Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Annu Rev Cell Dev Biol ; 31: 647-67, 2015.
Article in English | MEDLINE | ID: mdl-26566119

ABSTRACT

Myelinated axons are divided into polarized subdomains including axon initial segments and nodes of Ranvier. These domains initiate and propagate action potentials and regulate the trafficking and localization of somatodendritic and axonal proteins. Formation of axon initial segments and nodes of Ranvier depends on intrinsic (neuronal) and extrinsic (glial) interactions. Several levels of redundancy in both mechanisms and molecules also exist to ensure efficient node formation. Furthermore, the establishment of polarized domains at and near nodes of Ranvier reflects the intrinsic polarity of the myelinating glia responsible for node assembly. Here, we discuss the various polarized domains of myelinated axons, how they are established by both intrinsic and extrinsic interactions, and the polarity of myelinating glia.


Subject(s)
Axons/physiology , Cell Polarity/physiology , Action Potentials/physiology , Animals , Humans , Myelin Sheath/physiology , Neuroglia/physiology , Neurons/physiology
2.
Cell ; 149(5): 1125-39, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632975

ABSTRACT

AnkyrinG (ankG) is highly enriched in neurons at axon initial segments (AISs) where it clusters Na(+) and K(+) channels and maintains neuronal polarity. How ankG becomes concentrated at the AIS is unknown. Here, we show that as neurons break symmetry, they assemble a distal axonal submembranous cytoskeleton, comprised of ankyrinB (ankB), αII-spectrin, and ßII-spectrin, that defines a boundary limiting ankG to the proximal axon. Experimentally moving this boundary altered the length of ankG staining in the proximal axon, whereas disruption of the boundary through silencing of ankB, αII-spectrin, or ßII-spectrin expression blocked AIS assembly and permitted ankG to redistribute throughout the distal axon. In support of an essential role for the distal cytoskeleton in ankG clustering, we also found that αII and ßII-spectrin-deficient mice had disrupted AIS. Thus, the distal axonal cytoskeleton functions as an intra-axonal boundary restricting ankG to the AIS.


Subject(s)
Axons/metabolism , Cytoskeleton/metabolism , Neurons/metabolism , Animals , Ankyrins/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Mice , Neurons/cytology , Spectrin/metabolism
3.
Nat Rev Neurosci ; 22(1): 7-20, 2021 01.
Article in English | MEDLINE | ID: mdl-33239761

ABSTRACT

The nodes of Ranvier have clustered Na+ and K+ channels necessary for rapid and efficient axonal action potential conduction. However, detailed mechanisms of channel clustering have only recently been identified: they include two independent axon-glia interactions that converge on distinct axonal cytoskeletons. Here, we discuss how glial cell adhesion molecules and the extracellular matrix molecules that bind them assemble combinations of ankyrins, spectrins and other cytoskeletal scaffolding proteins, which cluster ion channels. We present a detailed molecular model, incorporating these overlapping mechanisms, to explain how the nodes of Ranvier are assembled in both the peripheral and central nervous systems.


Subject(s)
Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Ranvier's Nodes/metabolism , Animals , Ankyrins/metabolism , Axons/metabolism , Humans , Ion Channels/metabolism , Spectrin/metabolism
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074918

ABSTRACT

MeCP2 is associated with Rett syndrome (RTT), MECP2 duplication syndrome, and a number of conditions with isolated features of these diseases, including autism, intellectual disability, and motor dysfunction. MeCP2 is known to broadly bind methylated DNA, but the precise molecular mechanism driving disease pathogenesis remains to be determined. Using proximity-dependent biotinylation (BioID), we identified a transcription factor 20 (TCF20) complex that interacts with MeCP2 at the chromatin interface. Importantly, RTT-causing mutations in MECP2 disrupt this interaction. TCF20 and MeCP2 are highly coexpressed in neurons and coregulate the expression of key neuronal genes. Reducing Tcf20 partially rescued the behavioral deficits caused by MECP2 overexpression, demonstrating a functional relationship between MeCP2 and TCF20 in MECP2 duplication syndrome pathogenesis. We identified a patient exhibiting RTT-like neurological features with a missense mutation in the PHF14 subunit of the TCF20 complex that abolishes the MeCP2-PHF14-TCF20 interaction. Our data demonstrate the critical role of the MeCP2-TCF20 complex for brain function.


