Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(19): e2215590120, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37126693

RESUMO

Chronic stress induces depression- and anxiety-related behaviors, which are common mental disorders accompanied not only by dysfunction of the brain but also of the intestine. Activating transcription factor 4 (ATF4) is a stress-induced gene, and we previously show that it is important for gut functions; however, the contribution of the intestinal ATF4 to stress-related behaviors is not known. Here, we show that chronic stress inhibits the expression of ATF4 in gut epithelial cells. ATF4 overexpression in the colon relieves stress-related behavioral alterations in male mice, as measured by open-field test, elevated plus-maze test, and tail suspension test, whereas intestine-specific ATF4 knockout induces stress-related behavioral alterations in male mice. Furthermore, glutamatergic neurons are inhibited in the paraventricular thalamus (PVT) of two strains of intestinal ATF4-deficient mice, and selective activation of these neurons alleviates stress-related behavioral alterations in intestinal ATF4-deficient mice. The highly expressed gut-secreted peptide trefoil factor 3 (TFF3) is chosen from RNA-Seq data from ATF4 deletion mice and demonstrated decreased in gut epithelial cells, which is directly regulated by ATF4. Injection of TFF3 reverses stress-related behaviors in ATF4 knockout mice, and the beneficial effects of TFF3 are blocked by inhibiting PVT glutamatergic neurons using DREADDs. In summary, this study demonstrates the function of ATF4 in the gut-brain regulation of stress-related behavioral alterations, via TFF3 modulating PVT neural activity. This research provides evidence of gut signals regulating stress-related behavioral alterations and identifies possible drug targets for the treatment of stress-related behavioral disorders.


Assuntos
Fator 4 Ativador da Transcrição , Tálamo , Masculino , Animais , Camundongos , Fator 4 Ativador da Transcrição/metabolismo , Tálamo/metabolismo , Neurônios/metabolismo , Camundongos Knockout , Colo/metabolismo
2.
Hum Brain Mapp ; 45(4): e26586, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38433651

RESUMO

The assessment of consciousness states, especially distinguishing minimally conscious states (MCS) from unresponsive wakefulness states (UWS), constitutes a pivotal role in clinical therapies. Despite that numerous neural signatures of consciousness have been proposed, the effectiveness and reliability of such signatures for clinical consciousness assessment still remains an intense debate. Through a comprehensive review of the literature, inconsistent findings are observed about the effectiveness of diverse neural signatures. Notably, the majority of existing studies have evaluated neural signatures on a limited number of subjects (usually below 30), which may result in uncertain conclusions due to small data bias. This study presents a systematic evaluation of neural signatures with large-scale clinical resting-state electroencephalography (EEG) signals containing 99 UWS, 129 MCS, 36 emergence from the minimally conscious state, and 32 healthy subjects (296 total) collected over 3 years. A total of 380 EEG-based metrics for consciousness detection, including spectrum features, nonlinear measures, functional connectivity, and graph-based measures, are summarized and evaluated. To further mitigate the effect of data bias, the evaluation is performed with bootstrap sampling so that reliable measures can be obtained. The results of this study suggest that relative power in alpha and delta serve as dependable indicators of consciousness. With the MCS group, there is a notable increase in the phase lag index-related connectivity measures and enhanced functional connectivity between brain regions in comparison to the UWS group. A combination of features enables the development of an automatic detector of conscious states.


Assuntos
Estado de Consciência , Vigília , Humanos , Reprodutibilidade dos Testes , Benchmarking , Eletroencefalografia , Estado Vegetativo Persistente
3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33649213

