Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Alzheimers Dis ; 93(4): 1425-1441, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37182881

RESUMO

BACKGROUND: In Alzheimer's disease (AD) brain, neuronal polarity and synaptic connectivity are compromised. A key structure for regulating polarity and functions of neurons is the axon initial segment (AIS), which segregates somatodendritic from axonal proteins and initiates action potentials. Toxic tau species, including extracellular oligomers (xcTauOs), spread tau pathology from neuron to neuron by a prion-like process, but few other cell biological effects of xcTauOs have been described. OBJECTIVE: Test the hypothesis that AIS structure is sensitive to xcTauOs. METHODS: Cultured wild type (WT) and tau knockout (KO) mouse cortical neurons were exposed to xcTauOs, and quantitative western blotting and immunofluorescence microscopy with anti-TRIM46 monitored effects on the AIS. The same methods were used to compare TRIM46 and two other resident AIS proteins in human hippocampal tissue obtained from AD and age-matched non-AD donors. RESULTS: Without affecting total TRIM46 levels, xcTauOs reduce the concentration of TRIM46 within the AIS and cause AIS shortening in cultured WT, but not TKO neurons. Lentiviral-driven tau expression in tau KO neurons rescues AIS length sensitivity to xcTauOs. In human AD hippocampus, the overall protein levels of multiple resident AIS proteins are unchanged compared to non-AD brain, but TRIM46 concentration within the AIS and AIS length are reduced in neurons containing neurofibrillary tangles. CONCLUSION: xcTauOs cause partial AIS damage in cultured neurons by a mechanism dependent on intracellular tau, thereby raising the possibility that the observed AIS reduction in AD neurons in vivo is caused by xcTauOs working in concert with endogenous neuronal tau.


Assuntos
Doença de Alzheimer , Segmento Inicial do Axônio , Camundongos , Animais , Humanos , Segmento Inicial do Axônio/metabolismo , Segmento Inicial do Axônio/patologia , Axônios/patologia , Neurônios/metabolismo , Doença de Alzheimer/patologia , Hipocampo/patologia , Camundongos Knockout , Proteínas tau/genética , Proteínas tau/metabolismo
2.
Dev Biol ; 489: 47-54, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35640681

RESUMO

Neurons are highly polarized cells with extensive axonal and dendritic projections that send and receive signals over long distances. Neuronal polarity requires sorting and maintaining a unique set of proteins to the neuron's distinct axonal and somatodendritic domains. The axon initial segment (AIS) is a specialized subcellular region located between these two domains and is critical for neuronal polarity. The AIS has a complex and elaborately organized molecular structure that enables its functions in neuronal polarity. Disruption of the AIS is associated with neurodevelopmental and neuropsychiatric disease pathologies, thus highlighting the importance of the AIS in neuronal physiology. This review discusses recent progress toward understanding the molecular architecture of the AIS and its importance in neuronal polarity through regulating protein diffusion and vesicular trafficking.


Assuntos
Segmento Inicial do Axônio , Segmento Inicial do Axônio/metabolismo , Segmento Inicial do Axônio/patologia , Axônios/metabolismo , Polaridade Celular/fisiologia , Neurônios/metabolismo , Transporte Proteico/fisiologia
3.
PLoS One ; 16(11): e0259918, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34797870

RESUMO

The axon initial segment (AIS) responsible for action potential initiation is a dynamic structure that varies and changes together with neuronal excitability. Like other neuron types, alpha motoneurons in the mammalian spinal cord express heterogeneity and plasticity in AIS geometry, including length (AISl) and distance from soma (AISd). The present study aimed to establish the relationship of AIS geometry with a measure of intrinsic excitability, rheobase current, that varies by 20-fold or more among normal motoneurons. We began by determining whether AIS length or distance differed for motoneurons in motor pools that exhibit different activity profiles. Motoneurons sampled from the medial gastrocnemius (MG) motor pool exhibited values for average AISd that were significantly greater than that for motoneurons from the soleus (SOL) motor pool, which is more readily recruited in low-level activities. Next, we tested whether AISd covaried with intrinsic excitability of individual motoneurons. In anesthetized rats, we measured rheobase current intracellularly from MG motoneurons in vivo before labeling them for immunohistochemical study of AIS structure. For 16 motoneurons sampled from the MG motor pool, this combinatory approach revealed that AISd, but not AISl, was significantly related to rheobase, as AIS tended to be located further from the soma on motoneurons that were less excitable. Although a causal relation with excitability seems unlikely, AISd falls among a constellation of properties related to the recruitability of motor units and their parent motoneurons.


Assuntos
Segmento Inicial do Axônio/metabolismo , Segmento Inicial do Axônio/fisiologia , Neurônios Motores/fisiologia , Potenciais de Ação/fisiologia , Animais , Segmento Inicial do Axônio/patologia , Axônios/metabolismo , Axônios/patologia , Eletrofisiologia , Masculino , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Músculos/fisiologia , Condução Nervosa , Neurônios Eferentes/fisiologia , Ratos , Ratos Wistar , Medula Espinal/fisiologia
4.
Cells ; 10(8)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34440880

RESUMO

The 20-60 µm axon initial segment (AIS) is proximally located at the interface between the axon and cell body. AIS has characteristic molecular and structural properties regulated by the crucial protein, ankyrin-G. The AIS contains a high density of Na+ channels relative to the cell body, which allows low thresholds for the initiation of action potential (AP). Molecular and physiological studies have shown that the AIS is also a key domain for the control of neuronal excitability by homeostatic mechanisms. The AIS has high plasticity in normal developmental processes and pathological activities, such as injury, neurodegeneration, and neurodevelopmental disorders (NDDs). In the first half of this review, we provide an overview of the molecular, structural, and ion-channel characteristics of AIS, AIS regulation through axo-axonic synapses, and axo-glial interactions. In the second half, to understand the relationship between NDDs and AIS, we discuss the activity-dependent plasticity of AIS, the human mutation of AIS regulatory genes, and the pathophysiological role of an abnormal AIS in NDD model animals and patients. We propose that the AIS may provide a potentially valuable structural biomarker in response to abnormal network activity in vivo as well as a new treatment concept at the neural circuit level.


Assuntos
Segmento Inicial do Axônio/patologia , Transtornos do Neurodesenvolvimento/fisiopatologia , Potenciais de Ação , Anquirinas/genética , Anquirinas/metabolismo , Segmento Inicial do Axônio/metabolismo , Humanos , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Mutação , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/metabolismo , Neuroglia/metabolismo , Plasticidade Neuronal , Espectrina/genética , Espectrina/metabolismo , Sinapses/metabolismo
5.
J Neurosci ; 41(10): 2135-2151, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33483429

RESUMO

Can alterations in experience trigger different plastic modifications in neuronal structure and function, and if so, how do they integrate at the cellular level? To address this question, we interrogated circuitry in the mouse olfactory bulb responsible for the earliest steps in odor processing. We induced experience-dependent plasticity in mice of either sex by blocking one nostril for one day, a minimally invasive manipulation that leaves the sensory organ undamaged and is akin to the natural transient blockage suffered during common mild rhinal infections. We found that such brief sensory deprivation produced structural and functional plasticity in one highly specialized bulbar cell type: axon-bearing dopaminergic neurons in the glomerular layer. After 24 h naris occlusion, the axon initial segment (AIS) in bulbar dopaminergic neurons became significantly shorter, a structural modification that was also associated with a decrease in intrinsic excitability. These effects were specific to the AIS-positive dopaminergic subpopulation because no experience-dependent alterations in intrinsic excitability were observed in AIS-negative dopaminergic cells. Moreover, 24 h naris occlusion produced no structural changes at the AIS of bulbar excitatory neurons, mitral/tufted and external tufted cells, nor did it alter their intrinsic excitability. By targeting excitability in one specialized dopaminergic subpopulation, experience-dependent plasticity in early olfactory networks might act to fine-tune sensory processing in the face of continually fluctuating inputs.SIGNIFICANCE STATEMENT Sensory networks need to be plastic so they can adapt to changes in incoming stimuli. To see how cells in mouse olfactory circuits can change in response to sensory challenges, we blocked a nostril for just one day, a naturally relevant manipulation akin to the deprivation that occurs with a mild cold. We found that this brief deprivation induces forms of axonal and intrinsic functional plasticity in one specific olfactory bulb cell subtype: axon-bearing dopaminergic interneurons. In contrast, intrinsic properties of axon-lacking bulbar dopaminergic neurons and neighboring excitatory neurons remained unchanged. Within the same sensory circuits, specific cell types can therefore make distinct plastic changes in response to an ever-changing external landscape.