Subject(s)
Methyl-CpG-Binding Protein 2/metabolism , Multiprotein Complexes/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Transcription Factors/metabolism , Alleles , Animals , Biomarkers , Brain/metabolism , Disease Models, Animal , Disease Susceptibility , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Mutation , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Synapses/metabolism , Transcription Factors/genetics
5.
J Physiol ; 602(6): 1127-1145, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38441922

ABSTRACT

Spectrins function together with actin as obligatory subunits of the submembranous cytoskeleton. Spectrins maintain cell shape, resist mechanical forces, and stabilize ion channel and transporter protein complexes through binding to scaffolding proteins. Recently, pathogenic variants of SPTBN4 (ß4 spectrin) were reported to cause both neuropathy and myopathy. Although the role of ß4 spectrin in neurons is mostly understood, its function in skeletal muscle, another excitable tissue subject to large forces, is unknown. Here, using a muscle specific ß4 spectrin conditional knockout mouse, we show that ß4 spectrin does not contribute to muscle function. In addition, we show ß4 spectrin is not present in muscle, indicating the previously reported myopathy associated with pathogenic SPTBN4 variants is neurogenic in origin. More broadly, we show that α2, ß1 and ß2 spectrins are found in skeletal muscle, with α2 and ß1 spectrins being enriched at the postsynaptic neuromuscular junction (NMJ). Surprisingly, using muscle specific conditional knockout mice, we show that loss of α2 and ß2 spectrins had no effect on muscle health, function or the enrichment of ß1 spectrin at the NMJ. Muscle specific deletion of ß1 spectrin also had no effect on muscle health, but, with increasing age, resulted in the loss of clustered NMJ Na+ channels. Together, our results suggest that muscle ß1 spectrin functions independently of an associated α spectrin to maintain Na+ channel clustering at the postsynaptic NMJ. Furthermore, despite repeated exposure to strong forces and in contrast to neurons, muscles do not require spectrin cytoskeletons to maintain cell shape or integrity. KEY POINTS: The myopathy found in pathogenic human SPTBN4 variants (where SPTBN4 is the gene encoding ß4 spectrin) is neurogenic in origin. ß1 spectrin plays essential roles in maintaining the density of neuromuscular junction Nav1.4 Na+ channels. By contrast to the canonical view of spectrin organization and function, we show that ß1 spectrin can function independently of an associated α spectrin. Despite the large mechanical forces experienced by muscle, we show that spectrins are not required for muscle cell integrity. This is in stark contrast to red blood cells and the axons of neurons.


Subject(s)
Muscular Diseases , Spectrin , Mice , Animals , Humans , Spectrin/genetics , Spectrin/analysis , Spectrin/metabolism , Actin Cytoskeleton/metabolism , Neuromuscular Junction/metabolism , Muscle, Skeletal/metabolism
6.
J Neurosci ; 42(43): 8054-8065, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36096668

ABSTRACT

The axon initial segment (AIS) generates action potentials and maintains neuronal polarity by regulating the differential trafficking and distribution of proteins, transport vesicles, and organelles. Injury and disease can disrupt the AIS, and the subsequent loss of clustered ion channels and polarity mechanisms may alter neuronal excitability and function. However, the impact of AIS disruption on axon regeneration after injury is unknown. We generated male and female mice with AIS-deficient multipolar motor neurons by deleting AnkyrinG, the master scaffolding protein required for AIS assembly and maintenance. We found that after nerve crush, neuromuscular junction reinnervation was significantly delayed in AIS-deficient motor neurons compared with control mice. In contrast, loss of AnkyrinG from pseudo-unipolar sensory neurons did not impair axon regeneration into the intraepidermal nerve fiber layer. Even after AIS-deficient motor neurons reinnervated the neuromuscular junction, they failed to functionally recover because of reduced synaptic vesicle protein 2 at presynaptic terminals. In addition, mRNA trafficking was disrupted in AIS-deficient axons. Our results show that, after nerve injury, an intact AIS is essential for efficient regeneration and functional recovery of axons in multipolar motor neurons. Our results also suggest that loss of polarity in AIS-deficient motor neurons impairs the delivery of axonal proteins, mRNAs, and other cargoes necessary for regeneration. Thus, therapeutic strategies for axon regeneration must consider preservation or reassembly of the AIS.SIGNIFICANCE STATEMENT Disruption of the axon initial segment is a common event after nervous system injury. For multipolar motor neurons, we show that axon initial segments are essential for axon regeneration and functional recovery after injury. Our results may help explain injuries where axon regeneration fails, and suggest strategies to promote more efficient axon regeneration.