RESUMO

Various neuromodulation approaches have been employed to alter neuronal spiking activity and thus regulate brain functions and alleviate neurological disorders. Infrared neural stimulation (INS) could be a potential approach for neuromodulation because it requires no tissue contact and possesses a high spatial resolution. However, the risk of overheating and an unclear mechanism hamper its application. Here we show that midinfrared stimulation (MIRS) with a specific wavelength exerts nonthermal, long-distance, and reversible modulatory effects on ion channel activity, neuronal signaling, and sensorimotor behavior. Patch-clamp recording from mouse neocortical pyramidal cells revealed that MIRS readily provides gain control over spiking activities, inhibiting spiking responses to weak inputs but enhancing those to strong inputs. MIRS also shortens action potential (AP) waveforms by accelerating its repolarization, through an increase in voltage-gated K+ (but not Na+) currents. Molecular dynamics simulations further revealed that MIRS-induced resonance vibration of -C=O bonds at the K+ channel ion selectivity filter contributes to the K+ current increase. Importantly, these effects are readily reversible and independent of temperature increase. At the behavioral level in larval zebrafish, MIRS modulates startle responses by sharply increasing the slope of the sensorimotor input-output curve. Therefore, MIRS represents a promising neuromodulation approach suitable for clinical application.


Assuntos
Comportamento Animal/efeitos da radiação , Raios Infravermelhos , Neurônios/metabolismo , Transdução de Sinais/efeitos da radiação , Transmissão Sináptica/efeitos da radiação , Peixe-Zebra/metabolismo , Potenciais de Ação/efeitos da radiação , Animais , Camundongos
4.
Mol Psychiatry ; 27(11): 4790-4799, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36138130

RESUMO

As a prime mover in Alzheimer's disease (AD), microglial activation requires membrane translocation, integration, and activation of the metamorphic protein chloride intracellular channel 1 (CLIC1), which is primarily cytoplasmic under physiological conditions. However, the formation and activation mechanisms of functional CLIC1 are unknown. Here, we found that the human antimicrobial peptide (AMP) LL-37 promoted CLIC1 membrane translocation and integration. It also activates CLIC1 to cause microglial hyperactivation, neuroinflammation, and excitotoxicity. In mouse and monkey models, LL-37 caused significant pathological phenotypes linked to AD, including elevated amyloid-ß, increased neurofibrillary tangles, enhanced neuronal death and brain atrophy, enlargement of lateral ventricles, and impairment of synaptic plasticity and cognition, while Clic1 knockout and blockade of LL-37-CLIC1 interactions inhibited these phenotypes. Given AD's association with infection and that overloading AMP may exacerbate AD, this study suggests that LL-37, which is up-regulated upon infection, may be a driving force behind AD by acting as an endogenous agonist of CLIC1.


Assuntos
Doença de Alzheimer , Catelicidinas , Canais de Cloreto , Animais , Humanos , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Catelicidinas/metabolismo , Catelicidinas/farmacologia , Canais de Cloreto/metabolismo , Microglia/metabolismo
5.
Cereb Cortex ; 31(4): 2278-2290, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33279969

RESUMO

Autapses are self-synapses of a neuron. Inhibitory autapses in the neocortex release GABA in 2 modes, synchronous release and asynchronous release (AR), providing precise and prolonged self-inhibition, respectively. A subpopulation of neocortical pyramidal cells (PCs) also forms functional autapses, activation of which promotes burst firing by strong unitary autaptic response that reflects synchronous glutamate release. However, it remains unclear whether AR occurs at PC autapses and plays a role in neuronal signaling. We performed whole-cell recordings from layer-5 PCs in slices of mouse prefrontal cortex (PFC). In response to action potential (AP) burst, 63% of PCs showed robust long-lasting autaptic AR, much stronger than synaptic AR between neighboring PCs. The autaptic AR is mediated predominantly by P/Q-type Ca2+ channels, and its strength depends on the intensity of PC activity and the level of residual Ca2+. Further experiments revealed that autaptic AR enhances spiking activities but reduces the temporal precision of post-burst APs. Together, the results show the occurrence of AR at PC autapses, the delayed and persistent glutamate AR causes self-excitation in individual PCs but may desynchronize the autaptic PC population. Thus, glutamatergic autapses should be essential elements in PFC and contribute to cortical information processing.


