Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 529(16): 3621-3632, 2021 11.
Article in English | MEDLINE | ID: mdl-34235750

ABSTRACT

The axon initial segment (AIS) is structurally and functionally distinct from other regions of the axon, yet alterations in the milieu of the AIS after brain injury have not been well characterized. In this study, we have examined extracellular and intracellular changes in the AIS after hypoglossal nerve injury. Microglial adhesions to the AIS were rarely observed in healthy controls, whereas microglial adhesions to the AIS became apparent in the axonal injury model. Regarding intra-AIS morphology, we focused on mitochondria because mitochondrial flow into the injured axon appears critical for axonal regeneration. To visualize mitochondria specifically in injured axons, we used Atf3:BAC transgenic mice whose mitochondria were labeled with GFP in response to nerve injury. These mice clearly showed mitochondrial localization in the AIS after nerve injury. To precisely confirm the light microscopic observations, we performed three-dimensional ultrastructural analysis using focused ion beam/scanning electron microscopy (FIB/SEM). Although the healthy AIS was not surrounded by microglia, tight microglial adhesions with thick processes adhering to the AIS were observed after injury. FIB/SEM simultaneously allowed the observation of mitochondrial localization in the AIS. In the AIS of non-injured neurons, few mitochondria were observed, whereas mitochondria were abundantly localized in the cell body, axon hillock, and axon. Intriguingly, in the injured AIS, numerous mitochondria were observed throughout the AIS. Taken together, axonal injury changes the extracellular glial environment surrounding the AIS and intracellular mitochondrial localization in the AIS. These changes would be crucial responses, perhaps for injured neurons to regenerate after axonal injury.


Subject(s)
Axon Initial Segment/physiology , Axons/physiology , Extracellular Space/physiology , Mitochondria/physiology , Neuroglia/physiology , Activating Transcription Factor 3/genetics , Animals , Axon Initial Segment/ultrastructure , Axons/ultrastructure , Cell Adhesion , Female , Humans , Imaging, Three-Dimensional , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/ultrastructure , Nerve Crush , Neuroglia/ultrastructure
2.
J Comp Neurol ; 529(16): 3593-3620, 2021 11.
Article in English | MEDLINE | ID: mdl-34219229

ABSTRACT

Signal processing within the retina is generally mediated by graded potentials, whereas output is conveyed by action potentials transmitted along optic nerve axons. Among retinal neurons, amacrine cells seem to be an exception to this general rule, as several types generate voltage-gated Na+ (Nav ) channel-dependent action potentials. The AII, a narrow-field, bistratified axon-less amacrine cell found in mammalian retinas, displays a unique process that resembles an axon initial segment (AIS), with expression of Nav channels colocalized with the cytoskeletal protein ankyrin-G, and generates action potentials. As the role of spiking in AIIs is uncertain, we hypothesized that the morphological properties of the AIS-like process could provide information relevant for its functional importance, including potential pre- and/or postsynaptic connectivity. For morphological analysis, we injected AII amacrine cells in slices with fluorescent dye and immunolabeled the slices for ankyrin-G. Subsequently, this enabled us to reliably identify AII-type processes among ankyrin-G-labeled processes in wholemount retina. We systematically analyzed the laminar localization, spatial orientation, and distribution of the AIS-like processes as a function of retinal eccentricity. In the horizontal plane, the processes displayed no preferred orientation and terminal endings were randomly distributed. In the vertical plane, the processes displayed a horizontal preference, but also ascended and descended into the inner nuclear layer and proximal inner plexiform layer, respectively. These results suggest that the AII amacrine AIS-like process is unlikely to take part in conventional synaptic connections, but may instead be adapted to respond to volume neurotransmission by means of extrasynaptic receptors.


Subject(s)
Amacrine Cells/ultrastructure , Axon Initial Segment/ultrastructure , Axons/ultrastructure , Retina/ultrastructure , Action Potentials/physiology , Animals , Ankyrins/physiology , Dendrites , Female , Male , Rats , Rats, Wistar , Sodium Channels/physiology , Synaptic Transmission
3.
J Neurosci ; 41(17): 3764-3776, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33731449

ABSTRACT

The axon initial segment (AIS) is a specialized neuronal compartment in which synaptic input is converted into action potential (AP) output. This process is supported by a diverse complement of sodium, potassium, and calcium channels (CaV). Different classes of sodium and potassium channels are scaffolded at specific sites within the AIS, conferring unique functions, but how calcium channels are functionally distributed within the AIS is unclear. Here, we use conventional two-photon laser scanning and diffraction-limited, high-speed spot two-photon imaging to resolve AP-evoked calcium dynamics in the AIS with high spatiotemporal resolution. In mouse layer 5 prefrontal pyramidal neurons, calcium influx was mediated by a mix of CaV2 and CaV3 channels that differentially localized to discrete regions. CaV3 functionally localized to produce nanodomain hotspots of calcium influx that coupled to ryanodine-sensitive stores, whereas CaV2 localized to non-hotspot regions. Thus, different pools of CaVs appear to play distinct roles in AIS function.SIGNIFICANCE STATEMENT The axon initial segment (AIS) is the site where synaptic input is transformed into action potential (AP) output. It achieves this function through a diverse complement of sodium, potassium, and calcium channels (CaV). While the localization and function of sodium channels and potassium channels at the AIS is well described, less is known about the functional distribution of CaVs. We used high-speed two-photon imaging to understand activity-dependent calcium dynamics in the AIS of mouse neocortical pyramidal neurons. Surprisingly, we found that calcium influx occurred in two distinct domains: CaV3 generates hotspot regions of calcium influx coupled to calcium stores, whereas CaV2 channels underlie diffuse calcium influx between hotspots. Therefore, different CaV classes localize to distinct AIS subdomains, possibly regulating distinct cellular processes.


Subject(s)
Axon Initial Segment/physiology , Axon Initial Segment/ultrastructure , Calcium Channels/physiology , Calcium Signaling/physiology , Action Potentials/physiology , Animals , Axons , Caveolin 2/drug effects , Caveolin 2/physiology , Caveolin 3/drug effects , Caveolin 3/physiology , Female , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects
4.
Cell Rep ; 30(10): 3506-3519.e6, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160553

ABSTRACT

Genetic variations in TMEM106B, coding for a lysosomal membrane protein, affect frontotemporal lobar degeneration (FTLD) in GRN- (coding for progranulin) and C9orf72-expansion carriers and might play a role in aging. To determine the physiological function of TMEM106B, we generated TMEM106B-deficient mice. These mice develop proximal axonal swellings caused by drastically enlarged LAMP1-positive vacuoles, increased retrograde axonal transport of lysosomes, and accumulation of lipofuscin and autophagosomes. Giant vacuoles specifically accumulate at the distal end and within the axon initial segment, but not in peripheral nerves or at axon terminals, resulting in an impaired facial-nerve-dependent motor performance. These data implicate TMEM106B in mediating the axonal transport of LAMP1-positive organelles in motoneurons and axonal sorting at the initial segment. Our data provide mechanistic insight into how TMEM106B affects lysosomal proteolysis and degradative capacity in neurons.


Subject(s)
Axon Initial Segment/metabolism , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease , Lysosomes/metabolism , Membrane Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Axon Initial Segment/ultrastructure , Axonal Transport , Brain Stem/pathology , Cell Nucleus/metabolism , Facial Nerve/pathology , Lysosomes/ultrastructure , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/ultrastructure , Muscles/innervation , Nerve Tissue Proteins/deficiency , Risk Factors
5.
Neuron ; 104(2): 305-321.e8, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31474508

ABSTRACT

The axon initial segment (AIS) is a unique neuronal compartment that plays a crucial role in the generation of action potential and neuronal polarity. The assembly of the AIS requires membrane, scaffolding, and cytoskeletal proteins, including Ankyrin-G and TRIM46. How these components cooperate in AIS formation is currently poorly understood. Here, we show that Ankyrin-G acts as a scaffold interacting with End-Binding (EB) proteins and membrane proteins such as Neurofascin-186 to recruit TRIM46-positive microtubules to the plasma membrane. Using in vitro reconstitution and cellular assays, we demonstrate that TRIM46 forms parallel microtubule bundles and stabilizes them by acting as a rescue factor. TRIM46-labeled microtubules drive retrograde transport of Neurofascin-186 to the proximal axon, where Ankyrin-G prevents its endocytosis, resulting in stable accumulation of Neurofascin-186 at the AIS. Neurofascin-186 enrichment in turn reinforces membrane anchoring of Ankyrin-G and subsequent recruitment of TRIM46-decorated microtubules. Our study reveals feedback-based mechanisms driving AIS assembly.


Subject(s)
Ankyrins/metabolism , Axon Initial Segment/metabolism , Cell Adhesion Molecules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Animals , Axon Initial Segment/ultrastructure , Axonal Transport , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cytoskeleton , Endocytosis , Feedback, Physiological , HEK293 Cells , Hippocampus/cytology , Humans , Microtubules/ultrastructure , Neurons/ultrastructure , Rats , Tripartite Motif Proteins/metabolism
6.
Biochem Biophys Res Commun ; 516(1): 15-21, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31186137

ABSTRACT

Cilia, as key integrators of extracellular ligand-based signaling, play a critical role in signaling in the neuronal system. However, the function of cilia in neurons is largely unknown. In this study, we discovered that cilia morphology and ciliary protein localization were associated with axon initial segment (AIS) morphology, including length and location. Cilia morphological changes induced by the serotonin (5-HT) receptor 5-HT6R, intraflagellar transport 88 (Ift88) and kinesin family member 3A (KIF3A) altered AIS length and location. The change in cilia morphology was associated with aberrant localization of ankyrin G (AnkG) and voltage-gated sodium channel 1.2 (Nav1.2). Cilia morphology altered action potential (AP) amplitude and spike firing. Taken together, our data strongly suggest that cilia function might have a marked impact on neuron excitability by regulating AIS morphology and ion channel localization. Our findings highlight a novel aspect linking cilia function and neuron excitability.


Subject(s)
Axon Initial Segment/ultrastructure , Cilia/metabolism , Neurons/cytology , Receptors, Serotonin/metabolism , Action Potentials , Animals , Axon Initial Segment/metabolism , Cells, Cultured , Cilia/ultrastructure , Kinesins/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Tumor Suppressor Proteins/metabolism
7.
Ann N Y Acad Sci ; 1420(1): 46-61, 2018 05.
Article in English | MEDLINE | ID: mdl-29749636

ABSTRACT

The axon initial segment (AIS) is located at the proximal axon and is the site of action potential initiation. This reflects the high density of ion channels found at the AIS. Adaptive changes to the location and length of the AIS can fine-tune the excitability of neurons and modulate plasticity in response to activity. The AIS plays an important role in maintaining neuronal polarity by regulating the trafficking and distribution of proteins that function in somatodendritic or axonal compartments of the neuron. In this review, we provide an overview of the AIS cytoarchitecture, mechanism of assembly, and recent studies revealing mechanisms of differential transport at the AIS that maintain axon and dendrite identities. We further discuss how genetic mutations in AIS components (i.e., ankyrins, ion channels, and spectrins) and injuries may cause neurological disorders.


Subject(s)
Ankyrins/genetics , Axon Initial Segment/pathology , Nervous System Diseases/pathology , Protein Transport/physiology , Action Potentials/physiology , Axon Initial Segment/ultrastructure , Cell Polarity/physiology , Humans , Ion Channels/genetics , Ion Channels/physiology , Neurons/physiology , Ranvier's Nodes/physiology
8.
J Neurosci ; 38(9): 2135-2145, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29378864

ABSTRACT

At the base of axons sits a unique compartment called the axon initial segment (AIS). The AIS generates and shapes the action potential before it is propagated along the axon. Neuronal excitability thus depends crucially on the AIS composition and position, and these adapt to developmental and physiological conditions. The AIS also demarcates the boundary between the somatodendritic and axonal compartments. Recent studies have brought insights into the molecular architecture of the AIS and how it regulates protein trafficking. This Viewpoints article summarizes current knowledge about the AIS and highlights future challenges in understanding this key actor of neuronal physiology.


Subject(s)
Axon Initial Segment/physiology , Axon Initial Segment/ultrastructure , Animals , Humans
9.
J Neurosci ; 38(3): 733-744, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29217687

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.


Subject(s)
Axon Initial Segment/ultrastructure , Dopaminergic Neurons/physiology , Dopaminergic Neurons/ultrastructure , Models, Neurological , Animals , Axon Initial Segment/physiology , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Substantia Nigra/ultrastructure
10.
J Cell Sci ; 130(21): 3663-3675, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28935671

ABSTRACT

Central nervous system (CNS) axons lose their intrinsic ability to regenerate upon maturity, whereas peripheral nervous system (PNS) axons do not. A key difference between these neuronal types is their ability to transport integrins into axons. Integrins can mediate PNS regeneration, but are excluded from adult CNS axons along with their Rab11 carriers. We reasoned that exclusion of the contents of Rab11 vesicles including integrins might contribute to the intrinsic inability of CNS neurons to regenerate, and investigated this by performing laser axotomy. We identify a novel regulator of selective axon transport and regeneration, the ARF6 guanine-nucleotide-exchange factor (GEF) EFA6 (also known as PSD). EFA6 exerts its effects from a location within the axon initial segment (AIS). EFA6 does not localise at the AIS in dorsal root ganglion (DRG) axons, and in these neurons, ARF6 activation is counteracted by an ARF GTPase-activating protein (GAP), which is absent from the CNS, ACAP1. Depleting EFA6 from cortical neurons permits endosomal integrin transport and enhances regeneration, whereas overexpressing EFA6 prevents DRG regeneration. Our results demonstrate that ARF6 is an intrinsic regulator of regenerative capacity, implicating EFA6 as a focal molecule linking the AIS, signalling and transport.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Axon Initial Segment/metabolism , Axonal Transport/genetics , Cerebral Cortex/metabolism , Dendrites/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Integrin alpha Chains/metabolism , Neurons/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Axon Initial Segment/ultrastructure , Cerebral Cortex/ultrastructure , Dendrites/ultrastructure , Embryo, Mammalian , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Integrin alpha Chains/genetics , Male , Microtubules , Neurons/ultrastructure , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
11.
J Comp Neurol ; 525(16): 3529-3542, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28734032

ABSTRACT

The axon initial segment (AIS) is the site of initiation of action potentials and influences action potential waveform, firing pattern, and rate. In view of the fundamental aspects of motor function and behavior that depend on the firing of substantia nigra pars compacta (SNc) dopaminergic neurons, we identified and characterized their AIS in the mouse. Immunostaining for tyrosine hydroxylase (TH), sodium channels (Nav ) and ankyrin-G (Ank-G) was used to visualize the AIS of dopaminergic neurons. Reconstructions of sampled AIS of dopaminergic neurons revealed variable lengths (12-60 µm) and diameters (0.2-0.8 µm), and an average of 50% reduction in diameter between their widest and thinnest parts. Ultrastructural analysis revealed submembranous localization of Ank-G at nodes of Ranvier and AIS. Serial ultrathin section analysis and 3D reconstructions revealed that Ank-G colocalized with TH only at the AIS. Few cases of synaptic innervation of the AIS of dopaminergic neurons were observed. mRNA in situ hybridization of brain-specific Nav subunits revealed the expression of Nav 1.2 by most SNc neurons and a small proportion expressing Nav 1.6. The presence of sodium channels, along with the submembranous location of Ank-G is consistent with the role of AIS in action potential generation. Differences in the size of the AIS likely underlie differences in firing pattern, while the tapering diameter of AIS may define a trigger zone for action potentials. Finally, the conspicuous expression of Nav 1.2 by the majority of dopaminergic neurons may explain their high threshold for firing and their low discharge rate.


Subject(s)
Axon Initial Segment/physiology , Dopaminergic Neurons/cytology , Substantia Nigra/cytology , Action Potentials/physiology , Animals , Ankyrins/metabolism , Ankyrins/ultrastructure , Axon Initial Segment/ultrastructure , Gene Expression/physiology , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/metabolism , NAV1.2 Voltage-Gated Sodium Channel/ultrastructure , NAV1.6 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/metabolism , NAV1.6 Voltage-Gated Sodium Channel/ultrastructure , Neuroimaging , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/ultrastructure
12.
Nat Commun ; 8(1): 33, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28652571

ABSTRACT

A long-standing question in neurodevelopment is how neurons develop a single axon and multiple dendrites from common immature neurites. Long-range inhibitory signaling from the growing axon is hypothesized to prevent outgrowth of other immature neurites and to differentiate them into dendrites, but the existence and nature of this inhibitory signaling remains unknown. Here, we demonstrate that axonal growth triggered by neurotrophin-3 remotely inhibits neurite outgrowth through long-range Ca2+ waves, which are delivered from the growing axon to the cell body. These Ca2+ waves increase RhoA activity in the cell body through calcium/calmodulin-dependent protein kinase I. Optogenetic control of Rho-kinase combined with computational modeling reveals that active Rho-kinase diffuses to growing other immature neurites and inhibits their outgrowth. Mechanistically, calmodulin-dependent protein kinase I phosphorylates a RhoA-specific GEF, GEF-H1, whose phosphorylation enhances its GEF activity. Thus, our results reveal that long-range inhibitory signaling mediated by Ca2+ wave is responsible for neuronal polarization.Emerging evidence suggests that gut microbiota influences immune function in the brain and may play a role in neurological diseases. Here, the authors offer in vivo evidence from a Drosophila model that supports a role for gut microbiota in modulating the progression of Alzheimer's disease.


Subject(s)
Axon Initial Segment/metabolism , Calcium Signaling , Calcium/metabolism , Growth Cones/metabolism , Neurites/metabolism , rho GTP-Binding Proteins/genetics , Animals , Axon Initial Segment/ultrastructure , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Communication , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental , Growth Cones/ultrastructure , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred ICR , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurites/ultrastructure , Neurogenesis/genetics , Optical Imaging , Optogenetics , Primary Cell Culture , Protein Transport , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
13.
Nat Commun ; 7: 12278, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27452526

ABSTRACT

Tau, an intrinsically disordered protein confined to neuronal axons, binds to and regulates microtubule dynamics. Although there have been observations of string-like microtubule fascicles in the axon initial segment (AIS) and hexagonal bundles in neurite-like processes in non-neuronal cells overexpressing Tau, cell-free reconstitutions have not replicated either geometry. Here we map out the energy landscape of Tau-mediated, GTP-dependent 'active' microtubule bundles at 37 °C, as revealed by synchrotron SAXS and TEM. Widely spaced bundles (wall-to-wall distance Dw-w≈25-41 nm) with hexagonal and string-like symmetry are observed, the latter mimicking bundles found in the AIS. A second energy minimum (Dw-w≈16-23 nm) is revealed under osmotic pressure. The wide spacing results from a balance between repulsive forces, due to Tau's projection domain (PD), and a stabilizing sum of transient sub-kBT cationic/anionic charge-charge attractions mediated by weakly penetrating opposing PDs. This landscape would be significantly affected by charge-altering modifications of Tau associated with neurodegeneration.


Subject(s)
Axon Initial Segment/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Animals , Axon Initial Segment/ultrastructure , Cattle , Microtubules/ultrastructure , Osmotic Pressure , Protein Domains , Scattering, Small Angle , Thermodynamics , X-Ray Diffraction , tau Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...