1
|
Wang H, Li Y, Li X, Sun Z, Yu F, Pashang A, Kulasiri D, Li HW, Chen H, Hou H, Zhang Y. The Primary Cilia are Associated with the Axon Initial Segment in Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2407405. [PMID: 39804991 DOI: 10.1002/advs.202407405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/16/2024] [Indexed: 01/16/2025]
Abstract
The primary cilia serve as pivotal mediators of environmental signals and play crucial roles in neuronal responses. Disruption of ciliary function has been implicated in neuronal circuit disorders and aberrant neuronal excitability. However, the precise mechanisms remain elusive. To study the link between the primary cilia and neuronal excitability, manipulation of somatostatin receptor 3 (SSTR3) is investigated, as an example of how alterations in ciliary signaling may affect neuronal activity. It is found that aberrant SSTR3 expression perturbed not only ciliary morphology but also disrupted ciliary signaling cascades. Genetic deletion of SSTR3 resulted in perturbed spatial memory and synaptic plasticity. The axon initial segment (AIS) is a specialized region in the axon where action potentials are initiated. Interestingly, loss of ciliary SSTR3 led to decrease of Akt-dependent cyclic AMP-response element binding protein (CREB)-mediated transcription at the AIS, specifically downregulating AIS master organizer adaptor protein ankyrin G (AnkG) expression. In addition, alterations of other ciliary proteins serotonin 6 receptor (5-HT6R)and intraflagellar transport protein 88 (IFT88) also induced length changes of the AIS. The findings elucidate a specific interaction between the primary cilia and AIS, providing insight into the impact of the primary cilia on neuronal excitability and circuit integrity.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yu Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xin Li
- Beijing Life Science Academy, Beijing, 102200, China
| | - Zehui Sun
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Fengdan Yu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C-fACS), AGLS faculty, Lincoln University, Canterbury, 7647, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), AGLS faculty, Lincoln University, Canterbury, 7647, New Zealand
| | - Hung Wing Li
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Huan Chen
- Beijing Life Science Academy, Beijing, 102200, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing, 102200, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| |
Collapse
|
2
|
Eichel K. Endocytosis in the axon initial segment: Roles in neuronal polarity and plasticity. Curr Opin Neurobiol 2024; 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] [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
|
3
|
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
|
4
|
Liu Y, Xia D, Zhong L, Chen L, Zhang L, Ai M, Mei R, Pang R. Casein Kinase 2 Affects Epilepsy by Regulating Ion Channels: A Potential Mechanism. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:894-905. [PMID: 37350003 DOI: 10.2174/1871527322666230622124618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 06/24/2023]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal brain discharges, is the third most common chronic disorder of the Central Nervous System (CNS). Although significant progress has been made in the research on antiepileptic drugs (AEDs), approximately one-third of patients with epilepsy are refractory to these drugs. Thus, research on the pathogenesis of epilepsy is ongoing to find more effective treatments. Many pathological mechanisms are involved in epilepsy, including neuronal apoptosis, mossy fiber sprouting, neuroinflammation, and dysfunction of neuronal ion channels, leading to abnormal neuronal excitatory networks in the brain. CK2 (Casein kinase 2), which plays a critical role in modulating neuronal excitability and synaptic transmission, has been shown to be associated with epilepsy. However, there is limited research on the mechanisms involved. Recent studies have suggested that CK2 is involved in regulating the function of neuronal ion channels by directly phosphorylating them or their binding partners. Therefore, in this review, we will summarize recent research advances regarding the potential role of CK2 regulating ion channels in epilepsy, aiming to provide more evidence for future studies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Di Xia
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lianmei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ling Chen
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan, 650032, China
| | - Linming Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Mingda Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Rong Mei
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650034, China
| | - Ruijing Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
5
|
Needs HI, Wilkinson KA, Henley JM, Collinson I. Aggregation-prone Tau impairs mitochondrial import, which affects organelle morphology and neuronal complexity. J Cell Sci 2023; 136:jcs260993. [PMID: 37303235 PMCID: PMC10357015 DOI: 10.1242/jcs.260993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Mitochondrial protein import is essential for organellar biogenesis, and thereby for the sufficient supply of cytosolic ATP - which is particularly important for cells with high energy demands like neurons. This study explores the prospect of import machinery perturbation as a cause of neurodegeneration instigated by the accumulation of aggregating proteins linked to disease. We found that the aggregation-prone Tau variant (TauP301L) reduces the levels of components of the import machinery of the outer (TOM20, encoded by TOMM20) and inner membrane (TIM23, encoded by TIMM23) while associating with TOM40 (TOMM40). Intriguingly, this interaction affects mitochondrial morphology, but not protein import or respiratory function; raising the prospect of an intrinsic rescue mechanism. Indeed, TauP301L induced the formation of tunnelling nanotubes (TNTs), potentially for the recruitment of healthy mitochondria from neighbouring cells and/or the disposal of mitochondria incapacitated by aggregated Tau. Consistent with this, inhibition of TNT formation (and rescue) reveals Tau-induced import impairment. In primary neuronal cultures, TauP301L induced morphological changes characteristic of neurodegeneration. Interestingly, these effects were mirrored in cells where the import sites were blocked artificially. Our results reveal a link between aggregation-prone Tau and defective mitochondrial import relevant to disease.
Collapse
Affiliation(s)
- Hope I. Needs
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | - Jeremy M. Henley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
6
|
Compans B, Burrone J. Chandelier cells shine a light on the formation of GABAergic synapses. Curr Opin Neurobiol 2023; 80:102697. [PMID: 36907075 PMCID: PMC10682383 DOI: 10.1016/j.conb.2023.102697] [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: 11/09/2022] [Accepted: 02/05/2023] [Indexed: 03/12/2023]
Abstract
Uncovering the wiring rules employed by neurons during development represents a formidable challenge with important repercussions for neurodevelopmental disorders. Chandelier cells (ChCs) are a singular GABAergic interneuron type, with a unique morphology, that have recently begun to shed light on the rules that drive the formation and plasticity of inhibitory synapses. This review will focus on the wealth of recent data charting the emergence of synapses formed by ChCs onto pyramidal cells, from the molecules involved to the plasticity of these connections during development.
Collapse
Affiliation(s)
- Benjamin Compans
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Hospital Campus, London, SE1 1UL, London, UK. https://twitter.com/jbneuro
| | - Juan Burrone
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Hospital Campus, London, SE1 1UL, London, UK.
| |
Collapse
|
7
|
Ma F, Akolkar H, Xu J, Liu Y, Popova D, Xie J, Youssef MM, Benosman R, Hart RP, Herrup K. The Amyloid Precursor Protein Modulates the Position and Length of the Axon Initial Segment. J Neurosci 2023; 43:1830-1844. [PMID: 36717226 PMCID: PMC10010458 DOI: 10.1523/jneurosci.0172-22.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The amyloid precursor protein (APP) is linked to the genetics and pathogenesis of Alzheimer's disease (AD). It is the parent protein of the β-amyloid (Aβ) peptide, the main constituent of the amyloid plaques found in an AD brain. The pathways from APP to Aβ are intensively studied, yet the normal functions of APP itself have generated less interest. We report here that glutamate stimulation of neuronal activity leads to a rapid increase in App gene expression. In mouse and human neurons, elevated APP protein changes the structure of the axon initial segment (AIS) where action potentials are initiated. The AIS is shortened in length and shifts away from the cell body. The GCaMP8f Ca2+ reporter confirms the predicted decrease in neuronal activity. NMDA antagonists or knockdown of App block the glutamate effects. The actions of APP on the AIS are cell-autonomous; exogenous Aβ, either fibrillar or oligomeric, has no effect. In culture, APPSwe (a familial AD mutation) induces larger AIS changes than wild type APP. Ankyrin G and βIV-spectrin, scaffolding proteins of the AIS, both physically associate with APP, more so in AD brains. Finally, in humans with sporadic AD or in the R1.40 AD mouse model, both females and males, neurons have elevated levels of APP protein that invade the AIS. In vivo as in vitro, this increased APP is associated with a significant shortening of the AIS. The findings outline a new role for the APP and encourage a reconsideration of its relationship to AD.SIGNIFICANCE STATEMENT While the amyloid precursor protein (APP) has long been associated with Alzheimer's disease (AD), the normal functions of the full-length Type I membrane protein have been largely unexplored. We report here that the levels of APP protein increase with neuronal activity. In vivo and in vitro, modest amounts of excess APP alter the properties of the axon initial segment. The β-amyloid peptide derived from APP is without effect. Consistent with the observed changes in the axon initial segment which would be expected to decrease action potential firing, we show that APP expression depresses neuronal activity. In mouse AD models and human sporadic AD, APP physically associates with the scaffolding proteins of the axon initial segment, suggesting a relationship with AD dementia.
Collapse
Affiliation(s)
- Fulin Ma
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Himanshu Akolkar
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Jianquan Xu
- Departments of Medicine and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Yang Liu
- Departments of Medicine and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Dina Popova
- Human Genetics Institute, Rutgers University, Piscataway, NJ 08854
| | - Jiaan Xie
- Departments of Medicine and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Mark M Youssef
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Ryad Benosman
- Robotics Institute, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Ronald P Hart
- Human Genetics Institute, Rutgers University, Piscataway, NJ 08854
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
- Departments of Medicine and Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
8
|
Dorrego-Rivas A, Ezan J, Moreau MM, Poirault-Chassac S, Aubailly N, De Neve J, Blanchard C, Castets F, Fréal A, Battefeld A, Sans N, Montcouquiol M. The core PCP protein Prickle2 regulates axon number and AIS maturation by binding to AnkG and modulating microtubule bundling. SCIENCE ADVANCES 2022; 8:eabo6333. [PMID: 36083912 PMCID: PMC9462691 DOI: 10.1126/sciadv.abo6333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Core planar cell polarity (PCP) genes, which are involved in various neurodevelopmental disorders such as neural tube closure, epilepsy, and autism spectrum disorder, have poorly defined molecular signatures in neurons, mostly synapse-centric. Here, we show that the core PCP protein Prickle-like protein 2 (Prickle2) controls neuronal polarity and is a previously unidentified member of the axonal initial segment (AIS) proteome. We found that Prickle2 is present and colocalizes with AnkG480, the AIS master organizer, in the earliest stages of axonal specification and AIS formation. Furthermore, by binding to and regulating AnkG480, Prickle2 modulates its ability to bundle microtubules, a crucial mechanism for establishing neuronal polarity and AIS formation. Prickle2 depletion alters cytoskeleton organization, and Prickle2 levels determine both axon number and AIS maturation. Last, early Prickle2 depletion produces impaired action potential firing.
Collapse
Affiliation(s)
- Ana Dorrego-Rivas
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33077 Bordeaux, France
- Corresponding author.
| | - Jerome Ezan
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33077 Bordeaux, France
| | - Maïté M Moreau
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33077 Bordeaux, France
| | | | | | - Julie De Neve
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33077 Bordeaux, France
| | | | - Francis Castets
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, Case 907, 13288 Marseille Cedex 09, France
| | - Amélie Fréal
- Department of Functional Genomics, Vrije Universiteit (VU), Amsterdam, Netherlands
| | - Arne Battefeld
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Nathalie Sans
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33077 Bordeaux, France
- Corresponding author.
| | | |
Collapse
|
9
|
Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int J Mol Sci 2022; 23:ijms23084413. [PMID: 35457230 PMCID: PMC9028019 DOI: 10.3390/ijms23084413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3β is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3β is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3β acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3β. Here, we discuss the direct and indirect mechanisms by which GSK3β phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3β activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3β-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.
Collapse
|
10
|
Dorrego-Rivas A, Grubb MS. OUP accepted manuscript. Brain 2022; 145:1574-1575. [PMID: 35661860 PMCID: PMC9166549 DOI: 10.1093/brain/awac156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ana Dorrego-Rivas
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
| | - Matthew S. Grubb
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
- Correspondence to: Matthew S. Grubb E-mail:
| |
Collapse
|
11
|
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: 3] [Impact Index Per Article: 0.8] [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
|
12
|
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
|
13
|
Stone MC, Kothe GO, Rolls MM, Jegla T. Cytoskeletal and synaptic polarity of LWamide-like+ ganglion neurons in the sea anemone Nematostella vectensis. J Exp Biol 2020; 223:jeb233197. [PMID: 32968001 PMCID: PMC7673360 DOI: 10.1242/jeb.233197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022]
Abstract
The centralized nervous systems of bilaterian animals rely on directional signaling facilitated by polarized neurons with specialized axons and dendrites. It is not known whether axo-dendritic polarity is exclusive to bilaterians or was already present in early metazoans. We therefore examined neurite polarity in the starlet sea anemone Nematostella vectensis (Cnidaria). Cnidarians form a sister clade to bilaterians and share many neuronal building blocks characteristic of bilaterians, including channels, receptors and synaptic proteins, but their nervous systems comprise a comparatively simple net distributed throughout the body. We developed a tool kit of fluorescent polarity markers for live imaging analysis of polarity in an identified neuron type, large ganglion cells of the body column nerve net that express the LWamide-like neuropeptide. Microtubule polarity differs in bilaterian axons and dendrites, and this in part underlies polarized distribution of cargo to the two types of processes. However, in LWamide-like+ neurons, all neurites had axon-like microtubule polarity suggesting that they may have similar contents. Indeed, presynaptic and postsynaptic markers trafficked to all neurites and accumulated at varicosities where neurites from different neurons often crossed, suggesting the presence of bidirectional synaptic contacts. Furthermore, we could not identify a diffusion barrier in the plasma membrane of any of the neurites like the axon initial segment barrier that separates the axonal and somatodendritic compartments in bilaterian neurons. We conclude that at least one type of neuron in Nematostella vectensis lacks the axo-dendritic polarity characteristic of bilaterian neurons.
Collapse
Affiliation(s)
- Michelle C Stone
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gregory O Kothe
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Melissa M Rolls
- Department of Biochemistry and Molecular Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Timothy Jegla
- Department of Biology and the Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
14
|
Montenarh M, Götz C. Protein kinase CK2 and ion channels (Review). Biomed Rep 2020; 13:55. [PMID: 33082952 PMCID: PMC7560519 DOI: 10.3892/br.2020.1362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Protein kinase CK2 appears as a tetramer or higher molecular weight oligomer composed of catalytic CK2α, CK2α' subunits and non-catalytic regulatory CK2β subunits or as individual subunits. It is implicated in a variety of different regulatory processes, such as Akt signalling, splicing and DNA repair within eukaryotic cells. The present review evaluates the influence of CK2 on ion channels in the plasma membrane. CK2 phosphorylates platform proteins such as calmodulin and ankyrin G, which bind to channel proteins for a physiological transport to and positioning into the membrane. In addition, CK2 directly phosphorylates a variety of channel proteins directly to regulate opening and closing of the channels. Thus, modulation of CK2 activities by specific inhibitors, by siRNA technology or by CRISPR/Cas technology has an influence on intracellular ion concentrations and thereby on cellular signalling. The physiological regulation of the intracellular ion concentration is important for cell survival and correct intracellular signalling. Disturbance of this regulation results in a variety of different diseases including epilepsy, heart failure, cystic fibrosis and diabetes. Therefore, these effects should be considered when using CK2 inhibition as a treatment option for cancer.
Collapse
Affiliation(s)
- Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| |
Collapse
|
15
|
Werginz P, Raghuram V, Fried SI. The relationship between morphological properties and thresholds to extracellular electric stimulation in α RGCs. J Neural Eng 2020; 17:045015. [PMID: 32736374 DOI: 10.1088/1741-2552/abab47] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Retinal prostheses strive to restore vison to patients that are blind from retinal degeneration by electrically stimulating surviving retinal ganglion cells (RGCs). The quality of elicited percepts remains limited however and it is desirable to develop improved stimulation strategies. Here, we examine how the anatomical and biophysical properties of RGCs influence activation thresholds, including the effects of variations found naturally. APPROACH Detailed reconstructions were made of a large number of mouse α RGCs and were used to create an array of model cells; the models were used to study the effects of individual anatomical features on activation threshold to electric stimulation. Stimulation was delivered epiretinally from a point-source or disk electrode and consisted of monophasic or biphasic rectangular pulses. MAIN RESULTS Modeling results show that the region of minimum threshold always is within the axon initial segment (AIS). The properties of this region as well as the absolute value of the minimum threshold are dependent on the length of the AIS as well as on the relative composition of sodium channels within the AIS. Other morphological features, including cell size, dendritic field size and the distance between the AIS and the soma had only a minimal influence on thresholds. Introducing even a small number of low-threshold Nav1.6 channels into the AIS was sufficient to lower minimum thresholds substantially although further increases in Nav1.6 had diminishing effects. The distance between the AIS and the electrode affects threshold levels while alignment of the electrode with the axon or dendritic parts of the RGC can result in lower thresholds, even if the distance to the cell remains the same. SIGNIFICANCE Intrinsic morphological features can influence activation thresholds with the AIS having the strongest influence. However, the combined influence remains limited and may not be large enough to allow for selective activation between different RGC types.
Collapse
Affiliation(s)
- Paul Werginz
- Institute for Analysis and Scientific Computing, Vienna University of Technology, Vienna, Austria. Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America. Author to whom any correspondence should be addressed
| | | | | |
Collapse
|
16
|
Di Re J, Kayasandik C, Botello-Lins G, Labate D, Laezza F. Imaging of the Axon Initial Segment. ACTA ACUST UNITED AC 2020; 89:e78. [PMID: 31532918 DOI: 10.1002/cpns.78] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The axon initial segment (AIS) is the first 20- to 60-μm segment of the axon proximal to the soma of a neuron. This highly specialized subcellular domain is the initiation site of the action potential and contains a high concentration of voltage-gated ion channels held in place by a complex nexus of scaffolding and regulatory proteins that ensure proper electrical activity of the neuron. Studies have shown that dysfunction of many AIS channels and scaffolding proteins occurs in a variety of neuropsychiatric and neurodegenerative diseases, raising the need to develop accurate methods for visualization and quantification of the AIS and its protein content in models of normal and disease conditions. In this article, we describe methods for immunolabeling AIS proteins in cultured neurons and brain slices as well as methods for quantifying protein expression and pattern distribution using fluorescent labeling of these proteins. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Jessica Di Re
- Neuroscience Graduate Program, University of Texas Medical Branch, Galveston, Texas.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| | - Cihan Kayasandik
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey
| | - Gonzalo Botello-Lins
- Biotechnology Program, Clear Falls High School, Clear Creek Independent School District, League City, Texas
| | - Demetrio Labate
- Department of Mathematics, University of Houston, Houston, Texas
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
17
|
Goaillard JM, Moubarak E, Tapia M, Tell F. Diversity of Axonal and Dendritic Contributions to Neuronal Output. Front Cell Neurosci 2020; 13:570. [PMID: 32038171 PMCID: PMC6987044 DOI: 10.3389/fncel.2019.00570] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/09/2019] [Indexed: 11/13/2022] Open
Abstract
Our general understanding of neuronal function is that dendrites receive information that is transmitted to the axon, where action potentials (APs) are initiated and propagated to eventually trigger neurotransmitter release at synaptic terminals. Even though this canonical division of labor is true for a number of neuronal types in the mammalian brain (including neocortical and hippocampal pyramidal neurons or cerebellar Purkinje neurons), many neuronal types do not comply with this classical polarity scheme. In fact, dendrites can be the site of AP initiation and propagation, and even neurotransmitter release. In several interneuron types, all functions are carried out by dendrites as these neurons are devoid of a canonical axon. In this article, we present a few examples of "misbehaving" neurons (with a non-canonical polarity scheme) to highlight the diversity of solutions that are used by mammalian neurons to transmit information. Moreover, we discuss how the contribution of dendrites and axons to neuronal excitability may impose constraints on the morphology of these compartments in specific functional contexts.