Assuntos
Potenciais de Ação/fisiologia , Ácido Glutâmico/metabolismo , Neocórtex/metabolismo , Inibição Neural/fisiologia , Células Piramidais/metabolismo , Sinapses/metabolismo , Animais , Estimulação Elétrica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia
6.
Proc Natl Acad Sci U S A ; 116(27): 13404-13413, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31213539

RESUMO

BRUCE/Apollon is a membrane-associated inhibitor of apoptosis protein that is essential for viability and has ubiquitin-conjugating activity. On initiation of apoptosis, the ubiquitin ligase Nrdp1/RNF41 promotes proteasomal degradation of BRUCE. Here we demonstrate that BRUCE together with the proteasome activator PA28γ causes proteasomal degradation of LC3-I and thus inhibits autophagy. LC3-I on the phagophore membrane is conjugated to phosphatidylethanolamine to form LC3-II, which is required for the formation of autophagosomes and selective recruitment of substrates. SIP/CacyBP is a ubiquitination-related protein that is highly expressed in neurons and various tumors. Under normal conditions, SIP inhibits the ubiquitination and degradation of BRUCE, probably by blocking the binding of Nrdp1 to BRUCE. On DNA damage by topoisomerase inhibitors, Nrdp1 causes monoubiquitination of SIP and thus promotes apoptosis. However, on starvation, SIP together with Rab8 enhances the translocation of BRUCE into the recycling endosome, formation of autophagosomes, and degradation of BRUCE by optineurin-mediated autophagy. Accordingly, deletion of SIP in cultured cells reduces the autophagic degradation of damaged mitochondria and cytosolic protein aggregates. Thus, by stimulating proteasomal degradation of LC3-I, BRUCE also inhibits autophagy. Conversely, SIP promotes autophagy by blocking BRUCE-dependent degradation of LC3-I and by enhancing autophagosome formation and autophagic destruction of BRUCE. These actions of BRUCE and SIP represent mechanisms that link the regulation of autophagy and apoptosis under different conditions.


Assuntos
Autofagia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Apoptose , Autofagossomos/metabolismo , Dano ao DNA , Fibroblastos , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Ubiquitinação
7.
Sheng Li Xue Bao ; 72(3): 382-390, 2020 Jun 25.
Artigo em Zh | MEDLINE | ID: mdl-32572435

RESUMO

Enteric nervous system (ENS) is composed of intestinal submucosal and myenteric plexuses. ENS may independently regulate intestinal digestive and absorptive function, and it is also known as "the second brain" or gut brain. ENS has significant specificity relative to central nervous system (CNS) in properties and functional activities of neurons and neural circuits. ENS is connected with CNS through the feedback pathway (brain-gut-axis) of sympathetic and parasympathetic nerves and peripheral primary sensory afferent nerves to form the bidirectional brain-gut-axis, which may affect emotion, appetite and behavioral states of individuals. Gastrointestinal functional disorder (GIFD) induced by ENS dysfunction may not only cause abnormal gastrointestinal function but also has been implicated in cognitive and mood disorders, such as irritable bowel syndrome (IBS). GIFD would influence deeply the quality of life in patients. Nevertheless, in the worldwide, ENS has so far received much less attention as compared with CNS. The depth of research and scale of investment in ENS studies have been much lower than those in CNS studies. The situation in China is even more evident. From ENS research history, an outstanding problem is to ignore largely the unique properties of ENS and apply mechanically the hypotheses formed in CNS studies to ENS researches. In this review, the structure and function of ENS are briefly introduced, and the importance of extraordinary characteristics of ENS is illustrated by the problems encountered in our studies.


Assuntos
Sistema Nervoso Entérico , Qualidade de Vida , Encéfalo , China , Humanos
9.
Cereb Cortex ; 27(1): 509-521, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26494800

RESUMO

Serotonergic innervation of the prefrontal cortex (PFC) modulates neuronal activity and PFC functions. However, the cellular mechanism for serotonergic modulation of neuronal excitability remains unclear. We performed patch-clamp recording at the axon of layer-5 pyramidal neurons in rodent PFC slices. We found surprisingly that the activation of 5-HT1A receptors selectively inhibits Na+ currents obtained at the axon initial segment (AIS) but not those at the axon trunk. In addition, Na+ channel subtype NaV1.2 but not NaV1.6 at the AIS is selectively modulated by 5-HT1A receptors. Further experiments revealed that the inhibitory effect is attributable to a depolarizing shift of the activation curve and a facilitation of slow inactivation of AIS Na+ currents. Consistently, dual somatic and axonal recording and simulation results demonstrate that the activation of 5-HT1A receptors could decrease the success rate of action potential (AP) backpropagation toward the somatodendritic compartments, enhancing the segregation of axonal and dendritic activities. Together, our results reveal a selective modulation of NaV1.2 distributed at the proximal AIS region and AP backpropagation by 5-HT1A receptors, suggesting a potential mechanism for serotonergic regulation of functional polarization in the dendro-axonal axis, synaptic plasticity and PFC functions.