Assuntos
Segmento Inicial do Axônio/patologia , Neurônios Dopaminérgicos/patologia , Plasticidade Neuronal/fisiologia , Bulbo Olfatório/fisiopatologia , Privação Sensorial/fisiologia , Animais , Segmento Inicial do Axônio/fisiologia , Neurônios Dopaminérgicos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Curr Protoc Neurosci ; 89(1): e78, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31532918

RESUMO

The axon initial segment (AIS) is the first 20- to 60-µm segment of the axon proximal to the soma of a neuron. This highly specialized subcellular domain is the initiation site of the action potential and contains a high concentration of voltage-gated ion channels held in place by a complex nexus of scaffolding and regulatory proteins that ensure proper electrical activity of the neuron. Studies have shown that dysfunction of many AIS channels and scaffolding proteins occurs in a variety of neuropsychiatric and neurodegenerative diseases, raising the need to develop accurate methods for visualization and quantification of the AIS and its protein content in models of normal and disease conditions. In this article, we describe methods for immunolabeling AIS proteins in cultured neurons and brain slices as well as methods for quantifying protein expression and pattern distribution using fluorescent labeling of these proteins. © 2019 by John Wiley & Sons, Inc.


Assuntos
Potenciais de Ação/fisiologia , Segmento Inicial do Axônio/patologia , Axônios/patologia , Neuroimagem , Neurônios/fisiologia , Animais , Segmento Inicial do Axônio/fisiologia , Axônios/fisiologia , Encéfalo/fisiologia , Células Cultivadas , Neuroimagem/métodos , Neurônios/patologia
7.
Neuron ; 104(3): 458-470.e5, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31542321

RESUMO

Dysregulation of neuronal excitability underlies the pathogenesis of tauopathies, including frontotemporal dementia (FTD) with tau inclusions. A majority of FTD-causing tau mutations are located in the microtubule-binding domain, but how these mutations alter neuronal excitability is largely unknown. Here, using CRISPR/Cas9-based gene editing in human pluripotent stem cell (iPSC)-derived neurons and isogenic controls, we show that the FTD-causing V337M tau mutation impairs activity-dependent plasticity of the cytoskeleton in the axon initial segment (AIS). Extracellular recordings by multi-electrode arrays (MEAs) revealed that the V337M tau mutation in human neurons leads to an abnormal increase in neuronal activity in response to chronic depolarization. Stochastic optical reconstruction microscopy of human neurons with this mutation showed that AIS plasticity is impaired by the abnormal accumulation of end-binding protein 3 (EB3) in the AIS submembrane region. These findings expand our understanding of how FTD-causing tau mutations dysregulate components of the neuronal cytoskeleton, leading to network dysfunction.


Assuntos
Segmento Inicial do Axônio/metabolismo , Demência Frontotemporal/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Plasticidade Neuronal/genética , Agregação Patológica de Proteínas/genética , Proteínas tau/genética , Segmento Inicial do Axônio/patologia , Citoesqueleto/metabolismo , Fenômenos Eletrofisiológicos , Espaço Extracelular , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Homeostase , Humanos , Células-Tronco Pluripotentes Induzidas , Mutação , Neurônios/metabolismo , Neurônios/patologia , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Proteínas tau/metabolismo
8.
Neuroreport ; 29(18): 1537-1543, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30320703

RESUMO

Recent studies have indicated that the structure of the axon initial segment (AIS) of neurons is highly plastic in response to changes in neuronal activity. Whether an age-related enhancement of neuronal responses in the visual cortex is coupled with plasticity of AISs is unknown. Here, we compare the AIS length and the distribution of Nav1.6, a key Na ion channel in action potential (AP) initiation, along the AIS of layer II/III neurons in the primary visual cortex (V1) of young adult and aged rats, which were examined previously in a single-unit recording study. In that study, we found that V1 neurons in aged rats showed a significantly higher spontaneous activity and stronger visually evoked responses than did neurons in young rats. Our present study shows that the mean AIS length of layer II/III neurons in the V1 area of aged rats was significantly shorter than that of young adult rats. Further, the proportion of AIS with the Nav1.6 distribution was also reduced significantly in aged rats relative to young rats, as indicated by a decrease in the mean Nav1.6 immunofluorescence optical density within AISs and a specific decrease in Nav1.6 immunofluorescence optical density near the proximal region of the AIS. Our results indicate that aging results in both shortening of AISs and reduction of Nav1.6 Na ion channel distribution along AISs, which accompanies enhanced neuronal activity. This age-related morphological plasticity may lower the AP amplitude by reducing Na ion entry during AP initiation, spare ATPs consumed by Na ion pumps during membrane potential restoration, and thus balance the energy expenditure caused by an increased firing rate of cortical neurons during the aging process.


Assuntos
Envelhecimento/fisiologia , Segmento Inicial do Axônio/patologia , Segmento Inicial do Axônio/fisiologia , Plasticidade Neuronal/fisiologia , Córtex Visual/fisiopatologia , Potenciais de Ação/fisiologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Segmento Inicial do Axônio/metabolismo , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Córtex Visual/metabolismo
9.
Ann N Y Acad Sci ; 1420(1): 46-61, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29749636

RESUMO

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.


Assuntos
Anquirinas/genética , Segmento Inicial do Axônio/patologia , Doenças do Sistema Nervoso/patologia , Transporte Proteico/fisiologia , Potenciais de Ação/fisiologia , Segmento Inicial do Axônio/ultraestrutura , Polaridade Celular/fisiologia , Humanos , Canais Iônicos/genética , Canais Iônicos/fisiologia , Neurônios/fisiologia , Nós Neurofibrosos/fisiologia
10.
Cereb Cortex ; 28(1): 395-410, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29136108

RESUMO

GABAergic synapses in the basolateral amygdala (BLA) play an important role in fear memory generation. We have previously reported that reduction in GABAergic synapses innervating specifically at the axon initial segment (AIS) of principal neurons of BLA, by neurofascin (NF) knockdown, impairs fear extinction. BLA is bidirectionally connected with the medial prefrontal cortex (mPFC), which is a key region involved in extinction of acquired fear memory. Here, we showed that reducing AIS GABAergic synapses within the BLA leads to impairment of synaptic plasticity in the BLA-mPFC pathway, as well as in the ventral subiculum (vSub)-mPFC pathway, which is independent of BLA involvement. The results suggest that the alteration within the BLA subsequently resulted in a form of trans-regional metaplasticity in the mPFC. In support of that notion, we observed that NF knockdown induced a severe deficit in behavioral flexibility as measured by reversal learning. Interestingly, reversal learning similar to extinction learning is an mPFC-dependent behavior. In agreement with that, measurement of the immediate-early gene, c-Fos immunoreactivity after reversal learning was reduced in the mPFC and BLA, supporting further the notion that the BLA GABAergic manipulation resulted in trans-regional metaplastic alterations within the mPFC.


Assuntos
Segmento Inicial do Axônio/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal/fisiologia , Sinapses/fisiologia , Vicia faba/metabolismo , Animais , Ansiedade/patologia , Ansiedade/fisiopatologia , Segmento Inicial do Axônio/efeitos dos fármacos , Segmento Inicial do Axônio/patologia , Complexo Nuclear Basolateral da Amígdala/citologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/patologia , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Condicionamento Psicológico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Hipocampo/citologia , Hipocampo/patologia , Hipocampo/fisiologia , Masculino , Memória/fisiologia , Atividade Motora/fisiologia , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/patologia , Vias Neurais/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Reversão de Aprendizagem/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/patologia
11.
eNeuro ; 4(4)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28785724

RESUMO

Little is known about the specific contributions of aging to the neuron dysfunction and death in Alzheimer's disease (AD). AD is characterized by the pathological accumulation of abnormal tau (a microtubule-associated protein), and the mislocalization of tau from the axon to the somatodendritic compartment is thought to play an important role in disease pathogenesis. The axon initial segment (AIS) is thought to play a role in the selective localization of tau in the axonal compartment. Thus, disruption in the AIS barrier may allow tau to diffuse freely back into the somatodendritic compartment and potentially lead to neurotoxicity. Here, we analyzed AISs using stereological methods and protein immunoblotting, and the localization of tau was assessed with immunofluorescence optical density measurements and protein immunoblotting. None of the outcome measurements assessed, including AIS structure, AIS protein levels, the distribution of tau in neurons of the hippocampus (HP), and total tau or phospho-tau protein levels were different in young, middle-, and old-age Fischer 344 rats. The outcome measurements assessed, including AIS structure, AIS protein levels, the distribution of tau in neurons of the HP, and total tau or phospho-tau protein levels were not different in young, middle-, and old-age Fischer 344 rats, with the exception of a small reduction in AIS volume and diameter in the CA2 region of aged animals. These data suggest that aging largely has no effect on these properties of the AIS or tau distribution, and thus, may not contribute directly to tau mislocalization.


Assuntos
Envelhecimento/metabolismo , Envelhecimento/patologia , Segmento Inicial do Axônio/metabolismo , Segmento Inicial do Axônio/patologia , Proteínas tau/metabolismo , Animais , Imunofluorescência , Hipocampo/metabolismo , Hipocampo/patologia , Immunoblotting , Masculino , Ratos Endogâmicos F344
12.
Mol Med Rep ; 16(3): 3169-3178, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713955

RESUMO

Hippocampal neurogenesis in temporal lobe epilepsy (TLE) may result in alteration of the excitability of neurons, which contributes to spontaneous recurrent seizures. Axon initial segment (AIS) structural and functional plasticity is important in the control of neuronal excitability. It remains to be elucidated whether the plasticity of AIS occurs in hippocampal newly­generated neurons that are involved in recurrent seizures following pilocarpine­induced status epilepticus (SE). The present study first established a pilocarpine­induced TLE rat model to assess the features of newborn neurons and AIS plasticity alterations using double immunofluorescence staining of Ankyrin G and doublecortin (DCX). AIS plasticity alterations include length and distance from soma in the hippocampal newly­generated neurons post­SE. The results of the present study demonstrated that pilocarpine­induced epileptic rats exhibited aberrant hippocampal neurogenesis and longer DCX­labeled cell dendrites in the dentate gyrus. Pilocarpine­induced epileptic rats demonstrated shortened lengths of AIS and an increased distance from the soma in hippocampal newborn neurons. Mibefradil, a T/L­type calcium blocker, reversed the alterations in length and position of AIS in hippocampal newborn neurons post­SE, accompanied by decreased long­term seizure activity without increased aberrant neurogenesis. These findings indicate that the plasticity of AIS in hippocampal neurogenesis may have profound consequences in epilepsy, at least in animals.


Assuntos
Segmento Inicial do Axônio/patologia , Epilepsia do Lobo Temporal/patologia , Hipocampo/patologia , Neurogênese , Animais , Segmento Inicial do Axônio/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Dendritos/efeitos dos fármacos , Dendritos/patologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Proteína Duplacortina , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Masculino , Mibefradil/farmacologia , Mibefradil/uso terapêutico , Neurogênese/efeitos dos fármacos , Pilocarpina , Ratos Sprague-Dawley , Recidiva
13.
J Neuroinflammation ; 14(1): 116, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28595650

RESUMO

BACKGROUND: Chronic microglia-mediated inflammation and oxidative stress are well-characterized underlying factors in neurodegenerative disease, whereby reactive inflammatory microglia enhance ROS production and impact neuronal integrity. Recently, it has been shown that during chronic inflammation, neuronal integrity is compromised through targeted disruption of the axon initial segment (AIS), the axonal domain critical for action potential initiation. AIS disruption was associated with contact by reactive inflammatory microglia which wrap around the AIS, increasing association with disease progression. While it is clear that chronic microglial inflammation and enhanced ROS production impact neuronal integrity, little is known about how acute microglial inflammation influences AIS stability. Here, we demonstrate that acute neuroinflammation induces AIS structural plasticity in a ROS-mediated and calpain-dependent manner. METHODS: C57BL/6J and NOX2-/- mice were given a single injection of lipopolysaccharide (LPS; 5 mg/kg) or vehicle (0.9% saline, 10 mL/kg) and analyzed at 6 h-2 weeks post-injection. Anti-inflammatory Didox (250 mg/kg) or vehicle (0.9% saline, 10 mL/kg) was administered beginning 24 h post-LPS injection and continued for 5 days; animals were analyzed 1 week post-injection. Microglial inflammation was assessed using immunohistochemistry (IHC) and RT-qPCR, and AIS integrity was quantitatively analyzed using ankyrinG immunolabeling. Data were statistically compared by one-way or two-way ANOVA where mean differences were significant as assessed using Tukey's post hoc analysis. RESULTS: LPS-induced neuroinflammation, characterized by enhanced microglial inflammation and increased expression of ROS-producing enzymes, altered AIS protein clustering. Importantly, inflammation-induced AIS changes were reversed following resolution of microglial inflammation. Modulation of the inflammatory response using anti-inflammatory Didox, even after significant AIS disruption occurred, increased the rate of AIS recovery. qPCR and IHC analysis revealed that expression of microglial NOX2, a ROS-producing enzyme, was significantly increased correlating with AIS disruption. Furthermore, ablation of NOX2 prevented inflammation-induced AIS plasticity, suggesting that ROS drive AIS structural plasticity. CONCLUSIONS: In the presence of acute microglial inflammation, the AIS undergoes an adaptive change that is capable of spontaneous recovery. Moreover, recovery can be therapeutically accelerated. Together, these findings underscore the dynamic capabilities of this domain in the presence of a pathological insult and provide evidence that the AIS is a viable therapeutic target.


Assuntos
Segmento Inicial do Axônio/enzimologia , Segmento Inicial do Axônio/patologia , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , NADPH Oxidase 2/biossíntese , Plasticidade Neuronal/fisiologia , Animais , Segmento Inicial do Axônio/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Feminino , Inflamação/induzido quimicamente , Inflamação/enzimologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/enzimologia , Microglia/patologia , Plasticidade Neuronal/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
14.
J Biol Chem ; 292(29): 12192-12207, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28536263

RESUMO

Subcellular mislocalization of the microtubule-associated protein Tau is a hallmark of Alzheimer disease (AD) and other tauopathies. Six Tau isoforms, differentiated by the presence or absence of a second repeat or of N-terminal inserts, exist in the human CNS, but their physiological and pathological differences have long remained elusive. Here, we investigated the properties and distributions of human and rodent Tau isoforms in primary forebrain rodent neurons. We found that the Tau diffusion barrier (TDB), located within the axon initial segment (AIS), controls retrograde (axon-to-soma) and anterograde (soma-to-axon) traffic of Tau. Tau isoforms without the N-terminal inserts were sorted efficiently into the axon. However, the longest isoform (2N4R-Tau) was partially retained in cell bodies and dendrites, where it accelerated spine and dendrite growth. The TDB (located within the AIS) was impaired when AIS components (ankyrin G, EB1) were knocked down or when glycogen synthase kinase-3ß (GSK3ß; an AD-associated kinase tethered to the AIS) was overexpressed. Using superresolution nanoscopy and live-cell imaging, we observed that microtubules within the AIS appeared highly dynamic, a feature essential for the TDB. Pathomechanistically, amyloid-ß insult caused cofilin activation and F-actin remodeling and decreased microtubule dynamics in the AIS. Concomitantly with these amyloid-ß-induced disruptions, the AIS/TDB sorting function failed, causing AD-like Tau missorting. In summary, we provide evidence that the human and rodent Tau isoforms differ in axodendritic sorting and amyloid-ß-induced missorting and that the axodendritic distribution of Tau depends on AIS integrity.


Assuntos
Segmento Inicial do Axônio/metabolismo , Córtex Cerebral/metabolismo , Dendritos/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Segmento Inicial do Axônio/patologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Dendritos/patologia , Difusão , Embrião de Mamíferos/citologia , Deleção de Genes , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microtúbulos/patologia , Mutagênese Insercional , Neurônios/citologia , Neurônios/patologia , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Interferência de RNA , Ratos Wistar , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sequências Repetitivas de Aminoácidos , Proteínas tau/antagonistas & inibidores , Proteínas tau/química , Proteínas tau/genética
15.
Acta Neuropathol ; 133(5): 717-730, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28091722

RESUMO

Hyperphosphorylated tau has a critical role in tauopathies such as Alzheimer's disease and frontotemporal dementia, impairing neuronal function and eventually leading to neurodegeneration. A critical role for tau is supported by studies in transgenic mouse models that express the P301L tau mutation found in cases of familial frontotemporal dementia, with the accumulation of hyperphosphorylated tau in the hippocampus causing reductions in hippocampal long-term potentiation and impairments in spatial learning and memory. However, what has remained unexplored is the role of hyperphosphorylated tau in reducing neuronal excitability. Here, we show in two complementary P301L tau transgenic mouse models that hyperphosphorylated tau induces a more depolarized threshold for action potential initiation and reduces firing in hippocampal CA1 neurons, which was rescued by the suppression of transgenic tau. Furthermore, using mutagenesis and primary hippocampal neuronal cultures, we reveal that this reduction in neuronal excitability results from the relocation of the axon initial segment (AIS) down the axon in a tau phosphorylation-dependent manner. We also demonstrate that this effect is microtubule-dependent. In addition, pharmacological stabilization was found to prevent both the structural and functional deficits caused by tau hyperphosphorylation. Finally, we demonstrate that the AIS of neurons from tau transgenic mice is further down the axon, which correlates with a reduction in excitability. We therefore propose that a reduction in hippocampal excitability due to a tau-mediated distal relocalization of the AIS contributes to the hippocampal dysfunction observed in tauopathies.


Assuntos
Segmento Inicial do Axônio/patologia , Hipocampo/metabolismo , Potenciação de Longa Duração/fisiologia , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Segmento Inicial do Axônio/metabolismo , Modelos Animais de Doenças , Hipocampo/patologia , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação
16.
Mol Neurodegener ; 11(1): 47, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27356871

RESUMO

BACKGROUND: Neurons are highly polarized cells in which asymmetric axonal-dendritic distribution of proteins is crucial for neuronal function. Loss of polarized distribution of the axonal protein tau is an early sign of Alzheimer's disease (AD) and other neurodegenerative disorders. The cytoskeletal network in the axon initial segment (AIS) forms a barrier between the axon and the somatodentritic compartment, contributing to axonal retention of tau. Although perturbation of the AIS cytoskeleton has been implicated in neurological disorders, the molecular triggers and functional consequence of AIS perturbation are incompletely understood. RESULTS: Here we report that tau acetylation and consequent destabilization of the AIS cytoskeleton promote the somatodendritic mislocalization of tau. AIS cytoskeletal proteins, including ankyrin G and ßIV-spectrin, were downregulated in AD brains and negatively correlated with an increase in tau acetylated at K274 and K281. AIS proteins were also diminished in transgenic mice expressing tauK274/281Q, a tau mutant that mimics K274 and K281 acetylation. In primary neuronal cultures, the tauK274/281Q mutant caused hyperdynamic microtubules (MTs) in the AIS, shown by live-imaging of MT mobility and fluorescence recovery after photobleaching. Using photoconvertible tau constructs, we found that axonal tauK274/281Q was missorted into the somatodendritic compartment. Stabilizing MTs with epothilone D to restore the cytoskeletal barrier in the AIS prevented tau mislocalization in primary neuronal cultures. CONCLUSIONS: Together, these findings demonstrate that tau acetylation contributes to the pathogenesis of neurodegenerative disease by compromising the cytoskeletal sorting machinery in the AIS.


Assuntos
Doença de Alzheimer/patologia , Segmento Inicial do Axônio/metabolismo , Polaridade Celular , Citoesqueleto/patologia , Proteínas tau/metabolismo , Acetilação , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Segmento Inicial do Axônio/patologia , Western Blotting , Polaridade Celular/fisiologia , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Neurônios/metabolismo , Neurônios/patologia , Ratos
17.
Exp Neurol ; 281: 93-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27109181

RESUMO

UNLABELLED: Amyloid-ß (Aß) plaques are one of the central pathologies of Alzheimer's disease (AD). Plaque formation in animal models of AD coincides with the appearance of synaptic abnormalities, aberrant neuronal excitability, and cognitive decline. Aß plaques may disrupt neuronal excitability since they have been proposed to be synaptotoxic, to induce axonal varicosities and neurite breakage, and to significantly decrease spine density. Axon initial segments (AIS) also regulate neuronal excitability and help maintain neuronal polarity. Despite these essential functions, the effects of plaques on AIS structure have not been fully determined. Using a mouse AD model, we measured a significant decrease in the density of AIS up to 75µm away from the center of fibrillar, thioflavin-labeled plaques. The reduction was observed in animals with both moderate and severe plaque loads, and was associated with increased densities of microglia near the plaques. Furthermore, animals with severe plaque loads had significantly reduced AIS lengths adjacent to Aß plaques. These results suggest the local environment surrounding Aß plaques may be harmful to the AIS. We propose that AIS loss is a previously unappreciated consequence of AD that could significantly impact brain function. SIGNIFICANCE STATEMENT: This paper demonstrates that neurons near Aß plaques have disrupted axon initial segments. Loss or disruption of AIS is predicted to have detrimental consequences for brain function.


Assuntos
Doença de Alzheimer/patologia , Segmento Inicial do Axônio/patologia , Placa Amiloide/patologia , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Segmento Inicial do Axônio/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo , Moléculas de Adesão Celular/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA