Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.512
Filtrar
1.
Mol Vis ; 30: 67-73, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38586606

RESUMO

Purpose: Light-induced neural retinal insult leads to alterations in the visual cortex neurons. We examined light-induced neuronal alterations in the visual cortex layer 5 pyramidal neurons (V1-L5PNs) of adult male Wistar rats. Methods: A total of 24 rats were divided into the following groups (n=6 each): control (NC), blue (BL), white (WL), and yellow (YL). The exposure groups were subjected to light-emitting diodes (LED) exposure (450-500 lx) of differing wavelengths for 90 days (12:12 16 light-dark cycle). After LED exposure, the animals were sacrificed, and the brain tissues were removed and impregnated in freshly prepared Golgi-Cox stain for 21 days. Sholl's grading analysis was used to quantify the apical and basal dendritic branching points and intersections of the V1-L5PNs. Results: There was a significant difference in the number of apical branching points among all groups (p<0.001), with a particularly notable difference between the BL and WL groups (p<0.001). A post hoc test revealed that all exposure groups (BL, WL, and YL) had fewer apical branching points (p<0.001) on an average of 3.6 µm and a significant reduction in the dendritic intersections (p<0.001) compared to the number of branching points extending from layer Va (V1) neurons. Conclusions: Chronic and cumulative exposure to blue and white LEDs led to the pruning of V1-L5PNs, which might impair visual processing.


Assuntos
Dendritos , Córtex Visual , Masculino , Ratos , Animais , Roedores , Ratos Wistar , Células Piramidais/fisiologia , Córtex Visual/fisiologia
2.
Nat Commun ; 15(1): 2965, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38580652

RESUMO

VGluT3-expressing mouse retinal amacrine cells (VG3s) respond to small-object motion and connect to multiple types of bipolar cells (inputs) and retinal ganglion cells (RGCs, outputs). Because these input and output connections are intermixed on the same dendrites, making sense of VG3 circuitry requires comparing the distribution of synapses across their arbors to the subcellular flow of signals. Here, we combine subcellular calcium imaging and electron microscopic connectomic reconstruction to analyze how VG3s integrate and transmit visual information. VG3s receive inputs from all nearby bipolar cell types but exhibit a strong preference for the fast type 3a bipolar cells. By comparing input distributions to VG3 dendrite responses, we show that VG3 dendrites have a short functional length constant that likely depends on inhibitory shunting. This model predicts that RGCs that extend dendrites into the middle layers of the inner plexiform encounter VG3 dendrites whose responses vary according to the local bipolar cell response type.


Assuntos
Células Amácrinas , Retina , Camundongos , Animais , Células Amácrinas/fisiologia , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Sinapses/metabolismo , Microscopia Eletrônica , Dendritos/fisiologia
3.
Biochem Biophys Res Commun ; 710: 149874, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38581950

RESUMO

Synaptic plasticity is crucial as it dynamically molds the strength and connectivity of neural circuits, influencing learning, memory, and the development of neurological disorders. Metformin, a widely prescribed anti-diabetic medication, has been shown to readily cross the blood-brain barrier (BBB) and the placenta. However, its prolonged impact on neuronal morphology and functions remains underexplored. In this study, we investigated the influence of metformin on dendrite development and synaptic plasticity in embryonic brains and primary rat cortical neurons. Our findings reveal a negative modulation of dendrite development by metformin, as evidenced by altered dendritic arborization, impaired dendritic spine morphology and disruptions in synaptic plasticity, suggesting a potential link between metformin exposure and aberrations in neuronal connectivity. In addition, we extend our insights to the impact of maternal metformin exposure on embryonic brains, revealing a significant inhibition of dendrite development in E18.5 rat brains. In conclusion, this study adds to the expanding knowledge base on the non-metabolic effects of metformin, emphasizing the significance of assessing its potential influence on both neuronal structure and function. There is an urgent need for further investigations into the enduring impact of prolonged metformin administration on the structural and functional aspects of neurons.