Assuntos
Segmento Inicial do Axônio/metabolismo , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Canais de Sódio/metabolismo , Animais , Camundongos , Camundongos Mutantes , Ratos , Ratos Sprague-Dawley
10.
PLoS Biol ; 12(9): e1001944, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25203314

RESUMO

Action potential (AP) generation in inhibitory interneurons is critical for cortical excitation-inhibition balance and information processing. However, it remains unclear what determines AP initiation in different interneurons. We focused on two predominant interneuron types in neocortex: parvalbumin (PV)- and somatostatin (SST)-expressing neurons. Patch-clamp recording from mouse prefrontal cortical slices showed that axonal but not somatic Na+ channels exhibit different voltage-dependent properties. The minimal activation voltage of axonal channels in SST was substantially higher (∼7 mV) than in PV cells, consistent with differences in AP thresholds. A more mixed distribution of high- and low-threshold channel subtypes at the axon initial segment (AIS) of SST cells may lead to these differences. Surprisingly, NaV1.2 was found accumulated at AIS of SST but not PV cells; reducing NaV1.2-mediated currents in interneurons promoted recurrent network activity. Together, our results reveal the molecular identity of axonal Na+ channels in interneurons and their contribution to AP generation and regulation of network activity.


Assuntos
Potenciais de Ação/fisiologia , Interneurônios/metabolismo , Neocórtex/fisiologia , Rede Nervosa/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Axônios/metabolismo , Expressão Gênica , Interneurônios/citologia , Camundongos , Camundongos Transgênicos , Microtomia , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Neocórtex/citologia , Rede Nervosa/citologia , Parvalbuminas/genética , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Córtex Pré-Frontal/citologia , Somatostatina/genética , Somatostatina/metabolismo , Técnicas de Cultura de Tecidos
11.
Cereb Cortex ; 25(1): 258-70, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23968835

RESUMO

Delayed asynchronous release (AR) evoked by bursts of presynaptic action potentials (APs) occurs in certain types of hippocampal and neocortical inhibitory interneurons. Previous studies showed that AR provides long-lasting inhibition and desynchronizes the activity in postsynaptic cells. However, whether AR undergoes developmental change remains unknown. In this study, we performed whole-cell recording from fast-spiking (FS) interneurons and pyramidal cells (PCs) in prefrontal cortical slices obtained from juvenile and adult rats. In response to AP trains in FS neurons, AR occurred at their output synapses during both age periods, including FS autapses and FS-PC synapses; however, the AR strength was significantly weaker in adults than that in juveniles. Further experiments suggested that the reduction of AR in adult animals could be attributable to the rapid clearance of residual Ca(2+) from presynaptic terminals. Together, our results revealed that the AR strength was stronger at juvenile but weaker in adult, possibly resulting from changes in presynaptic Ca(2+) dynamics. AR changes may meet the needs of the neural network to generate different types of oscillations for cortical processing at distinct behavioral states.


Assuntos
Potenciais de Ação/fisiologia , Interneurônios/fisiologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Ácido gama-Aminobutírico/metabolismo , Fatores Etários , Animais , Sinalização do Cálcio , Potenciais Pós-Sinápticos Inibidores , Rede Nervosa/fisiologia , Ratos , Ratos Sprague-Dawley
12.
PLoS Biol ; 10(5): e1001324, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22589699

RESUMO

Down-regulation of GABAergic inhibition may result in the generation of epileptiform activities. Besides spike-triggered synchronous GABA release, changes in asynchronous release (AR) following high-frequency discharges may further regulate epileptiform activities. In brain slices obtained from surgically removed human neocortical tissues of patients with intractable epilepsy and brain tumor, we found that AR occurred at GABAergic output synapses of fast-spiking (FS) neurons and its strength depended on the type of connections, with FS autapses showing the strongest AR. In addition, we found that AR depended on residual Ca²âº at presynaptic terminals but was independent of postsynaptic firing. Furthermore, AR at FS autapses was markedly elevated in human epileptic tissue as compared to non-epileptic tissue. In a rat model of epilepsy, we found similar elevation of AR at both FS autapses and synapses onto excitatory neurons. Further experiments and analysis showed that AR elevation in epileptic tissue may result from an increase in action potential amplitude in the FS neurons and elevation of residual Ca²âº concentration. Together, these results revealed that GABAergic AR occurred at both human and rat neocortex, and its elevation in epileptic tissue may contribute to the regulation of epileptiform activities.


Assuntos
Epilepsia/metabolismo , Interneurônios/metabolismo , Neocórtex/patologia , Adolescente , Adulto , Idoso , Animais , Cálcio/metabolismo , Criança , Pré-Escolar , Fenômenos Eletrofisiológicos , Epilepsia/patologia , Feminino , Humanos , Técnicas In Vitro , Interneurônios/patologia , Masculino , Pessoa de Meia-Idade , Neocórtex/efeitos dos fármacos , Neocórtex/metabolismo , Inibição Neural , Técnicas de Patch-Clamp , Pilocarpina/administração & dosagem , Pilocarpina/farmacologia , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica , Adulto Jovem , Ácido gama-Aminobutírico/metabolismo
13.
J Neurosci ; 33(45): 17884-96, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24198377

RESUMO

Nav1.8 is a tetrodotoxin-resistant voltage-gated sodium channel selectively expressed in primary sensory neurons. Peripheral inflammation and nerve injury induce Nav1.8 accumulation in peripheral nerves. However, the mechanisms and related significance of channel accumulation in nerves remains unclear. Here we report that KIF5B promotes the forward transport of Nav1.8 to the plasma membrane and axons in dorsal root ganglion (DRG) neurons of the rat. In peripheral inflammation induced through the intraplantar injection of complete Freund's adjuvant, increased KIF5 and Nav1.8 accumulation were observed in the sciatic nerve. The knock-down of KIF5B, a highly expressed member of the KIF5 family in DRGs, reduced the current density of Nav1.8 in both cultured DRG neurons and ND7-23 cells. Overexpression of KIF5B in ND7-23 cells increased the current density and surface expression of Nav1.8, which were abolished through brefeldin A treatment, whereas the increases were lost in KIF5B mutants defective in ATP hydrolysis or cargo binding. Overexpression of KIF5B also decreased the proteasome-associated degradation of Nav1.8. In addition, coimmunoprecipitation experiments showed interactions between the N terminus of Nav1.8 and the 511-620 aa sequence in the stalk domain of KIF5B. Furthermore, KIF5B increased Nav1.8 accumulation, Nav1.8 current, and neuronal excitability detected in the axons of cultured DRG neurons, which were completely abolished by the disruption of interactions between KIF5B and the N terminus of Nav1.8. Therefore, our results reveal that KIF5B is required for the forward transport and axonal function of Nav1.8, suggesting a mechanism for axonal accumulation of Nav1.8 in inflammatory pain.


Assuntos
Axônios/metabolismo , Gânglios Espinais/metabolismo , Cinesinas/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neurônios/metabolismo , Potenciais de Ação/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Gânglios Espinais/citologia , Inflamação/metabolismo , Cinesinas/genética , Masculino , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Neurônios/citologia , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
14.
PLoS Biol ; 9(3): e1001032, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21445327