Collapse
Affiliation(s)
- Jean-Marc Goaillard
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Estelle Moubarak
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Mónica Tapia
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| | - Fabien Tell
- UMR_S 1072, Aix Marseille Université, INSERM, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
18
|
Rathour RK, Narayanan R. Degeneracy in hippocampal physiology and plasticity. Hippocampus 2019; 29:980-1022. [PMID: 31301166 PMCID: PMC6771840 DOI: 10.1002/hipo.23139] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/27/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
Degeneracy, defined as the ability of structurally disparate elements to perform analogous function, has largely been assessed from the perspective of maintaining robustness of physiology or plasticity. How does the framework of degeneracy assimilate into an encoding system where the ability to change is an essential ingredient for storing new incoming information? Could degeneracy maintain the balance between the apparently contradictory goals of the need to change for encoding and the need to resist change towards maintaining homeostasis? In this review, we explore these fundamental questions with the mammalian hippocampus as an example encoding system. We systematically catalog lines of evidence, spanning multiple scales of analysis that point to the expression of degeneracy in hippocampal physiology and plasticity. We assess the potential of degeneracy as a framework to achieve the conjoint goals of encoding and homeostasis without cross-interferences. We postulate that biological complexity, involving interactions among the numerous parameters spanning different scales of analysis, could establish disparate routes towards accomplishing these conjoint goals. These disparate routes then provide several degrees of freedom to the encoding-homeostasis system in accomplishing its tasks in an input- and state-dependent manner. Finally, the expression of degeneracy spanning multiple scales offers an ideal reconciliation to several outstanding controversies, through the recognition that the seemingly contradictory disparate observations are merely alternate routes that the system might recruit towards accomplishment of its goals.
Collapse
Affiliation(s)
- Rahul K. Rathour
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| | - Rishikesh Narayanan
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| |
Collapse
|
19
|
Khalaf B, Roncador A, Pischedda F, Casini A, Thomas S, Piccoli G, Kiebler M, Macchi P. Ankyrin-G induces nucleoporin Nup358 to associate with the axon initial segment of neurons. J Cell Sci 2019; 132:jcs.222802. [PMID: 31427429 DOI: 10.1242/jcs.222802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Nup358 (also known as RanBP2) is a member of the large nucleoporin family that constitutes the nuclear pore complex. Depending on the cell type and the physiological state, Nup358 interacts with specific partner proteins and influences distinct mechanisms independent of its role in nucleocytoplasmic transport. Here, we provide evidence that Nup358 associates selectively with the axon initial segment (AIS) of mature neurons, mediated by the AIS scaffold protein ankyrin-G (AnkG, also known as Ank3). The N-terminus of Nup358 is demonstrated to be sufficient for its localization at the AIS. Further, we show that Nup358 is expressed as two isoforms, one full-length and another shorter form of Nup358. These isoforms differ in their subcellular distribution in neurons and expression level during neuronal development. Overall, the present study highlights an unprecedented localization of Nup358 within the AIS and suggests its involvement in neuronal function.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Bouchra Khalaf
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Alessandro Roncador
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Francesca Pischedda
- Dulbecco Telethon Laboratory of Biology of Synapses, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Antonio Casini
- Laboratory of Molecular Virology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Sabine Thomas
- Department for Cell Biology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University of Munich, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Giovanni Piccoli
- Dulbecco Telethon Laboratory of Biology of Synapses, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| | - Michael Kiebler
- Department for Cell Biology, Biomedical Center, Medical Faculty, Ludwig-Maximilian University of Munich, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Paolo Macchi
- Laboratory of Molecular and Cellular Neurobiology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy
| |
Collapse
|
20
|
Klinman E, Tokito M, Holzbaur ELF. CDK5-dependent activation of dynein in the axon initial segment regulates polarized cargo transport in neurons. Traffic 2018; 18:808-824. [PMID: 28941293 DOI: 10.1111/tra.12529] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 02/03/2023]
Abstract
The unique polarization of neurons depends on selective sorting of axonal and somatodendritic cargos to their correct compartments. Axodendritic sorting and filtering occurs within the axon initial segment (AIS). However, the underlying molecular mechanisms responsible for this filter are not well understood. Here, we show that local activation of the neuronal-specific kinase cyclin-dependent kinase 5 (CDK5) is required to maintain AIS integrity, as depletion or inhibition of CDK5 induces disordered microtubule polarity and loss of AIS cytoskeletal structure. Furthermore, CDK5-dependent phosphorylation of the dynein regulator Ndel1 is required for proper re-routing of mislocalized somatodendritic cargo out of the AIS; inhibition of this pathway induces profound mis-sorting defects. While inhibition of the CDK5-Ndel1-Lis1-dynein pathway alters both axonal microtubule polarity and axodendritic sorting, we found that these defects occur on distinct timescales; brief inhibition of dynein disrupts axonal cargo sorting before loss of microtubule polarity becomes evident. Together, these studies identify CDK5 as a master upstream regulator of trafficking in vertebrate neurons, required for both AIS microtubule organization and polarized dynein-dependent sorting of axodendritic cargos, and support an ongoing and essential role for dynein at the AIS.
Collapse
Affiliation(s)
- Eva Klinman
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mariko Tokito
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erika L F Holzbaur
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Vascak M, Jin X, Jacobs KM, Povlishock JT. Mild Traumatic Brain Injury Induces Structural and Functional Disconnection of Local Neocortical Inhibitory Networks via Parvalbumin Interneuron Diffuse Axonal Injury. Cereb Cortex 2018; 28:1625-1644. [PMID: 28334184 PMCID: PMC5907353 DOI: 10.1093/cercor/bhx058] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/20/2017] [Indexed: 12/18/2022] Open
Abstract
Diffuse axonal injury (DAI) plays a major role in cortical network dysfunction posited to cause excitatory/inhibitory imbalance after mild traumatic brain injury (mTBI). Current thought holds that white matter (WM) is uniquely vulnerable to DAI. However, clinically diagnosed mTBI is not always associated with WM DAI. This suggests an undetected neocortical pathophysiology, implicating GABAergic interneurons. To evaluate this possibility, we used mild central fluid percussion injury to generate DAI in mice with Cre-driven tdTomato labeling of parvalbumin (PV) interneurons. We followed tdTomato+ profiles using confocal and electron microscopy, together with patch-clamp analysis to probe for DAI-mediated neocortical GABAergic interneuron disruption. Within 3 h post-mTBI tdTomato+ perisomatic axonal injury (PSAI) was found across somatosensory layers 2-6. The DAI marker amyloid precursor protein colocalized with GAD67 immunoreactivity within tdTomato+ PSAI, representing the majority of GABAergic interneuron DAI. At 24 h post-mTBI, we used phospho-c-Jun, a surrogate DAI marker, for retrograde assessments of sustaining somas. Via this approach, we estimated DAI occurs in ~9% of total tdTomato+ interneurons, representing ~14% of pan-neuronal DAI. Patch-clamp recordings of tdTomato+ interneurons revealed decreased inhibitory transmission. Overall, these data show that PV interneuron DAI is a consistent and significant feature of experimental mTBI with important implications for cortical network dysfunction.
Collapse
Affiliation(s)
- Michal Vascak
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, PO Box 980709, Richmond, VA 23298-0709, USA
| | - Xiaotao Jin
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, PO Box 980709, Richmond, VA 23298-0709, USA
| | - Kimberle M Jacobs
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, PO Box 980709, Richmond, VA 23298-0709, USA
| | - John T Povlishock
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, PO Box 980709, Richmond, VA 23298-0709, USA
| |
Collapse
|
22
|
Yue ZW, Wang YL, Xiao B, Feng L. Axon Initial Segment Structural Plasticity is Involved in Seizure Susceptibility in a Rat Model of Cortical Dysplasia. Neurochem Res 2018; 43:878-885. [PMID: 29468458 DOI: 10.1007/s11064-018-2493-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 11/28/2022]
Abstract
Cortical dysplasia is the most common etiology of intractable epilepsy. Both excitability changes in cortical neurons and neural network reconstitution play a role in cortical dysplasia epileptogenesis. Recent research shows that the axon initial segment, a subcompartment of the neuron important to the shaping of action potentials, adjusts its position in response to changes in input, which contributes to neuronal excitability and local circuit balance. It is unknown whether axon initial segment plasticity occurs in neurons involved in seizure susceptibility in cortical dysplasia. Here, we developed a "Carmustine"- "pilocarpine" rat model of cortical dysplasia and show that it exhibits a lower seizure threshold, as indicated by behavior studies and electroencephalogram monitoring. Using immunofluorescence, we measured the axon initial segment positions of deep L5 somatosensory neurons and show that it is positioned closer to the soma after acute seizure, and that this displacement is sustained in the chronic phase. We then show that Nifedipine has a dose-dependent protective effect against axon initial segment displacement and increased seizure susceptibility. These findings further our understanding of the pathophysiology of seizures in cortical dysplasia and suggests Nifedipine as a potential therapeutic agent.
Collapse
Affiliation(s)
- Zong-Wei Yue
- 1Neurology Department, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, Hunan, China
| | - Ye-Lan Wang
- 1Neurology Department, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, Hunan, China
| | - Bo Xiao
- 1Neurology Department, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, Hunan, China.
| | - Li Feng
- 1Neurology Department, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, Hunan, China.
| |
Collapse
|
23
|
Weiland JD, Walston ST, Humayun MS. Electrical Stimulation of the Retina to Produce Artificial Vision. Annu Rev Vis Sci 2018; 2:273-294. [PMID: 28532361 DOI: 10.1146/annurev-vision-111815-114425] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Retinal prostheses aim to restore vision to blind individuals suffering from retinal diseases such as retinitis pigmentosa and age-related macular degeneration. These devices function by electrically stimulating surviving retinal neurons, whose activation is interpreted by the brain as a visual percept. Many prostheses are currently under development. They are categorized as epiretinal, subretinal, and suprachoroidal prostheses on the basis of the placement of the stimulating microelectrode array. Each can activate ganglion cells through direct or indirect stimulation. The response of retinal neurons to these modes of stimulation are discussed in detail and are placed in context of the visual percept they are likely to evoke. This article further reviews challenges faced by retinal prosthesis and discusses potential solutions to address them.
Collapse
Affiliation(s)
- James D Weiland
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90007; .,USC Roski Eye Institute, University of Southern California, Los Angeles, California 90033.,Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California 90033
| | - Steven T Walston
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90007;
| | - Mark S Humayun
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90007; .,USC Roski Eye Institute, University of Southern California, Los Angeles, California 90033.,Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California 90033
| |
Collapse
|
24
|
Abstract
Schizophrenia is a complex disorder lacking an effective treatment option for the pervasive and debilitating cognitive impairments experienced by patients. Working memory is a core cognitive function impaired in schizophrenia that depends upon activation of distributed neural network, including the circuitry of the dorsolateral prefrontal cortex (DLPFC). Accordingly, individuals diagnosed with schizophrenia show reduced DLPFC activation while performing working-memory tasks. This lower DLPFC activation appears to be an integral part of the disease pathophysiology, and not simply a reflection of poor performance. Thus, the cellular and circuitry alterations that underlie lower DLPFC neuronal activity in schizophrenia must be determined in order to identify appropriate therapeutic targets. Studies using human postmortem brain tissue provide a robust way to investigate and characterize these cellular and circuitry alterations at multiple levels of resolution, and such studies provide essential information that cannot be obtained either through in vivo studies in humans or through experimental animal models. Studies examining neuronal morphology, protein expression and localization, and transcript levels indicate that a microcircuit composed of excitatory pyramidal cells and inhibitory interneurons containing the calcium-binding protein parvalbumin is altered in the DLPFC of subjects with schizophrenia and likely contributes to DLPFC dysfunction.
Collapse
Affiliation(s)
- Jill R Glausier
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
25
|
Role of the Axon Initial Segment in the Control of Spontaneous Frequency of Nigral Dopaminergic Neurons In Vivo. J Neurosci 2017; 38:733-744. [PMID: 29217687 DOI: 10.1523/jneurosci.1432-17.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 10/24/2017] [Accepted: 11/20/2017] [Indexed: 11/21/2022] Open
Abstract
The spontaneous tonic discharge activity of nigral dopamine neurons plays a fundamental role in dopaminergic signaling. To investigate the role of neuronal morphology and architecture with respect to spontaneous activity in this population, we visualized the 3D structure of the axon initial segment (AIS) along with the entire somatodendritic domain of adult male mouse dopaminergic neurons, previously recorded in vivo We observed a positive correlation of the firing rate with both proximity and size of the AIS. Computational modeling showed that the size of the AIS, but not its position within the somatodendritic domain, is the major causal determinant of the tonic firing rate in the intact model, by virtue of the higher intrinsic frequency of the isolated AIS. Further mechanistic analysis of the relationship between neuronal morphology and firing rate showed that dopaminergic neurons function as a coupled oscillator whose frequency of discharge results from a compromise between AIS and somatodendritic oscillators. Thus, morphology plays a critical role in setting the basal tonic firing rate, which in turn could control striatal dopaminergic signaling that mediates motivation and movement.SIGNIFICANCE STATEMENT The frequency at which nigral dopamine neurons discharge action potentials sets baseline dopamine levels in the brain, which enables activity in motor, cognitive, and motivational systems. Here, we demonstrate that the size of the axon initial segment, a subcellular compartment responsible for initiating action potentials, is a key determinant of the firing rate in these neurons. The axon initial segment and all the molecular components that underlie its critical function may provide a novel target for the regulation of dopamine levels in the brain.
Collapse
|
26
|
Liu TT, Feng L, Liu HF, Shu Y, Xiao B. Altered axon initial segment in hippocampal newborn neurons, associated with recurrence of temporal lobe epilepsy in rats. Mol Med Rep 2017; 16:3169-3178. [PMID: 28713955 PMCID: PMC5547972 DOI: 10.3892/mmr.2017.7017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/06/2017] [Indexed: 01/23/2023] Open
Abstract
Hippocampal neurogenesis in temporal lobe epilepsy (TLE) may result in alteration of the excitability of neurons, which contributes to spontaneous recurrent seizures. Axon initial segment (AIS) structural and functional plasticity is important in the control of neuronal excitability. It remains to be elucidated whether the plasticity of AIS occurs in hippocampal newly-generated neurons that are involved in recurrent seizures following pilocarpine-induced status epilepticus (SE). The present study first established a pilocarpine-induced TLE rat model to assess the features of newborn neurons and AIS plasticity alterations using double immunofluorescence staining of Ankyrin G and doublecortin (DCX). AIS plasticity alterations include length and distance from soma in the hippocampal newly-generated neurons post-SE. The results of the present study demonstrated that pilocarpine-induced epileptic rats exhibited aberrant hippocampal neurogenesis and longer DCX-labeled cell dendrites in the dentate gyrus. Pilocarpine-induced epileptic rats demonstrated shortened lengths of AIS and an increased distance from the soma in hippocampal newborn neurons. Mibefradil, a T/L-type calcium blocker, reversed the alterations in length and position of AIS in hippocampal newborn neurons post-SE, accompanied by decreased long-term seizure activity without increased aberrant neurogenesis. These findings indicate that the plasticity of AIS in hippocampal neurogenesis may have profound consequences in epilepsy, at least in animals.
Collapse
Affiliation(s)
- Tian-Tian Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Heng-Fang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
27
|
Dong J, Song H, Wang Y, Li M, Yu Y, Wang Y, Chen J. Maternal Different Degrees of Iodine Deficiency during Pregnant and Lactation Impair the Development of Cerebellar Pinceau in Offspring. Front Neurosci 2017; 11:298. [PMID: 28611576 PMCID: PMC5446996 DOI: 10.3389/fnins.2017.00298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/12/2017] [Indexed: 11/13/2022] Open
Abstract
Aims: Iodine is critical for synthesis of thyroid hormones (TH). And iodine deficiency (ID) is one of the most significant reasons of intellectual disability and motor memory impairment, although the potential mechanisms are still under investigation. Presently, mild ID and marginal ID are largely ignored problems for women of child bearing age. Mild ID is a subtle form of TH deficiency, which shows low levels of free thyroxine (FT4) and relatively normal free triiodothyronine (FT3) or thyroid stimulation hormone (TSH). And marginal ID is a milder form of ID with decreased total T4 (TT4) but relatively normal FT3, FT4, and TSH. Therefore, we investigated the effects of maternal different degrees of ID on the development of pinceau in cerebellar purkinje cells (PCs) and studied the expression of pinceau related protein, which is crucial for the development and maturation of pinceau. Methods and Results: Three developmental iodine deficient rat models were created by feeding dam rats with an iodine-deficient diet and deionized water supplemented with potassiumiodide. Our study showed that different degrees of ID inhibited cerebellar pinceau synapse development and maturation on postnatal day (PN) 14 and PN21. What's more, mild and severe ID reduced the expression of AnkG, β4-spectrin, neurofascin186 and NrCAM on PN7, PN14, and PN21. However, marginal ID rarely altered expression of these proteins in the offspring. Conclusion: These results suggested that maternal mild and severe ID impaired the development and maturation of cerebellar pinceau, which may be attributed to the decrease of AnkG, β4-spectrin, neurofascin 186, and NrCAM. And the alteration of development and maturation in cerebellar pinceau in the offspring were also observed following maternal marginal ID, which is slighter than that of mild ID.
Collapse
Affiliation(s)
- Jing Dong
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Heling Song
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yuan Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Min Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Ye Yu
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
28
|
Evans MD, Tufo C, Dumitrescu AS, Grubb MS. Myosin II activity is required for structural plasticity at the axon initial segment. Eur J Neurosci 2017; 46:1751-1757. [PMID: 28452088 PMCID: PMC5573965 DOI: 10.1111/ejn.13597] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/29/2017] [Accepted: 04/20/2017] [Indexed: 12/21/2022]
Abstract
In neurons, axons possess a molecularly defined and highly organised proximal region – the axon initial segment (AIS) – that is a key regulator of both electrical excitability and cellular polarity. Despite existing as a large, dense structure with specialised cytoskeletal architecture, the AIS is surprisingly plastic, with sustained alterations in neuronal activity bringing about significant alterations to its position, length or molecular composition. However, although the upstream activity‐dependent signalling pathways that lead to such plasticity have begun to be elucidated, the downstream mechanisms that produce structural changes at the AIS are completely unknown. Here, we use dissociated cultures of rat hippocampus to show that two forms of AIS plasticity in dentate granule cells – long‐term relocation, and more rapid shortening – are completely blocked by treatment with blebbistatin, a potent and selective myosin II ATPase inhibitor. These data establish a link between myosin II and AIS function, and suggest that myosin II's primary role at the structure may be to effect activity‐dependent morphological alterations.
Collapse
Affiliation(s)
- Mark D Evans
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | - Candida Tufo
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | - Adna S Dumitrescu
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK.,FENS-Kavli Network of Excellence, Europe-wide
| |
Collapse
|
29
|
Hsu WCJ, Wildburger NC, Haidacher SJ, Nenov MN, Folorunso O, Singh AK, Chesson BC, Franklin WF, Cortez I, Sadygov RG, Dineley KT, Rudra JS, Taglialatela G, Lichti CF, Denner L, Laezza F. PPARgamma agonists rescue increased phosphorylation of FGF14 at S226 in the Tg2576 mouse model of Alzheimer's disease. Exp Neurol 2017; 295:1-17. [PMID: 28522250 DOI: 10.1016/j.expneurol.2017.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/13/2017] [Accepted: 05/13/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cognitive impairment in humans with Alzheimer's disease (AD) and in animal models of Aβ-pathology can be ameliorated by treatments with the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARγ) agonists, such as rosiglitazone (RSG). Previously, we demonstrated that in the Tg2576 animal model of AD, RSG treatment rescued cognitive deficits and reduced aberrant activity of granule neurons in the dentate gyrus (DG), an area critical for memory formation. METHODS We used a combination of mass spectrometry, confocal imaging, electrophysiology and split-luciferase assay and in vitro phosphorylation and Ingenuity Pathway Analysis. RESULTS Using an unbiased, quantitative nano-LC-MS/MS screening, we searched for potential molecular targets of the RSG-dependent rescue of DG granule neurons. We found that S226 phosphorylation of fibroblast growth factor 14 (FGF14), an accessory protein of the voltage-gated Na+ (Nav) channels required for neuronal firing, was reduced in Tg2576 mice upon treatment with RSG. Using confocal microscopy, we confirmed that the Tg2576 condition decreased PanNav channels at the AIS of the DG, and that RSG treatment of Tg2576 mice reversed the reduction in PanNav channels. Analysis from previously published data sets identified correlative changes in action potential kinetics in RSG-treated T2576 compared to untreated and wildtype controls. In vitro phosphorylation and mass spectrometry confirmed that the multifunctional kinase GSK-3β, a downstream target of insulin signaling highly implicated in AD, phosphorylated FGF14 at S226. Assembly of the FGF14:Nav1.6 channel complex and functional regulation of Nav1.6-mediated currents by FGF14 was impaired by a phosphosilent S226A mutation. Bioinformatics pathway analysis of mass spectrometry and biochemistry data revealed a highly interconnected network encompassing PPARγ, FGF14, SCN8A (Nav 1.6), and the kinases GSK-3 β, casein kinase 2β, and ERK1/2. CONCLUSIONS These results identify FGF14 as a potential PPARγ-sensitive target controlling Aβ-induced dysfunctions of neuronal activity in the DG underlying memory loss in early AD.
Collapse
Affiliation(s)
- Wei-Chun J Hsu
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Biochemistry and Molecular Biology Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; M.D./Ph.D. Combined Degree Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Norelle C Wildburger
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, United States
| | - Sigmund J Haidacher
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Miroslav N Nenov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Oluwarotimi Folorunso
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Brent C Chesson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Whitney F Franklin
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Ibdanelo Cortez
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Rovshan G Sadygov
- Biochemistry and Molecular Biology Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Kelly T Dineley
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Jay S Rudra
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Neurology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Cheryl F Lichti
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Larry Denner
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States; Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, United States.
| |
Collapse
|
30
|
Satake T, Yamashita K, Hayashi K, Miyatake S, Tamura-Nakano M, Doi H, Furuta Y, Shioi G, Miura E, Takeo YH, Yoshida K, Yahikozawa H, Matsumoto N, Yuzaki M, Suzuki A. MTCL1 plays an essential role in maintaining Purkinje neuron axon initial segment. EMBO J 2017; 36:1227-1242. [PMID: 28283581 DOI: 10.15252/embj.201695630] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 11/09/2022] Open
Abstract
The axon initial segment (AIS) is a specialized domain essential for neuronal function, the formation of which begins with localization of an ankyrin-G (AnkG) scaffold. However, the mechanism directing and maintaining AnkG localization is largely unknown. In this study, we demonstrate that in vivo knockdown of microtubule cross-linking factor 1 (MTCL1) in cerebellar Purkinje cells causes loss of axonal polarity coupled with AnkG mislocalization. MTCL1 lacking MT-stabilizing activity failed to restore these defects, and stable MT bundles spanning the AIS were disorganized in knockdown cells. Interestingly, during early postnatal development, colocalization of MTCL1 with these stable MT bundles was observed prominently in the axon hillock and proximal axon. These results indicate that MTCL1-mediated formation of stable MT bundles is crucial for maintenance of AnkG localization. We also demonstrate that Mtcl1 gene disruption results in abnormal motor coordination with Purkinje cell degeneration, and provide evidence suggesting possible involvement of MTCL1 dysfunction in the pathogenesis of spinocerebellar ataxia.
Collapse
Affiliation(s)
- Tomoko Satake
- Molecular Cellular Biology Laboratory, Yokohama City University Graduate School of Medical Life Science, Tsurumi-ku Yokohama, Japan
| | - Kazunari Yamashita
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Kanazawa-ku Yokohama, Japan
| | - Kenji Hayashi
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Kanazawa-ku Yokohama, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medical Science, Kanazawa-ku Yokohama, Japan
| | - Miwa Tamura-Nakano
- Communal Laboratory, Research Institute, National Center for Global Health and Medicine, Toyama Shinjuku-ku Tokyo, Japan
| | - Hiroshi Doi
- Department of Neurology, Yokohama City University Graduate School of Medical Science, Kanazawa-ku Yokohama, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit, RIKEN Center for Life Science Technologies, Chuou-ku Kobe, Japan.,Genetic Engineering Team, RIKEN Center for Life Science Technologies, Chuou-ku Kobe, Japan
| | - Go Shioi
- Genetic Engineering Team, RIKEN Center for Life Science Technologies, Chuou-ku Kobe, Japan
| | - Eriko Miura
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku Tokyo, Japan
| | - Yukari H Takeo
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku Tokyo, Japan
| | - Kunihiro Yoshida
- Division of Neurogenetics, Department of Brain Disease Research, Shinshu University School of Medicine, Asahi Matsumoto, Japan
| | | | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medical Science, Kanazawa-ku Yokohama, Japan
| | - Michisuke Yuzaki
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku Tokyo, Japan
| | - Atsushi Suzuki
- Molecular Cellular Biology Laboratory, Yokohama City University Graduate School of Medical Life Science, Tsurumi-ku Yokohama, Japan
| |
Collapse
|
31
|
Jiang L, Ni H, Wang QY, Huang L, Zhao SD, Yu JD, Ge RJ. Dual face of axonal inhibitory inputs in the modulation of neuronal excitability in cortical pyramidal neurons. Neural Regen Res 2017; 12:1079-1085. [PMID: 28852389 PMCID: PMC5558486 DOI: 10.4103/1673-5374.211186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Limited by the tiny structure of axons, the effects of these axonal hyperpolarizing inputs on neuronal activity have not been directly elucidated. Here, we imitated these processes by simultaneously recording the activities of the somas and proximal axons of cortical pyramidal neurons. We found that spikes and subthreshold potentials propagate between somas and axons with high fidelity. Furthermore, inhibitory inputs on axons have opposite effects on neuronal activity according to their temporal integration with upstream signals. Concurrent with somatic depolarization, inhibitory inputs on axons decrease neuronal excitability and impede spike generation. In addition, following action potentials, inhibitory inputs on an axon increase neuronal spike capacity and improve spike precision. These results indicate that inhibitory inputs on proximal axons have dual regulatory functions in neuronal activity (suppression or facilitation) according to neuronal network patterns.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui Province, China.,Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, China
| | - Hong Ni
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Qi-Yi Wang
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Shi-di Zhao
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jian-Dong Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration, Ministry of Education, Jinan University, Guangzhou, Guangdong Province, China
| | - Rong-Jing Ge
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui Province, China
| |
Collapse
|
32
|
Jegla T, Nguyen MM, Feng C, Goetschius DJ, Luna E, van Rossum DB, Kamel B, Pisupati A, Milner ES, Rolls MM. Bilaterian Giant Ankyrins Have a Common Evolutionary Origin and Play a Conserved Role in Patterning the Axon Initial Segment. PLoS Genet 2016; 12:e1006457. [PMID: 27911898 PMCID: PMC5135030 DOI: 10.1371/journal.pgen.1006457] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 11/03/2016] [Indexed: 12/03/2022] Open
Abstract
In vertebrate neurons, the axon initial segment (AIS) is specialized for action potential initiation. It is organized by a giant 480 Kd variant of ankyrin G (AnkG) that serves as an anchor for ion channels and is required for a plasma membrane diffusion barrier that excludes somatodendritic proteins from the axon. An unusually long exon required to encode this 480Kd variant is thought to have been inserted only recently during vertebrate evolution, so the giant ankyrin-based AIS scaffold has been viewed as a vertebrate adaptation for fast, precise signaling. We re-examined AIS evolution through phylogenomic analysis of ankyrins and by testing the role of ankyrins in proximal axon organization in a model multipolar Drosophila neuron (ddaE). We find giant isoforms of ankyrin in all major bilaterian phyla, and present evidence in favor of a single common origin for giant ankyrins and the corresponding long exon in a bilaterian ancestor. This finding raises the question of whether giant ankyrin isoforms play a conserved role in AIS organization throughout the Bilateria. We examined this possibility by looking for conserved ankyrin-dependent AIS features in Drosophila ddaE neurons via live imaging. We found that ddaE neurons have an axonal diffusion barrier proximal to the cell body that requires a giant isoform of the neuronal ankyrin Ank2. Furthermore, the potassium channel shal concentrates in the proximal axon in an Ank2-dependent manner. Our results indicate that the giant ankyrin-based cytoskeleton of the AIS may have evolved prior to the radiation of extant bilaterian lineages, much earlier than previously thought. The axon initial segment (AIS) is currently thought to be a distinguishing feature of vertebrate neurons that adapts them for rapid, precise signaling. It serves as a hub for the regulation of neuronal excitability as the site of action potential initiation and also acts as the boundary between the highly-specialized axon and the rest of the cell. Here we show that the giant ankyrins that structurally organize the AIS, and were thought to be vertebrate-specific, instead have an ancient origin in a bilaterian ancestor. We further show the presence of a giant ankyrin-dependent AIS-like plasma membrane boundary between the axon and soma in a Drosophila sensory neuron. These results suggest that the cytoskeletal backbone for the AIS is not unique to vertebrates, but instead may be an evolutionarily conserved feature of bilaterian neurons.
Collapse
Affiliation(s)
- Timothy Jegla
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (MMR); (TJ)
| | - Michelle M. Nguyen
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Chengye Feng
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Daniel J. Goetschius
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Esteban Luna
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Damian B. van Rossum
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Bishoy Kamel
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Aditya Pisupati
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Elliott S. Milner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Melissa M. Rolls
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (MMR); (TJ)
| |
Collapse
|
33
|
Ankyrin G expression is associated with androgen receptor stability, invasiveness, and lethal outcome in prostate cancer patients. J Mol Med (Berl) 2016; 94:1411-1422. [PMID: 27534968 DOI: 10.1007/s00109-016-1458-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 07/27/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Ankyrin G (ANK3) is a member of the Ankyrin family, which functions to provide cellular stability by anchoring the cytoskeleton to the plasma membrane. Deregulation of ANK3 expression has been observed in multiple human cancers but its mechanism remains unknown. ANK3 expression in relation to disease progression and patients' outcome was investigated in two cohorts of prostate cancer (PCA). Mechanistic studies were carried out in vitro and in vivo using several PCA cell lines and the avian embryo model. Silencing ANK3 resulted in significant reduction of cell proliferation through an AR-independent mechanism. Decreased ANK3 expression delayed S phase to G2/M cell cycle transition and reduced the expression of cyclins A and B. However, cells with knocked-down ANK3 exhibited significant increase in cell invasion through an AR-dependent mechanism. Furthermore, we found that ANK3 is a regulator of AR protein stability. ANK3 knockdown also promoted cancer cell invasion and extravasations in vivo using the avian embryo model (p < 0.01). In human samples, ANK3 expression was dramatically upregulated in high grade intraepithelial neoplasia (HGPIN) and localized PCA (p < 0.0001). However, it was downregulated castration resistant stage (p < 0.0001) and showed inverse relation to Gleason score (p < 0.0001). In addition, increased expression of ANK3 in cancer tissues was correlated with better cancer-specific survival of PCA patients (p = 0.012). KEY MESSAGE Silencing ANK3 results in significant reduction of cell proliferation through an AR-independent mechanism. ANK3 knockdown results in significant increase in cell invasion through an AR-dependent mechanism. ANK3 is a regulator of AR protein stability. ANK3 knockdown also promotes cancer cell invasion and extravasation in vivo using the avian embryo model.
Collapse
|
34
|
Wang Y, Sun D, Yue Z, Tang W, Xiao B, Feng L. Rats with Malformations of Cortical Development Exhibit Decreased Length of AIS and Hypersensitivity to Pilocarpine-Induced Status Epilepticus. Neurochem Res 2016; 41:2215-22. [PMID: 27286680 DOI: 10.1007/s11064-016-1936-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 03/24/2016] [Accepted: 04/25/2016] [Indexed: 11/28/2022]
Abstract
Malformations of cortical development (MCD) are critical brain development disorders associated with varied abnormalities in both anatomic structures and neural functioning. It is also a very common etiology to the epilepsy, in which the alteration on excitability of cortical neurons is hypothesized as one of important causes to the epileptic seizures. Due to the key role in regulating neuron firing properties, the plasticity of axon initial segment (AIS) was investigated in present study to further determine the relation between MCD and epilepsy. Our results showed a prolonged decrease in the length of AIS occurred in MCD animal models. Besides, the AIS was also found greatly shortened in MCD models during the acute, but not chronic phase of status epileptics compared with intact controls. Our findings of identification of AIS plasticity in MCD animal models and its hypersensitivity to status epilepsy are significant in furthering our understanding of the pathophysiological mechanisms involved in this disorder.
Collapse
Affiliation(s)
- Yelan Wang
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China
| | - Danni Sun
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China
| | - Zongwei Yue
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China
| | - Weiting Tang
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China.
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu Area, Changsha, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
35
|
Petersen AV, Cotel F, Perrier JF. Plasticity of the Axon Initial Segment: Fast and Slow Processes with Multiple Functional Roles. Neuroscientist 2016; 23:364-373. [PMID: 27143656 DOI: 10.1177/1073858416648311] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The axon initial segment (AIS) is a key neuronal compartment because it is responsible for action potential initiation. The local density of Na+ channels, the biophysical properties of K+ channels, as well as the length and diameter of the AIS determine the spiking of neurons. These parameters undergo important modifications during development. The development of the AIS is governed by intrinsic mechanisms. In addition, surrounding neuronal networks modify its maturation. As a result, neurons get tuned to particular physiological functions. Neuronal activity also influences the morphology of the mature AIS. When excitatory neurons are hyperactive, their AIS undergo structural changes that decrease their excitability and thereby maintain the activity within a given range. These slow homeostatic regulatory mechanisms occur on a time scale of hours or days. In contrast, the activation of metabotropic receptors modulates the properties of ion channels expressed at the AIS within seconds and consequently produces fast adjustments of neuronal excitability. Recent results suggest that this plasticity plays important roles in physiological functions as diverse as memory formation, hearing, and motor control.
Collapse
Affiliation(s)
- Anders Victor Petersen
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Florence Cotel
- 2 Queensland Brain Institute, University of Queensland, St Lucia, Australia
| | - Jean-François Perrier
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Stoler O, Fleidervish IA. Functional implications of axon initial segment cytoskeletal disruption in stroke. Acta Pharmacol Sin 2016; 37:75-81. [PMID: 26687934 DOI: 10.1038/aps.2015.107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Axon initial segment (AIS) is the proximal part of the axon, which is not covered with a myelin sheath and possesses a distinctive, specialized assembly of voltage-gated ion channels and associated proteins. AIS plays critical roles in synaptic integration and action potential generation in central neurons. Recent evidence shows that stroke causes rapid, irreversible calpain-mediated proteolysis of the AIS cytoskeleton of neurons surrounding the ischemic necrotic core. A better understanding of the molecular mechanisms underlying this "non-lethal" neuronal damage might provide new therapeutic strategies for improving stroke outcome. Here, we present a brief overview of the structure and function of the AIS. We then discuss possible mechanisms underlying stroke-induced AIS damage, including the roles of calpains and possible sources of Ca(2+) ions, which are necessary for the activation of calpains. Finally, we discuss the potential functional implications of the loss of the AIS cytoskeleton and ion channel clusters for neuronal excitability.
Collapse
|
37
|
Leterrier C, Potier J, Caillol G, Debarnot C, Rueda Boroni F, Dargent B. Nanoscale Architecture of the Axon Initial Segment Reveals an Organized and Robust Scaffold. Cell Rep 2015; 13:2781-93. [DOI: 10.1016/j.celrep.2015.11.051] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/14/2015] [Accepted: 11/13/2015] [Indexed: 12/17/2022] Open
|
38
|
Abstract
Polarized distribution of signaling molecules to axons and dendrites facilitates directional information flow in complex vertebrate nervous systems. The topic we address here is when the key aspects of neuronal polarity evolved. All neurons have a central cell body with thin processes that extend from it to cover long distances, and they also all rely on voltage-gated ion channels to propagate signals along their length. The most familiar neurons, those in vertebrates, have additional cellular features that allow them to send directional signals efficiently. In these neurons, dendrites typically receive signals and axons send signals. It has been suggested that many of the distinct features of axons and dendrites, including the axon initial segment, are found only in vertebrates. However, it is now becoming clear that two key cytoskeletal features that underlie polarized sorting, a specialized region at the base of the axon and polarized microtubules, are found in invertebrate neurons as well. It thus seems likely that all bilaterians generate axons and dendrites in the same way. As a next step, it will be extremely interesting to determine whether the nerve nets of cnidarians and ctenophores also contain polarized neurons with true axons and dendrites, or whether polarity evolved in concert with the more centralized nervous systems found in bilaterians.
Collapse
Affiliation(s)
- Melissa M Rolls
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Timothy J Jegla
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
39
|
Exchange Protein Directly Activated by cAMP (EPAC) Regulates Neuronal Polarization through Rap1B. J Neurosci 2015; 35:11315-29. [PMID: 26269639 DOI: 10.1523/jneurosci.3645-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Acquisition of neuronal polarity is a complex process involving cellular and molecular events. The second messenger cAMP is involved in axonal specification through activation of protein kinase A. However, an alternative cAMP-dependent mechanism involves the exchange protein directly activated by cAMP (EPAC), which also responds to physiological changes in cAMP concentration, promoting activation of the small Rap GTPases. Here, we present evidence that EPAC signaling contributes to axon specification and elongation. In primary rat hippocampal neurons, EPAC isoforms were expressed differentially during axon specification. Furthermore, 8-pCPT, an EPAC pharmacological activator, and genetic manipulations of EPAC in neurons induced supernumerary axons indicative of Rap1b activation. Moreover, 8-pCPT-treated neurons expressed ankyrin G and other markers of mature axons such as synaptophysin and axonal accumulation of vGLUT1. In contrast, pharmacological inhibition of EPAC delayed neuronal polarity. Genetic manipulations to inactivate EPAC1 using either shRNA or neurons derived from EPAC1 knock-out (KO) mice led to axon elongation and polarization defects. Interestingly, multiaxonic neurons generated by 8-pCPT treatments in wild-type neurons were not found in EPAC1 KO mice neurons. Altogether, these results propose that EPAC signaling is an alternative and complementary mechanism for cAMP-dependent axon determination. SIGNIFICANCE STATEMENT This study identifies the guanine exchange factor responsible for Rap1b activation during neuronal polarization and provides an alternate explanation for cAMP-dependent acquisition of neuronal polarity.
Collapse
|
40
|
Kevenaar JT, Hoogenraad CC. The axonal cytoskeleton: from organization to function. Front Mol Neurosci 2015; 8:44. [PMID: 26321907 PMCID: PMC4536388 DOI: 10.3389/fnmol.2015.00044] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/31/2015] [Indexed: 01/20/2023] Open
Abstract
The axon is the single long fiber that extends from the neuron and transmits electrical signals away from the cell body. The neuronal cytoskeleton, composed of microtubules (MTs), actin filaments and neurofilaments, is not only required for axon formation and axonal transport but also provides the structural basis for several specialized axonal structures, such as the axon initial segment (AIS) and presynaptic boutons. Emerging evidence suggest that the unique cytoskeleton organization in the axon is essential for its structure and integrity. In addition, the increasing number of neurodevelopmental and neurodegenerative diseases linked to defect in actin- and microtubule-dependent processes emphasizes the importance of a properly regulated cytoskeleton for normal axonal functioning. Here, we provide an overview of the current understanding of actin and microtubule organization within the axon and discuss models for the functional role of the cytoskeleton at specialized axonal structures.
Collapse
Affiliation(s)
- Josta T. Kevenaar
- Cell Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| | | |
Collapse
|
41
|
Activity-dependent mismatch between axo-axonic synapses and the axon initial segment controls neuronal output. Proc Natl Acad Sci U S A 2015. [PMID: 26195803 DOI: 10.1073/pnas.1502902112] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The axon initial segment (AIS) is a structure at the start of the axon with a high density of sodium and potassium channels that defines the site of action potential generation. It has recently been shown that this structure is plastic and can change its position along the axon, as well as its length, in a homeostatic manner. Chronic activity-deprivation paradigms in a chick auditory nucleus lead to a lengthening of the AIS and an increase in neuronal excitability. On the other hand, a long-term increase in activity in dissociated rat hippocampal neurons results in an outward movement of the AIS and a decrease in the cell's excitability. Here, we investigated whether the AIS is capable of undergoing structural plasticity in rat hippocampal organotypic slices, which retain the diversity of neuronal cell types present at postnatal ages, including chandelier cells. These interneurons exclusively target the AIS of pyramidal neurons and form rows of presynaptic boutons along them. Stimulating individual CA1 pyramidal neurons that express channelrhodopsin-2 for 48 h leads to an outward shift of the AIS. Intriguingly, both the pre- and postsynaptic components of the axo-axonic synapses did not change position after AIS relocation. We used computational modeling to explore the functional consequences of this partial mismatch and found that it allows the GABAergic synapses to strongly oppose action potential generation, and thus downregulate pyramidal cell excitability. We propose that this spatial arrangement is the optimal configuration for a homeostatic response to long-term stimulation.
Collapse
|
42
|
Action potential initiation in a multi-compartmental model with cooperatively gating Na channels in the axon initial segment. J Comput Neurosci 2015; 39:63-75. [PMID: 26001536 DOI: 10.1007/s10827-015-0561-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 03/11/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
Abstract
Somatic action potentials (AP) of cortical pyramidal neurons have characteristically high onset-rapidness. The onset of the AP waveform is an indirect measure for the ability of a neuron to respond to temporally fast-changing stimuli. Theoretical studies on the pyramidal neuron response usually involves a canonical Hodgkin-Huxley (HH) type ion channel gating model, which assumes statistically independent gating of each individual channel. However, cooperative activity of ion channels are observed for various cell types, meaning that the activity (e.g. opening) of one channel triggers the activity (e.g. opening) of a certain fraction of its neighbors and hence, these groups of channels behave as a unit. In this study, we describe a multi-compartmental conductance-based model with cooperatively gating voltage-gated Na channels in the axon initial segment. Our model successfully reproduced the somatic sharp AP onsets of cortical pyramidal neurons. The onset latencies from the initiation site to the soma and the conduction velocities were also in agreement with the previous experimental studies.
Collapse
|
43
|
Modelling the Effects of Electrical Coupling between Unmyelinated Axons of Brainstem Neurons Controlling Rhythmic Activity. PLoS Comput Biol 2015; 11:e1004240. [PMID: 25954930 PMCID: PMC4425518 DOI: 10.1371/journal.pcbi.1004240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/15/2015] [Indexed: 11/19/2022] Open
Abstract
Gap junctions between fine unmyelinated axons can electrically couple groups of brain neurons to synchronise firing and contribute to rhythmic activity. To explore the distribution and significance of electrical coupling, we modelled a well analysed, small population of brainstem neurons which drive swimming in young frog tadpoles. A passive network of 30 multicompartmental neurons with unmyelinated axons was used to infer that: axon-axon gap junctions close to the soma gave the best match to experimentally measured coupling coefficients; axon diameter had a strong influence on coupling; most neurons were coupled indirectly via the axons of other neurons. When active channels were added, gap junctions could make action potential propagation along the thin axons unreliable. Increased sodium and decreased potassium channel densities in the initial axon segment improved action potential propagation. Modelling suggested that the single spike firing to step current injection observed in whole-cell recordings is not a cellular property but a dynamic consequence of shunting resulting from electrical coupling. Without electrical coupling, firing of the population during depolarising current was unsynchronised; with coupling, the population showed synchronous recruitment and rhythmic firing. When activated instead by increasing levels of modelled sensory pathway input, the population without electrical coupling was recruited incrementally to unpatterned activity. However, when coupled, the population was recruited all-or-none at threshold into a rhythmic swimming pattern: the tadpole “decided” to swim. Modelling emphasises uncertainties about fine unmyelinated axon physiology but, when informed by biological data, makes general predictions about gap junctions: locations close to the soma; relatively small numbers; many indirect connections between neurons; cause of action potential propagation failure in fine axons; misleading alteration of intrinsic firing properties. Modelling also indicates that electrical coupling within a population can synchronize recruitment of neurons and their pacemaker firing during rhythmic activity. Some groups of nerve cells in the brain are connected to each other electrically where their processes make contact and form specialized “gap” junctions. The simplest function of electrical connections is to make activity propagate faster by avoiding the delays resulting from chemical messengers at synaptic connections. In other cases, especially in higher brain regions where more spread out nerve cells may be connected by their axons, the function of electrical coupling is less clear. To understand this type of electrical connection better we have built computer models of a group of electrically coupled nerve cells in the brain which control swimming in very young frog tadpoles. We show that the coupling can be indirect, via other members of the group, and can profoundly influence the properties of the nerve cells which would be recorded during real experiments. The main role of the coupling is to synchronise the firing of the group so they are all recruited together when the tadpole is stimulated and then fire in a rhythm suitable to drive swimming movements. The results from this simple animal raise issues which will help to understand coupling in more complex brains.
Collapse
|
44
|
A distinct subtype of dopaminergic interneuron displays inverted structural plasticity at the axon initial segment. J Neurosci 2015; 35:1573-90. [PMID: 25632134 DOI: 10.1523/jneurosci.3515-14.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The axon initial segment (AIS) is a specialized structure near the start of the axon that is a site of neuronal plasticity. Changes in activity levels in vitro and in vivo can produce structural AIS changes in excitatory cells that have been linked to alterations in excitability, but these effects have never been described in inhibitory interneurons. In the mammalian olfactory bulb (OB), dopaminergic interneurons are particularly plastic, undergoing constitutive turnover throughout life and regulating tyrosine hydroxylase expression in an activity-dependent manner. Here we used dissociated cultures of rat and mouse OB to show that a subset of bulbar dopaminergic neurons possess an AIS and that these AIS-positive cells are morphologically and functionally distinct from their AIS-negative counterparts. Under baseline conditions, OB dopaminergic AISs were short and located distally along the axon but, in response to chronic 24 h depolarization, lengthened and relocated proximally toward the soma. These activity-dependent changes were in the opposite direction to both those we saw in non-GABAergic OB neurons and those reported previously for excitatory cell types. Inverted AIS plasticity in OB dopaminergic cells was bidirectional, involved all major components of the structure, was dependent on the activity of L-type CaV1 calcium channels but not on the activity of the calcium-activated phosphatase calcineurin, and was opposed by the actions of cyclin-dependent kinase 5. Such distinct forms of AIS plasticity in inhibitory interneurons and excitatory projection neurons may allow considerable flexibility when neuronal networks must adapt to perturbations in their ongoing activity.
Collapse
|
45
|
Ankyrin-G regulates neurogenesis and Wnt signaling by altering the subcellular localization of β-catenin. Mol Psychiatry 2015; 20:388-97. [PMID: 24821222 PMCID: PMC4231016 DOI: 10.1038/mp.2014.42] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 03/17/2014] [Accepted: 03/20/2014] [Indexed: 12/29/2022]
Abstract
Ankyrin-G is a scaffolding protein required for the formation of the axon initial segment in neurons. Recent genome-wide association studies and whole-exome sequencing have identified ANK3, the gene coding for ankyrin-G, to be a risk gene for multiple neuropsychiatric disorders, such as bipolar disorder, schizophrenia and autism spectrum disorder. Here, we describe a novel role for ankyrin-G in neural progenitor proliferation in the developing cortex. We found that ankyrin-G regulates canonical Wnt signaling by altering the subcellular localization and availability of β-catenin in proliferating cells. Ankyrin-G loss-of-function increases β-catenin levels in the nucleus, thereby promoting neural progenitor proliferation. Importantly, abnormalities in proliferation can be rescued by reducing Wnt pathway signaling. Taken together, these results suggest that ankyrin-G is required for proper brain development.
Collapse
|
46
|
Wildburger NC, Ali SR, Hsu WCJ, Shavkunov AS, Nenov MN, Lichti CF, LeDuc RD, Mostovenko E, Panova-Elektronova NI, Emmett MR, Nilsson CL, Laezza F. Quantitative proteomics reveals protein-protein interactions with fibroblast growth factor 12 as a component of the voltage-gated sodium channel 1.2 (nav1.2) macromolecular complex in Mammalian brain. Mol Cell Proteomics 2015; 14:1288-300. [PMID: 25724910 PMCID: PMC4424400 DOI: 10.1074/mcp.m114.040055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Nav1.1–Nav1.9) are responsible for the initiation and propagation of action potentials in neurons, controlling firing patterns, synaptic transmission and plasticity of the brain circuit. Yet, it is the protein–protein interactions of the macromolecular complex that exert diverse modulatory actions on the channel, dictating its ultimate functional outcome. Despite the fundamental role of Nav channels in the brain, information on its proteome is still lacking. Here we used affinity purification from crude membrane extracts of whole brain followed by quantitative high-resolution mass spectrometry to resolve the identity of Nav1.2 protein interactors. Of the identified putative protein interactors, fibroblast growth factor 12 (FGF12), a member of the nonsecreted intracellular FGF family, exhibited 30-fold enrichment in Nav1.2 purifications compared with other identified proteins. Using confocal microscopy, we visualized native FGF12 in the brain tissue and confirmed that FGF12 forms a complex with Nav1.2 channels at the axonal initial segment, the subcellular specialized domain of neurons required for action potential initiation. Co-immunoprecipitation studies in a heterologous expression system validate Nav1.2 and FGF12 as interactors, whereas patch-clamp electrophysiology reveals that FGF12 acts synergistically with CaMKII, a known kinase regulator of Nav channels, to modulate Nav1.2-encoded currents. In the presence of CaMKII inhibitors we found that FGF12 produces a bidirectional shift in the voltage-dependence of activation (more depolarized) and the steady-state inactivation (more hyperpolarized) of Nav1.2, increasing the channel availability. Although providing the first characterization of the Nav1.2 CNS proteome, we identify FGF12 as a new functionally relevant interactor. Our studies will provide invaluable information to parse out the molecular determinant underlying neuronal excitability and plasticity, and extending the relevance of iFGFs signaling in the normal and diseased brain.
Collapse
Affiliation(s)
- Norelle C Wildburger
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; §Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074;
| | - Syed R Ali
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Wei-Chun J Hsu
- ‖Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617
| | - Alexander S Shavkunov
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Miroslav N Nenov
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Cheryl F Lichti
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Richard D LeDuc
- **National Center for Genome Analysis Support, Indiana University, 107 S Indiana Ave., Bloomington, Indiana, 47408
| | - Ekaterina Mostovenko
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Neli I Panova-Elektronova
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Mark R Emmett
- ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074; ‖Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617
| | - Carol L Nilsson
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Fernanda Laezza
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617;
| |
Collapse
|
47
|
King AN, Manning CF, Trimmer JS. A unique ion channel clustering domain on the axon initial segment of mammalian neurons. J Comp Neurol 2015; 522:2594-608. [PMID: 24477962 DOI: 10.1002/cne.23551] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 01/11/2023]
Abstract
The axon initial segment (AIS) plays a key role in initiation of action potentials and neuronal output. The plasma membrane of the AIS contains high densities of voltage-gated ion channels required for these electrical events, and much recent work has focused on defining the mechanisms for generating and maintaining this unique neuronal plasma membrane domain. The Kv2.1 voltage-gated potassium channel is abundantly present in large clusters on the soma and proximal dendrites of mammalian brain neurons. Kv2.1 is also a component of the ion channel repertoire at the AIS. Here we show that Kv2.1 clusters on the AIS of brain neurons across diverse mammalian species including humans define a noncanonical ion channel clustering domain deficient in Ankyrin-G. The sites of Kv2.1 clustering on the AIS are sites where cisternal organelles, specialized intracellular calcium release membranes, come into close apposition with the plasma membrane, and are also sites of clustering of γ-aminobutyric acid (GABA)ergic synapses. Using an antibody specific for a single Kv2.1 phosphorylation site, we find that the phosphorylation state differs between Kv2.1 clusters on the proximal and distal portions of the AIS. Together, these studies show that the sites of Kv2.1 clustering on the AIS represent specialized domains containing components of diverse neuronal signaling pathways that may contribute to local regulation of Kv2.1 function and AIS membrane excitability.
Collapse
Affiliation(s)
- Anna N King
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, 95616
| | | | | |
Collapse
|
48
|
Martinello K, Huang Z, Lujan R, Tran B, Watanabe M, Cooper EC, Brown DA, Shah MM. Cholinergic afferent stimulation induces axonal function plasticity in adult hippocampal granule cells. Neuron 2015; 85:346-63. [PMID: 25578363 PMCID: PMC4306544 DOI: 10.1016/j.neuron.2014.12.030] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2014] [Indexed: 01/25/2023]
Abstract
Acetylcholine critically influences hippocampal-dependent learning. Cholinergic fibers innervate hippocampal neuron axons, dendrites, and somata. The effects of acetylcholine on axonal information processing, though, remain unknown. By stimulating cholinergic fibers and making electrophysiological recordings from hippocampal dentate gyrus granule cells, we show that synaptically released acetylcholine preferentially lowered the action potential threshold, enhancing intrinsic excitability and synaptic potential-spike coupling. These effects persisted for at least 30 min after the stimulation paradigm and were due to muscarinic receptor activation. This caused sustained elevation of axonal intracellular Ca2+ via T-type Ca2+ channels, as indicated by two-photon imaging. The enhanced Ca2+ levels inhibited an axonal KV7/M current, decreasing the spike threshold. In support, immunohistochemistry revealed muscarinic M1 receptor, CaV3.2, and KV7.2/7.3 subunit localization in granule cell axons. Since alterations in axonal signaling affect neuronal firing patterns and neurotransmitter release, this is an unreported cellular mechanism by which acetylcholine might, at least partly, enhance cognitive processing. Cholinergic fiber stimulation caused a persistent reduction in the spike threshold Post-synaptic muscarinic receptor activation enhanced axonal CaV3.2 channel activity The sustained Ca2+ entry inhibited axonal KV7 channels, lowering the spike threshold The lower spike threshold increased the propensity for action potential generation
Collapse
Affiliation(s)
| | - Zhuo Huang
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Rafael Lujan
- Departamento de Ciencias Medicas, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Baouyen Tran
- Neurology and Neuroscience, Baylor College of Medicine, TX 77030, USA
| | - Masahiko Watanabe
- Anatomy, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Edward C Cooper
- Neurology and Neuroscience, Baylor College of Medicine, TX 77030, USA
| | - David A Brown
- Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Mala M Shah
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
49
|
Dufour MA, Woodhouse A, Amendola J, Goaillard JM. Non-linear developmental trajectory of electrical phenotype in rat substantia nigra pars compacta dopaminergic neurons. eLife 2014; 3:e04059. [PMID: 25329344 PMCID: PMC4241557 DOI: 10.7554/elife.04059] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/19/2014] [Indexed: 12/12/2022] Open
Abstract
Neurons have complex electrophysiological properties, however, it is often difficult to determine which properties are the most relevant to neuronal function. By combining current-clamp measurements of electrophysiological properties with multi-variate analysis (hierarchical clustering, principal component analysis), we were able to characterize the postnatal development of substantia nigra dopaminergic neurons' electrical phenotype in an unbiased manner, such that subtle changes in phenotype could be analyzed. We show that the intrinsic electrical phenotype of these neurons follows a non-linear trajectory reaching maturity by postnatal day 14, with two developmental transitions occurring between postnatal days 3-5 and 9-11. This approach also predicted which parameters play a critical role in phenotypic variation, enabling us to determine (using pharmacology, dynamic-clamp) that changes in the leak, sodium and calcium-activated potassium currents are central to these two developmental transitions. This analysis enables an unbiased definition of neuronal type/phenotype that is applicable to a range of research questions.
Collapse
Affiliation(s)
- Martial A Dufour
- Inserm UMR 1072, Faculté de Médecine Secteur Nord, Université de la Méditerranée, Marseille, France
- Aix-Marseille Université, Marseille, France
| | - Adele Woodhouse
- Inserm UMR 1072, Faculté de Médecine Secteur Nord, Université de la Méditerranée, Marseille, France
- Aix-Marseille Université, Marseille, France
| | - Julien Amendola
- Inserm UMR 1072, Faculté de Médecine Secteur Nord, Université de la Méditerranée, Marseille, France
- Aix-Marseille Université, Marseille, France
| | - Jean-Marc Goaillard
- Inserm UMR 1072, Faculté de Médecine Secteur Nord, Université de la Méditerranée, Marseille, France
- Aix-Marseille Université, Marseille, France
| |
Collapse
|
50
|
Jones SL, Korobova F, Svitkina T. Axon initial segment cytoskeleton comprises a multiprotein submembranous coat containing sparse actin filaments. ACTA ACUST UNITED AC 2014; 205:67-81. [PMID: 24711503 PMCID: PMC3987141 DOI: 10.1083/jcb.201401045] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The axon initial segment of differentiated neurons contains a dense submembranous cytoskeleton that overlays microtubule bundles and includes two sparse actin populations: short, stable actin filaments and longer, dynamic non-oriented filaments. The axon initial segment (AIS) of differentiated neurons regulates action potential initiation and axon–dendritic polarity. The latter function depends on actin dynamics, but actin structure and functions at the AIS remain unclear. Using platinum replica electron microscopy (PREM), we have characterized the architecture of the AIS cytoskeleton in mature and developing hippocampal neurons. The AIS cytoskeleton assembly begins with bundling of microtubules and culminates in formation of a dense, fibrillar–globular coat over microtubule bundles. Immunogold PREM revealed that the coat contains a network of known AIS proteins, including ankyrin G, spectrin βIV, neurofascin, neuronal cell adhesion molecule, voltage-gated sodium channels, and actin filaments. Contrary to existing models, we find neither polarized actin arrays, nor dense actin meshworks in the AIS. Instead, the AIS contains two populations of sparse actin filaments: short, stable filaments and slightly longer dynamic filaments. We propose that stable actin filaments play a structural role for formation of the AIS diffusion barrier, whereas dynamic actin may promote AIS coat remodeling.
Collapse
Affiliation(s)
- Steven L Jones
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | | | | |
Collapse
|