Assuntos
Plasticidade Neuronal , Neurônios , Gravidez , Feminino , Ratos , Animais , Plasticidade Neuronal/fisiologia , Aprendizagem , Barreira Hematoencefálica , Dendritos
4.
Environ Int ; 186: 108643, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38615544

RESUMO

Exposure to bisphenol S (BPS) is known to adversely affect neuronal development. As pivotal components of neuronal polarization, axons and dendrites are indispensable structures within neurons, crucial for the maintenance of nervous system function. Here, we investigated the impact of BPS exposure on axonal and dendritic development both in vivo and in vitro. Our results revealed that exposure to BPS during pregnancy and lactation led to a reduction in the complexity, density, and length of axons and dendrites in the prefrontal cortex (PFC) of offspring. Employing RNA sequencing technology to elucidate the underlying mechanisms of axonal and dendritic damage induced by BPS, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis highlighted a significant alteration in the oxidative phosphorylation (OXPHOS) pathway, essential for mitochondrial function. Subsequent experiments demonstrate BPS-induced impairment in mitochondrial function, including damaged morphology, decreased adenosine triphosphate (ATP) and superoxide dismutase (SOD) levels, and increased reactive oxygen species and malondialdehyde (MDA). These alterations coincided with the downregulated expression of OXPHOS pathway-related genes (ATP6V1B1, ATP5K, NDUFC1, NDUFC2, NDUFA3, COX6B1) and Myosin 19 (Myo19). Notably, Myo19 overexpression restored the BPS-induced mitochondrial dysfunction by alleviating the inhibition of OXPHOS pathway. Consequently, this amelioration was associated with a reduction in BPS-induced axonal and dendritic injury observed in cultured neurons of the PFC.


Assuntos
Axônios , Dendritos , Mitocôndrias , Fosforilação Oxidativa , Fenóis , Sulfonas , Animais , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fenóis/toxicidade , Dendritos/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Feminino , Sulfonas/toxicidade , Axônios/efeitos dos fármacos , Gravidez , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Camundongos
5.
Nature ; 628(8009): 818-825, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38658687

RESUMO

Timothy syndrome (TS) is a severe, multisystem disorder characterized by autism, epilepsy, long-QT syndrome and other neuropsychiatric conditions1. TS type 1 (TS1) is caused by a gain-of-function variant in the alternatively spliced and developmentally enriched CACNA1C exon 8A, as opposed to its counterpart exon 8. We previously uncovered several phenotypes in neurons derived from patients with TS1, including delayed channel inactivation, prolonged depolarization-induced calcium rise, impaired interneuron migration, activity-dependent dendrite retraction and an unanticipated persistent expression of exon 8A2-6. We reasoned that switching CACNA1C exon utilization from 8A to 8 would represent a potential therapeutic strategy. Here we developed antisense oligonucleotides (ASOs) to effectively decrease the inclusion of exon 8A in human cells both in vitro and, following transplantation, in vivo. We discovered that the ASO-mediated switch from exon 8A to 8 robustly rescued defects in patient-derived cortical organoids and migration in forebrain assembloids. Leveraging a transplantation platform previously developed7, we found that a single intrathecal ASO administration rescued calcium changes and in vivo dendrite retraction of patient neurons, suggesting that suppression of CACNA1C exon 8A expression is a potential treatment for TS1. Broadly, these experiments illustrate how a multilevel, in vivo and in vitro stem cell model-based approach can identify strategies to reverse disease-relevant neural pathophysiology.


Assuntos
Transtorno Autístico , Canais de Cálcio Tipo L , Movimento Celular , Éxons , Síndrome do QT Longo , Neurônios , Oligonucleotídeos Antissenso , Sindactilia , Humanos , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/administração & dosagem , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Transtorno Autístico/genética , Transtorno Autístico/terapia , Transtorno Autístico/tratamento farmacológico , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Éxons/genética , Sindactilia/genética , Sindactilia/terapia , Animais , Síndrome do QT Longo/genética , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/terapia , Camundongos , Movimento Celular/efeitos dos fármacos , Cálcio/metabolismo , Organoides/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/citologia , Processamento Alternativo/genética , Masculino , Dendritos/metabolismo , Dendritos/efeitos dos fármacos , Feminino
6.
Nat Commun ; 15(1): 2001, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38443374