RESUMO

Dynamic balance of excitation and inhibition is crucial for network stability and cortical processing, but it is unclear how this balance is achieved at different membrane potentials (V(m)) of cortical neurons, as found during persistent activity or slow V(m) oscillation. Here we report that a V(m)-dependent modulation of recurrent inhibition between pyramidal cells (PCs) contributes to the excitation-inhibition balance. Whole-cell recording from paired layer-5 PCs in rat somatosensory cortical slices revealed that both the slow and the fast disynaptic IPSPs, presumably mediated by low-threshold spiking and fast spiking interneurons, respectively, were modulated by changes in presynaptic V(m). Somatic depolarization (>5 mV) of the presynaptic PC substantially increased the amplitude and shortened the onset latency of the slow disynaptic IPSPs in neighboring PCs, leading to a narrowed time window for EPSP integration. A similar increase in the amplitude of the fast disynaptic IPSPs in response to presynaptic depolarization was also observed. Further paired recording from PCs and interneurons revealed that PC depolarization increases EPSP amplitude and thus elevates interneuronal firing and inhibition of neighboring PCs, a reflection of the analog mode of excitatory synaptic transmission between PCs and interneurons. Together, these results revealed an immediate V(m)-dependent modulation of cortical inhibition, a key strategy through which the cortex dynamically maintains the balance of excitation and inhibition at different states of cortical activity.


Assuntos
Potenciais da Membrana/fisiologia , Neocórtex/fisiologia , Inibição Neural/fisiologia , Animais , Axônios/metabolismo , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/fisiologia , Interneurônios/fisiologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Superfamília Shaker de Canais de Potássio/metabolismo , Sinapses/metabolismo
15.
bioRxiv ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38659885

RESUMO

The stability of functional brain network is maintained by homeostatic plasticity, which restores equilibrium following perturbation. As the initiation site of action potentials, the axon initial segment (AIS) of glutamatergic projection neurons (PyNs) undergoes dynamic adjustment that exerts powerful control over neuronal firing properties in response to changes in network states. Although AIS plasticity has been reported to be coupled with the changes of network activity, it is poorly understood whether it involves direct synaptic input to the AIS. Here we show that changes of GABAergic synaptic input to the AIS of cortical PyNs, specifically from chandelier cells (ChCs), are sufficient to drive homeostatic tuning of the AIS within 1-2 weeks, while those from parvalbumin-positive basket cells do not. This tuning is reflected in the morphology of the AIS, the expression level of voltage-gated sodium channels, and the intrinsic neuronal excitability of PyNs. Interestingly, the timing of AIS tuning in PyNs of the prefrontal cortex corresponds to the recovery of changes in social behavior caused by alterations of ChC synaptic transmission. Thus, homeostatic plasticity of the AIS at postsynaptic PyNs may counteract deficits elicited by imbalanced ChC presynaptic input. Teaser: Axon initial segment dynamically responds to changes in local input from chandelier cells to prevent abnormal neuronal functions.

16.
Sci Adv ; 10(31): eadk4331, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093969

RESUMO

Homeostatic plasticity maintains the stability of functional brain networks. The axon initial segment (AIS), where action potentials start, undergoes dynamic adjustment to exert powerful control over neuronal firing properties in response to network activity changes. However, it is poorly understood whether this plasticity involves direct synaptic input to the AIS. Here, we show that changes of GABAergic synaptic input from chandelier cells (ChCs) drive homeostatic tuning of the AIS of principal neurons (PNs) in the prelimbic (PL) region, while those from parvalbumin-positive basket cells do not. This tuning is evident in AIS morphology, voltage-gated sodium channel expression, and PN excitability. Moreover, the impact of this homeostatic plasticity can be reflected in animal behavior. Social behavior, inversely linked to PL PN activity, shows time-dependent alterations tightly coupled to changes in AIS plasticity and PN excitability. Thus, AIS-originated homeostatic plasticity in PNs may counteract deficits elicited by imbalanced ChC presynaptic input at cellular and behavioral levels.


Assuntos
Segmento Inicial do Axônio , Axônios , Homeostase , Plasticidade Neuronal , Sinapses , Animais , Plasticidade Neuronal/fisiologia , Segmento Inicial do Axônio/metabolismo , Axônios/fisiologia , Axônios/metabolismo , Camundongos , Sinapses/fisiologia , Potenciais de Ação , Masculino , Neurônios GABAérgicos/fisiologia , Neurônios GABAérgicos/metabolismo
17.
J Physiol ; 591(13): 3233-51, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23568892

RESUMO

Voltage-gated K(+) (KV) channels play critical roles in shaping neuronal signals. KV channels distributed in the perisomatic regions and thick dendrites of cortical pyramidal neurons have been extensively studied. However, the properties and regulation of KV channels distributed in the thin axons remain unknown. In this study, by performing somatic and axonal patch-clamp recordings from layer 5 pyramidal neurons of prefrontal cortical slices, we showed that the rapidly inactivating A-currents mediated the transient K(+) currents evoked by action potential (AP) waveform command (KAP) at the soma, whereas the rapidly activating but slowly inactivating KV1-mediated D-currents dominated the KAP at the axon. In addition, activation of D1-like receptors for dopamine decreased the axonal K(+) currents, as a result of an increase in the activity of cAMP-PKA pathway. In contrast, activation of D2-like receptors showed an opposite effect on the axonal K(+) currents. Further experiments demonstrated that functional D1-like receptors were expressed at the main axon trunk and their activation could broaden the waveforms of axonal APs. Together, these results show that axonal KV channels were subjected to dopamine modulation, and this modulation could regulate the waveforms of propagating APs at the axon, suggesting an important role of dopaminergic modulation of axonal KV channels in regulating neuronal signalling.


Assuntos
Axônios/fisiologia , Dopamina/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Potenciais de Ação , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Técnicas In Vitro , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/fisiologia
18.
Nature ; 445(7123): E1-2; discussion E2-3, 2007 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17203021

RESUMO

Action potentials in cortical neurons show a variable threshold and a sudden rise in membrane potential at initiation. Naundorf et al. fail to explain these features using single- or double-compartment Hodgkin-Huxley-style models, suggesting instead that they could arise from cooperative opening of Na+ channels, although there is no direct biological evidence to support this. Here we show that these so-called unique features are to be expected from Hodgkin-Huxley models if the spatial geometry and spike initiation properties of cortical neurons are taken into account--it is therefore unnecessary to invoke exotic channel-gating properties as an explanation.


Assuntos
Potenciais de Ação , Modelos Neurológicos , Células Piramidais/metabolismo , Canais de Sódio/metabolismo , Axônios/metabolismo , Eletrofisiologia , Células Piramidais/citologia
19.
bioRxiv ; 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37502942

RESUMO

A fundamental feature of the cerebral cortex is the ability to rapidly turn on and off maintained activity within ensembles of neurons through recurrent excitation balanced by inhibition. Here we demonstrate that reduction of the h-current, which is especially prominent in pyramidal cell dendrites, strongly increases the ability of local cortical networks to generate maintained recurrent activity. Reduction of the h-current resulted in hyperpolarization and increase in input resistance of both the somata and apical dendrites of layer 5 pyramidal cells, while strongly increasing the dendrosomatic transfer of low (<20 Hz) frequencies, causing an increased responsiveness to dynamic clamp-induced recurrent network-like activity injected into the dendrites and substantially increasing the duration of spontaneous Up states. We propose that modulation of the h-current may strongly control the ability of cortical networks to generate recurrent persistent activity and the formation and dissolution of neuronal ensembles.

20.
Neuroscientist ; 29(4): 488-505, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35086369

RESUMO

Dysfunction in the prefrontal cortex is commonly implicated in anxiety disorders, but the mechanisms remain unclear. Approach-avoidance conflict tasks have been extensively used in animal research to better understand how changes in neural activity within the prefrontal cortex contribute to avoidance behaviors, which are believed to play a major role in the maintenance of anxiety disorders. In this article, we first review studies utilizing in vivo electrophysiology to reveal the relationship between changes in neural activity and avoidance behavior in rodents. We then review recent studies that take advantage of optical and genetic techniques to test the unique contribution of specific prefrontal cortex circuits and cell types to the control of anxiety-related avoidance behaviors. This new body of work reveals that behavior during approach-avoidance conflict is dynamically modulated by individual cell types, distinct neural pathways, and specific oscillatory frequencies. The integration of these different pathways, particularly as mediated by interactions between excitatory and inhibitory neurons, represents an exciting opportunity for the future of understanding anxiety.


Assuntos
Transtornos de Ansiedade , Ansiedade , Animais , Transtornos de Ansiedade/metabolismo , Córtex Pré-Frontal/fisiologia , Aprendizagem da Esquiva/fisiologia , Vias Neurais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA