmin Roohi
- Department of Clinical Genetics, Morgan Stanley Children’s Hospital of New York-Presbytarian, New York, NY, 10032, USA
- Clinical Genetics, Kaiser Permanente Mid-Atlantic Permanente Medical Group, Rockville, MD 20852, USA
| | - Amelie Piton
- Laboratoire de Diagnostic Génétique, Institut de Génétique Médicale d’Alsace (IGMA), Hôspitaux Universitaire de Strasbourg, Strasbourg, BP 426 67091, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France
| | - Anne de Saint Martin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France
- Department of Pediatric Neurology, Strasbourg University Hospital, Hôspital de Hautepierre, Strasbourg, BP 426 67091, France
| | - Sarah Baer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France
- Department of Pediatric Neurology, Strasbourg University Hospital, Hôspital de Hautepierre, Strasbourg, BP 426 67091, France
| | - Simone Seiffert
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Tuebingen, 72076, Germany
| | - Konrad Platzer
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig 04103, Germany
| | - Rami Abou Jamra
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig 04103, Germany
| | - Steffen Syrbe
- Division of Paediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Jan H Doering
- Division of Paediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Shenela Lakhani
- Department of neurogenetics, Weill Cornell Medicine, Brain and Mind Research Institute, New York, NY, 10065, USA
| | - Srishti Nangia
- Department of Pediatrics, Division of Child Neurology, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, 10032, USA
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, GA 6525, the Netherlands
| | - R Jeroen Vermeulen
- Department of Neurology, Maastricht University Medical Center, Maastricht, HX 6229, The Netherlands
| | - Rob P W Rouhl
- Department of Neurology, Maastricht University Medical Center, Maastricht, HX 6229, The Netherlands
- Academic Center for Epileptology Kempenhaeghe/Maastricht University Medical Center, Heeze 5591 VE, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, MD 6200, the Netherlands
| | - Han G Brunner
- Academic Center for Epileptology Kempenhaeghe/Maastricht University Medical Center, Heeze 5591 VE, The Netherlands
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, MD 6200, the Netherlands
- Department of Clinical Genetics and GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, MD 6299, the Netherlands
| | - Marjolein H Willemsen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, GA 6525, the Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, HB 6500, the Netherlands
| |
Collapse
|
23
|
Stajković N, Liu Y, Arsić A, Meng N, Lyu H, Zhang N, Grimm D, Lerche H, Nikić-Spiegel I. Direct fluorescent labeling of NF186 and NaV1.6 in living primary neurons using bioorthogonal click chemistry. J Cell Sci 2023; 136:jcs260600. [PMID: 37288813 PMCID: PMC10323244 DOI: 10.1242/jcs.260600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
The axon initial segment (AIS) is a highly specialized neuronal compartment that regulates the generation of action potentials and maintenance of neuronal polarity. Live imaging of the AIS is challenging due to the limited number of suitable labeling methods. To overcome this limitation, we established a novel approach for live labeling of the AIS using unnatural amino acids (UAAs) and click chemistry. The small size of UAAs and the possibility of introducing them virtually anywhere into target proteins make this method particularly suitable for labeling of complex and spatially restricted proteins. Using this approach, we labeled two large AIS components, the 186 kDa isoform of neurofascin (NF186; encoded by Nfasc) and the 260 kDa voltage-gated Na+ channel (NaV1.6, encoded by Scn8a) in primary neurons and performed conventional and super-resolution microscopy. We also studied the localization of epilepsy-causing NaV1.6 variants with a loss-of-function effect. Finally, to improve the efficiency of UAA incorporation, we developed adeno-associated viral (AAV) vectors for click labeling in neurons, an achievement that could be transferred to more complex systems such as organotypic slice cultures, organoids, and animal models.
Collapse
Affiliation(s)
- Nevena Stajković
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tübingen, 72076 Tübingen, Germany
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Aleksandra Arsić
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tübingen, 72076 Tübingen, Germany
| | - Ning Meng
- Virus-Host Interaction Group, Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Cluster of Excellence CellNetworks, BioQuant, 69120 Heidelberg, Germany
| | - Hang Lyu
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of Tübingen, 72076 Tübingen, Germany
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Nan Zhang
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Dirk Grimm
- Virus-Host Interaction Group, Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Cluster of Excellence CellNetworks, BioQuant, 69120 Heidelberg, Germany
- German Center for Infection Research and German Center for Cardiovascular Research, partner site Heidelberg, 69120 Heidelberg, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Ivana Nikić-Spiegel
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
24
|
Jungenitz T, Bird A, Engelhardt M, Jedlicka P, Schwarzacher SW, Deller T. Structural plasticity of the axon initial segment in rat hippocampal granule cells following high frequency stimulation and LTP induction. Front Neuroanat 2023; 17:1125623. [PMID: 37090138 PMCID: PMC10113456 DOI: 10.3389/fnana.2023.1125623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
The axon initial segment (AIS) is the site of action potential initiation and important for the integration of synaptic input. Length and localization of the AIS are dynamic, modulated by afferent activity and contribute to the homeostatic control of neuronal excitability. Synaptopodin is a plasticity-related protein expressed by the majority of telencephalic neurons. It is required for the formation of cisternal organelles within the AIS and an excellent marker to identify these enigmatic organelles at the light microscopic level. Here we applied 2 h of high frequency stimulation of the medial perforant path in rats in vivo to induce a strong long-term potentiation of dentate gyrus granule cells. Immunolabeling for βIV-spectrin and synaptopodin were performed to study structural changes of the AIS and its cisternal organelles. Three-dimensional analysis of the AIS revealed a shortening of the AIS and a corresponding reduction of the number of synaptopodin clusters. These data demonstrate a rapid structural plasticity of the AIS and its cisternal organelles to strong stimulation, indicating a homeostatic response of the entire AIS compartment.