RESUMO

The human subarachnoid space harbors the cerebrospinal fluid, which flows within a landscape of blood vessels and trabeculae. Functional implications of subarachnoid space anatomy remain far less understood. This study of 75 patients utilizes a cerebrospinal fluid tracer (gadobutrol) and consecutive magnetic resonance imaging to investigate features of early (i.e. within 2-3 h after injection) tracer propagation within the subarachnoid space. There is a time-dependent perivascular pattern of enrichment antegrade along the major cerebral artery trunks; the anterior-, middle-, and posterior cerebral arteries. The correlation between time of first enrichment around arteries and early enrichment in nearby cerebral cortex is significant. These observations suggest the existence of a compartmentalized subarachnoid space, where perivascular ensheathment of arteries facilitates antegrade tracer passage towards brain tissue. Periarterial transport is impaired in subjects with reduced intracranial pressure-volume reserve capacity and in idiopathic normal pressure hydrocephalus patients who also show increased perivascular space size.


Assuntos
Artérias , Espaço Subaracnóideo , Humanos , Espaço Subaracnóideo/diagnóstico por imagem , Osso Esponjoso , Córtex Cerebral , Dendritos
7.
Sci Adv ; 10(11): eadk0540, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38489360

RESUMO

During learning, synaptic connections between excitatory neurons in the brain display considerable dynamism, with new connections being added and old connections eliminated. Synapse elimination offers an opportunity to understand the features of synapses that the brain deems dispensable. However, with limited observations of synaptic activity and plasticity in vivo, the features of synapses subjected to elimination remain poorly understood. Here, we examined the functional basis of synapse elimination in the apical dendrites of L2/3 neurons in the primary motor cortex throughout motor learning. We found no evidence that synapse elimination is facilitated by a lack of activity or other local forms of plasticity. Instead, eliminated synapses display asynchronous activity with nearby synapses, suggesting that functional synaptic clustering is a critical component of synapse survival. In addition, eliminated synapses show delayed activity timing with respect to postsynaptic output. Thus, synaptic inputs that fail to be co-active with their neighboring synapses or are mistimed with neuronal output are targeted for elimination.


Assuntos
Plasticidade Neuronal , Sinapses , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Dendritos , Aprendizagem
8.
Nat Commun ; 15(1): 2142, 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38459070

RESUMO

Neuronal mitochondria play important roles beyond ATP generation, including Ca2+ uptake, and therefore have instructive roles in synaptic function and neuronal response properties. Mitochondrial morphology differs significantly between the axon and dendrites of a given neuronal subtype, but in CA1 pyramidal neurons (PNs) of the hippocampus, mitochondria within the dendritic arbor also display a remarkable degree of subcellular, layer-specific compartmentalization. In the dendrites of these neurons, mitochondria morphology ranges from highly fused and elongated in the apical tuft, to more fragmented in the apical oblique and basal dendritic compartments, and thus occupy a smaller fraction of dendritic volume than in the apical tuft. However, the molecular mechanisms underlying this striking degree of subcellular compartmentalization of mitochondria morphology are unknown, precluding the assessment of its impact on neuronal function. Here, we demonstrate that this compartment-specific morphology of dendritic mitochondria requires activity-dependent, Ca2+ and Camkk2-dependent activation of AMPK and its ability to phosphorylate two direct effectors: the pro-fission Drp1 receptor Mff and the recently identified anti-fusion, Opa1-inhibiting protein, Mtfr1l. Our study uncovers a signaling pathway underlying the subcellular compartmentalization of mitochondrial morphology in dendrites of neurons in vivo through spatially precise and activity-dependent regulation of mitochondria fission/fusion balance.