Subject(s)
Axon Initial Segment , Axons , Male , Female , Mice , Animals , Axons/physiology , Axon Initial Segment/metabolism , Ankyrins/metabolism , Nerve Regeneration , Synapses/metabolism , Ion Channels/metabolism , Motor Neurons/metabolism , RNA, Messenger/metabolism
7.
J Neurosci ; 42(1): 2-15, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34785580

ABSTRACT

Ankyrin scaffolding proteins are critical for membrane domain organization and protein stabilization in many different cell types including neurons. In the cerebellum, Ankyrin-R (AnkR) is highly enriched in Purkinje neurons, granule cells, and in the cerebellar nuclei (CN). Using male and female mice with a floxed allele for Ank1 in combination with Nestin-Cre and Pcp2-Cre mice, we found that ablation of AnkR from Purkinje neurons caused ataxia, regional and progressive neurodegeneration, and altered cerebellar output. We show that AnkR interacts with the cytoskeletal protein ß3 spectrin and the potassium channel Kv3.3. Loss of AnkR reduced somatic membrane levels of ß3 spectrin and Kv3.3 in Purkinje neurons. Thus, AnkR links Kv3.3 channels to the ß3 spectrin-based cytoskeleton. Our results may help explain why mutations in ß3 spectrin and Kv3.3 both cause spinocerebellar ataxia.SIGNIFICANCE STATEMENT Ankyrin scaffolding proteins localize and stabilize ion channels in the membrane by linking them to the spectrin-based cytoskeleton. Here, we show that Ankyrin-R (AnkR) links Kv3.3 K+ channels to the ß3 spectrin-based cytoskeleton in Purkinje neurons. Loss of AnkR causes Purkinje neuron degeneration, altered cerebellar physiology, and ataxia, which is consistent with mutations in Kv3.3 and ß3 spectrin causing spinocerebellar ataxia.


Subject(s)
Ankyrins/metabolism , Cytoskeleton/metabolism , Purkinje Cells/metabolism , Shaw Potassium Channels/metabolism , Spectrin/metabolism , Animals , Cell Survival/physiology , Female , Male , Mice , Spinocerebellar Ataxias/genetics
8.
J Cell Sci ; 134(6)2021 03 11.
Article in English | MEDLINE | ID: mdl-33536249

ABSTRACT

Ranbp2 (also known as Nup358) is a member of the nucleoporin family, which constitutes the nuclear pore complex. Ranbp2 localizes at the nuclear membrane and was recently reported at the axon initial segment (AIS). However, we show that the anti-Ranbp2 antibody used in previous studies is not specific for Ranbp2. We mapped the antibody binding site to the amino acid sequence KPLQG, which is present in both Ranbp2 and neurofascin (Nfasc), a well-known AIS protein. After silencing neurofascin expression in neurons, the AIS was not stained by the antibody. Surprisingly, an exogenously expressed N-terminal fragment of Ranbp2 localizes at the AIS. We show that this fragment interacts with stable microtubules. Finally, using CRISPR/Cas9 in primary cultured neurons, we inserted an HA-epitope tag at N-terminal, C-terminal or internal sites of the endogenously expressed Ranbp2. No matter the location of the HA-epitope, endogenous Ranbp2 was found at the nuclear membrane but not the AIS. These results show that endogenously expressed Ranbp2 is not found at AISs.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Axon Initial Segment , Nuclear Pore Complex Proteins , Humans , Neurons , Nuclear Envelope , Nuclear Pore , Nuclear Pore Complex Proteins/genetics
9.
Brain ; 144(8): 2527-2540, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34014281

ABSTRACT

Gene discovery efforts in autism spectrum disorder have identified heterozygous defects in chromatin remodeller genes, the 'readers, writers and erasers' of methyl marks on chromatin, as major contributors to this disease. Despite this advance, a convergent aetiology between these defects and aberrant chromatin architecture or gene expression has remained elusive. Recently, data have begun to emerge that chromatin remodellers also function directly on the cytoskeleton. Strongly associated with autism spectrum disorder, the SETD2 histone methyltransferase for example, has now been shown to directly methylate microtubules of the mitotic spindle. However, whether microtubule methylation occurs in post-mitotic cells, for example on the neuronal cytoskeleton, is not known. We found the SETD2 α-tubulin lysine 40 trimethyl mark occurs on microtubules in the brain and in primary neurons in culture, and that the SETD2 C-terminal SRI domain is required for binding and methylation of α-tubulin. A CRISPR knock-in of a pathogenic SRI domain mutation (Setd2SRI) that disables microtubule methylation revealed at least one wild-type allele was required in mice for survival, and while viable, heterozygous Setd2SRI/wtmice exhibited an anxiety-like phenotype. Finally, whereas RNA-sequencing (RNA-seq) and chromatin immunoprecipitation-sequencing (ChIP-seq) showed no concomitant changes in chromatin methylation or gene expression in Setd2SRI/wtmice, primary neurons exhibited structural deficits in axon length and dendritic arborization. These data provide the first demonstration that microtubules of neurons are methylated, and reveals a heterozygous chromatin remodeller defect that specifically disables microtubule methylation is sufficient to drive an autism-associated phenotype.