Collapse
Affiliation(s)
- Tassilo Jungenitz
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Alexander Bird
- Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University Giessen, Giessen, Germany
| | - Maren Engelhardt
- Institute of Anatomy and Cell Biology, Johannes Kepler University Linz, Linz, Austria
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt, Frankfurt am Main, Germany
- Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University Giessen, Giessen, Germany
| | | | - Thomas Deller
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Boal AM, McGrady NR, Chamling X, Kagitapalli BS, Zack DJ, Calkins DJ, Risner ML. Microfluidic Platforms Promote Polarization of Human-Derived Retinal Ganglion Cells That Model Axonopathy. Transl Vis Sci Technol 2023; 12:1. [PMID: 37010860 PMCID: PMC10080917 DOI: 10.1167/tvst.12.4.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023] Open
Abstract
Purpose Axons depend on long-range transport of proteins and organelles which increases susceptibility to metabolic stress in disease. The axon initial segment (AIS) is particularly vulnerable due to the high bioenergetic demand of action potential generation. Here, we prepared retinal ganglion cells derived from human embryonic stem cells (hRGCs) to probe how axonal stress alters AIS morphology. Methods hRGCs were cultured on coverslips or microfluidic platforms. We assayed AIS specification and morphology by immunolabeling against ankyrin G (ankG), an axon-specific protein, and postsynaptic density 95 (PSD-95), a dendrite-specific protein. Using microfluidic platforms that enable fluidic isolation, we added colchicine to the axon compartment to lesion axons. We verified axonopathy by measuring the anterograde axon transport of cholera toxin subunit B and immunolabeling against cleaved caspase 3 (CC3) and phosphorylated neurofilament H (SMI-34). We determined the influence of axon injury on AIS morphology by immunolabeling samples against ankG and measuring AIS distance from soma and length. Results Based on measurements of ankG and PSD-95 immunolabeling, microfluidic platforms promote the formation and separation of distinct somatic-dendritic versus axonal compartments in hRGCs compared to coverslip cultures. Chemical lesioning of axons by colchicine reduced hRGC anterograde axon transport, increased varicosity density, and enhanced expression of CC3 and SMI-34. Interestingly, we found that colchicine selectively affected hRGCs with axon-carrying dendrites by reducing AIS distance from somas and increasing length, thus suggesting reduced capacity to maintain excitability. Conclusions Thus, microfluidic platforms promote polarized hRGCs that enable modeling of axonopathy. Translational Relevance Microfluidic platforms may be used to assay compartmentalized degeneration that occurs during glaucoma.
Collapse
Affiliation(s)
- Andrew M. Boal
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nolan R. McGrady
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xitiz Chamling
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bhanu S. Kagitapalli
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Donald J. Zack
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J. Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L. Risner
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
26
|
CDK5/p35-Dependent Microtubule Reorganization Contributes to Homeostatic Shortening of the Axon Initial Segment. J Neurosci 2023; 43:359-372. [PMID: 36639893 PMCID: PMC9864565 DOI: 10.1523/jneurosci.0917-22.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/12/2022] Open
Abstract
The structural plasticity of the axon initial segment (AIS) contributes to the homeostatic control of activity and optimizes the function of neural circuits; however, the underlying mechanisms are not fully understood. In this study, we prepared a slice culture containing nucleus magnocellularis from chickens of both sexes that reproduces most features of AIS plasticity in vivo, regarding its effects on characteristics of AIS and cell-type specificity, and revealed that microtubule reorganization via activation of CDK5 underlies plasticity. Treating the culture with a high-K+ medium shortened the AIS and reduced sodium current and membrane excitability, specifically in neurons tuned to high-frequency sound, creating a tonotopic difference in AIS length in the nucleus. Pharmacological analyses revealed that this AIS shortening was driven by multiple Ca2+ pathways and subsequent signaling molecules that converge on CDK5 via the activation of ERK1/2. AIS shortening was suppressed by overexpression of dominant-negative CDK5, whereas it was facilitated by the overexpression of p35, an activator of CDK5. Notably, p35(T138A), a phosphorylation-inactive mutant of p35, did not shorten the AIS. Moreover, microtubule stabilizers occluded AIS shortening during the p35 overexpression, indicating that CDK5/p35 mediated AIS shortening by promoting disassembly of microtubules at distal AIS. This study highlights the importance of microtubule reorganization and regulation of CDK5 activity in structural AIS plasticity and the tuning of AIS characteristics in neurons.SIGNIFICANCE STATEMENT The structural plasticity of AIS has a strong impact on the output of neurons and plays a fundamental role in the physiology and pathology of the brain. However, the mechanisms linking neuronal activity to structural changes in AIS are not well understood. In this study, we prepared an organotypic culture of avian auditory brainstem, reproducing most AIS plasticity features in vivo, and we revealed that activity-dependent AIS shortening occurs through the disassembly of microtubules at distal AIS via activation of CDK5/p35 signals. This study emphasizes the importance of microtubule reorganization and regulation of CDK5 activity in structural AIS plasticity and tonotopic differentiation of AIS structures in the brainstem auditory circuit.
Collapse
|
27
|
Senol AD, Pinto G, Beau M, Guillemot V, Dupree JL, Stadelmann C, Ranft J, Lubetzki C, Davenne M. Alterations of the axon initial segment in multiple sclerosis grey matter. Brain Commun 2022; 4:fcac284. [PMID: 36451656 PMCID: PMC9700164 DOI: 10.1093/braincomms/fcac284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/14/2022] [Accepted: 11/02/2022] [Indexed: 07/22/2023] Open
Abstract
Grey matter damage has been established as a key contributor to disability progression in multiple sclerosis. Aside from neuronal loss and axonal transections, which predominate in cortical demyelinated lesions, synaptic alterations have been detected in both demyelinated plaques and normal-appearing grey matter, resulting in functional neuronal damage. The axon initial segment is a key element of neuronal function, responsible for action potential initiation and maintenance of neuronal polarity. Despite several reports of profound axon initial segment alterations in different pathological models, among which experimental auto-immune encephalomyelitis, whether the axon initial segment is affected in multiple sclerosis is still unknown. Using immunohistochemistry, we analysed axon initial segments from control and multiple sclerosis tissue, focusing on layer 5/6 pyramidal neurons in the neocortex and Purkinje cells in the cerebellum and performed analysis on the parameters known to control neuronal excitability, i.e. axon initial segment length and position. We found that the axon initial segment length was increased only in pyramidal neurons of inactive demyelinated lesions, compared with normal appearing grey matter tissue. In contrast, in both cell types, the axon initial segment position was altered, with an increased soma-axon initial segment gap, in both active and inactive demyelinated lesions. In addition, using a computational model, we show that this increased gap between soma and axon initial segment might increase neuronal excitability. Taken together, these results show, for the first time, changes of axon initial segments in multiple sclerosis, in active as well as inactive grey matter lesions in both neocortex and cerebellum, which might alter neuronal function.
Collapse
Affiliation(s)
- Aysegul Dilsizoglu Senol
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
| | - Giulia Pinto
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
| | - Maxime Beau
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris, France
| | - Vincent Guillemot
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
- Institut Pasteur, Université de Paris, Bioinformatics and Biostatistics Hub, Paris F-75015, France
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jonas Ranft
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, Inserm, PSL Research University, Paris, France
| | - Catherine Lubetzki
- Sorbonne University, Paris Brain Institute—ICM, Inserm, CNRS, Pitié-Salpêtrière Hospital, Paris, France
- Assistance Publique des Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, DMU Neurosciences, Paris, France
| | - Marc Davenne
- Correspondence to: Dr Marc Davenne Paris Brain Institute, Pitié-Salpêtrière Hospital 47, bd de l’hôpital, F-75013 Paris, France E-mail:
| |
Collapse
|
28
|
Bar L, Shalom L, Lezmy J, Peretz A, Attali B. Excitatory and inhibitory hippocampal neurons differ in their homeostatic adaptation to chronic M-channel modulation. Front Mol Neurosci 2022; 15:972023. [PMID: 36311018 PMCID: PMC9614320 DOI: 10.3389/fnmol.2022.972023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
A large body of studies has investigated bidirectional homeostatic plasticity both in vitro and in vivo using numerous pharmacological manipulations of activity or behavioral paradigms. However, these experiments rarely explored in the same cellular system the bidirectionality of the plasticity and simultaneously on excitatory and inhibitory neurons. M-channels are voltage-gated potassium channels that play a crucial role in regulating neuronal excitability and plasticity. In cultured hippocampal excitatory neurons, we previously showed that chronic exposure to the M-channel blocker XE991 leads to adaptative compensations, thereby triggering at different timescales intrinsic and synaptic homeostatic plasticity. This plastic adaptation barely occurs in hippocampal inhibitory neurons. In this study, we examined whether this homeostatic plasticity induced by M-channel inhibition was bidirectional by investigating the acute and chronic effects of the M-channel opener retigabine on hippocampal neuronal excitability. Acute retigabine exposure decreased excitability in both excitatory and inhibitory neurons. Chronic retigabine treatment triggered in excitatory neurons homeostatic adaptation of the threshold current and spontaneous firing rate at a time scale of 4–24 h. These plastic changes were accompanied by a substantial decrease in the M-current density and by a small, though significant, proximal relocation of Kv7.3-FGF14 segment along the axon initial segment. Thus, bidirectional homeostatic changes were observed in excitatory neurons though not symmetric in kinetics and mechanisms. Contrastingly, in inhibitory neurons, the compensatory changes in intrinsic excitability barely occurred after 48 h, while no homeostatic normalization of the spontaneous firing rate was observed. Our results indicate that excitatory and inhibitory hippocampal neurons differ in their adaptation to chronic alterations in neuronal excitability induced by M-channel bidirectional modulation.
Collapse
|
29
|
Harris AC, Jin XT, Greer JE, Povlishock JT, Jacobs KM. Somatostatin interneurons exhibit enhanced functional output and resilience to axotomy after mild traumatic brain injury. Neurobiol Dis 2022; 171:105801. [PMID: 35753625 PMCID: PMC9383472 DOI: 10.1016/j.nbd.2022.105801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/01/2022] Open
Abstract
Mild traumatic brain injury (mTBI) gives rise to a remarkable breadth of pathobiological consequences, principal among which are traumatic axonal injury and perturbation of the functional integrity of neuronal networks that may arise secondary to the elimination of the presynaptic contribution of axotomized neurons. Because there exists a vast diversity of neocortical neuron subtypes, it is imperative to elucidate the relative vulnerability to axotomy among different subtypes. Toward this end, we exploited SOM-IRES-Cre mice to investigate the consequences of the central fluid percussion model of mTBI on the microanatomical integrity and the functional efficacy of the somatostatin (SOM) interneuron population, one of the principal subtypes of neocortical interneuron. We found that the SOM population is resilient to axotomy, representing only 10% of the global burden of inhibitory interneuron axotomy, a result congruous with past work demonstrating that parvalbumin (PV) interneurons bear most of the burden of interneuron axotomy. However, the intact structure of SOM interneurons after injury did not translate to normal cellular function. One day after mTBI, the SOM population is more intrinsically excitable and demonstrates enhanced synaptic efficacy upon post-synaptic layer 5 pyramidal neurons as measured by optogenetics, yet the global evoked inhibitory tone within layer 5 is stable. Simultaneously, there exists a significant increase in the frequency of miniature inhibitory post-synaptic currents within layer 5 pyramidal neurons. These results are consistent with a scheme in which 1 day after mTBI, SOM interneurons are stimulated to compensate for the release from inhibition of layer 5 pyramidal neurons secondary to the disproportionate axotomy of PV interneurons. The enhancement of SOM interneuron intrinsic excitability and synaptic efficacy may represent the initial phase of a dynamic process of attempted autoregulation of neocortical network homeostasis secondary to mTBI.
Collapse
Affiliation(s)
- Alan C Harris
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - Xiao-Tao Jin
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - John E Greer
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - John T Povlishock
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - Kimberle M Jacobs
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| |
Collapse
|
30
|
Single-dose ethanol intoxication causes acute and lasting neuronal changes in the brain. Proc Natl Acad Sci U S A 2022; 119:e2122477119. [PMID: 35700362 DOI: 10.1073/pnas.2122477119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alcohol intoxication at early ages is a risk factor for the development of addictive behavior. To uncover neuronal molecular correlates of acute ethanol intoxication, we used stable-isotope-labeled mice combined with quantitative mass spectrometry to screen more than 2,000 hippocampal proteins, of which 72 changed synaptic abundance up to twofold after ethanol exposure. Among those were mitochondrial proteins and proteins important for neuronal morphology, including MAP6 and ankyrin-G. Based on these candidate proteins, we found acute and lasting molecular, cellular, and behavioral changes following a single intoxication in alcohol-naïve mice. Immunofluorescence analysis revealed a shortening of axon initial segments. Longitudinal two-photon in vivo imaging showed increased synaptic dynamics and mitochondrial trafficking in axons. Knockdown of mitochondrial trafficking in dopaminergic neurons abolished conditioned alcohol preference in Drosophila flies. This study introduces mitochondrial trafficking as a process implicated in reward learning and highlights the potential of high-resolution proteomics to identify cellular mechanisms relevant for addictive behavior.
Collapse
|
31
|
Pyramidal cell axon initial segment in Alzheimer´s disease. Sci Rep 2022; 12:8722. [PMID: 35610289 PMCID: PMC9130508 DOI: 10.1038/s41598-022-12700-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 05/09/2022] [Indexed: 11/08/2022] Open
Abstract
The axon initial segment (AIS) is a region of the neuron that is critical for action potential generation as well as for the regulation of neural activity. This specialized structure-characterized by the expression of different types of ion channels as well as adhesion, scaffolding and cytoskeleton proteins-is subjected to morpho-functional plastic changes in length and position upon variations in neural activity or in pathological conditions. In the present study, using immunocytochemistry with the AT8 antibody (phospho-tau S202/T205) and 3D confocal microscopy reconstruction techniques in brain tissue from Alzheimer's disease patients, we found that around half of the cortical pyramidal neurons with hyperphosphorylated tau showed changes in AIS length and position in comparison with AT8-negative neurons from the same cortical layers. We observed a wide variety of AIS alterations in neurons with hyperphosphorylated tau, although the most common changes were a proximal shift or a lengthening of the AISs. Similar results were found in neocortical tissue from non-demented cases with neurons containing hyperphosphorylated tau. These findings support the notion that the accumulation of phospho-tau is associated with structural alterations of the AIS that are likely to have an impact on normal neuronal activity, which might contribute to neuronal dysfunction in AD.
Collapse
|
32
|
Peña-Ortega F, Robles-Gómez ÁA, Xolalpa-Cueva L. Microtubules as Regulators of Neural Network Shape and Function: Focus on Excitability, Plasticity and Memory. Cells 2022; 11:cells11060923. [PMID: 35326374 PMCID: PMC8946818 DOI: 10.3390/cells11060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
Neuronal microtubules (MTs) are complex cytoskeletal protein arrays that undergo activity-dependent changes in their structure and function as a response to physiological demands throughout the lifespan of neurons. Many factors shape the allostatic dynamics of MTs and tubulin dimers in the cytosolic microenvironment, such as protein–protein interactions and activity-dependent shifts in these interactions that are responsible for their plastic capabilities. Recently, several findings have reinforced the role of MTs in behavioral and cognitive processes in normal and pathological conditions. In this review, we summarize the bidirectional relationships between MTs dynamics, neuronal processes, and brain and behavioral states. The outcomes of manipulating the dynamicity of MTs by genetic or pharmacological approaches on neuronal morphology, intrinsic and synaptic excitability, the state of the network, and behaviors are heterogeneous. We discuss the critical position of MTs as responders and adaptative elements of basic neuronal function whose impact on brain function is not fully understood, and we highlight the dilemma of artificially modulating MT dynamics for therapeutic purposes.
Collapse
|
33
|
Marosi M, Nenov MN, Di Re J, Dvorak NM, Alshammari M, Laezza F. Inhibition of the Akt/PKB Kinase Increases Na v1.6-Mediated Currents and Neuronal Excitability in CA1 Hippocampal Pyramidal Neurons. Int J Mol Sci 2022; 23:ijms23031700. [PMID: 35163623 PMCID: PMC8836202 DOI: 10.3390/ijms23031700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
In neurons, changes in Akt activity have been detected in response to the stimulation of transmembrane receptors. However, the mechanisms that lead to changes in neuronal function upon Akt inhibition are still poorly understood. In the present study, we interrogate how Akt inhibition could affect the activity of the neuronal Nav channels with while impacting intrinsic excitability. To that end, we employed voltage-clamp electrophysiological recordings in heterologous cells expressing the Nav1.6 channel isoform and in hippocampal CA1 pyramidal neurons in the presence of triciribine, an inhibitor of Akt. We showed that in both systems, Akt inhibition resulted in a potentiation of peak transient Na+ current (INa) density. Akt inhibition correspondingly led to an increase in the action potential firing of the CA1 pyramidal neurons that was accompanied by a decrease in the action potential current threshold. Complementary confocal analysis in the CA1 pyramidal neurons showed that the inhibition of Akt is associated with the lengthening of Nav1.6 fluorescent intensity along the axonal initial segment (AIS), providing a mechanism for augmented neuronal excitability. Taken together, these findings provide evidence that Akt-mediated signal transduction might affect neuronal excitability in a Nav1.6-dependent manner.
Collapse
Affiliation(s)
- Mate Marosi
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
| | - Miroslav N. Nenov
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
| | - Jessica Di Re
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
| | - Nolan M. Dvorak
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
| | - Musaad Alshammari
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
- Department of Pharmacology, College of Pharmacy, King Saud University, Riyadh P.O. Box 145111, Saudi Arabia
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA; (M.M.); (M.N.N.); (J.D.R.); (N.M.D.); (M.A.)
- Center for Addiction Research, Center for Biomedical Engineering and Mitchell, Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
- Correspondence: ; Tel.: +1-(409)-772-9672; Fax: +1-(409)-772-9642
| |
Collapse
|
34
|
Radulovic J, Ivkovic S, Adzic M. From chronic stress and anxiety to neurodegeneration: Focus on neuromodulation of the axon initial segment. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:481-495. [PMID: 35034756 DOI: 10.1016/b978-0-12-819410-2.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To adapt to the sustained demands of chronic stress, discrete brain circuits undergo structural and functional changes often resulting in anxiety disorders. In some individuals, anxiety disorders precede the development of motor symptoms of Parkinson's disease (PD) caused by degeneration of neurons in the substantia nigra (SN). Here, we present a circuit framework for probing a causal link between chronic stress, anxiety, and PD, which postulates a central role of abnormal neuromodulation of the SN's axon initial segment by brainstem inputs. It is grounded in findings demonstrating that the earliest PD pathologies occur in the stress-responsive, emotion regulation network of the brainstem, which provides the SN with dense aminergic and cholinergic innervation. SN's axon initial segment (AIS) has unique features that support the sustained and bidirectional propagation of activity in response to synaptic inputs. It is therefore, especially sensitive to circuit-mediated stress-induced imbalance of neuromodulation, and thus a plausible initiating site of neurodegeneration. This could explain why, although secondary to pathophysiologies in other brainstem nuclei, SN degeneration is the most extensive. Consequently, the cardinal symptom of PD, severe motor deficits, arise from degeneration of the nigrostriatal pathway rather than other brainstem nuclei. Understanding when and how circuit dysfunctions underlying anxiety can progress to neurodegeneration, raises the prospect of timed interventions for reversing, or at least impeding, the early pathophysiologies that lead to PD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Neuroscience, Albert Einstein Medical College, Bronx, NY, United States; Department of Psychiatry and Behavioral Sciences, Albert Einstein Medical College, Bronx, NY, United States.
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
35
|
Integrative genomic analysis of PPP3R1 in Alzheimer's disease: a potential biomarker for predictive, preventive, and personalized medical approach. EPMA J 2021; 12:647-658. [PMID: 34956428 DOI: 10.1007/s13167-021-00261-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is associated with abnormal calcium signaling, a pathway regulated by the calcium-dependent protein phosphatase. This study aimed to investigate the molecular function of protein phosphatase 3 regulatory subunit B (PPP3R1) underlying AD, which may provide novel insights for the predictive diagnostics, targeted prevention, and personalization of medical services in AD by targeting PPP3R1. A total of 1860 differentially expressed genes (DEGs) from 13,049 background genes were overlapped in AD/control and PPP3R1-low/high cohorts. Based on these DEGs, six co-expression modules were constructed by weight gene correlation network analysis (WGCNA). The turquoise module had the strongest correlation with AD and low PPP3R1, in which DEGs participated in axon guidance, glutamatergic synapse, long-term potentiation (LTP), mitogen-activated protein kinase (MAPK), Ras, and hypoxia-inducible factor 1 (HIF-1) signaling pathways. Furthermore, the cross-talking pathways of PPP3R1, such as axon guidance, glutamatergic synapse, LTP, and MAPK signaling pathways, were identified in the global regulatory network. The area under the curve (AUC) analysis showed that low PPP3R1 could accurately predict the onset of AD. Therefore, our findings highlight the involvement of PPP3R1 in the pathogenesis of AD via axon guidance, glutamatergic synapse, LTP, and MAPK signaling pathways, and identify downregulation of PPP3R1 as a potential biomarker for AD treatment in the context of 3P medicine-predictive diagnostics, targeted prevention, and personalization of medical services. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-021-00261-2.
Collapse
|
36
|
Usui N, Tian X, Harigai W, Togawa S, Utsunomiya R, Doi T, Miyoshi K, Shinoda K, Tanaka J, Shimada S, Katayama T, Yoshimura T. Length impairments of the axon initial segment in rodent models of attention-deficit hyperactivity disorder and autism spectrum disorder. Neurochem Int 2021; 153:105273. [PMID: 34971749 DOI: 10.1016/j.neuint.2021.105273] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022]
Abstract
The axon initial segment (AIS) is a structural neuronal compartment of the proximal axon that plays key roles in sodium channel clustering, action potential initiation, and signal propagation of neuronal outputs. Mutations in constitutive genes of the AIS, such as ANK3, have been identified in patients with neurodevelopmental disorders. Nevertheless, morphological changes in the AIS in neurodevelopmental disorders have not been characterized. In this study, we investigated the length of the AIS in rodent models of attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). We observed abnormalities in AIS length in both animal models. In ADHD model rodents, we observed shorter AIS length in layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC) and primary somatosensory barrel field (S1BF). Further, we observed shorter AIS length in S1BF L5 neurons. In ASD model mice, we observed shorter AIS length in L2/3 and L5 neurons of the S1BF. These results suggest that impairments in AIS length are common phenomena in neurodevelopmental disorders such as ADHD and ASD and may be conserved across species. Our findings provide novel insight into the potential contribution of the AIS to the pathophysiology and pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan.
| | - Xiaoye Tian
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Wakana Harigai
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Ryo Utsunomiya
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Tomomi Doi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Ko Miyoshi
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Takeshi Yoshimura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
37
|
Raghuram V, Werginz P, Fried SI, Timko BP. Morphological Factors that Underlie Neural Sensitivity to Stimulation in the Retina. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100069. [PMID: 35399546 PMCID: PMC8993153 DOI: 10.1002/anbr.202100069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Retinal prostheses are a promising therapeutic intervention for patients afflicted by outer retinal degenerative diseases like retinitis pigmentosa and age-related macular degeneration. While significant advances in the development of retinal implants have been made, the quality of vision elicited by these devices remains largely sub-optimal. The variability in the responses produced by retinal devices is most likely due to the differences between the natural cell type-specific signaling that occur in the healthy retina vs. the non-specific activation of multiple cell types arising from artificial stimulation. In order to replicate these natural signaling patterns, stimulation strategies must be capable of preferentially activating specific RGC types. To design more selective stimulation strategies, a better understanding of the morphological factors that underlie the sensitivity to prosthetic stimulation must be developed. This review will focus on the role that different anatomical components play in driving the direct activation of RGCs by extracellular stimulation. Briefly, it will (1) characterize the variability in morphological properties of α-RGCs, (2) detail the influence of morphology on the direct activation of RGCs by electric stimulation, and (3) describe some of the potential biophysical mechanisms that could explain differences in activation thresholds and electrically evoked responses between RGC types.
Collapse
Affiliation(s)
- Vineeth Raghuram
- Boston VA Healthcare System, 150 S Huntington Ave, Boston, MA 02130, USA
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Paul Werginz
- Institute for Analysis and Scientific Computing, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna, Austria
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Shelley I. Fried
- Boston VA Healthcare System, 150 S Huntington Ave, Boston, MA 02130, USA
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
- Dept. of Neurosurgery, Massachusetts General Hospital - Harvard Medical School, 50 Blossom Street, Boston, MA, 02114
| | - Brian P. Timko
- Dept. of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
38
|
Rotterman TM, Carrasco DI, Housley SN, Nardelli P, Powers RK, Cope TC. Axon initial segment geometry in relation to motoneuron excitability. PLoS One 2021; 16:e0259918. [PMID: 34797870 PMCID: PMC8604372 DOI: 10.1371/journal.pone.0259918] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
The axon initial segment (AIS) responsible for action potential initiation is a dynamic structure that varies and changes together with neuronal excitability. Like other neuron types, alpha motoneurons in the mammalian spinal cord express heterogeneity and plasticity in AIS geometry, including length (AISl) and distance from soma (AISd). The present study aimed to establish the relationship of AIS geometry with a measure of intrinsic excitability, rheobase current, that varies by 20-fold or more among normal motoneurons. We began by determining whether AIS length or distance differed for motoneurons in motor pools that exhibit different activity profiles. Motoneurons sampled from the medial gastrocnemius (MG) motor pool exhibited values for average AISd that were significantly greater than that for motoneurons from the soleus (SOL) motor pool, which is more readily recruited in low-level activities. Next, we tested whether AISd covaried with intrinsic excitability of individual motoneurons. In anesthetized rats, we measured rheobase current intracellularly from MG motoneurons in vivo before labeling them for immunohistochemical study of AIS structure. For 16 motoneurons sampled from the MG motor pool, this combinatory approach revealed that AISd, but not AISl, was significantly related to rheobase, as AIS tended to be located further from the soma on motoneurons that were less excitable. Although a causal relation with excitability seems unlikely, AISd falls among a constellation of properties related to the recruitability of motor units and their parent motoneurons.
Collapse
Affiliation(s)
- Travis M. Rotterman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- * E-mail: (TMR); (TCC)
| | - Darío I. Carrasco
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Stephen N. Housley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Paul Nardelli
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Randall K. Powers
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States of America
| | - Timothy C. Cope
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
- W.H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States of America
- * E-mail: (TMR); (TCC)
| |
Collapse
|
39
|
A general principle of dendritic constancy: A neuron's size- and shape-invariant excitability. Neuron 2021; 109:3647-3662.e7. [PMID: 34555313 DOI: 10.1016/j.neuron.2021.08.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/29/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022]
Abstract
Reducing neuronal size results in less membrane and therefore lower input conductance. Smaller neurons are thus more excitable, as seen in their responses to somatic current injections. However, the impact of a neuron's size and shape on its voltage responses to dendritic synaptic activation is much less understood. Here we use analytical cable theory to predict voltage responses to distributed synaptic inputs in unbranched cables, showing that these are entirely independent of dendritic length. For a given synaptic density, neuronal responses depend only on the average dendritic diameter and intrinsic conductivity. This remains valid for a wide range of morphologies irrespective of their arborization complexity. Spiking models indicate that morphology-invariant numbers of spikes approximate the percentage of active synapses. In contrast to spike rate, spike times do depend on dendrite morphology. In summary, neuronal excitability in response to distributed synaptic inputs is largely unaffected by dendrite length or complexity.
Collapse
|
40
|
Chang CW, Evans MD, Yu X, Yu GQ, Mucke L. Tau reduction affects excitatory and inhibitory neurons differently, reduces excitation/inhibition ratios, and counteracts network hypersynchrony. Cell Rep 2021; 37:109855. [PMID: 34686344 PMCID: PMC8648275 DOI: 10.1016/j.celrep.2021.109855] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/04/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022] Open
Abstract
The protein tau has been implicated in many brain disorders. In animal models, tau reduction suppresses epileptogenesis of diverse causes and ameliorates synaptic and behavioral abnormalities in various conditions associated with excessive excitation-inhibition (E/I) ratios. However, the underlying mechanisms are unknown. Global genetic ablation of tau in mice reduces the action potential (AP) firing and E/I ratio of pyramidal cells in acute cortical slices without affecting the excitability of these cells. Tau ablation reduces the excitatory inputs to inhibitory neurons, increases the excitability of these cells, and structurally alters their axon initial segments (AISs). In primary neuronal cultures subjected to prolonged overstimulation, tau ablation diminishes the homeostatic response of AISs in inhibitory neurons, promotes inhibition, and suppresses hypersynchrony. Together, these differential alterations in excitatory and inhibitory neurons help explain how tau reduction prevents network hypersynchrony and counteracts brain disorders causing abnormally increased E/I ratios.
Collapse
Affiliation(s)
- Che-Wei Chang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mark D Evans
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Xinxing Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
41
|
The Type 2 Diabetes Factor Methylglyoxal Mediates Axon Initial Segment Shortening and Alters Neuronal Function at the Cellular and Network Levels. eNeuro 2021; 8:ENEURO.0201-21.2021. [PMID: 34531281 PMCID: PMC8496204 DOI: 10.1523/eneuro.0201-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023] Open
Abstract
Recent evidence suggests that alteration of axon initial segment (AIS) geometry (i.e., length or location along the axon) contributes to CNS dysfunction in neurological diseases. For example, AIS length is shorter in the prefrontal cortex of type 2 diabetic mice with cognitive impairment. To determine the key type 2 diabetes-related factor that produces AIS shortening we modified levels of insulin, glucose, or the reactive glucose metabolite methylglyoxal in cultures of dissociated cortices from male and female mice and quantified AIS geometry using immunofluorescent imaging of the AIS proteins AnkyrinG and βIV spectrin. Neither insulin nor glucose modification altered AIS length. Exposure to 100 but not 1 or 10 μm methylglyoxal for 24 h resulted in accumulation of the methylglyoxal-derived advanced glycation end-product hydroimidazolone and produced reversible AIS shortening without cell death. Methylglyoxal-evoked AIS shortening occurred in both excitatory and putative inhibitory neuron populations and in the presence of tetrodotoxin (TTX). In single-cell recordings resting membrane potential was depolarized at 0.5-3 h and returned to normal at 24 h. In multielectrode array (MEA) recordings methylglyoxal produced an immediate ∼300% increase in spiking and bursting rates that returned to normal within 2 min, followed by a ∼20% reduction of network activity at 0.5-3 h and restoration of activity to baseline levels at 24 h. AIS length was unchanged at 0.5-3 h despite the presence of depolarization and network activity reduction. Nevertheless, these results suggest that methylglyoxal could be a key mediator of AIS shortening and disruptor of neuronal function during type 2 diabetes.
Collapse
|
42
|
Neural excitability increases with axonal resistance between soma and axon initial segment. Proc Natl Acad Sci U S A 2021; 118:2102217118. [PMID: 34389672 DOI: 10.1073/pnas.2102217118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The position of the axon initial segment (AIS) is thought to play a critical role in neuronal excitability. Previous experimental studies have found that a distal shift in AIS position correlates with a reduction in excitability. Yet theoretical work has suggested the opposite, because of increased electrical isolation. A distal shift in AIS position corresponds to an elevation of axial resistance R a We therefore examined how changes in R a at the axon hillock impact the voltage threshold (Vth) of the somatic action potential in L5 pyramidal neurons. Increasing R a by mechanically pinching the axon between the soma and the AIS was found to lower Vth by ∼6 mV. Conversely, decreasing R a by substituting internal ions with higher mobility elevated Vth All R a -dependent changes in Vth could be reproduced in a Hodgkin-Huxley compartmental model. We conclude that in L5 pyramidal neurons, excitability increases with axial resistance and therefore with a distal shift of the AIS.
Collapse
|
43
|
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
|
44
|
Quistgaard EM, Nissen JD, Hansen S, Nissen P. Mind the Gap: Molecular Architecture of the Axon Initial Segment - From Fold Prediction to a Mechanistic Model of Function? J Mol Biol 2021; 433:167176. [PMID: 34303720 DOI: 10.1016/j.jmb.2021.167176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 11/28/2022]
Abstract
The axon initial segment (AIS) is a distinct neuronal domain, which is responsible for initiating action potentials, and therefore of key importance to neuronal signaling. To determine how it functions, it is necessary to establish which proteins reside there, how they are organized, and what the dynamic features are. Great strides have been made in recent years, and it is now clear that several AIS cytoskeletal and membrane proteins interact to form a higher-order periodic structure. Here we briefly describe AIS function, protein composition and molecular architecture, and discuss perspectives for future structural characterization, and if structure predictions will be able to model complex higher-order assemblies.
Collapse
Affiliation(s)
- Esben M Quistgaard
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Dept. Molecular Biology and Genetics, DK - 8000 Aarhus C, Denmark
| | - Josephine Dannersø Nissen
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Dept. Molecular Biology and Genetics, DK - 8000 Aarhus C, Denmark
| | - Sean Hansen
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Dept. Molecular Biology and Genetics, DK - 8000 Aarhus C, Denmark
| | - Poul Nissen
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Dept. Molecular Biology and Genetics, DK - 8000 Aarhus C, Denmark.
| |
Collapse
|
45
|
Plasmalogens regulate the AKT-ULK1 signaling pathway to control the position of the axon initial segment. Prog Neurobiol 2021; 205:102123. [PMID: 34302896 DOI: 10.1016/j.pneurobio.2021.102123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/11/2021] [Accepted: 07/14/2021] [Indexed: 01/04/2023]
Abstract
The axon initial segment (AIS) is a specialized region in neurons that encompasses two essential functions, the generation of action potentials and the regulation of the axodendritic polarity. The mechanism controlling the position of the axon initial segment to allow plasticity and regulation of neuron excitability is unclear. Here we demonstrate that plasmalogens, the most abundant ether-phospholipid, are essential for the homeostatic positioning of the AIS. Plasmalogen deficiency is a hallmark of Rhizomelic Chondrodysplasia Punctata (RCDP) and Zellweger spectrum disorders, but Alzheimer's and Parkinson's disease, are also characterized by plasmalogen defects. Neurons lacking plasmalogens displaced the AIS to more distal positions and were characterized by reduced excitability. Treatment with a short-chain alkyl glycerol was able to rescue AIS positioning. Plasmalogen deficiency impaired AKT activation, and we show that inhibition of AKT phosphorylation at Ser473 and Thr308 is sufficient to induce a distal relocation of the AIS. Pathway analysis revealed that downstream of AKT, overtly active ULK1 mediates AIS repositioning. Rescuing the impaired AKT signaling pathway was able to normalize AIS position independently of the biochemical defect. These results unveil a previously unknown mechanism that couples the phospholipid composition of the neuronal membrane to the positional assembly of the AIS.
Collapse
|
46
|
Bülow P, Wenner PA, Faundez V, Bassell GJ. Mitochondrial Structure and Polarity in Dendrites and the Axon Initial Segment Are Regulated by Homeostatic Plasticity and Dysregulated in Fragile X Syndrome. Front Cell Dev Biol 2021; 9:702020. [PMID: 34350185 PMCID: PMC8327182 DOI: 10.3389/fcell.2021.702020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction has long been overlooked in neurodevelopmental disorders, but recent studies have provided new links to genetic forms of autism, including Rett syndrome and fragile X syndrome (FXS). Mitochondria show plasticity in morphology and function in response to neuronal activity, and previous research has reported impairments in mitochondrial morphology and function in disease. We and others have previously reported abnormalities in distinct types of homeostatic plasticity in FXS. It remains unknown if or how activity deprivation triggering homeostatic plasticity affects mitochondria in axons and/or dendrites and whether impairments occur in neurodevelopmental disorders. Here, we test the hypothesis that mitochondria are structurally and functionally modified in a compartment-specific manner during homeostatic plasticity using a model of activity deprivation in cortical neurons from wild-type mice and that this plasticity-induced regulation is altered in Fmr1-knockout (KO) neurons. We uncovered dendrite-specific regulation of the mitochondrial surface area, whereas axon initial segment (AIS) mitochondria show changes in polarity; both responses are lost in the Fmr1 KO. Taken together, our results demonstrate impairments in mitochondrial plasticity in FXS, which has not previously been reported. These results suggest that mitochondrial dysregulation in FXS could contribute to abnormal neuronal plasticity, with broader implications to other neurodevelopmental disorders and therapeutic strategies.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Peter A Wenner
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
47
|
Purkinje cell axonal swellings enhance action potential fidelity and cerebellar function. Nat Commun 2021; 12:4129. [PMID: 34226561 PMCID: PMC8257784 DOI: 10.1038/s41467-021-24390-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/11/2021] [Indexed: 11/08/2022] Open
Abstract
Axonal plasticity allows neurons to control their output, which critically determines the flow of information in the brain. Axon diameter can be regulated by activity, yet how morphological changes in an axon impact its function remains poorly understood. Axonal swellings have been found on Purkinje cell axons in the cerebellum both in healthy development and in neurodegenerative diseases, and computational models predicts that axonal swellings impair axonal function. Here we report that in young Purkinje cells, axons with swellings propagated action potentials with higher fidelity than those without, and that axonal swellings form when axonal failures are high. Furthermore, we observed that healthy young adult mice with more axonal swellings learn better on cerebellar-related tasks than mice with fewer swellings. Our findings suggest that axonal swellings underlie a form of axonal plasticity that optimizes the fidelity of action potential propagation in axons, resulting in enhanced learning.
Collapse
|
48
|
Di Re J, Hsu WCJ, Kayasandik CB, Fularczyk N, James TF, Nenov MN, Negi P, Marosi M, Scala F, Prasad S, Labate D, Laezza F. Inhibition of AKT Signaling Alters βIV Spectrin Distribution at the AIS and Increases Neuronal Excitability. Front Mol Neurosci 2021; 14:643860. [PMID: 34276302 PMCID: PMC8278006 DOI: 10.3389/fnmol.2021.643860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 05/27/2021] [Indexed: 11/24/2022] Open
Abstract
The axon initial segment (AIS) is a highly regulated subcellular domain required for neuronal firing. Changes in the AIS protein composition and distribution are a form of structural plasticity, which powerfully regulates neuronal activity and may underlie several neuropsychiatric and neurodegenerative disorders. Despite its physiological and pathophysiological relevance, the signaling pathways mediating AIS protein distribution are still poorly studied. Here, we used confocal imaging and whole-cell patch clamp electrophysiology in primary hippocampal neurons to study how AIS protein composition and neuronal firing varied in response to selected kinase inhibitors targeting the AKT/GSK3 pathway, which has previously been shown to phosphorylate AIS proteins. Image-based features representing the cellular pattern distribution of the voltage-gated Na+ (Nav) channel, ankyrin G, βIV spectrin, and the cell-adhesion molecule neurofascin were analyzed, revealing βIV spectrin as the most sensitive AIS protein to AKT/GSK3 pathway inhibition. Within this pathway, inhibition of AKT by triciribine has the greatest effect on βIV spectrin localization to the AIS and its subcellular distribution within neurons, a phenotype that Support Vector Machine classification was able to accurately distinguish from control. Treatment with triciribine also resulted in increased excitability in primary hippocampal neurons. Thus, perturbations to signaling mechanisms within the AKT pathway contribute to changes in βIV spectrin distribution and neuronal firing that may be associated with neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jessica Di Re
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Wei-Chun J. Hsu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
- Biochemistry and Molecular Biology Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, Galveston, TX, United States
- M.D./Ph.D. Combined Degree Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, Galveston, TX, United States
| | - Cihan B. Kayasandik
- Department of Mathematics, University of Houston, Houston, TX, United States
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey
| | - Nickolas Fularczyk
- Department of Mathematics, University of Houston, Houston, TX, United States
| | - T. F. James
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Miroslav N. Nenov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Pooran Negi
- Department of Mathematics, University of Houston, Houston, TX, United States
| | - Mate Marosi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Federico Scala
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Saurabh Prasad
- Department of Electrical and Computer Engineering, University of Houston, Houston, TX, United States
| | - Demetrio Labate
- Department of Mathematics, University of Houston, Houston, TX, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
49
|
Coviello S, Benedetti B, Jakubecova D, Belles M, Klimczak P, Gramuntell Y, Couillard-Despres S, Nacher J. PSA Depletion Induces the Differentiation of Immature Neurons in the Piriform Cortex of Adult Mice. Int J Mol Sci 2021; 22:5733. [PMID: 34072166 PMCID: PMC8198564 DOI: 10.3390/ijms22115733] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Immature neurons are maintained in cortical regions of the adult mammalian brain. In rodents, many of these immature neurons can be identified in the piriform cortex based on their high expression of early neuronal markers, such as doublecortin (DCX) and the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). This molecule plays critical roles in different neurodevelopmental events. Taking advantage of a DCX-CreERT2/Flox-EGFP reporter mice, we investigated the impact of targeted PSA enzymatic depletion in the piriform cortex on the fate of immature neurons. We report here that the removal of PSA accelerated the final development of immature neurons. This was revealed by a higher frequency of NeuN expression, an increase in the number of cells carrying an axon initial segment (AIS), and an increase in the number of dendrites and dendritic spines on the immature neurons. Taken together, our results demonstrated the crucial role of the PSA moiety in the protracted development of immature neurons residing outside of the neurogenic niches. More studies will be required to understand the intrinsic and extrinsic factors affecting PSA-NCAM expression to understand how the brain regulates the incorporation of these immature neurons to the established neuronal circuits of the adult brain.
Collapse
Affiliation(s)
- Simona Coviello
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100 Burjassot, Spain; (S.C.); (M.B.); (P.K.); (Y.G.)
| | - Bruno Benedetti
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (D.J.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Dominika Jakubecova
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (D.J.)
| | - Maria Belles
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100 Burjassot, Spain; (S.C.); (M.B.); (P.K.); (Y.G.)
| | - Patrycja Klimczak
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100 Burjassot, Spain; (S.C.); (M.B.); (P.K.); (Y.G.)
| | - Yaiza Gramuntell
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100 Burjassot, Spain; (S.C.); (M.B.); (P.K.); (Y.G.)
| | - Sebastien Couillard-Despres
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (D.J.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100 Burjassot, Spain; (S.C.); (M.B.); (P.K.); (Y.G.)
- Spanish National Network for Research in Mental Health (CIBERSAM), 28029 Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
50
|
Damenti M, Coceano G, Pennacchietti F, Bodén A, Testa I. STED and parallelized RESOLFT optical nanoscopy of the tubular endoplasmic reticulum and its mitochondrial contacts in neuronal cells. Neurobiol Dis 2021; 155:105361. [PMID: 33857635 DOI: 10.1016/j.nbd.2021.105361] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 01/19/2023] Open
Abstract
The classic view of organelle cell biology is undergoing a constant revision fueled by the new insights unraveled by fluorescence nanoscopy, which enable sensitive, faster and gentler observation of specific proteins in situ. The endoplasmic reticulum (ER) is one of the most challenging structure to capture due the rapid and constant restructuring of fine sheets and tubules across the full 3D cell volume. Here we apply STED and parallelized 2D and 3D RESOLFT live imaging to uncover the tubular ER organization in the fine processes of neuronal cells with focus on mitochondria-ER contacts, which recently gained medical attention due to their role in neurodegeneration. Multi-color STED nanoscopy enables the simultaneous visualization of small transversal ER tubules crossing and constricting mitochondria all along axons and dendrites. Parallelized RESOLFT allows for dynamic studies of multiple contact sites within seconds and minutes with prolonged time-lapse imaging at ~50 nm spatial resolution. When operated in 3D super resolution mode it enables a new isotropic visualization of such contacts extending our understanding of the three-dimensional architecture of these packed structures in axons and dendrites.
Collapse
Affiliation(s)
- Martina Damenti
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Giovanna Coceano
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Francesca Pennacchietti
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Andreas Bodén
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden.
| |
Collapse
|