Assuntos
Neurônios , Células Piramidais , Neurônios/metabolismo , Células Piramidais/fisiologia , Hipocampo , Axônios/metabolismo , Mitocôndrias/metabolismo , Dendritos/fisiologia
9.
J Comput Neurosci ; 52(2): 125-131, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38470534

RESUMO

Long-term potentiation (LTP) is a synaptic mechanism involved in learning and memory. Experiments have shown that dendritic sodium spikes (Na-dSpikes) are required for LTP in the distal apical dendrites of CA1 pyramidal cells. On the other hand, LTP in perisomatic dendrites can be induced by synaptic input patterns that can be both subthreshold and suprathreshold for Na-dSpikes. It is unclear whether these results can be explained by one unifying plasticity mechanism. Here, we show in biophysically and morphologically realistic compartmental models of the CA1 pyramidal cell that these forms of LTP can be fully accounted for by a simple plasticity rule. We call it the voltage-based Event-Timing-Dependent Plasticity (ETDP) rule. The presynaptic event is the presynaptic spike or release of glutamate. The postsynaptic event is the local depolarization that exceeds a certain plasticity threshold. Our model reproduced the experimentally observed LTP in a variety of protocols, including local pharmacological inhibition of dendritic spikes by tetrodotoxin (TTX). In summary, we have provided a validation of the voltage-based ETDP, suggesting that this simple plasticity rule can be used to model even complex spatiotemporal patterns of long-term synaptic plasticity in neuronal dendrites.


Assuntos
Potenciais de Ação , Região CA1 Hipocampal , Dendritos , Potenciação de Longa Duração , Modelos Neurológicos , Células Piramidais , Dendritos/fisiologia , Potenciação de Longa Duração/fisiologia , Células Piramidais/fisiologia , Animais , Região CA1 Hipocampal/fisiologia , Região CA1 Hipocampal/citologia , Potenciais de Ação/fisiologia , Plasticidade Neuronal/fisiologia , Tetrodotoxina/farmacologia , Simulação por Computador
10.
Front Neural Circuits ; 18: 1280604, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505865

RESUMO

A feature of the brains of intelligent animals is the ability to learn to respond to an ensemble of active neuronal inputs with a behaviorally appropriate ensemble of active neuronal outputs. Previously, a hypothesis was proposed on how this mechanism is implemented at the cellular level within the neocortical pyramidal neuron: the apical tuft or perisomatic inputs initiate "guess" neuron firings, while the basal dendrites identify input patterns based on excited synaptic clusters, with the cluster excitation strength adjusted based on reward feedback. This simple mechanism allows neurons to learn to classify their inputs in a surprisingly intelligent manner. Here, we revise and extend this hypothesis. We modify synaptic plasticity rules to align with behavioral time scale synaptic plasticity (BTSP) observed in hippocampal area CA1, making the framework more biophysically and behaviorally plausible. The neurons for the guess firings are selected in a voluntary manner via feedback connections to apical tufts in the neocortical layer 1, leading to dendritic Ca2+ spikes with burst firing, which are postulated to be neural correlates of attentional, aware processing. Once learned, the neuronal input classification is executed without voluntary or conscious control, enabling hierarchical incremental learning of classifications that is effective in our inherently classifiable world. In addition to voluntary, we propose that pyramidal neuron burst firing can be involuntary, also initiated via apical tuft inputs, drawing attention toward important cues such as novelty and noxious stimuli. We classify the excitations of neocortical pyramidal neurons into four categories based on their excitation pathway: attentional versus automatic and voluntary/acquired versus involuntary. Additionally, we hypothesize that dendrites within pyramidal neuron minicolumn bundles are coupled via depolarization cross-induction, enabling minicolumn functions such as the creation of powerful hierarchical "hyperneurons" and the internal representation of the external world. We suggest building blocks to extend the microcircuit theory to network-level processing, which, interestingly, yields variants resembling the artificial neural networks currently in use. On a more speculative note, we conjecture that principles of intelligence in universes governed by certain types of physical laws might resemble ours.


Assuntos
Neocórtex , Sinapses , Animais , Potenciais de Ação/fisiologia , Sinapses/fisiologia , Células Piramidais/fisiologia , Dendritos/fisiologia , Neocórtex/fisiologia , Atenção
11.
Methods Mol Biol ; 2761: 57-66, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38427229

RESUMO

The objective of this chapter is to provide an overview of the methods used to investigate the connectivity and structure of the nervous system. These methods allow neuronal cells to be categorized according to their location, shape, and connections to other cells. The Golgi-Cox staining gives a thorough picture of all significant neuronal structures found in the brain that may be distinguished from one another. The most significant characteristic is its three-dimensional integrity since all neuronal structures may be followed continuously from one part to the next. Successions of sections of the brain's neurons are seen with the Golgi stain. The Golgi method is used to serially segment chosen brain parts, and the resulting neurons are produced from those sections.


Assuntos
Dendritos , Espinhas Dendríticas , Espinhas Dendríticas/fisiologia , Dendritos/fisiologia , Neurônios/fisiologia , Lobo Temporal , Coloração pela Prata , Hipocampo
12.
Mol Autism ; 15(1): 10, 2024 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383466

RESUMO

BACKGROUND: A growing body of evidence suggests that immune dysfunction and inflammation in the peripheral tissues as well as the central nervous system are associated with the neurodevelopmental deficits observed in autism spectrum disorder (ASD). Elevated expression of pro-inflammatory cytokines in the plasma, serum, and peripheral blood mononuclear cells of ASD has been reported. These cytokine expression levels are associated with the severity of behavioral impairments and symptoms in ASD. In a prior study, our group reported that tumor necrosis factor-α (TNF-α) expression in granulocyte-macrophage colony-stimulating factor-induced macrophages (GM-CSF MΦ) and the TNF-α expression ratio in GM-CSF MΦ/M-CSF MΦ (macrophage colony-stimulating factor-induced macrophages) was markedly higher in individuals with ASD than in typically developed (TD) individuals. However, the mechanisms of how the macrophages and the highly expressed cytokines affect neurons remain to be addressed. METHODS: To elucidate the effect of macrophages on human neurons, we used a co-culture system of control human-induced pluripotent stem cell-derived neurons and differentiated macrophages obtained from the peripheral blood mononuclear cells of five TD individuals and five individuals with ASD. All participants were male and ethnically Japanese. RESULTS: Our results of co-culture experiments showed that GM-CSF MΦ affect the dendritic outgrowth of neurons through the secretion of pro-inflammatory cytokines, interleukin-1α and TNF-α. Macrophages derived from individuals with ASD exerted more severe effects than those derived from TD individuals. LIMITATIONS: The main limitations of our study were the small sample size with a gender bias toward males, the use of artificially polarized macrophages, and the inability to directly observe the interaction between neurons and macrophages from the same individuals. CONCLUSIONS: Our co-culture system revealed the non-cell autonomous adverse effects of GM-CSF MΦ in individuals with ASD on neurons, mediated by interleukin-1α and TNF-α. These results may support the immune dysfunction hypothesis of ASD, providing new insights into its pathology.


Assuntos
Transtorno do Espectro Autista , Citocinas , Feminino , Masculino , Humanos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Leucócitos Mononucleares/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1alfa/farmacologia , Transtorno do Espectro Autista/metabolismo , Células Cultivadas , Sexismo , Macrófagos/metabolismo , Granulócitos/metabolismo , Dendritos/metabolismo
13.
J Neural Eng ; 21(1)2024 02 29.
Artigo em Inglês | MEDLINE | ID: mdl-38382101

RESUMO

Objective.Transcranial alternating current stimulation (tACS) is a non-invasive brain stimulation technique that directly interacts with ongoing brain oscillations in a frequency-dependent manner. However, it remains largely unclear how the cellular effects of tACS vary between cell types and subcellular elements.Approach.In this study, we use a set of morphologically realistic models of neocortical neurons to simulate the cellular response to uniform oscillating electric fields (EFs). We systematically characterize the membrane polarization in the soma, axons, and dendrites with varying field directions, intensities, and frequencies.Main results.Pyramidal cells are more sensitive to axial EF that is roughly parallel to the cortical column, while interneurons are sensitive to axial EF and transverse EF that is tangent to the cortical surface. Membrane polarization in each subcellular element increases linearly with EF intensity, and its slope, i.e. polarization length, highly depends on the stimulation frequency. At each frequency, pyramidal cells are more polarized than interneurons. Axons usually experience the highest polarization, followed by the dendrites and soma. Moreover, a visible frequency resonance presents in the apical dendrites of pyramidal cells, while the other subcellular elements primarily exhibit low-pass filtering properties. In contrast, each subcellular element of interneurons exhibits complex frequency-dependent polarization. Polarization phase in each subcellular element of cortical neurons lags that of field and exhibits high-pass filtering properties. These results demonstrate that the membrane polarization is not only frequency-dependent, but also cell type- and subcellular element-specific. Through relating effective length and ion mechanism with polarization, we emphasize the crucial role of cell morphology and biophysics in determining the frequency-dependent membrane polarization.Significance.Our findings highlight the diverse polarization patterns across cell types as well as subcellular elements, which provide some insights into the tACS cellular effects and should be considered when understanding the neural spiking activity by tACS.


Assuntos
Neocórtex , Estimulação Transcraniana por Corrente Contínua , Células Piramidais/fisiologia , Neurônios/fisiologia , Dendritos/fisiologia
14.
Neurotoxicology ; 101: 82-92, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38346645

RESUMO

Recent evidence showed that general anesthesia produces long-term neurotoxicity and cognitive dysfunction. However, it remains unclear whether maternal non-obstetric surgery under ketamine anesthesia during second trimester causes cognitive impairment in offspring. The present study assigned pregnant rats into three groups: 1) normal control group receiving no anesthesia and no surgery, 2) ketamine group receiving ketamine anesthesia for 2 h on the 14th day of gestation but no surgery, and 3) surgery group receiving abdominal surgery under ketamine anesthesia on the 14th day of gestation. On postnatal day 1, the offspring rats in Ketamine group and surgery group were assigned to receive intra-peritoneal injection of Senegenin (15 mg/kg), once per day for consecutive 14 days. The offspring's spatial perception, anxiety-like behavior, and learning and memory were evaluated. Then the offspring's hippocampal tissues were collected. The offspring of the surgery group were impaired in the spatial perception in the cliff avoidance test and the spatial learning and memory in the Morris water maze test. Accordingly, the activity of histone deacetylases increased, the protein levels of NEDD9, BDNF, p-TrkB, Syn and PSD-95 decreased, and the density of dendritic spines reduced in the hippocampus of the offspring of the surgery group, and such effects were not seen in the offspring of the ketamine group, neither in the offspring of control group. Senegenin alleviated the learning and memory impairment, and increased the protein levels of NEDD9, BDNF, p-TrkB, Syn and PSD-95 and the density of dendritic spines in the offspring of the surgery group. ketamine anesthesia plus surgery during second trimester impairs hippocampus-dependent learning and memory, and the deficits could be rescued by treatment with Senegenin.


Assuntos
Anestesia , Ketamina , Gravidez , Feminino , Ratos , Animais , Ketamina/toxicidade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Aprendizagem Espacial , Hipocampo , Dendritos , Aprendizagem em Labirinto
15.
Nat Commun ; 15(1): 1819, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418467

RESUMO

Dendritic mechanisms driving input-output transformation in starburst amacrine cells (SACs) are not fully understood. Here, we combine two-photon subcellular voltage and calcium imaging and electrophysiological recording to determine the computational architecture of mouse SAC dendrites. We found that the perisomatic region integrates motion signals over the entire dendritic field, providing a low-pass-filtered global depolarization to dendrites. Dendrites integrate local synaptic inputs with this global signal in a direction-selective manner. Coincidental local synaptic inputs and the global motion signal in the outward motion direction generate local suprathreshold calcium transients. Moreover, metabotropic glutamate receptor 2 (mGluR2) signaling in SACs modulates the initiation of calcium transients in dendrites but not at the soma. In contrast, voltage-gated potassium channel 3 (Kv3) dampens fast voltage transients at the soma. Together, complementary mGluR2 and Kv3 signaling in different subcellular regions leads to dendritic compartmentalization and direction selectivity, highlighting the importance of these mechanisms in dendritic computation.


Assuntos
Células Amácrinas , Receptores de Glutamato Metabotrópico , Animais , Camundongos , Células Amácrinas/fisiologia , Cálcio , Transdução de Sinais , Dendritos/fisiologia
16.
PLoS Comput Biol ; 20(2): e1011267, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38394339

RESUMO

Investigating and modelling the functionality of human neurons remains challenging due to the technical limitations, resulting in scarce and incomplete 3D anatomical reconstructions. Here we used a morphological modelling approach based on optimal wiring to repair the parts of a dendritic morphology that were lost due to incomplete tissue samples. In Drosophila, where dendritic regrowth has been studied experimentally using laser ablation, we found that modelling the regrowth reproduced a bimodal distribution between regeneration of cut branches and invasion by neighbouring branches. Interestingly, our repair model followed growth rules similar to those for the generation of a new dendritic tree. To generalise the repair algorithm from Drosophila to mammalian neurons, we artificially sectioned reconstructed dendrites from mouse and human hippocampal pyramidal cell morphologies, and showed that the regrown dendrites were morphologically similar to the original ones. Furthermore, we were able to restore their electrophysiological functionality, as evidenced by the recovery of their firing behaviour. Importantly, we show that such repairs also apply to other neuron types including hippocampal granule cells and cerebellar Purkinje cells. We then extrapolated the repair to incomplete human CA1 pyramidal neurons, where the anatomical boundaries of the particular brain areas innervated by the neurons in question were known. Interestingly, the repair of incomplete human dendrites helped to simulate the recently observed increased synaptic thresholds for dendritic NMDA spikes in human versus mouse dendrites. To make the repair tool available to the neuroscience community, we have developed an intuitive and simple graphical user interface (GUI), which is available in the TREES toolbox (www.treestoolbox.org).


Assuntos
Dendritos , Neurônios , Humanos , Camundongos , Animais , Dendritos/fisiologia , Neurônios/fisiologia , Células Piramidais/fisiologia , Hipocampo/fisiologia , Drosophila , Mamíferos
17.
Proc Natl Acad Sci U S A ; 121(10): e2310740121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38408233

RESUMO

Autophagy is essential for the turnover of damaged organelles and long-lived proteins. It is responsible for many biological processes such as maintaining brain functions and aging. Impaired autophagy is often linked to neurodevelopmental and neurodegenerative diseases in humans. However, the role of autophagy in neuronal pruning during development remains poorly understood. Here, we report that autophagy regulates dendrite-specific pruning of ddaC sensory neurons in parallel to local caspase activation. Impaired autophagy causes the formation of ubiquitinated protein aggregates in ddaC neurons, dependent on the autophagic receptor Ref(2)P. Furthermore, the metabolic regulator AMP-activated protein kinase and the insulin-target of rapamycin pathway act upstream to regulate autophagy during dendrite pruning. Importantly, autophagy is required to activate the transcription factor CncC (Cap "n" collar isoform C), thereby promoting dendrite pruning. Conversely, CncC also indirectly affects autophagic activity via proteasomal degradation, as impaired CncC results in the inhibition of autophagy through sequestration of Atg8a into ubiquitinated protein aggregates. Thus, this study demonstrates the important role of autophagy in activating CncC prior to dendrite pruning, and further reveals an interplay between autophagy and CncC in neuronal pruning.


