51
|
Benusa SD, George NM, Sword BA, DeVries GH, Dupree JL. Acute neuroinflammation induces AIS structural plasticity in a NOX2-dependent manner. J Neuroinflammation 2017; 14:116. [PMID: 28595650 PMCID: PMC5465457 DOI: 10.1186/s12974-017-0889-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/25/2017] [Indexed: 02/08/2023] Open
Abstract
Background Chronic microglia-mediated inflammation and oxidative stress are well-characterized underlying factors in neurodegenerative disease, whereby reactive inflammatory microglia enhance ROS production and impact neuronal integrity. Recently, it has been shown that during chronic inflammation, neuronal integrity is compromised through targeted disruption of the axon initial segment (AIS), the axonal domain critical for action potential initiation. AIS disruption was associated with contact by reactive inflammatory microglia which wrap around the AIS, increasing association with disease progression. While it is clear that chronic microglial inflammation and enhanced ROS production impact neuronal integrity, little is known about how acute microglial inflammation influences AIS stability. Here, we demonstrate that acute neuroinflammation induces AIS structural plasticity in a ROS-mediated and calpain-dependent manner. Methods C57BL/6J and NOX2−/− mice were given a single injection of lipopolysaccharide (LPS; 5 mg/kg) or vehicle (0.9% saline, 10 mL/kg) and analyzed at 6 h–2 weeks post-injection. Anti-inflammatory Didox (250 mg/kg) or vehicle (0.9% saline, 10 mL/kg) was administered beginning 24 h post-LPS injection and continued for 5 days; animals were analyzed 1 week post-injection. Microglial inflammation was assessed using immunohistochemistry (IHC) and RT-qPCR, and AIS integrity was quantitatively analyzed using ankyrinG immunolabeling. Data were statistically compared by one-way or two-way ANOVA where mean differences were significant as assessed using Tukey’s post hoc analysis. Results LPS-induced neuroinflammation, characterized by enhanced microglial inflammation and increased expression of ROS-producing enzymes, altered AIS protein clustering. Importantly, inflammation-induced AIS changes were reversed following resolution of microglial inflammation. Modulation of the inflammatory response using anti-inflammatory Didox, even after significant AIS disruption occurred, increased the rate of AIS recovery. qPCR and IHC analysis revealed that expression of microglial NOX2, a ROS-producing enzyme, was significantly increased correlating with AIS disruption. Furthermore, ablation of NOX2 prevented inflammation-induced AIS plasticity, suggesting that ROS drive AIS structural plasticity. Conclusions In the presence of acute microglial inflammation, the AIS undergoes an adaptive change that is capable of spontaneous recovery. Moreover, recovery can be therapeutically accelerated. Together, these findings underscore the dynamic capabilities of this domain in the presence of a pathological insult and provide evidence that the AIS is a viable therapeutic target.
Collapse
Affiliation(s)
- S D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Curriculum, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - N M George
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Graduate Program, University of Colorado, Denver, CO, 80204, USA
| | - B A Sword
- Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - G H DeVries
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - J L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA. .,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA.
| |
Collapse
|
52
|
Vascak M, Sun J, Baer M, Jacobs KM, Povlishock JT. Mild Traumatic Brain Injury Evokes Pyramidal Neuron Axon Initial Segment Plasticity and Diffuse Presynaptic Inhibitory Terminal Loss. Front Cell Neurosci 2017. [PMID: 28634442 PMCID: PMC5459898 DOI: 10.3389/fncel.2017.00157] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The axon initial segment (AIS) is the site of action potential (AP) initiation, thus a crucial regulator of neuronal activity. In excitatory pyramidal neurons, the high density of voltage-gated sodium channels (NaV1.6) at the distal AIS regulates AP initiation. A surrogate AIS marker, ankyrin-G (ankG) is a structural protein regulating neuronal functional via clustering voltage-gated ion channels. In neuronal circuits, changes in presynaptic input can alter postsynaptic output via AIS structural-functional plasticity. Recently, we showed experimental mild traumatic brain injury (mTBI) evokes neocortical circuit disruption via diffuse axonal injury (DAI) of excitatory and inhibitory neuronal systems. A key finding was that mTBI-induced neocortical electrophysiological changes involved non-DAI/ intact excitatory pyramidal neurons consistent with AIS-specific alterations. In the current study we employed Thy1-yellow fluorescent protein (YFP)-H mice to test if mTBI induces AIS structural and/or functional plasticity within intact pyramidal neurons 2 days after mTBI. We used confocal microscopy to assess intact YFP+ pyramidal neurons in layer 5 of primary somatosensory barrel field (S1BF), whose axons were continuous from the soma of origin to the subcortical white matter (SCWM). YFP+ axonal traces were superimposed on ankG and NaV1.6 immunofluorescent profiles to determine AIS position and length. We found that while mTBI had no effect on ankG start position, the length significantly decreased from the distal end, consistent with the site of AP initiation at the AIS. However, NaV1.6 structure did not change after mTBI, suggesting uncoupling from ankG. Parallel quantitative analysis of presynaptic inhibitory terminals along the postsynaptic perisomatic domain of these same intact YFP+ excitatory pyramidal neurons revealed a significant decrease in GABAergic bouton density. Also within this non-DAI population, patch-clamp recordings of intact YFP+ pyramidal neurons showed AP acceleration decreased 2 days post-mTBI, consistent with AIS functional plasticity. Simulations of realistic pyramidal neuron computational models using experimentally determined AIS lengths showed a subtle decrease is NaV1.6 density is sufficient to attenuate AP acceleration. Collectively, these findings highlight the complexity of mTBI-induced neocortical circuit disruption, involving changes in extrinsic/presynaptic inhibitory perisomatic input interfaced with intrinsic/postsynaptic intact excitatory neuron AIS output.
Collapse
Affiliation(s)
- Michal Vascak
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of MedicineRichmond, VA, United States
| | - Jianli Sun
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of MedicineRichmond, VA, United States
| | - Matthew Baer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of MedicineRichmond, VA, United States
| | - Kimberle M Jacobs
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of MedicineRichmond, VA, United States
| | - John T Povlishock
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of MedicineRichmond, VA, United States
| |
Collapse
|
53
|
Zempel H, Dennissen FJA, Kumar Y, Luedtke J, Biernat J, Mandelkow EM, Mandelkow E. Axodendritic sorting and pathological missorting of Tau are isoform-specific and determined by axon initial segment architecture. J Biol Chem 2017; 292:12192-12207. [PMID: 28536263 DOI: 10.1074/jbc.m117.784702] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 05/21/2017] [Indexed: 11/06/2022] Open
Abstract
Subcellular mislocalization of the microtubule-associated protein Tau is a hallmark of Alzheimer disease (AD) and other tauopathies. Six Tau isoforms, differentiated by the presence or absence of a second repeat or of N-terminal inserts, exist in the human CNS, but their physiological and pathological differences have long remained elusive. Here, we investigated the properties and distributions of human and rodent Tau isoforms in primary forebrain rodent neurons. We found that the Tau diffusion barrier (TDB), located within the axon initial segment (AIS), controls retrograde (axon-to-soma) and anterograde (soma-to-axon) traffic of Tau. Tau isoforms without the N-terminal inserts were sorted efficiently into the axon. However, the longest isoform (2N4R-Tau) was partially retained in cell bodies and dendrites, where it accelerated spine and dendrite growth. The TDB (located within the AIS) was impaired when AIS components (ankyrin G, EB1) were knocked down or when glycogen synthase kinase-3β (GSK3β; an AD-associated kinase tethered to the AIS) was overexpressed. Using superresolution nanoscopy and live-cell imaging, we observed that microtubules within the AIS appeared highly dynamic, a feature essential for the TDB. Pathomechanistically, amyloid-β insult caused cofilin activation and F-actin remodeling and decreased microtubule dynamics in the AIS. Concomitantly with these amyloid-β-induced disruptions, the AIS/TDB sorting function failed, causing AD-like Tau missorting. In summary, we provide evidence that the human and rodent Tau isoforms differ in axodendritic sorting and amyloid-β-induced missorting and that the axodendritic distribution of Tau depends on AIS integrity.
Collapse
Affiliation(s)
- Hans Zempel
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany; Max Planck Institute for Metabolism Research, 50931 Cologne, Germany; University of Bonn, 53113 Bonn, Germany.
| | - Frank J A Dennissen
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany
| | - Yatender Kumar
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Julia Luedtke
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany
| | - Jacek Biernat
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany
| | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany; Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; CAESAR Research Center, 53175 Bonn, Germany; Max Planck Institute for Metabolism Research, 50931 Cologne, Germany.
| |
Collapse
|
54
|
Abstract
Structural plasticity of the axon initial segment (AIS), the site of action potential initiation, is observed as part of the normal early development of the cortex, as well as in association with injury and disease. Here, we show that structural AIS plasticity also occurs with normal aging in adult marmosets. Immunohistochemical techniques were used to reveal the extent of the AIS of layer 2/3A pyramidal cells in 8 neocortical areas. We found that the AIS length varied significantly between areas in young adult (2-3 years old) marmosets, with neurons in frontal area 14C having the longest AIS, and those in the primary visual cortex the shortest. Similar interareal differences were observed in aged (12-14 year old) monkeys, but the AIS was significantly shortened in many areas, relative to the corresponding length in young adults. Shortening of the AIS is likely to represent a compensatory response to changes in the excitation-inhibition balance, associated with the loss of GABAergic interneurons in the aged cortex.
Collapse
Affiliation(s)
- Nafiseh Atapour
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia; Australian Research Council, Centre of Excellence for Integrative Brain Function, Monash University Node, Melbourne, Victoria, Australia.
| | - Marcello G P Rosa
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia; Australian Research Council, Centre of Excellence for Integrative Brain Function, Monash University Node, Melbourne, Victoria, Australia
| |
Collapse
|
55
|
Duménieu M, Oulé M, Kreutz MR, Lopez-Rojas J. The Segregated Expression of Voltage-Gated Potassium and Sodium Channels in Neuronal Membranes: Functional Implications and Regulatory Mechanisms. Front Cell Neurosci 2017; 11:115. [PMID: 28484374 PMCID: PMC5403416 DOI: 10.3389/fncel.2017.00115] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/05/2017] [Indexed: 01/25/2023] Open
Abstract
Neurons are highly polarized cells with apparent functional and morphological differences between dendrites and axon. A critical determinant for the molecular and functional identity of axonal and dendritic segments is the restricted expression of voltage-gated ion channels (VGCs). Several studies show an uneven distribution of ion channels and their differential regulation within dendrites and axons, which is a prerequisite for an appropriate integration of synaptic inputs and the generation of adequate action potential (AP) firing patterns. This review article will focus on the signaling pathways leading to segmented expression of voltage-gated potassium and sodium ion channels at the neuronal plasma membrane and the regulatory mechanisms ensuring segregated functions. We will also discuss the relevance of proper ion channel targeting for neuronal physiology and how alterations in polarized distribution contribute to neuronal pathology.
Collapse
Affiliation(s)
- Maël Duménieu
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Marie Oulé
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH)Hamburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| |
Collapse
|
56
|
Yamada R, Kuba H. Structural and Functional Plasticity at the Axon Initial Segment. Front Cell Neurosci 2016; 10:250. [PMID: 27826229 PMCID: PMC5078684 DOI: 10.3389/fncel.2016.00250] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/12/2016] [Indexed: 11/13/2022] Open
Abstract
The axon initial segment (AIS) is positioned between the axonal and somato-dendritic compartments and plays a pivotal role in triggering action potentials (APs) and determining neuronal output. It is now widely accepted that structural properties of the AIS, such as length and/or location relative to the soma, change in an activity-dependent manner. This structural plasticity of the AIS is known to be crucial for homeostatic control of neuronal excitability. However, it is obvious that the impact of the AIS on neuronal excitability is critically dependent on the biophysical properties of the AIS, which are primarily determined by the composition and characteristics of ion channels in this domain. Moreover, these properties can be altered via phosphorylation and/or redistribution of the channels. Recently, studies in auditory neurons showed that alterations in the composition of voltage-gated K+ (Kv) channels at the AIS coincide with elongation of the AIS, thereby enhancing the neuronal excitability, suggesting that the interaction between structural and functional plasticities of the AIS is important in the control of neuronal excitability. In this review, we will summarize the current knowledge regarding structural and functional alterations of the AIS and discuss how they interact and contribute to regulating the neuronal output.
Collapse
Affiliation(s)
- Rei Yamada
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| |
Collapse
|
57
|
Griggs RB, Yermakov LM, Susuki K. Formation and disruption of functional domains in myelinated CNS axons. Neurosci Res 2016; 116:77-87. [PMID: 27717670 DOI: 10.1016/j.neures.2016.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022]
Abstract
Communication in the central nervous system (CNS) occurs through initiation and propagation of action potentials at excitable domains along axons. Action potentials generated at the axon initial segment (AIS) are regenerated at nodes of Ranvier through the process of saltatory conduction. Proper formation and maintenance of the molecular structure at the AIS and nodes are required for sustaining conduction fidelity. In myelinated CNS axons, paranodal junctions between the axolemma and myelinating oligodendrocytes delineate nodes of Ranvier and regulate the distribution and localization of specialized functional elements, such as voltage-gated sodium channels and mitochondria. Disruption of excitable domains and altered distribution of functional elements in CNS axons is associated with demyelinating diseases such as multiple sclerosis, and is likely a mechanism common to other neurological disorders. This review will provide a brief overview of the molecular structure of the AIS and nodes of Ranvier, as well as the distribution of mitochondria in myelinated axons. In addition, this review highlights important structural and functional changes within myelinated CNS axons that are associated with neurological dysfunction.
Collapse
Affiliation(s)
- Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States.
| |
Collapse
|
58
|
Axon Initial Segment Cytoskeleton: Architecture, Development, and Role in Neuron Polarity. Neural Plast 2016; 2016:6808293. [PMID: 27493806 PMCID: PMC4967436 DOI: 10.1155/2016/6808293] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/22/2016] [Indexed: 12/28/2022] Open
Abstract
The axon initial segment (AIS) is a specialized structure in neurons that resides in between axonal and somatodendritic domains. The localization of the AIS in neurons is ideal for its two major functions: it serves as the site of action potential firing and helps to maintain neuron polarity. It has become increasingly clear that the AIS cytoskeleton is fundamental to AIS functions. In this review, we discuss current understanding of the AIS cytoskeleton with particular interest in its unique architecture and role in maintenance of neuron polarity. The AIS cytoskeleton is divided into two parts, submembrane and cytoplasmic, based on localization, function, and molecular composition. Recent studies using electron and subdiffraction fluorescence microscopy indicate that submembrane cytoskeletal components (ankyrin G, βIV-spectrin, and actin filaments) form a sophisticated network in the AIS that is conceptually similar to the polygonal/triangular network of erythrocytes, with some important differences. Components of the AIS cytoplasmic cytoskeleton (microtubules, actin filaments, and neurofilaments) reside deeper within the AIS shaft and display structural features distinct from other neuronal domains. We discuss how the AIS submembrane and cytoplasmic cytoskeletons contribute to different aspects of AIS polarity function and highlight recent advances in understanding their AIS cytoskeletal assembly and stability.
Collapse
|
59
|
Sohn PD, Tracy TE, Son HI, Zhou Y, Leite REP, Miller BL, Seeley WW, Grinberg LT, Gan L. Acetylated tau destabilizes the cytoskeleton in the axon initial segment and is mislocalized to the somatodendritic compartment. Mol Neurodegener 2016; 11:47. [PMID: 27356871 PMCID: PMC4928318 DOI: 10.1186/s13024-016-0109-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/01/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Neurons are highly polarized cells in which asymmetric axonal-dendritic distribution of proteins is crucial for neuronal function. Loss of polarized distribution of the axonal protein tau is an early sign of Alzheimer's disease (AD) and other neurodegenerative disorders. The cytoskeletal network in the axon initial segment (AIS) forms a barrier between the axon and the somatodentritic compartment, contributing to axonal retention of tau. Although perturbation of the AIS cytoskeleton has been implicated in neurological disorders, the molecular triggers and functional consequence of AIS perturbation are incompletely understood. RESULTS Here we report that tau acetylation and consequent destabilization of the AIS cytoskeleton promote the somatodendritic mislocalization of tau. AIS cytoskeletal proteins, including ankyrin G and βIV-spectrin, were downregulated in AD brains and negatively correlated with an increase in tau acetylated at K274 and K281. AIS proteins were also diminished in transgenic mice expressing tauK274/281Q, a tau mutant that mimics K274 and K281 acetylation. In primary neuronal cultures, the tauK274/281Q mutant caused hyperdynamic microtubules (MTs) in the AIS, shown by live-imaging of MT mobility and fluorescence recovery after photobleaching. Using photoconvertible tau constructs, we found that axonal tauK274/281Q was missorted into the somatodendritic compartment. Stabilizing MTs with epothilone D to restore the cytoskeletal barrier in the AIS prevented tau mislocalization in primary neuronal cultures. CONCLUSIONS Together, these findings demonstrate that tau acetylation contributes to the pathogenesis of neurodegenerative disease by compromising the cytoskeletal sorting machinery in the AIS.
Collapse
Affiliation(s)
- Peter Dongmin Sohn
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA
| | - Tara E Tracy
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Hye-In Son
- Gladstone Institute of Virology & Immunology, San Francisco, CA, 94158, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA
| | - Renata E P Leite
- Physiopathology in Aging Lab/Brazilian Aging Brain Study Group-LIM22, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology University of California, San Francisco, CA, 94158, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, 94158, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
60
|
Clark KC, Josephson A, Benusa SD, Hartley RK, Baer M, Thummala S, Joslyn M, Sword BA, Elford H, Oh U, Dilsizoglu-Senol A, Lubetzki C, Davenne M, DeVries GH, Dupree JL. Compromised axon initial segment integrity in EAE is preceded by microglial reactivity and contact. Glia 2016; 64:1190-209. [PMID: 27100937 DOI: 10.1002/glia.22991] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/11/2022]
Abstract
Axonal pathology is a key contributor to long-term disability in multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS), but the mechanisms that underlie axonal pathology in MS remain elusive. Evidence suggests that axonal pathology is a direct consequence of demyelination, as we and others have shown that the node of Ranvier disassembles following loss of myelin. In contrast to the node of Ranvier, we now show that the axon initial segment (AIS), the axonal domain responsible for action potential initiation, remains intact following cuprizone-induced cortical demyelination. Instead, we find that the AIS is disrupted in the neocortex of mice that develop experimental autoimmune encephalomyelitis (EAE) independent of local demyelination. EAE-induced mice demonstrate profound compromise of AIS integrity with a progressive disruption that corresponds to EAE clinical disease severity and duration, in addition to cortical microglial reactivity. Furthermore, treatment with the drug didox results in attenuation of AIS pathology concomitantly with microglial reversion to a less reactive state. Together, our findings suggest that inflammation, but not demyelination, disrupts AIS integrity and that therapeutic intervention may protect and reverse this pathology. GLIA 2016;64:1190-1209.
Collapse
Affiliation(s)
- Kareem C Clark
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia.,VCU, Neuroscience Curriculum, Richmond, Virginia
| | - Anna Josephson
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Savannah D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia.,VCU, Neuroscience Curriculum, Richmond, Virginia
| | - Rebecca K Hartley
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Matthew Baer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Suneel Thummala
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Martha Joslyn
- Department of Research,, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia
| | - Brooke A Sword
- Department of Research,, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia
| | | | - Unsong Oh
- Department of Neurology, VCU, Richmond, Virginia
| | - Aysegul Dilsizoglu-Senol
- UPMC/Univ Paris 06 UMR S 1127, Institut Du Cerveau Et De La Moelle Épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, Paris, F-75013, France
| | - Catherine Lubetzki
- UPMC/Univ Paris 06 UMR S 1127, Institut Du Cerveau Et De La Moelle Épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, Paris, F-75013, France.,AP-HP, Hôpital De La Pitié Salpêtrière, Paris, F-75013, France
| | - Marc Davenne
- UPMC/Univ Paris 06 UMR S 1127, Institut Du Cerveau Et De La Moelle Épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, Paris, F-75013, France
| | - George H DeVries
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia.,Department of Research,, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia.,Department of Research,, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia
| |
Collapse
|
61
|
Marin MA, Ziburkus J, Jankowsky J, Rasband MN. Amyloid-β plaques disrupt axon initial segments. Exp Neurol 2016; 281:93-8. [PMID: 27109181 DOI: 10.1016/j.expneurol.2016.04.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/14/2016] [Accepted: 04/20/2016] [Indexed: 11/12/2022]
Abstract
UNLABELLED Amyloid-β (Aβ) plaques are one of the central pathologies of Alzheimer's disease (AD). Plaque formation in animal models of AD coincides with the appearance of synaptic abnormalities, aberrant neuronal excitability, and cognitive decline. Aβ plaques may disrupt neuronal excitability since they have been proposed to be synaptotoxic, to induce axonal varicosities and neurite breakage, and to significantly decrease spine density. Axon initial segments (AIS) also regulate neuronal excitability and help maintain neuronal polarity. Despite these essential functions, the effects of plaques on AIS structure have not been fully determined. Using a mouse AD model, we measured a significant decrease in the density of AIS up to 75μm away from the center of fibrillar, thioflavin-labeled plaques. The reduction was observed in animals with both moderate and severe plaque loads, and was associated with increased densities of microglia near the plaques. Furthermore, animals with severe plaque loads had significantly reduced AIS lengths adjacent to Aβ plaques. These results suggest the local environment surrounding Aβ plaques may be harmful to the AIS. We propose that AIS loss is a previously unappreciated consequence of AD that could significantly impact brain function. SIGNIFICANCE STATEMENT This paper demonstrates that neurons near Aβ plaques have disrupted axon initial segments. Loss or disruption of AIS is predicted to have detrimental consequences for brain function.
Collapse
Affiliation(s)
- Miguel A Marin
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jokubus Ziburkus
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Joanna Jankowsky
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
62
|
Susuki K, Kuba H. Activity-dependent regulation of excitable axonal domains. J Physiol Sci 2016; 66:99-104. [PMID: 26464228 PMCID: PMC10717305 DOI: 10.1007/s12576-015-0413-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 09/30/2015] [Indexed: 01/04/2023]
Abstract
Rapid action potential propagation along myelinated axons requires voltage-gated Na(+) channel clustering at the axon initial segments (AISs) and nodes of Ranvier. The AIS is intrinsically defined by cytoskeletal proteins expressed in axons, whereas nodes of Ranvier are formed by interaction between neurons and myelinating glia. These axonal domains have long been considered stable structures, but recent studies revealed that they are plastic and contribute to fine adjustment of neuronal activities and circuit function. The AIS changes its distribution and maintains neural circuit activity at a constant level. Morphological changes in myelinated nerve structures presumably modulate the excitability of nodal regions and regulate the timing of activity, thereby optimizing signal processing in a neural circuit. This review highlights recent findings on the structural plasticity of these excitable axonal domains.
Collapse
Affiliation(s)
- Keiichiro Susuki
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH, 45435, USA.
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
- PRESTO, JST, Saitama, Japan.
| |
Collapse
|
63
|
Alshammari MA, Alshammari TK, Laezza F. Improved Methods for Fluorescence Microscopy Detection of Macromolecules at the Axon Initial Segment. Front Cell Neurosci 2016; 10:5. [PMID: 26909021 PMCID: PMC4754416 DOI: 10.3389/fncel.2016.00005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
Abstract
The axonal initial segment (AIS) is the subcellular compartment required for initiation of the action potential in neurons. Scaffolding and regulatory proteins at the AIS cluster with ion channels ensuring the integrity of electrical signaling. Interference with the configuration of this protein network can lead to profound effects on neuronal polarity, excitability, cell-to-cell connectivity and brain circuit plasticity. As such, the ability to visualize AIS components with precision provides an invaluable opportunity for parsing out key molecular determinants of neuronal function. Fluorescence-based immunolabeling is a sensitive method for morphological and molecular characterization of fine structures in neurons. Yet, even when combined with confocal microscopy, detection of AIS elements with immunofluorescence has been limited by the loss of antigenicity caused by fixative materials. This technical barrier has posed significant limitations in detecting AIS components alone or in combination with other markers. Here, we designed improved protocols targeted to confocal immunofluorescence detection of the AIS marker fibroblast growth factor 14 (FGF14) in combination with the cytoskeletal-associated protein Ankyrin-G, the scaffolding protein βIV-spectrin, voltage-gated Na+ (Nav) channels (especially the Nav1.6 isoform) and critical cell type-specific neuronal markers such as parvalbumin, calbindin, and NeuN in the mouse brain. Notably, we demonstrate that intracardiac perfusion of animals with a commercially available solution containing 1% formaldehyde and 0.5% methanol, followed by brief fixation with cold acetone is an optimal and sensitive protocol for FGF14 and other AIS marker detection that guarantees excellent tissue integrity. With variations in the procedure, we also significantly improved the detection of Nav1.6, a Nav isoform known for its fixative-sensitivity. Overall, this study provides an ensemble of immunohistochemical recipes that permit excellent staining of otherwise invisible molecules within well-preserved tissue architecture. While improving the specific investigation of AIS physiology and cell biology, our thorough study can also serve as a roadmap for optimizing immunodetection of other fixative-sensitive proteins expanding the repertoire of enabling methods for brain studies.
Collapse
Affiliation(s)
- Musaad A Alshammari
- Graduate Studies Abroad Program, King Saud UniversityRiyadh, Saudi Arabia; Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA
| | - Tahani K Alshammari
- Graduate Studies Abroad Program, King Saud UniversityRiyadh, Saudi Arabia; Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA; Mitchell Center for Neurodegenerative Diseases, University of Texas Medical BranchGalveston, TX, USA; Center for Addiction Research, University of Texas Medical BranchGalveston, TX, USA; Center for Biomedical Engineering, University of Texas Medical BranchGalveston, TX, USA
| |
Collapse
|
64
|
|
65
|
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
|
66
|
Miras-Portugal MT, Gomez-Villafuertes R, Gualix J, Diaz-Hernandez JI, Artalejo AR, Ortega F, Delicado EG, Perez-Sen R. Nucleotides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:243-54. [PMID: 26359530 DOI: 10.1016/j.neuropharm.2015.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 02/03/2023]
Abstract
Brain injury generates the release of a multitude of factors including extracellular nucleotides, which exhibit bi-functional properties and contribute to both detrimental actions in the acute phase and also protective and reparative actions in the later recovery phase to allow neuroregeneration. A promising strategy toward restoration of neuronal function is based on activation of endogenous adult neural stem/progenitor cells. The implication of purinergic signaling in stem cell biology, including regulation of proliferation, differentiation, and cell death has become evident in the last decade. In this regard, current strategies of acute transplantation of ependymal stem/progenitor cells after spinal cord injury restore altered expression of P2X4 and P2X7 receptors and improve functional locomotor recovery. The expression of both receptors is transcriptionally regulated by Sp1 factor, which plays a key role in the startup of the transcription machinery to induce regeneration-associated genes expression. Finally, general signaling pathways triggered by nucleotide receptors in neuronal populations converge on several intracellular kinases, such as PI3K/Akt, GSK3 and ERK1,2, as well as the Nrf-2/heme oxigenase-1 axis, which specifically link them to neuroprotection. In this regard, regulation of dual specificity protein phosphatases can become novel mechanism of actions for nucleotide receptors that associate them to cell homeostasis regulation. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- M Teresa Miras-Portugal
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Rosa Gomez-Villafuertes
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain.
| | - Javier Gualix
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Juan Ignacio Diaz-Hernandez
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Antonio R Artalejo
- Department of Toxicology and Pharmacology, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Felipe Ortega
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Esmerilda G Delicado
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| | - Raquel Perez-Sen
- Department of Biochemistry and Molecular Biology IV, Veterinary School, Universidad Complutense of Madrid, 28040 Madrid, Spain
| |
Collapse
|
67
|
Myelin loss and axonal ion channel adaptations associated with gray matter neuronal hyperexcitability. J Neurosci 2015; 35:7272-86. [PMID: 25948275 DOI: 10.1523/jneurosci.4747-14.2015] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Myelination and voltage-gated ion channel clustering at the nodes of Ranvier are essential for the rapid saltatory conduction of action potentials. Whether myelination influences the structural organization of the axon initial segment (AIS) and action potential initiation is poorly understood. Using the cuprizone mouse model, we combined electrophysiological recordings with immunofluorescence of the voltage-gated Nav1.6 and Kv7.3 subunits and anchoring proteins to analyze the functional and structural properties of single demyelinated neocortical L5 axons. Whole-cell recordings demonstrated that neurons with demyelinated axons were intrinsically more excitable, characterized by increased spontaneous suprathreshold depolarizations as well as antidromically propagating action potentials ectopically generated in distal parts of the axon. Immunofluorescence examination of demyelinated axons showed that βIV-spectrin, Nav1.6, and the Kv7.3 channels in nodes of Ranvier either dissolved or extended into the paranodal domains. In contrast, while the AIS in demyelinated axons started more closely to the soma, ankyrin G, βIV-spectrin, and the ion channel expression were maintained. Structure-function analysis and computational modeling, constrained by the AIS location and realistic dendritic and axonal morphologies, confirmed that a more proximal onset of the AIS slightly reduced the efficacy of action potential generation, suggesting a compensatory role. These results suggest that oligodendroglial myelination is not only important for maximizing conduction velocity, but also for limiting hyperexcitability of pyramidal neurons.
Collapse
|
68
|
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
|
69
|
Trimmer JS. Subcellular localization of K+ channels in mammalian brain neurons: remarkable precision in the midst of extraordinary complexity. Neuron 2015; 85:238-56. [PMID: 25611506 DOI: 10.1016/j.neuron.2014.12.042] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium channels (KChs) are the most diverse ion channels, in part due to extensive combinatorial assembly of a large number of principal and auxiliary subunits into an assortment of KCh complexes. Their structural and functional diversity allows KChs to play diverse roles in neuronal function. Localization of KChs within specialized neuronal compartments defines their physiological role and also fundamentally impacts their activity, due to localized exposure to diverse cellular determinants of channel function. Recent studies in mammalian brain reveal an exquisite refinement of KCh subcellular localization. This includes axonal KChs at the initial segment, and near/within nodes of Ranvier and presynaptic terminals, dendritic KChs found at sites reflecting specific synaptic input, and KChs defining novel neuronal compartments. Painting the remarkable diversity of KChs onto the complex architecture of mammalian neurons creates an elegant picture of electrical signal processing underlying the sophisticated function of individual neuronal compartments, and ultimately neurotransmission and behavior.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
70
|
Balaraman Y, Lahiri DK, Nurnberger JI. Variants in Ion Channel Genes Link Phenotypic Features of Bipolar Illness to Specific Neurobiological Process Domains. MOLECULAR NEUROPSYCHIATRY 2015; 1:23-35. [PMID: 27602355 DOI: 10.1159/000371886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022]
Abstract
Recent advances in genome-wide association studies are pointing towards a major role for voltage-gated ion channels in neuropsychiatric disorders and, in particular, bipolar disorder (BD). The phenotype of BD is complex, with symptoms during mood episodes and deficits persisting between episodes. We have tried to elucidate the common neurobiological mechanisms associated with ion channel signaling in order to provide a new perspective on the clinical symptoms and possible endophenotypes seen in BD patients. We propose a model in which the multiple variants in genes coding for ion channel proteins would perturb motivational circuits, synaptic plasticity, myelination, hypothalamic-pituitary-adrenal axis function, circadian neuronal rhythms, and energy regulation. These changes in neurobiological mechanisms would manifest in endophenotypes of aberrant reward processing, white matter hyperintensities, deficits in executive function, altered frontolimbic connectivity, increased amygdala activity, increased melatonin suppression, decreased REM latency, and aberrant myo-inositol/ATP shuttling. The endophenotypes result in behaviors of poor impulse control, motivational changes, cognitive deficits, abnormal stress response, sleep disturbances, and energy changes involving different neurobiological process domains. The hypothesis is that these disturbances start with altered neural circuitry during development, following which multiple environmental triggers may disrupt the neuronal excitability balance through an activity-dependent molecular process, resulting in clinical mood episodes.
Collapse
Affiliation(s)
- Yokesh Balaraman
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - Debomoy K Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - John I Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| |
Collapse
|
71
|
Identifying a kinase network regulating FGF14:Nav1.6 complex assembly using split-luciferase complementation. PLoS One 2015; 10:e0117246. [PMID: 25659151 PMCID: PMC4319734 DOI: 10.1371/journal.pone.0117246] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022] Open
Abstract
Kinases play fundamental roles in the brain. Through complex signaling pathways, kinases regulate the strength of protein:protein interactions (PPI) influencing cell cycle, signal transduction, and electrical activity of neurons. Changes induced by kinases on neuronal excitability, synaptic plasticity and brain connectivity are linked to complex brain disorders, but the molecular mechanisms underlying these cellular events remain for the most part elusive. To further our understanding of brain disease, new methods for rapidly surveying kinase pathways in the cellular context are needed. The bioluminescence-based luciferase complementation assay (LCA) is a powerful, versatile toolkit for the exploration of PPI. LCA relies on the complementation of two firefly luciferase protein fragments that are functionally reconstituted into the full luciferase enzyme by two interacting binding partners. Here, we applied LCA in live cells to assay 12 kinase pathways as regulators of the PPI complex formed by the voltage-gated sodium channel, Nav1.6, a transmembrane ion channel that elicits the action potential in neurons and mediates synaptic transmission, and its multivalent accessory protein, the fibroblast growth factor 14 (FGF14). Through extensive dose-dependent validations of structurally-diverse kinase inhibitors and hierarchical clustering, we identified the PI3K/Akt pathway, the cell-cycle regulator Wee1 kinase, and protein kinase C (PKC) as prospective regulatory nodes of neuronal excitability through modulation of the FGF14:Nav1.6 complex. Ingenuity Pathway Analysis shows convergence of these pathways on glycogen synthase kinase 3 (GSK3) and functional assays demonstrate that inhibition of GSK3 impairs excitability of hippocampal neurons. This combined approach provides a versatile toolkit for rapidly surveying PPI signaling, allowing the discovery of new modular pathways centered on GSK3 that might be the basis for functional alterations between the normal and diseased brain.
Collapse
|
72
|
Liu Y, Zhang Y, Wang JH. Crystal structure of human Ankyrin G death domain. Proteins 2014; 82:3476-82. [PMID: 25307106 DOI: 10.1002/prot.24702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/29/2014] [Accepted: 10/04/2014] [Indexed: 11/07/2022]
Abstract
Ankyrins (Ank) are a ubiquitously expressed family of multifunctional membrane adapter proteins. Ankyrin G (AnkG) is critical for assembling and maintenance of the axon initial segment. Here we present the 2.1 Å crystal structure of human AnkG death domain (hAnkG-DD). The core death domain is composed of six α-helices and three 3₁₀-helices. It forms a hydrophobic pocket on the surface of the molecule. The C-terminal tail of the hAnkG-DD curves back to have the aromatic ring of a phenylalanine residue, Phe100 insert into this pocket, which anchors the flexible tail onto the core domain. Related DDs were selected for structure comparison. The major variations are at the C-terminal region, including the α6 and the long C-terminal extension. The results of size exclusion chromatography and analytical ultracentrifugation suggest that hAnkG-DD exists as monomer in solution. Our work should help for the future investigation of the structure-function of AnkG.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China
| | | | | |
Collapse
|
73
|
Selective filtering defect at the axon initial segment in Alzheimer's disease mouse models. Proc Natl Acad Sci U S A 2014; 111:14271-6. [PMID: 25232037 DOI: 10.1073/pnas.1411837111] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Axon pathology has been widely reported in Alzheimer's disease (AD) patients and AD mouse models. Herein we report that increased miR-342-5p down-regulates the expression of ankyrin G (AnkG), a protein known to play a critical role in establishing selective filtering machinery at the axon initial segment (AIS). Diminished AnkG expression leads to defective AIS filtering in cultured hippocampal neurons from AD mouse models, as monitored by selective exclusion of large macromolecules from the axons. Furthermore, AnkG-deficiency impairs AIS localization of Nav 1.6 channels and confines NR2B to the somatodendritic compartments. The expression of exogenous AnkG improved the cognitive performance of 12-mo-old APP/PS1 mice; thus, our data suggest that AnkG and impairment of AIS filtering may play important roles in AD pathology.
Collapse
|
74
|
Abstract
The axon initial segment (AIS) is a specialized axonal compartment that is involved in conversion of synaptic potentials into action potentials. Recent studies revealed that structural properties of the AIS, such as length and position relative to the soma, are differentiated in a cell-specific manner and shape signal processing of individual neurons. Moreover, these structural properties are not fixed but vary in response to prolonged changes of neuronal activity, which readjusts action potential threshold and compensates for the changes of activity, indicating that this structural plasticity of the AIS works as a homeostatic mechanism and contributes to maintain neuronal activity. Neuronal activity plays a crucial role in formation, maintenance, and refinement of neural circuits as well as in pathogenesis and/or pathophysiology of diseases. Thus, this plasticity should be a key to understand physiology and pathology of the brain.
Collapse
Affiliation(s)
- Ryota Adachi
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Rei Yamada
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- PRESTO, JST, Saitama, Japan
| |
Collapse
|
75
|
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
|
76
|
Del Puerto A, Fronzaroli-Molinieres L, Perez-Alvarez MJ, Giraud P, Carlier E, Wandosell F, Debanne D, Garrido JJ. ATP-P2X7 Receptor Modulates Axon Initial Segment Composition and Function in Physiological Conditions and Brain Injury. Cereb Cortex 2014; 25:2282-94. [PMID: 24610121 DOI: 10.1093/cercor/bhu035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Axon properties, including action potential initiation and modulation, depend on both AIS integrity and the regulation of ion channel expression in the AIS. Alteration of the axon initial segment (AIS) has been implicated in neurodegenerative, psychiatric, and brain trauma diseases, thus identification of the physiological mechanisms that regulate the AIS is required to understand and circumvent AIS alterations in pathological conditions. Here, we show that the purinergic P2X7 receptor and its agonist, adenosine triphosphate (ATP), modulate both structural proteins and ion channel density at the AIS in cultured neurons and brain slices. In cultured hippocampal neurons, an increment of extracellular ATP concentration or P2X7-green fluorescent protein (GFP) expression reduced the density of ankyrin G and voltage-gated sodium channels at the AIS. This effect is mediated by P2X7-regulated calcium influx and calpain activation, and impaired by P2X7 inhibition with Brilliant Blue G (BBG), or P2X7 suppression. Electrophysiological studies in brain slices showed that P2X7-GFP transfection decreased both sodium current amplitude and intrinsic neuronal excitability, while P2X7 inhibition had the opposite effect. Finally, inhibition of P2X7 with BBG prevented AIS disruption after ischemia/reperfusion in rats. In conclusion, our study demonstrates an involvement of P2X7 receptors in the regulation of AIS mediated neuronal excitability in physiological and pathological conditions.
Collapse
Affiliation(s)
- Ana Del Puerto
- Instituto Cajal, CSIC, Department of Cellular, Molecular and Developmental Neurobiology, Madrid 28002, Spain Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laure Fronzaroli-Molinieres
- Institut National de la Santé et de la Recherche Médicale, U1072, Marseille F-13344 France Aix-Marseille Université, Faculté de Médecine Secteur Nord, Marseille F-13344 France
| | - María José Perez-Alvarez
- Departamento de Biología (Unidad Docente Fisiología Animal), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Pierre Giraud
- Institut National de la Santé et de la Recherche Médicale, U1072, Marseille F-13344 France Aix-Marseille Université, Faculté de Médecine Secteur Nord, Marseille F-13344 France
| | - Edmond Carlier
- Institut National de la Santé et de la Recherche Médicale, U1072, Marseille F-13344 France Aix-Marseille Université, Faculté de Médecine Secteur Nord, Marseille F-13344 France
| | - Francisco Wandosell
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain Centro de Biología Molecular, CSIC-UAM, Madrid 28049 Spain
| | - Dominique Debanne
- Institut National de la Santé et de la Recherche Médicale, U1072, Marseille F-13344 France Aix-Marseille Université, Faculté de Médecine Secteur Nord, Marseille F-13344 France
| | - Juan José Garrido
- Instituto Cajal, CSIC, Department of Cellular, Molecular and Developmental Neurobiology, Madrid 28002, Spain Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
77
|
Integrative biological analysis for neuropsychopharmacology. Neuropsychopharmacology 2014; 39:5-23. [PMID: 23800968 PMCID: PMC3857644 DOI: 10.1038/npp.2013.156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 04/18/2013] [Accepted: 04/19/2013] [Indexed: 01/24/2023]
Abstract
Although advances in psychotherapy have been made in recent years, drug discovery for brain diseases such as schizophrenia and mood disorders has stagnated. The need for new biomarkers and validated therapeutic targets in the field of neuropsychopharmacology is widely unmet. The brain is the most complex part of human anatomy from the standpoint of number and types of cells, their interconnections, and circuitry. To better meet patient needs, improved methods to approach brain studies by understanding functional networks that interact with the genome are being developed. The integrated biological approaches--proteomics, transcriptomics, metabolomics, and glycomics--have a strong record in several areas of biomedicine, including neurochemistry and neuro-oncology. Published applications of an integrated approach to projects of neurological, psychiatric, and pharmacological natures are still few but show promise to provide deep biological knowledge derived from cells, animal models, and clinical materials. Future studies that yield insights based on integrated analyses promise to deliver new therapeutic targets and biomarkers for personalized medicine.
Collapse
|
78
|
Sun X, Wu Y, Gu M, Zhang Y. miR-342-5p Decreases Ankyrin G Levels in Alzheimer’s Disease Transgenic Mouse Models. Cell Rep 2014; 6:264-70. [DOI: 10.1016/j.celrep.2013.12.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/22/2013] [Accepted: 12/17/2013] [Indexed: 12/14/2022] Open
|
79
|
Abstract
Dysfunction and/or disruption of nodes of Ranvier are now recognized as key contributors to the pathophysiology of various neurological diseases. One reason is that the excitable nodal axolemma contains a high density of Nav (voltage-gated Na+ channels) that are required for the rapid and efficient saltatory conduction of action potentials. Nodal physiology is disturbed by altered function, localization, and expression of voltage-gated ion channels clustered at nodes and juxtaparanodes, and by disrupted axon–glial interactions at paranodes. This paper reviews recent discoveries in molecular/cellular neuroscience, genetics, immunology, and neurology that highlight the critical roles of nodes of Ranvier in health and disease.
Collapse
|
80
|
Lin YC, Phua SC, Lin B, Inoue T. Visualizing molecular diffusion through passive permeability barriers in cells: conventional and novel approaches. Curr Opin Chem Biol 2013; 17:663-71. [PMID: 23731778 DOI: 10.1016/j.cbpa.2013.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 04/23/2013] [Indexed: 12/19/2022]
Abstract
Diffusion barriers are universal solutions for cells to achieve distinct organizations, compositions, and activities within a limited space. The influence of diffusion barriers on the spatiotemporal dynamics of signaling molecules often determines cellular physiology and functions. Over the years, the passive permeability barriers in various subcellular locales have been characterized using elaborate analytical techniques. In this review, we will summarize the current state of knowledge on the various passive permeability barriers present in mammalian cells. We will conclude with a description of several conventional techniques and one new approach based on chemically inducible diffusion trap (CIDT) for probing permeable barriers.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, United States.
| | | | | | | |
Collapse
|
81
|
Koike M, Tanida I, Nanao T, Tada N, Iwata JI, Ueno T, Kominami E, Uchiyama Y. Enrichment of GABARAP relative to LC3 in the axonal initial segments of neurons. PLoS One 2013; 8:e63568. [PMID: 23671684 PMCID: PMC3650058 DOI: 10.1371/journal.pone.0063568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 04/03/2013] [Indexed: 01/19/2023] Open
Abstract
GABAA receptor-associated protein (GABARAP) was initially identified as a protein that interacts with GABAA receptor. Although LC3 (microtubule-associated protein 1 light chain 3), a GABARAP homolog, has been localized in the dendrites and cell bodies of neurons under normal conditions, the subcellular distribution of GABARAP in neurons remains unclear. Subcellular fractionation indicated that endogenous GABARAP was localized to the microsome-enriched and synaptic vesicle-enriched fractions of mouse brain as GABARAP-I, an unlipidated form. To investigate the distribution of GABARAP in neurons, we generated GFP-GABARAP transgenic mice. Immunohistochemistry in these transgenic mice showed that positive signals for GFP-GABARAP were widely distributed in neurons in various brain regions, including the hippocampus and cerebellum. Interestingly, intense diffuse and/or fibrillary expression of GFP-GABARAP was detected along the axonal initial segments (AIS) of hippocampal pyramidal neurons and cerebellar Purkinje cells, in addition to the cell bodies and dendrites of these neurons. In contrast, only slight amounts of LC3 were detected along the AIS of these neurons, while diffuse and/or fibrillary staining for LC3 was mainly detected in their cell bodies and dendrites. These results indicated that, compared with LC3, GABARAP is enriched in the AIS, in addition to the cell bodies and dendrites, of these hippocampal pyramidal neurons and cerebellar Purkinje cells.
Collapse
Affiliation(s)
- Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Baalman KL, Cotton RJ, Rasband SN, Rasband MN. Blast wave exposure impairs memory and decreases axon initial segment length. J Neurotrauma 2013; 30:741-51. [PMID: 23025758 DOI: 10.1089/neu.2012.2478] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Exposure to a blast wave has been proposed to cause mild traumatic brain injury (mTBI), with symptoms including altered cognition, memory, and behavior. This idea, however, remains controversial, and the mechanisms of blast-induced brain injury remain unknown. To begin to resolve these questions, we constructed a simple compressed air shock tube, placed rats inside the tube, and exposed them to a highly reproducible and controlled blast wave. Consistent with the generation of a mild injury, 2 weeks after exposure to the blast, we found that motor performance was unaffected, and a panel of common injury markers showed little or no significant changes in expression in the cortex, corpus callosum, or hippocampus. Similarly, we were unable to detect elevated spectrin breakdown products in brains collected from blast-exposed rats. Using an object recognition task, however, we found that rats exposed to a blast wave spent significantly less time exploring a novel object when compared with control rats. Intriguingly, we also observed a significant shortening of the axon initial segment (AIS) in both the cortex and hippocampus of blast-exposed rats, suggesting altered neuronal excitability after exposure to a blast. A computational model showed that shortening the AIS increased both threshold and the interspike interval of repetitively firing neurons. These results support the conclusion that exposure to a single blast wave can lead to mTBI with accompanying cognitive impairment and subcellular changes in the molecular organization of neurons.
Collapse
Affiliation(s)
- Kelli L Baalman
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
83
|
Chang KJ, Rasband MN. Excitable domains of myelinated nerves: axon initial segments and nodes of Ranvier. CURRENT TOPICS IN MEMBRANES 2013; 72:159-92. [PMID: 24210430 DOI: 10.1016/b978-0-12-417027-8.00005-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurons are highly polarized cells. They can be subdivided into at least two structurally and functionally distinct domains: somatodendritic and axonal domains. The somatodendritic domain receives and integrates upstream input signals, and the axonal domain generates and relays outputs in the form of action potentials to the downstream target. Demand for quick response to the harsh surroundings prompted evolution to equip vertebrates' neurons with a remarkable glia-derived structure called myelin. Not only Insulating the axon, myelinating glia also rearrange the axonal components and elaborate functional subdomains along the axon. Proper functioning of all theses domains and subdomains is vital for a normal, efficient nervous system.
Collapse
Affiliation(s)
- Kae-Jiun Chang
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
84
|
Abstract
Voltage-gated K(+) channels (Kv) represent the largest family of genes in the K(+) channel family. The Kv1 subfamily plays an essential role in the initiation and shaping of action potentials, influencing action potential firing patterns and controlling neuronal excitability. Overlapping patterns with differential expression and precise localization of Kv1.1 and Kv1.2 channels targeted to specialized subcellular compartments contribute to distinctive patterns of neuronal excitability. Dynamic regulation of the components in these subcellular domains help to finely tune the cellular and regional networks. Disruption of the expression, distribution, and density of these channels through deletion or mutation of the genes encoding these channels, Kcna1 and Kcna2, is associated with neurologic pathologies including epilepsy and ataxia in humans and in rodent models. Kv1.1 and Kv1.2 knockout mice both have seizures beginning early in development; however, each express a different seizure type (pathway), although the channels are from the same subfamily and are abundantly coexpressed. Voltage-gated ion channels clustered in specific locations may present a novel therapeutic target for influencing excitability in neurologic disorders associated with some channelopathies.
Collapse
Affiliation(s)
- Carol A Robbins
- Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | |
Collapse
|
85
|
|
86
|
Harkany T. Molecular mechanisms of neuronal specification. Eur J Neurosci 2011; 34:1513-5. [PMID: 22103409 DOI: 10.1111/j.1460-9568.2011.07912.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|