Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dis Model Mech ; 15(12)2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36398590

RESUMO

The proteosome inhibitor bortezomib has revolutionized the treatment of multiple hematologic malignancies, but in many cases, its efficacy is limited by a dose-dependent peripheral neuropathy. We show that human induced pluripotent stem cell (hiPSC)-derived motor neurons and sensory neurons provide a model system for the study of bortezomib-induced peripheral neuropathy, with promising implications for furthering the mechanistic understanding of and developing treatments for preventing axonal damage. Human neurons in tissue culture displayed distal-to-proximal neurite degeneration when exposed to bortezomib. This process coincided with disruptions in mitochondrial function and energy homeostasis, similar to those described in rodent models of bortezomib-induced neuropathy. Moreover, although the degenerative process was unaffected by inhibition of caspases, it was completely blocked by exogenous nicotinamide adenine dinucleotide (NAD+), a mediator of the SARM1-dependent axon degeneration pathway. We demonstrate that bortezomib-induced neurotoxicity in relevant human neurons proceeds through mitochondrial dysfunction and NAD+ depletion-mediated axon degeneration, raising the possibility that targeting these changes might provide effective therapeutics for the prevention of bortezomib-induced neuropathy and that modeling chemotherapy-induced neuropathy in human neurons has utility.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças do Sistema Nervoso Periférico , Humanos , NAD , Bortezomib/farmacologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente
2.
Elife ; 92020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32425157

RESUMO

Previously, we showed that a hierarchy of spectrin cytoskeletal proteins maintains nodal Na+ channels (Liu et al., 2020). Here, using mice lacking ß1, ß4, or ß1/ß4 spectrins, we show this hierarchy does not function at axon initial segments (AIS). Although ß1 spectrin, together with AnkyrinR (AnkR), compensates for loss of nodal ß4 spectrin, it cannot compensate at AIS. We show AnkR lacks the domain necessary for AIS localization. Whereas loss of ß4 spectrin causes motor impairment and disrupts AIS, loss of ß1 spectrin has no discernable effect on central nervous system structure or function. However, mice lacking both neuronal ß1 and ß4 spectrin show exacerbated nervous system dysfunction compared to mice lacking ß1 or ß4 spectrin alone, including profound disruption of AIS Na+ channel clustering, progressive loss of nodal Na+ channels, and seizures. These results further define the important role of AIS and nodal spectrins for nervous system function.


Assuntos
Segmento Inicial do Axônio/metabolismo , Proteínas de Transporte/metabolismo , Hipocampo/metabolismo , Proteínas dos Microfilamentos/metabolismo , Espectrina/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Anquirinas/metabolismo , Comportamento Animal , Proteínas de Transporte/genética , Células Cultivadas , Feminino , Hipocampo/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Atividade Motora , Domínios Proteicos , Teste de Desempenho do Rota-Rod , Convulsões/genética , Convulsões/metabolismo , Convulsões/fisiopatologia , Espectrina/deficiência , Espectrina/genética
3.
J Cell Biol ; 219(2)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31727776

RESUMO

Axon initial segments (AISs) initiate action potentials and regulate the trafficking of vesicles between somatodendritic and axonal compartments. However, the mechanisms controlling AIS assembly remain poorly defined. We performed differential proteomics and found nuclear mitotic apparatus protein 1 (NuMA1) is downregulated in AIS-deficient neonatal mouse brains and neurons. NuMA1 is transiently located at the AIS during development where it interacts with the scaffolding protein 4.1B and the dynein regulator lissencephaly 1 (Lis1). Silencing NuMA1 or protein 4.1B by shRNA disrupts AIS assembly, but not maintenance. Silencing Lis1 or overexpressing NuMA1 during AIS assembly increased the density of AIS proteins, including ankyrinG and neurofascin-186 (NF186). NuMA1 inhibits the endocytosis of AIS NF186 by impeding Lis1's interaction with doublecortin, a potent facilitator of NF186 endocytosis. Our results indicate the transient expression and AIS localization of NuMA1 stabilizes the developing AIS by inhibiting endocytosis and removal of AIS proteins.


Assuntos
Segmento Inicial do Axônio/metabolismo , Proteínas de Ciclo Celular/genética , Dineínas/genética , Endocitose/genética , Proteômica , 1-Alquil-2-acetilglicerofosfocolina Esterase/antagonistas & inibidores , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Anquirinas/genética , Axônios/metabolismo , Moléculas de Adesão Celular/genética , Citoesqueleto/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Fatores de Crescimento Neural/genética , Neurônios/metabolismo , Transporte Proteico/genética , RNA Interferente Pequeno/farmacologia
4.
Elife ; 82019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31461398

RESUMO

Dendritic arbor architecture profoundly impacts neuronal connectivity and function, and aberrant dendritic morphology characterizes neuropsychiatric disorders. Here, we identify the adhesion-GPCR BAI1 as an important regulator of dendritic arborization. BAI1 loss from mouse or rat hippocampal neurons causes dendritic hypertrophy, whereas BAI1 overexpression precipitates dendrite retraction. These defects specifically manifest as dendrites transition from growth to stability. BAI1-mediated growth arrest is independent of its Rac1-dependent synaptogenic function. Instead, BAI1 couples to the small GTPase RhoA, driving late RhoA activation in dendrites coincident with growth arrest. BAI1 loss lowers RhoA activation and uncouples it from dendrite dynamics, causing overgrowth. None of BAI1's known downstream effectors mediates BAI1-dependent growth arrest. Rather, BAI1 associates with the Rho-GTPase regulatory protein Bcr late in development and stimulates its cryptic RhoA-GEF activity, which functions together with its Rac1-GAP activity to terminate arborization. Our results reveal a late-acting signaling pathway mediating a key transition in dendrite development.


Assuntos
Proteínas Angiogênicas/metabolismo , Proliferação de Células , Dendritos/metabolismo , Proteínas Proto-Oncogênicas c-bcr/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Camundongos , Ratos
5.
Mol Neurobiol ; 56(6): 3948-3957, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30232777

RESUMO

Axotomy results in permanent loss of function after brain and spinal cord injuries due to the minimal regenerative propensity of the adult central nervous system (CNS). To identify pharmacological enhancers of axon regeneration, 960 compounds were screened for cortical neuron axonal regrowth using an in vitro cortical scrape assay. Diltiazem, verapamil, and bromopride were discovered to facilitate axon regeneration in rat cortical cultures, in the presence of chondroitin sulfate proteoglycans (CSPGs). Diltiazem, an L-type calcium channel blocker (L-CCB), also promotes axon outgrowth in adult primary mouse dorsal root ganglion (DRG) and induced human sensory (iSensory) neurons.


Assuntos
Axônios/fisiologia , Diltiazem/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Amidas/farmacologia , Animais , Axônios/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Sinergismo Farmacológico , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Ratos Sprague-Dawley
6.
Nat Commun ; 9(1): 3358, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30135423

RESUMO

Critical functions of intra-axonally synthesized proteins are thought to depend on regulated recruitment of mRNA from storage depots in axons. Here we show that axotomy of mammalian neurons induces translation of stored axonal mRNAs via regulation of the stress granule protein G3BP1, to support regeneration of peripheral nerves. G3BP1 aggregates within peripheral nerve axons in stress granule-like structures that decrease during regeneration, with a commensurate increase in phosphorylated G3BP1. Colocalization of G3BP1 with axonal mRNAs is also correlated with the growth state of the neuron. Disrupting G3BP functions by overexpressing a dominant-negative protein activates intra-axonal mRNA translation, increases axon growth in cultured neurons, disassembles axonal stress granule-like structures, and accelerates rat nerve regeneration in vivo.


Assuntos
Axônios/metabolismo , Grânulos Citoplasmáticos/metabolismo , Regeneração Nervosa/fisiologia , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Mensageiro/metabolismo , Animais , Células Cultivadas , Feminino , Recuperação de Fluorescência Após Fotodegradação , Células HEK293 , Humanos , Masculino , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Regeneração Nervosa/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
7.
J Neurosci ; 37(47): 11311-11322, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29038240

RESUMO

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.


Assuntos
Axônios/metabolismo , Citoesqueleto/metabolismo , Nós Neurofibrosos/metabolismo , Espectrina/metabolismo , Potenciais de Ação , Animais , Axônios/fisiologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Deleção de Genes , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Nós Neurofibrosos/fisiologia , Espectrina/genética
8.
Elife ; 52016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27901468

RESUMO

Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by mutations in Frataxin (FXN). Loss of FXN causes impaired mitochondrial function and iron homeostasis. An elevated production of reactive oxygen species (ROS) was previously proposed to contribute to the pathogenesis of FRDA. We recently showed that loss of frataxin homolog (fh), a Drosophila homolog of FXN, causes a ROS independent neurodegeneration in flies (Chen et al., 2016). In fh mutants, iron accumulation in the nervous system enhances the synthesis of sphingolipids, which in turn activates 3-phosphoinositide dependent protein kinase-1 (Pdk1) and myocyte enhancer factor-2 (Mef2) to trigger neurodegeneration of adult photoreceptors. Here, we show that loss of Fxn in the nervous system in mice also activates an iron/sphingolipid/PDK1/Mef2 pathway, indicating that the mechanism is evolutionarily conserved. Furthermore, sphingolipid levels and PDK1 activity are also increased in hearts of FRDA patients, suggesting that a similar pathway is affected in FRDA.


Assuntos
Proteínas de Ligação ao Ferro/metabolismo , Ferro/metabolismo , Fatores de Transcrição MEF2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Esfingolipídeos/metabolismo , Animais , Ataxia de Friedreich/patologia , Técnicas de Inativação de Genes , Humanos , Proteínas de Ligação ao Ferro/genética , Camundongos , Piruvato Desidrogenase Quinase de Transferência de Acetil , Frataxina
9.
Elife ; 52016 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-27343351

RESUMO

Mutations in Frataxin (FXN) cause Friedreich's ataxia (FRDA), a recessive neurodegenerative disorder. Previous studies have proposed that loss of FXN causes mitochondrial dysfunction, which triggers elevated reactive oxygen species (ROS) and leads to the demise of neurons. Here we describe a ROS independent mechanism that contributes to neurodegeneration in fly FXN mutants. We show that loss of frataxin homolog (fh) in Drosophila leads to iron toxicity, which in turn induces sphingolipid synthesis and ectopically activates 3-phosphoinositide dependent protein kinase-1 (Pdk1) and myocyte enhancer factor-2 (Mef2). Dampening iron toxicity, inhibiting sphingolipid synthesis by Myriocin, or reducing Pdk1 or Mef2 levels, all effectively suppress neurodegeneration in fh mutants. Moreover, increasing dihydrosphingosine activates Mef2 activity through PDK1 in mammalian neuronal cell line suggesting that the mechanisms are evolutionarily conserved. Our results indicate that an iron/sphingolipid/Pdk1/Mef2 pathway may play a role in FRDA.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Proteínas de Drosophila/metabolismo , Ataxia de Friedreich/fisiopatologia , Proteínas de Ligação ao Ferro/genética , Ferro/toxicidade , Fatores de Regulação Miogênica/metabolismo , Esfingolipídeos/biossíntese , Animais , Modelos Animais de Doenças , Drosophila , Frataxina
10.
J Neurosci ; 35(5): 2283-92, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653382

RESUMO

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.


Assuntos
Axônios/fisiologia , Microglia/citologia , Potenciais de Ação , Animais , Anquirinas/genética , Anquirinas/metabolismo , Axônios/metabolismo , Lesões Encefálicas/patologia , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Dendritos/fisiologia , Matriz Extracelular/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Quimiocinas/metabolismo
11.
Nat Neurosci ; 17(12): 1664-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25362473

RESUMO

The scaffolding protein ankyrin-G is required for Na(+) channel clustering at axon initial segments. It is also considered essential for Na(+) channel clustering at nodes of Ranvier to facilitate fast and efficient action potential propagation. However, notwithstanding these widely accepted roles, we show here that ankyrin-G is dispensable for nodal Na(+) channel clustering in vivo. Unexpectedly, in the absence of ankyrin-G, erythrocyte ankyrin (ankyrin-R) and its binding partner ßI spectrin substitute for and rescue nodal Na(+) channel clustering. In addition, channel clustering is also rescued after loss of nodal ßIV spectrin by ßI spectrin and ankyrin-R. In mice lacking both ankyrin-G and ankyrin-R, Na(+) channels fail to cluster at nodes. Thus, ankyrin R-ßI spectrin protein complexes function as secondary reserve Na(+) channel clustering machinery, and two independent ankyrin-spectrin protein complexes exist in myelinated axons to cluster Na(+) channels at nodes of Ranvier.


Assuntos
Anquirinas/análise , Nós Neurofibrosos/química , Canais de Sódio/análise , Espectrina/análise , Animais , Anquirinas/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Nós Neurofibrosos/metabolismo , Ratos , Canais de Sódio/metabolismo , Espectrina/metabolismo
12.
J Neurosci ; 33(16): 6964-78, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595754

RESUMO

Excitatory synapses are polarized structures that primarily reside on dendritic spines in the brain. The small GTPase Rac1 regulates the development and plasticity of synapses and spines by modulating actin dynamics. By restricting the Rac1-guanine nucleotide exchange factor Tiam1 to spines, the polarity protein Par3 promotes synapse development by spatially controlling Rac1 activation. However, the mechanism for recruiting Par3 to spines is unknown. Here, we identify brain-specific angiogenesis inhibitor 1 (BAI1) as a synaptic adhesion GPCR that is required for spinogenesis and synaptogenesis in mice and rats. We show that BAI1 interacts with Par3/Tiam1 and recruits these proteins to synaptic sites. BAI1 knockdown results in Par3/Tiam1 mislocalization and loss of activated Rac1 and filamentous actin from spines. Interestingly, BAI1 also mediates Rac-dependent engulfment in professional phagocytes through its interaction with a different Rac1-guanine nucleotide exchange factor module, ELMO/DOCK180. However, this interaction is dispensable for BAI1's role in synapse development because a BAI1 mutant that cannot interact with ELMO/DOCK180 rescues spine defects in BAI1-knockdown neurons, whereas a mutant that cannot interact with Par3/Tiam1 rescues neither spine defects nor Par3 localization. Further, overexpression of Tiam1 rescues BAI1 knockdown spine phenotypes. These results indicate that BAI1 plays an important role in synaptogenesis that is mechanistically distinct from its role in phagocytosis. Furthermore, our results provide the first example of a cell surface receptor that targets members of the PAR polarity complex to synapses.


Assuntos
Proteínas Angiogênicas/metabolismo , Proteínas de Transporte/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas de Neoplasias/metabolismo , Neurônios/fisiologia , Sinapses/metabolismo , Actinas/metabolismo , Análise de Variância , Proteínas Angiogênicas/genética , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Polaridade Celular/genética , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica , Eletroporação , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Humanos , Imageamento Tridimensional , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso , Técnicas de Patch-Clamp , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Long-Evans , Receptores Acoplados a Proteínas G , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Transfecção , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
13.
J Neurosci ; 32(21): 7232-43, 2012 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-22623668

RESUMO

Neurons are highly polarized cells with functionally distinct axonal and somatodendritic compartments. Voltage-gated sodium channels Na(v)1.2 and Na(v)1.6 are highly enriched at axon initial segments (AISs) and nodes of Ranvier, where they are necessary for generation and propagation of action potentials. Previous studies using reporter proteins in unmyelinated cultured neurons suggest that an ankyrinG-binding motif within intracellular loop 2 (L2) of sodium channels is sufficient for targeting these channels to the AIS, but mechanisms of channel targeting to nodes remain poorly understood. Using a CD4-Na(v)1.2/L2 reporter protein in rat dorsal root ganglion neuron-Schwann cell myelinating cocultures, we show that the ankyrinG-binding motif is sufficient for protein targeting to nodes of Ranvier. However, reporter proteins cannot capture the complexity of full-length channels. To determine how native, full-length sodium channels are clustered in axons, and to show the feasibility of studying these channels in vivo, we constructed fluorescently tagged and functional mouse Na(v)1.6 channels for in vivo analysis using in utero brain electroporation. We show here that wild-type tagged-Na(v)1.6 channels are efficiently clustered at nodes and AISs in vivo. Furthermore, we show that mutation of a single invariant glutamic acid residue (E1100) within the ankyrinG-binding motif blocked Na(v)1.6 targeting in neurons both in vitro and in vivo. Additionally, we show that caseine kinase phosphorylation sites within this motif, while not essential for targeting, can modulate clustering at the AIS. Thus, the ankyrinG-binding motif is both necessary and sufficient for the clustering of sodium channels at nodes of Ranvier and the AIS.


Assuntos
Anquirinas/fisiologia , Axônios/metabolismo , Transporte Proteico/genética , Transporte Proteico/fisiologia , Nós Neurofibrosos/metabolismo , Canais de Sódio/metabolismo , Animais , Anquirinas/genética , Técnicas de Cocultura , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Hipocampo/metabolismo , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Imagem Molecular/métodos , Mutação , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Ratos , Ratos Sprague-Dawley
14.
Cell ; 149(5): 1125-39, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22632975

RESUMO

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.


Assuntos
Axônios/metabolismo , Citoesqueleto/metabolismo , Neurônios/metabolismo , Animais , Anquirinas/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Neurônios/citologia , Espectrina/metabolismo
15.
Cereb Cortex ; 21(4): 884-95, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20802242

RESUMO

When rat fetuses grew from embryonic day (E) 18 to the day of birth (P0), the corticothalamic (CT) neurons, as identified by back labeling with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine (DiI), in the somatosensory cortex underwent gradual changes in the shape of their cell bodies, in their distribution in the cortical plate and in the complexity of dendritic branching. Fluorescence immunocytochemical studies indicated that in the marginal zone (MZ) the apical dendrites of the CT neurons formed contacts with horizontally oriented axons and contained putative glutamatergic, as clusters exhibiting both synaptophysin and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor GluR1 subunit immunoreactivities, and γ-aminobutyric acid (GABA)-ergic synapses, as clusters exhibiting both synaptophysin and gephyrin immunoreactivities. Quantitative analyses further revealed that during this perinatal period, the proportion of CT neurons containing glutamatergic synapses increased significantly, whereas the proportion of CT neurons containing GABAergic synapses remained virtually unchanged. Our results indicate that glutamatergic and GABAergic synapses between the CT neurons and the axons in the MZ are already formed in rat cortices as early as E18 and further suggest that the activities of the neural networks in the somatosensory cortex could be conveyed to their targets in the thalamus in rat brains at least 3 days before birth.


Assuntos
Neurogênese , Neurônios/citologia , Córtex Somatossensorial/crescimento & desenvolvimento , Sinapses/ultraestrutura , Animais , Imuno-Histoquímica , Microscopia Confocal , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...