Assuntos
Proteínas de Drosophila , Drosophila , Compostos de Amônio Quaternário , Animais , Humanos , Autofagia/fisiologia , Dendritos/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Plasticidade Neuronal , Proteínas Ubiquitinadas/metabolismo
18.
J Comput Neurosci ; 52(1): 1-19, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38349479

RESUMO

The vast majority of excitatory synaptic connections occur on dendritic spines. Due to their extremely small volume and spatial segregation from the dendrite, even moderate synaptic currents can significantly alter ionic concentrations. This results in chemical potential gradients between the dendrite and the spine head, leading to measurable electrical currents. In modeling electric signals in spines, different formalisms were previously used. While the cable equation is fundamental for understanding the electrical potential along dendrites, it only considers electrical currents as a result of gradients in electrical potential. The Poisson-Nernst-Planck (PNP) equations offer a more accurate description for spines by incorporating both electrical and chemical potential. However, solving PNP equations is computationally complex. In this work, diffusion currents are incorporated into the cable equation, leveraging an analogy between chemical and electrical potential. For simulating electric signals based on this extension of the cable equation, a straightforward numerical solver is introduced. The study demonstrates that this set of equations can be accurately solved using an explicit finite difference scheme. Through numerical simulations, this study unveils a previously unrecognized mechanism involving diffusion currents that amplify electric signals in spines. This discovery holds crucial implications for both numerical simulations and experimental studies focused on spine neck resistance and calcium signaling in dendritic spines.


Assuntos
Espinhas Dendríticas , Modelos Neurológicos , Sinalização do Cálcio , Dendritos , Sinapses
19.
J Neurosci ; 44(13)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38286625

RESUMO

Modern, high-density neuronal recordings reveal at ever higher precision how information is represented by neural populations. Still, we lack the tools to understand these processes bottom-up, emerging from the biophysical properties of neurons, synapses, and network structure. The concept of the dynamic gain function, a spectrally resolved approximation of a population's coding capability, has the potential to link cell-level properties to network-level performance. However, the concept is not only useful but also very complex because the dynamic gain's shape is co-determined by axonal and somato-dendritic parameters and the population's operating regime. Previously, this complexity precluded an understanding of any individual parameter's impact. Here, we decomposed the dynamic gain function into three components corresponding to separate signal transformations. This allowed attribution of network-level encoding features to specific cell-level parameters. Applying the method to data from real neurons and biophysically plausible models, we found: (1) The encoding bandwidth of real neurons, approximately 400 Hz, is constrained by the voltage dependence of axonal currents during early action potential initiation. (2) State-of-the-art models only achieve encoding bandwidths around 100 Hz and are limited mainly by subthreshold processes instead. (3) Large dendrites and low-threshold potassium currents modulate the bandwidth by shaping the subthreshold stimulus-to-voltage transformation. Our decomposition provides physiological interpretations when the dynamic gain curve changes, for instance during spectrinopathies and neurodegeneration. By pinpointing shortcomings of current models, it also guides inference of neuron models best suited for large-scale network simulations.


Assuntos
Dendritos , Neurônios , Dendritos/fisiologia , Neurônios/fisiologia , Canais Iônicos/fisiologia , Potenciais de Ação/fisiologia , Axônios , Modelos Neurológicos
20.
Neuron ; 112(7): 1133-1149.e6, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38290518

RESUMO

Dysfunction in sodium channels and their ankyrin scaffolding partners have both been implicated in neurodevelopmental disorders, including autism spectrum disorder (ASD). In particular, the genes SCN2A, which encodes the sodium channel NaV1.2, and ANK2, which encodes ankyrin-B, have strong ASD association. Recent studies indicate that ASD-associated haploinsufficiency in Scn2a impairs dendritic excitability and synaptic function in neocortical pyramidal cells, but how NaV1.2 is anchored within dendritic regions is unknown. Here, we show that ankyrin-B is essential for scaffolding NaV1.2 to the dendritic membrane of mouse neocortical neurons and that haploinsufficiency of Ank2 phenocopies intrinsic dendritic excitability and synaptic deficits observed in Scn2a+/- conditions. These results establish a direct, convergent link between two major ASD risk genes and reinforce an emerging framework suggesting that neocortical pyramidal cell dendritic dysfunction can contribute to neurodevelopmental disorder pathophysiology.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Neocórtex , Animais , Camundongos , Anquirinas/genética , Anquirinas/metabolismo , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno Autístico/metabolismo , Dendritos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Neocórtex/metabolismo , Células Piramidais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...