Subject(s)
Anxiety/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Microtubules/metabolism , Neurons/metabolism , Animals , Brain/metabolism , Histones/metabolism , Methylation , Mice , Phenotype
10.
Am J Hum Genet ; 102(6): 1158-1168, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861105

ABSTRACT

ßIV spectrin links ankyrinG (AnkG) and clustered ion channels at axon initial segments (AISs) and nodes of Ranvier to the axonal cytoskeleton. Here, we report bi-allelic pathogenic SPTBN4 variants (three homozygous and two compound heterozygous) that cause a severe neurological syndrome that includes congenital hypotonia, intellectual disability, and motor axonal and auditory neuropathy. We introduced these variants into ßIV spectrin, expressed these in neurons, and found that 5/7 were loss-of-function variants disrupting AIS localization or abolishing phosphoinositide binding. Nerve biopsies from an individual with a loss-of-function variant had reduced nodal Na+ channels and no nodal KCNQ2 K+ channels. Modeling the disease in mice revealed that although ankyrinR (AnkR) and ßI spectrin can cluster Na+ channels and partially compensate for the loss of AnkG and ßIV spectrin at nodes of Ranvier, AnkR and ßI spectrin cannot cluster KCNQ2- and KCNQ3-subunit-containing K+ channels. Our findings define a class of spectrinopathies and reveal the molecular pathologies causing nervous-system dysfunction.


Subject(s)
Axons/pathology , Intellectual Disability/genetics , Motor Neuron Disease/genetics , Muscle Hypotonia/congenital , Muscle Hypotonia/genetics , Nerve Tissue Proteins/genetics , Spectrin/genetics , Alleles , Animals , Axons/metabolism , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Female , HEK293 Cells , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/physiopathology , Lipids , Male , Mice, Knockout , Motor Neuron Disease/complications , Motor Neuron Disease/physiopathology , Muscle Hypotonia/complications , Muscle Hypotonia/physiopathology , Mutant Proteins/metabolism , Mutation/genetics , Rats, Sprague-Dawley
11.
J Biol Chem ; 294(24): 9576-9591, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31064843

ABSTRACT

Spectrins are cytoskeletal proteins essential for membrane biogenesis and regulation and serve critical roles in protein targeting and cellular signaling. αII spectrin (SPTAN1) is one of two α spectrin genes and αII spectrin dysfunction is linked to alterations in axon initial segment formation, cortical lamination, and neuronal excitability. Furthermore, human αII spectrin loss-of-function variants cause neurological disease. As global αII spectrin knockout mice are embryonic lethal, the in vivo roles of αII spectrin in adult heart are unknown and untested. Here, based on pronounced alterations in αII spectrin regulation in human heart failure we tested the in vivo roles of αII spectrin in the vertebrate heart. We created a mouse model of cardiomyocyte-selective αII spectrin-deficiency (cKO) and used this model to define the roles of αII spectrin in cardiac function. αII spectrin cKO mice displayed significant structural, cellular, and electrical phenotypes that resulted in accelerated structural remodeling, fibrosis, arrhythmia, and mortality in response to stress. At the molecular level, we demonstrate that αII spectrin plays a nodal role for global cardiac spectrin regulation, as αII spectrin cKO hearts exhibited remodeling of αI spectrin and altered ß-spectrin expression and localization. At the cellular level, αII spectrin deficiency resulted in altered expression, targeting, and regulation of cardiac ion channels NaV1.5 and KV4.3. In summary, our findings define critical and unexpected roles for the multifunctional αII spectrin protein in the heart. Furthermore, our work provides a new in vivo animal model to study the roles of αII spectrin in the cardiomyocyte.


Subject(s)
Arrhythmias, Cardiac/pathology , Disease Models, Animal , Heart Failure/pathology , Ischemia/pathology , Myocytes, Cardiac/pathology , Spectrin/physiology , Animals , Arrhythmias, Cardiac/etiology , Cells, Cultured , Female , Heart Failure/etiology , Humans , Ischemia/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
12.
J Neurosci ; 38(27): 6063-6075, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29853631

ABSTRACT

Action potential conduction along myelinated axons depends on high densities of voltage-gated Na+ channels at the nodes of Ranvier. Flanking each node, paranodal junctions (paranodes) are formed between axons and Schwann cells in the peripheral nervous system (PNS) or oligodendrocytes in the CNS. Paranodal junctions contribute to both node assembly and maintenance. Despite their importance, the molecular mechanisms responsible for paranode assembly and maintenance remain poorly understood. ßII spectrin is expressed in diverse cells and is an essential part of the submembranous cytoskeleton. Here, we show that Schwann cell ßII spectrin is highly enriched at paranodes. To elucidate the roles of glial ßII spectrin, we generated mutant mice lacking ßII spectrin in myelinating glial cells by crossing mice with a floxed allele of Sptbn1 with Cnp-Cre mice, and analyzed both male and female mice. Juvenile (4 weeks) and middle-aged (60 weeks) mutant mice showed reduced grip strength and sciatic nerve conduction slowing, whereas no phenotype was observed between 8 and 24 weeks of age. Consistent with these findings, immunofluorescence microscopy revealed disorganized paranodes in the PNS and CNS of both postnatal day 13 and middle-aged mutant mice, but not in young adult mutant mice. Electron microscopy confirmed partial loss of transverse bands at the paranodal axoglial junction in the middle-aged mutant mice in both the PNS and CNS. These findings demonstrate that a spectrin-based cytoskeleton in myelinating glia contributes to formation and maintenance of paranodal junctions.SIGNIFICANCE STATEMENT Myelinating glia form paranodal axoglial junctions that flank both sides of the nodes of Ranvier. These junctions contribute to node formation and maintenance and are essential for proper nervous system function. We found that a submembranous spectrin cytoskeleton is highly enriched at paranodes in Schwann cells. Ablation of ßII spectrin in myelinating glial cells disrupted the paranodal cell adhesion complex in both peripheral and CNSs, resulting in muscle weakness and sciatic nerve conduction slowing in juvenile and middle-aged mice. Our data show that a spectrin-based submembranous cytoskeleton in myelinating glia plays important roles in paranode formation and maintenance.


Subject(s)
Axons/metabolism , Cytoskeleton/metabolism , Neuroglia/metabolism , Spectrin/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Ranvier's Nodes
13.
J Neurosci ; 37(47): 11323-11334, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29038243

ABSTRACT

Axons must withstand mechanical forces, including tension, torsion, and compression. Spectrins and actin form a periodic cytoskeleton proposed to protect axons against these forces. However, because spectrins also participate in assembly of axon initial segments (AISs) and nodes of Ranvier, it is difficult to uncouple their roles in maintaining axon integrity from their functions at AIS and nodes. To overcome this problem and to determine the importance of spectrin cytoskeletons for axon integrity, we generated mice with αII spectrin-deficient peripheral sensory neurons. The axons of these neurons are very long and exposed to the mechanical forces associated with limb movement; most lack an AIS, and some are unmyelinated and have no nodes. We analyzed αII spectrin-deficient mice of both sexes and found that, in myelinated axons, αII spectrin forms a periodic cytoskeleton with ßIV and ßII spectrin at nodes of Ranvier and paranodes, respectively, but that loss of αII spectrin disrupts this organization. Avil-cre;Sptan1f/f mice have reduced numbers of nodes, disrupted paranodal junctions, and mislocalized Kv1 K+ channels. We show that the density of nodal ßIV spectrin is constant among axons, but the density of nodal αII spectrin increases with axon diameter. Remarkably, Avil-cre;Sptan1f/f mice have intact nociception and small-diameter axons, but severe ataxia due to preferential degeneration of large-diameter myelinated axons. Our results suggest that nodal αII spectrin helps resist the mechanical forces experienced by large-diameter axons, and that αII spectrin-dependent cytoskeletons are also required for assembly of nodes of Ranvier.SIGNIFICANCE STATEMENT A periodic axonal cytoskeleton consisting of actin and spectrin has been proposed to help axons resist the mechanical forces to which they are exposed (e.g., compression, torsion, and stretch). However, until now, no vertebrate animal model has tested the requirement of the spectrin cytoskeleton in maintenance of axon integrity. We demonstrate the role of the periodic spectrin-dependent cytoskeleton in axons and show that loss of αII spectrin from PNS axons causes preferential degeneration of large-diameter myelinated axons. We show that nodal αII spectrin is found at greater densities in large-diameter myelinated axons, suggesting that nodes are particularly vulnerable domains requiring a specialized cytoskeleton to protect against axon degeneration.


Subject(s)
Axons/metabolism , Cytoskeleton/metabolism , Demyelinating Diseases/metabolism , Ranvier's Nodes/metabolism , Spectrin/metabolism , Animals , Axons/pathology , Axons/physiology , Demyelinating Diseases/genetics , Female , Male , Mice , Mice, Inbred C57BL , Ranvier's Nodes/pathology , Ranvier's Nodes/physiology , Spectrin/genetics
14.
J Neurosci ; 37(47): 11311-11322, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29038240

ABSTRACT

Spectrins form a submembranous cytoskeleton proposed to confer strength and flexibility to neurons and to participate in ion channel clustering at axon initial segments (AIS) and nodes of Ranvier. Neuronal spectrin cytoskeletons consist of diverse ß subunits and αII spectrin. Although αII spectrin is found in neurons in both axonal and somatodendritic domains, using proteomics, biochemistry, and superresolution microscopy, we show that αII and ßIV spectrin interact and form a periodic AIS cytoskeleton. To determine the role of spectrins in the nervous system, we generated Sptan1f/f mice for deletion of CNS αII spectrin. We analyzed αII spectrin-deficient mice of both sexes and found that loss of αII spectrin causes profound reductions in all ß spectrins. αII spectrin-deficient mice die before 1 month of age and have disrupted AIS and many other neurological impairments including seizures, disrupted cortical lamination, and widespread neurodegeneration. These results demonstrate the importance of the spectrin cytoskeleton both at the AIS and throughout the nervous system.SIGNIFICANCE STATEMENT Spectrin cytoskeletons play diverse roles in neurons, including assembly of excitable domains such as the axon initial segment (AIS) and nodes of Ranvier. However, the molecular composition and structure of these cytoskeletons remain poorly understood. Here, we show that αII spectrin partners with ßIV spectrin to form a periodic cytoskeleton at the AIS. Using a new αII spectrin conditional knock-out mouse, we show that αII spectrin is required for AIS assembly, neuronal excitability, cortical lamination, and to protect against neurodegeneration. These results demonstrate the broad importance of spectrin cytoskeletons for nervous system function and development and have important implications for nervous system injuries and diseases because disruption of the spectrin cytoskeleton is a common molecular pathology.


Subject(s)
Axons/metabolism , Cytoskeleton/metabolism , Ranvier's Nodes/metabolism , Spectrin/metabolism , Action Potentials , Animals , Axons/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Deletion , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Mice , Mice, Inbred C57BL , Ranvier's Nodes/physiology , Spectrin/genetics
15.
Mol Cell Proteomics ; 15(2): 355-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26342039

ABSTRACT

The nervous system consists of neurons and glial cells. Neurons generate and propagate electrical and chemical signals, whereas glia function mainly to modulate neuron function and signaling. Just as there are many different kinds of neurons with different roles, there are also many types of glia that perform diverse functions. For example, glia make myelin; modulate synapse formation, function, and elimination; regulate blood flow and metabolism; and maintain ionic and water homeostasis to name only a few. Although proteomic approaches have been used extensively to understand neurons, the same cannot be said for glia. Importantly, like neurons, glial cells have unique protein compositions that reflect their diverse functions, and these compositions can change depending on activity or disease. Here, I discuss the major classes and functions of glial cells in the central and peripheral nervous systems. I describe proteomic approaches that have been used to investigate glial cell function and composition and the experimental limitations faced by investigators working with glia.


Subject(s)
Neurogenesis/genetics , Neuroglia/metabolism , Neurons/metabolism , Proteomics , Axons/metabolism , Axons/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Signal Transduction , Synapses/genetics , Synapses/metabolism
16.
J Neurosci ; 36(35): 9148-60, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27581456

ABSTRACT

UNLABELLED: Action potential initiation and propagation in myelinated axons require ion channel clustering at axon initial segments (AIS) and nodes of Ranvier. Disruption of these domains after injury impairs nervous system function. Traditionally, injured CNS axons are considered refractory to regeneration, but some recent approaches challenge this view by showing robust long-distance regeneration. However, whether these approaches allow remyelination and promote the reestablishment of AIS and nodes of Ranvier is unknown. Using mouse optic nerve crush as a model for CNS traumatic injury, we performed a detailed analysis of AIS and node disruption after nerve crush. We found significant disruption of AIS and loss of nodes within days of the crush, and complete loss of nodes 1 week after injury. Genetic deletion of the tumor suppressor phosphatase and tensin homolog (Pten) in retinal ganglion cells (RGCs), coupled with stimulation of RGCs by inflammation and cAMP, dramatically enhanced regeneration. With this treatment, we found significant reestablishment of RGC AIS, remyelination, and even reassembly of nodes in regions proximal, within, and distal to the crush site. Remyelination began near the retina, progressed distally, and was confirmed by electron microscopy. Although axons grew rapidly, remyelination and nodal ion channel clustering was much slower. Finally, genetic deletion of ankyrinG from RGCs to block AIS reassembly did not affect axon regeneration, indicating that preservation of neuronal polarity is not required for axon regeneration. Together, our results demonstrate, for the first time, that regenerating CNS axons can be remyelinated and reassemble new AIS and nodes of Ranvier. SIGNIFICANCE STATEMENT: We show, for the first time, that regenerated CNS axons have the capacity to both remyelinate and reassemble the axon initial segments and nodes of Ranvier necessary for rapid and efficient action potential propagation.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Optic Nerve Diseases/pathology , Optic Nerve Diseases/physiopathology , Animals , Ankyrins/genetics , Ankyrins/metabolism , Axons/ultrastructure , Cell Adhesion Molecules, Neuronal , Cholera Toxin/metabolism , Disease Models, Animal , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Nerve Regeneration/genetics , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Ranvier's Nodes/metabolism , Ranvier's Nodes/pathology , Ranvier's Nodes/ultrastructure , Spectrin/metabolism , Statistics, Nonparametric , Time Factors
17.
J Neurosci ; 35(29): 10474-84, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26203142

ABSTRACT

Myelin is essential for rapid and efficient action potential propagation in vertebrates. However, the molecular mechanisms regulating myelination remain incompletely characterized. For example, even before myelination begins in the PNS, Schwann cells must radially sort axons to form 1:1 associations. Schwann cells then ensheathe and wrap axons, and establish polarized, subcellular domains, including apical and basolateral domains, paranodes, and Schmidt-Lanterman incisures. Intriguingly, polarity proteins, such as Pals1/Mpp5, are highly enriched in some of these domains, suggesting that they may regulate the polarity of Schwann cells and myelination. To test this, we generated mice with Schwann cells and oligodendrocytes that lack Pals1. During early development of the PNS, Pals1-deficient mice had impaired radial sorting of axons, delayed myelination, and reduced nerve conduction velocities. Although myelination and conduction velocities eventually recovered, polyaxonal myelination remained a prominent feature of adult Pals1-deficient nerves. Despite the enrichment of Pals1 at paranodes and incisures of control mice, nodes of Ranvier and paranodes were unaffected in Pals1-deficient mice, although we measured a significant increase in the number of incisures. As in other polarized cells, we found that Pals1 interacts with Par3 and loss of Pals1 reduced levels of Par3 in Schwann cells. In the CNS, loss of Pals1 affected neither myelination nor the establishment of polarized membrane domains. These results demonstrate that Schwann cells and oligodendrocytes use distinct mechanisms to control their polarity, and that radial sorting in the PNS is a key polarization event that requires Pals1. Significance statement: This paper reveals the role of the canonical polarity protein Pals1 in radial sorting of axons by Schwann cells. Radial sorting is essential for efficient and proper myelination and is disrupted in some types of congenital muscular dystrophy.


Subject(s)
Axons/metabolism , Cell Polarity/physiology , Membrane Proteins/metabolism , Neurogenesis/physiology , Nucleoside-Phosphate Kinase/metabolism , Schwann Cells/metabolism , Animals , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
J Neurosci ; 35(18): 7082-94, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25948259

ABSTRACT

In myelinated axons, K(+) channels are clustered in distinct membrane domains to regulate action potentials (APs). At nodes of Ranvier, Kv7 channels are expressed with Na(+) channels, whereas Kv1 channels flank nodes at juxtaparanodes. Regulation of axonal APs by K(+) channels would be particularly important in fast-spiking projection neurons such as cerebellar Purkinje cells. Here, we show that BK/Slo1 channels are clustered at the paranodal junctions of myelinated Purkinje cell axons of rat and mouse. The paranodal junction is formed by a set of cell-adhesion molecules, including Caspr, between the node and juxtaparanodes in which it separates nodal from internodal membrane domains. Remarkably, only Purkinje cell axons have detectable paranodal BK channels, whose clustering requires the formation of the paranodal junction via Caspr. Thus, BK channels occupy this unique domain in Purkinje cell axons along with the other K(+) channel complexes at nodes and juxtaparanodes. To investigate the physiological role of novel paranodal BK channels, we examined the effect of BK channel blockers on antidromic AP conduction. We found that local application of blockers to the axon resulted in a significant increase in antidromic AP failure at frequencies above 100 Hz. We also found that Ni(2+) elicited a similar effect on APs, indicating the involvement of Ni(2+)-sensitive Ca(2+) channels. Furthermore, axonal application of BK channel blockers decreased the inhibitory synaptic response in the deep cerebellar nuclei. Thus, paranodal BK channels uniquely support high-fidelity firing of APs in myelinated Purkinje cell axons, thereby underpinning the output of the cerebellar cortex.


Subject(s)
Action Potentials/physiology , Axons/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Nerve Fibers, Myelinated/physiology , Purkinje Cells/physiology , Ranvier's Nodes/physiology , Animals , Female , Intercellular Junctions/physiology , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
19.
J Neurosci ; 35(5): 2283-92, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25653382

ABSTRACT

Microglia are the brain's resident immune cells and function as the main defense against pathogens or injury. However, in the absence of disease, microglia have other functions in the normal brain. For example, previous studies showed that microglia contribute to circuit refinement and synaptic plasticity in the developing and adult brain, respectively. Thus, microglia actively participate in regulating neuronal excitability and function. Here, we report that in the cortex, but not other brain regions, a subset of microglia extend a single process that specifically associates and overlaps with the axon initial segment (AIS), the site where action potentials are generated. Similar associations were not observed with dendrites or distal axons. Microglia-AIS interactions appear early in development, persist throughout adulthood, and are conserved across species including mice, rats, and primates. However, these interactions are lost after microglial activation following brain injury, suggesting that such interactions may be part of healthy brain function. Loss of microglial CX3CR1 receptors, or the specialized extracellular matrix surrounding the AIS, did not disrupt the interaction. However, loss of AIS proteins by the neuron-specific deletion of the master AIS scaffold AnkyrinG disrupted microglia-AIS interactions. These results reveal a unique population of microglia that specifically interact with the AIS in the adult cortex.


Subject(s)
Axons/physiology , Microglia/cytology , Action Potentials , Animals , Ankyrins/genetics , Ankyrins/metabolism , Axons/metabolism , Brain Injuries/pathology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Dendrites/physiology , Extracellular Matrix/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/physiology , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/metabolism
20.
Circulation ; 131(8): 695-708, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25632041

ABSTRACT

BACKGROUND: The cardiac cytoskeleton plays key roles in maintaining myocyte structural integrity in health and disease. In fact, human mutations in cardiac cytoskeletal elements are tightly linked to cardiac pathologies, including myopathies, aortopathies, and dystrophies. Conversely, the link between cytoskeletal protein dysfunction and cardiac electric activity is not well understood and often overlooked in the cardiac arrhythmia field. METHODS AND RESULTS: Here, we uncover a new mechanism for the regulation of cardiac membrane excitability. We report that ßII spectrin, an actin-associated molecule, is essential for the posttranslational targeting and localization of critical membrane proteins in heart. ßII spectrin recruits ankyrin-B to the cardiac dyad, and a novel human mutation in the ankyrin-B gene disrupts the ankyrin-B/ßII spectrin interaction, leading to severe human arrhythmia phenotypes. Mice lacking cardiac ßII spectrin display lethal arrhythmias, aberrant electric and calcium handling phenotypes, and abnormal expression/localization of cardiac membrane proteins. Mechanistically, ßII spectrin regulates the localization of cytoskeletal and plasma membrane/sarcoplasmic reticulum protein complexes, including the Na/Ca exchanger, ryanodine receptor 2, ankyrin-B, actin, and αII spectrin. Finally, we observe accelerated heart failure phenotypes in ßII spectrin-deficient mice. CONCLUSIONS: Our findings identify ßII spectrin as critical for normal myocyte electric activity, link this molecule to human disease, and provide new insight into the mechanisms underlying cardiac myocyte biology.


Subject(s)
Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cytoskeleton/physiology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Spectrin/physiology , Amino Acid Sequence , Animals , Ankyrins/genetics , Ankyrins/physiology , Arrhythmias, Cardiac/genetics , Carrier Proteins/genetics , Carrier Proteins/physiology , Disease Models, Animal , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Membrane Proteins/physiology , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Microtubules/physiology , Molecular Sequence Data , Mutation/genetics , Phenotype , Spectrin/analysis , Spectrin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL