Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.903
Filter
1.
Methods Mol Biol ; 2799: 107-138, 2024.
Article in English | MEDLINE | ID: mdl-38727905

ABSTRACT

NMDAR-dependent forms of synaptic plasticity in brain regions like the hippocampus are widely believed to provide the neural substrate for long-term associative memory formation. However, the experimental data are equivocal at best and may suggest a more nuanced role for NMDARs and synaptic plasticity in memory. Much of the experimental data available comes from studies in genetically modified mice in which NMDAR subunits have been deleted or mutated in order to disrupt NMDAR function. Behavioral assessment of long-term memory in these mice has involved tests like the Morris watermaze and the radial arm maze. Here we describe these behavioral tests and some of the different testing protocols that can be used to assess memory performance. We discuss the importance of distinguishing selective effects on learning and memory processes from nonspecific effects on sensorimotor or motivational aspects of performance.


Subject(s)
Maze Learning , Memory, Long-Term , Receptors, N-Methyl-D-Aspartate , Spatial Memory , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Mice , Memory, Long-Term/physiology , Maze Learning/physiology , Spatial Memory/physiology , Hippocampus/physiology , Hippocampus/metabolism , Behavior, Animal/physiology , Neuronal Plasticity/physiology
2.
Cereb Cortex ; 34(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38752980

ABSTRACT

The effects of hypoxia on brain function remain largely unknown. This study aimed to clarify this issue by visual-stimulated functional magnetic resonance imaging design. Twenty-three college students with a 30-d high-altitude exposure were tested before, 1 week and 3 months after returning to sea level. Brain functional magnetic resonance imaging and retinal electroretinogram were acquired. One week after returning to sea level, decreased blood oxygenation level dependent in the right lingual gyrus accompanied with increased blood oxygenation level dependent in the frontal cortex and insular cortex, and decreased amplitude of electroretinogram a-wave in right eye; moreover, the bilateral lingual gyri showed increased functional connectivity within the dorsal visual stream pathway, and the blood oxygenation level dependent signals in the right lingual gyrus showed positive correlation with right retinal electroretinogram a-wave. Three months after returning to sea level, the blood oxygenation level dependent signals recovered to normal level, while intensively increased blood oxygenation level dependent signals in a broad of brain regions and decreased retinal electroretinogram were also existed. In conclusion, hypoxic exposure has long-term effects on visual cortex, and the impaired retinal electroretinogram may contribute to it. The increased functional connectivity of dorsal stream may compensate for the decreased function of retinal photoreceptor cells to maintain normal visual function.


Subject(s)
Electroretinography , Magnetic Resonance Imaging , Neuronal Plasticity , Visual Pathways , Humans , Male , Young Adult , Female , Neuronal Plasticity/physiology , Visual Pathways/physiology , Visual Pathways/diagnostic imaging , Hypoxia/physiopathology , Adult , Oxygen/blood , Visual Cortex/diagnostic imaging , Visual Cortex/physiology , Brain/physiology , Brain/diagnostic imaging , Photic Stimulation/methods , Retina/physiology , Retina/diagnostic imaging , Brain Mapping/methods
3.
Neuroimage ; 293: 120633, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704057

ABSTRACT

Video games are a valuable tool for studying the effects of training and neural plasticity on the brain. However, the underlying mechanisms related to plasticity-associated brain structural changes and their impact on brain dynamics are unknown. Here, we used a semi-empirical whole-brain model to study structural neural plasticity mechanisms linked to video game expertise. We hypothesized that video game expertise is associated with neural plasticity-mediated changes in structural connectivity that manifest at the meso­scale level, resulting in a more segregated functional network topology. To test this hypothesis, we combined structural connectivity data of StarCraft II video game players (VGPs, n = 31) and non-players (NVGPs, n = 31), with generic fMRI data from the Human Connectome Project and computational models, to generate simulated fMRI recordings. Graph theory analysis on simulated data was performed during both resting-state conditions and external stimulation. VGPs' simulated functional connectivity was characterized by a meso­scale integration, with increased local connectivity in frontal, parietal, and occipital brain regions. The same analyses at the level of structural connectivity showed no differences between VGPs and NVGPs. Regions that increased their connectivity strength in VGPs are known to be involved in cognitive processes crucial for task performance such as attention, reasoning, and inference. In-silico stimulation suggested that differences in FC between VGPs and NVGPs emerge in noisy contexts, specifically when the noisy level of stimulation is increased. This indicates that the connectomes of VGPs may facilitate the filtering of noise from stimuli. These structural alterations drive the meso­scale functional changes observed in individuals with gaming expertise. Overall, our work sheds light on the mechanisms underlying structural neural plasticity triggered by video game experiences.


Subject(s)
Brain , Connectome , Magnetic Resonance Imaging , Neuronal Plasticity , Video Games , Humans , Neuronal Plasticity/physiology , Connectome/methods , Male , Adult , Brain/physiology , Brain/diagnostic imaging , Young Adult , Female , Nerve Net/physiology , Nerve Net/diagnostic imaging , Models, Neurological
4.
Commun Biol ; 7(1): 555, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724614

ABSTRACT

Spatio-temporal activity patterns have been observed in a variety of brain areas in spontaneous activity, prior to or during action, or in response to stimuli. Biological mechanisms endowing neurons with the ability to distinguish between different sequences remain largely unknown. Learning sequences of spikes raises multiple challenges, such as maintaining in memory spike history and discriminating partially overlapping sequences. Here, we show that anti-Hebbian spike-timing dependent plasticity (STDP), as observed at cortico-striatal synapses, can naturally lead to learning spike sequences. We design a spiking model of the striatal output neuron receiving spike patterns defined as sequential input from a fixed set of cortical neurons. We use a simple synaptic plasticity rule that combines anti-Hebbian STDP and non-associative potentiation for a subset of the presented patterns called rewarded patterns. We study the ability of striatal output neurons to discriminate rewarded from non-rewarded patterns by firing only after the presentation of a rewarded pattern. In particular, we show that two biological properties of striatal networks, spiking latency and collateral inhibition, contribute to an increase in accuracy, by allowing a better discrimination of partially overlapping sequences. These results suggest that anti-Hebbian STDP may serve as a biological substrate for learning sequences of spikes.


Subject(s)
Corpus Striatum , Learning , Neuronal Plasticity , Neuronal Plasticity/physiology , Learning/physiology , Corpus Striatum/physiology , Models, Neurological , Animals , Action Potentials/physiology , Neurons/physiology , Humans
5.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38695719

ABSTRACT

Microglia sense the changes in their environment. How microglia actively translate these changes into suitable cues to adapt brain physiology is unknown. We reveal an activity-dependent regulation of cortical inhibitory synapses by microglia, driven by purinergic signaling acting on P2RX7 and mediated by microglia-derived TNFα. We demonstrate that sleep induces microglia-dependent synaptic enrichment of GABAARs in a manner dependent on microglial TNFα and P2RX7. We further show that microglia-specific depletion of TNFα alters slow waves during NREM sleep and blunt memory consolidation in sleep-dependent learning tasks. Together, our results reveal that microglia orchestrate sleep-intrinsic plasticity of synaptic GABAARs, sculpt sleep slow waves, and support memory consolidation.


Subject(s)
Microglia , Receptors, GABA-A , Sleep, Slow-Wave , Synapses , Tumor Necrosis Factor-alpha , Animals , Male , Mice , Memory Consolidation , Mice, Inbred C57BL , Microglia/metabolism , Neuronal Plasticity/physiology , Receptors, GABA-A/metabolism , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Signal Transduction , Sleep/physiology , Synapses/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Nat Commun ; 15(1): 3722, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697981

ABSTRACT

An important difference between brains and deep neural networks is the way they learn. Nervous systems learn online where a stream of noisy data points are presented in a non-independent, identically distributed way. Further, synaptic plasticity in the brain depends only on information local to synapses. Deep networks, on the other hand, typically use non-local learning algorithms and are trained in an offline, non-noisy, independent, identically distributed setting. Understanding how neural networks learn under the same constraints as the brain is an open problem for neuroscience and neuromorphic computing. A standard approach to this problem has yet to be established. In this paper, we propose that discrete graphical models that learn via an online maximum a posteriori learning algorithm could provide such an approach. We implement this kind of model in a neural network called the Sparse Quantized Hopfield Network. We show our model outperforms state-of-the-art neural networks on associative memory tasks, outperforms these networks in online, continual settings, learns efficiently with noisy inputs, and is better than baselines on an episodic memory task.


Subject(s)
Algorithms , Neural Networks, Computer , Humans , Memory/physiology , Models, Neurological , Brain/physiology , Neuronal Plasticity/physiology , Deep Learning
7.
F1000Res ; 13: 116, 2024.
Article in English | MEDLINE | ID: mdl-38779314

ABSTRACT

Background: Motor learning is central to human existence, such as learning to speak or walk, sports moves, or rehabilitation after injury. Evidence suggests that all forms of motor learning share an evolutionarily conserved molecular plasticity pathway. Here, we present novel insights into the neural processes underlying operant self-learning, a form of motor learning in the fruit fly Drosophila. Methods: We operantly trained wild type and transgenic Drosophila fruit flies, tethered at the torque meter, in a motor learning task that required them to initiate and maintain turning maneuvers around their vertical body axis (yaw torque). We combined this behavioral experiment with transgenic peptide expression, CRISPR/Cas9-mediated, spatio-temporally controlled gene knock-out and confocal microscopy. Results: We find that expression of atypical protein kinase C (aPKC) in direct wing steering motoneurons co-expressing the transcription factor FoxP is necessary for this type of motor learning and that aPKC likely acts via non-canonical pathways. We also found that it takes more than a week for CRISPR/Cas9-mediated knockout of FoxP in adult animals to impair motor learning, suggesting that adult FoxP expression is required for operant self-learning. Conclusions: Our experiments suggest that, for operant self-learning, a type of motor learning in Drosophila, co-expression of atypical protein kinase C (aPKC) and the transcription factor FoxP is necessary in direct wing steering motoneurons. Some of these neurons control the wing beat amplitude when generating optomotor responses, and we have discovered modulation of optomotor behavior after operant self-learning. We also discovered that aPKC likely acts via non-canonical pathways and that FoxP expression is also required in adult flies.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Motor Neurons , Protein Kinase C , Animals , Protein Kinase C/metabolism , Motor Neurons/physiology , Motor Neurons/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Learning/physiology , Forkhead Transcription Factors/metabolism , Wings, Animal/physiology , Animals, Genetically Modified , Neuronal Plasticity/physiology , Conditioning, Operant/physiology , CRISPR-Cas Systems , Drosophila/physiology
8.
Commun Biol ; 7(1): 635, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796622

ABSTRACT

The capacity to learn enabled the human species to adapt to various challenging environmental conditions and pass important achievements on to the next generation. A growing body of research suggests links between neocortical folding properties and numerous aspects of human behavior, but their impact on enhanced human learning capacity remains unexplored. Here we leverage three training cohorts to demonstrate that higher levels of premotor cortical folding reliably predict individual long-term learning gains in a challenging new motor task, above and beyond initial performance differences. Individual folding-related predisposition to motor learning was found to be independent of cortical thickness and intracortical microstructure, but dependent on larger cortical surface area in premotor regions. We further show that learning-relevant features of cortical folding occurred in close spatial proximity to practice-induced structural brain plasticity. Our results suggest a link between neocortical surface folding and human behavioral adaptability.


Subject(s)
Learning , Motor Cortex , Humans , Motor Cortex/physiology , Motor Cortex/anatomy & histology , Male , Learning/physiology , Female , Adult , Young Adult , Magnetic Resonance Imaging , Neuronal Plasticity/physiology
9.
J Vis ; 24(5): 7, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38771584

ABSTRACT

This study aimed to investigate the impact of eccentric-vision training on population receptive field (pRF) estimates to provide insights into brain plasticity processes driven by practice. Fifteen participants underwent functional magnetic resonance imaging (fMRI) measurements before and after behavioral training on a visual crowding task, where the relative orientation of the opening (gap position: up/down, left/right) in a Landolt C optotype had to be discriminated in the presence of flanking ring stimuli. Drifting checkerboard bar stimuli were used for pRF size estimation in multiple regions of interest (ROIs): dorsal-V1 (dV1), dorsal-V2 (dV2), ventral-V1 (vV1), and ventral-V2 (vV2), including the visual cortex region corresponding to the trained retinal location. pRF estimates in V1 and V2 were obtained along eccentricities from 0.5° to 9°. Statistical analyses revealed a significant decrease of the crowding anisotropy index (p = 0.009) after training, indicating improvement on crowding task performance following training. Notably, pRF sizes at and near the trained location decreased significantly (p = 0.005). Dorsal and ventral V2 exhibited significant pRF size reductions, especially at eccentricities where the training stimuli were presented (p < 0.001). In contrast, no significant changes in pRF estimates were found in either vV1 (p = 0.181) or dV1 (p = 0.055) voxels. These findings suggest that practice on a crowding task can lead to a reduction of pRF sizes in trained visual cortex, particularly in V2, highlighting the plasticity and adaptability of the adult visual system induced by prolonged training.


Subject(s)
Magnetic Resonance Imaging , Neuronal Plasticity , Visual Cortex , Visual Fields , Humans , Male , Female , Visual Cortex/physiology , Adult , Visual Fields/physiology , Magnetic Resonance Imaging/methods , Young Adult , Neuronal Plasticity/physiology , Photic Stimulation/methods
10.
CNS Neurosci Ther ; 30(5): e14743, 2024 May.
Article in English | MEDLINE | ID: mdl-38780008

ABSTRACT

AIMS: Nerve growth factor (NGF) loss is a potential factor for the degeneration of basal forebrain cholinergic neurons (BFCNs) in Alzheimer's disease (AD), and Rab5a is a key regulatory molecule of NGF signaling transduction. Here, we investigated the changes of Rab5a in 5 × FAD mice and further explored the mechanism of Electroacupuncture (EA) treatment in improving cognition in the early stage of AD. METHODS: The total Rab5a and Rab5a-GTP in 5-month-old 5 × FAD mice and wild-type mice were detected using WB and IP technologies. 5 × FAD mice were treated with EA at the Bai hui (DU20) and Shen ting (DU24) acupoints for 4 weeks and CRE/LOXP technology was used to confirm the role of Rab5a in AD mediated by EA stimulation. The Novel Object Recognition and Morris water maze tests were used to evaluate the cognitive function of 5 × FAD mice. The Nissl, immunohistochemistry, and Thioflavin S staining were used to observe pathological morphological changes in the basal forebrain circuit. The Golgi staining was used to investigate the synaptic plasticity of the basal forebrain circuit and WB technology was used to detect the expression levels of cholinergic-related and NGF signal-related proteins. RESULTS: The total Rab5a was unaltered, but Rab5a-GTP increased and the rab5a-positive early endosomes appeared enlarged in the hippocampus of 5 × FAD mice. Notably, EA reduced Rab5a-GTP in the hippocampus in the early stage of 5 × FAD mice. EA could improve object recognition memory and spatial learning memory by reducing Rab5a activity in the early stage of 5 × FAD mice. Moreover, EA could reduce Rab5a activity to increase NGF transduction and increase the levels of phosphorylated TrkA, AKT, and ERK in the basal forebrain and hippocampus, and increase the expression of cholinergic-related proteins, such as ChAT, vAchT, ChT1, m1AchR, and m2AchR in the basal forebrain and ChAT, m1AchR, and m2AchR in the hippocampus, improving synaptic plasticity in the basal forebrain hippocampal circuit in the early stage of 5 × FAD mice. CONCLUSIONS: Rab5a hyperactivation is an early pathological manifestation of 5 × FAD mice. EA could suppress Rab5a-GTP to promote the transduction of NGF signaling, and enhance the synaptic plasticity of the basal forebrain hippocampal circuit improving cognitive impairment in the early stage of 5 × FAD mice.


Subject(s)
Alzheimer Disease , Electroacupuncture , Mice, Transgenic , Nerve Growth Factor , rab5 GTP-Binding Proteins , Animals , rab5 GTP-Binding Proteins/metabolism , Nerve Growth Factor/metabolism , Mice , Electroacupuncture/methods , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Signal Transduction/physiology , Male , Memory/physiology , Learning/physiology , Maze Learning/physiology , Mice, Inbred C57BL , Neuronal Plasticity/physiology
11.
Synapse ; 78(4): e22292, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38813758

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) at hippocampal excitatory synapses undergo a late postnatal shift in subunit composition, from an initial prevalence of GluN2B subunit incorporation to a later predominance of GluN2A. This GluN2B to GluN2A shift alters NMDAR calcium conductance dynamics and intracellular molecular signaling that are individually regulated by distinct GluN2 signaling domains and temporally align with developmental alterations in dendritic and synaptic plasticity. However, the impacts of individual GluN2B to GluN2A signaling domains on neuronal development remain unknown. Ionotropic and intracellular signaling domains of GluN2 subunits were separated by creating chimeric GluN2 subunits that were expressed in two transgenic mouse lines. Western blot and immunoprecipitation revealed that roughly one third of native synaptic NMDARs were replaced by transformed NMDARs without altering total synaptic NMDAR content. Schaffer collateral synaptic strength was transiently increased in acutely prepared hippocampal slices at just over 3 weeks of age in animals overexpressing the GluN2B carboxy terminus. Long-term potentiation (LTP) induction following lower frequency stimulation was regulated by GluN2 ionotropic signaling domains in an age-dependent manner and LTP maintenance was enhanced by overexpression of the GluN2B CTD in mature animals. After higher frequency stimulation, the induction and maintenance of LTP were increased in young adult animals overexpressing the GluN2B ionotropic signaling domains but reduced in juveniles just over 3 weeks of age. Confocal imaging of green fluorescent protein (GFP)- labeled CA1 pyramidal neurons revealed no alterations in dendritic morphology or spine density in mice expressing chimeric GluN2 subunits. These results illustrate how individual GluN2 subunit signaling domains do or do not control physiological and morphological development of hippocampal excitatory neurons and better clarify the neurobiological factors that govern hippocampal maturation. SIGNIFICANCE STATEMENT: A developmental reduction in the magnitude of hippocampal long-term synaptic potentiation (LTP) and a concomitant improvement in spatial maze performance coincide with greater incorporation of GluN2A subunits into synaptic NMDARs. Corroborating our prior discovery that overexpression of GluN2A-type ionotropic signaling domains enables context-based navigation in immature mice, GluN2A-type ionotropic signaling domain overexpression reduces LTP induction threshold and magnitude in immature mice. Also, we previously found that GluN2B carboxy terminal domain (CTD) overexpression enhances long-term spatial memory in mature mice and now report that the GluN2B CTD is associated with greater amplitude of LTP after induction in mature mice. Thus, the late postnatal maturation of context encoding likely relies on a shift toward GluN2A-type ionotropic signaling and a reduction in the threshold to induce LTP while memory consolidation and LTP maintenance are regulated by GluN2B subunit CTD signaling.


Subject(s)
Dendrites , Hippocampus , Mice, Transgenic , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Hippocampus/metabolism , Hippocampus/growth & development , Hippocampus/physiology , Dendrites/physiology , Dendrites/metabolism , Mice , Neuronal Plasticity/physiology , Long-Term Potentiation/physiology , Synaptic Transmission/physiology , Excitatory Postsynaptic Potentials/physiology , Signal Transduction/physiology , Mice, Inbred C57BL , Male
12.
Nat Commun ; 15(1): 4645, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821918

ABSTRACT

Non-synaptic (intrinsic) plasticity of membrane excitability contributes to aspects of memory formation, but it remains unclear whether it merely facilitates synaptic long-term potentiation or plays a permissive role in determining the impact of synaptic weight increase. We use tactile stimulation and electrical activation of parallel fibers to probe intrinsic and synaptic contributions to receptive field plasticity in awake mice during two-photon calcium imaging of cerebellar Purkinje cells. Repetitive activation of both stimuli induced response potentiation that is impaired in mice with selective deficits in either synaptic or intrinsic plasticity. Spatial analysis of calcium signals demonstrated that intrinsic, but not synaptic plasticity, enhances the spread of dendritic parallel fiber response potentiation. Simultaneous dendrite and axon initial segment recordings confirm these dendritic events affect axonal output. Our findings support the hypothesis that intrinsic plasticity provides an amplification mechanism that exerts a permissive control over the impact of long-term potentiation on neuronal responsiveness.


Subject(s)
Cerebellum , Dendrites , Long-Term Potentiation , Neuronal Plasticity , Purkinje Cells , Synapses , Animals , Purkinje Cells/physiology , Mice , Neuronal Plasticity/physiology , Cerebellum/physiology , Cerebellum/cytology , Long-Term Potentiation/physiology , Dendrites/physiology , Synapses/physiology , Calcium/metabolism , Male , Axons/physiology , Mice, Inbred C57BL , Electric Stimulation , Female
13.
Biomed Pharmacother ; 175: 116688, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692060

ABSTRACT

Metabolic syndrome (MetS) is characterized by insulin resistance, hyperglycemia, excessive fat accumulation and dyslipidemia, and is known to be accompanied by neuropathological symptoms such as memory loss, anxiety, and depression. As the number of MetS patients is rapidly increasing globally, studies on the mechanisms of metabolic imbalance-related neuropathology are emerging as an important issue. Ca2+/calmodulin-dependent kinase II (CaMKII) is the main Ca2+ sensor and contributes to diverse intracellular signaling in peripheral organs and the central nervous system (CNS). CaMKII exerts diverse functions in cells, related to mechanisms such as RNA splicing, reactive oxygen species (ROS) generation, cytoskeleton, and protein-protein interactions. In the CNS, CaMKII regulates vascular function, neuronal circuits, neurotransmission, synaptic plasticity, amyloid beta toxicity, lipid metabolism, and mitochondrial function. Here, we review recent evidence for the role of CaMKII in neuropathologic issues associated with metabolic disorders.


Subject(s)
Amyloid beta-Peptides , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Lipid Metabolism , Nervous System Diseases , Neuronal Plasticity , Humans , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neuronal Plasticity/physiology , Animals , Lipid Metabolism/physiology , Amyloid beta-Peptides/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology
14.
Neuropharmacology ; 254: 109987, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38705570

ABSTRACT

Age is the largest risk factor for developing Alzheimer's disease (AD), a neurodegenerative disorder that causes a progressive and severe dementia. The underlying cause of cognitive deficits seen in AD is thought to be the disconnection of neural circuits that control memory and executive functions. Insight into the mechanisms by which AD diverges from normal aging will require identifying precisely which cellular events are driven by aging and which are impacted by AD-related pathologies. Since microglia, the brain-resident macrophages, are known to have critical roles in the formation and maintenance of neural circuits through synaptic pruning, they are well-positioned to modulate synaptic connectivity in circuits sensitive to aging or AD. In this review, we provide an overview of the current state of the field and on emerging technologies being employed to elucidate microglia-synaptic interactions in aging and AD. We also discuss the importance of leveraging genetic diversity to study how these interactions are shaped across more realistic contexts. We propose that these approaches will be essential to define specific aging- and disease-relevant trajectories for more personalized therapeutics aimed at reducing the effects of age or AD pathologies on the brain. This article is part of the Special Issue on "Microglia".


Subject(s)
Aging , Alzheimer Disease , Microglia , Synapses , Humans , Alzheimer Disease/pathology , Microglia/metabolism , Aging/physiology , Aging/pathology , Animals , Synapses/pathology , Synapses/physiology , Brain/pathology , Neuronal Plasticity/physiology
15.
Elife ; 122024 May 07.
Article in English | MEDLINE | ID: mdl-38712831

ABSTRACT

Representational drift refers to the dynamic nature of neural representations in the brain despite the behavior being seemingly stable. Although drift has been observed in many different brain regions, the mechanisms underlying it are not known. Since intrinsic neural excitability is suggested to play a key role in regulating memory allocation, fluctuations of excitability could bias the reactivation of previously stored memory ensembles and therefore act as a motor for drift. Here, we propose a rate-based plastic recurrent neural network with slow fluctuations of intrinsic excitability. We first show that subsequent reactivations of a neural ensemble can lead to drift of this ensemble. The model predicts that drift is induced by co-activation of previously active neurons along with neurons with high excitability which leads to remodeling of the recurrent weights. Consistent with previous experimental works, the drifting ensemble is informative about its temporal history. Crucially, we show that the gradual nature of the drift is necessary for decoding temporal information from the activity of the ensemble. Finally, we show that the memory is preserved and can be decoded by an output neuron having plastic synapses with the main region.


Subject(s)
Models, Neurological , Neuronal Plasticity , Neurons , Neurons/physiology , Neuronal Plasticity/physiology , Memory/physiology , Brain/physiology , Nerve Net/physiology , Animals , Humans , Action Potentials/physiology
16.
Invest Ophthalmol Vis Sci ; 65(5): 3, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691090

ABSTRACT

Purpose: Forty-hertz light flicker stimulation has been proven to reduce neurodegeneration, but its effect on optic nerve regeneration is unclear. This study explores the effect of 40-Hz light flicker in promoting optic nerve regeneration in zebrafish and investigates the underlying mechanisms. Methods: Wild-type and mpeg1:EGFP zebrafish were used to establish a model of optic nerve crush. Biocytin tracing and hematoxylin and eosin staining were employed to observe whether 40-Hz light flicker promotes regeneration of retinal ganglion cell axons and dendrites. Optomotor and optokinetic responses were evaluated to assess recovery of visual function. Immunofluorescence staining of mpeg1:EGFP zebrafish was performed to observe changes in microglia. Differentially expressed genes that promote optic nerve regeneration following 40-Hz light flicker stimulation were identified and validated through RNA-sequencing analysis and quantitative real-time PCR (qRT-PCR). Results: Zebrafish exhibited spontaneous optic nerve regeneration after optic nerve injury and restored visual function. We observed that 40-Hz light flicker significantly activated microglia following optic nerve injury and promoted regeneration of retinal ganglion cell axons and dendrites, as well as recovery of visual function. Transcriptomics and qRT-PCR analyses revealed that 40-Hz light flicker increased the expression of genes associated with neuronal plasticity, including bdnf, npas4a, fosab, fosb, egr4, and ier2a. Conclusions: To our knowledge, this study is the first to demonstrate that 40-Hz light flicker stimulation promotes regeneration of retinal ganglion cell axons and dendrites and recovery of visual function in zebrafish, which is associated with microglial activation and enhancement of neural plasticity.


Subject(s)
Microglia , Nerve Regeneration , Neuronal Plasticity , Optic Nerve Injuries , Retinal Ganglion Cells , Zebrafish , Animals , Microglia/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/physiopathology , Neuronal Plasticity/physiology , Retinal Ganglion Cells/physiology , Photic Stimulation , Disease Models, Animal , Optic Nerve/physiology , Axons/physiology , Real-Time Polymerase Chain Reaction
17.
Article in English | MEDLINE | ID: mdl-38691431

ABSTRACT

In hippocampus, synaptic plasticity and rhythmic oscillations reflect the cytological basis and the intermediate level of cognition, respectively. Transcranial ultrasound stimulation (TUS) has demonstrated the ability to elicit changes in neural response. However, the modulatory effect of TUS on synaptic plasticity and rhythmic oscillations was insufficient in the present studies, which may be attributed to the fact that TUS acts mainly through mechanical forces. To enhance the modulatory effect on synaptic plasticity and rhythmic oscillations, transcranial magneto-acoustic stimulation (TMAS) which induced a coupled electric field together with TUS's ultrasound field was applied. The modulatory effect of TMAS and TUS with a pulse repetition frequency of 100 Hz were compared. TMAS/TUS were performed on C57 mice for 7 days at two different ultrasound intensities (3 W/cm2 and 5 W/cm [Formula: see text]. Behavioral tests, long-term potential (LTP) and local field potentials in vivo were performed to evaluate TUS/TMAS modulatory effect on cognition, synaptic plasticity and rhythmic oscillations. Protein expression based on western blotting were used to investigate the under- lying mechanisms of these beneficial effects. At 5 W/cm2, TMAS-induced LTP were 113.4% compared to the sham group and 110.5% compared to TUS. Moreover, the relative power of high gamma oscillations (50-100Hz) in the TMAS group ( 1.060±0.155 %) was markedly higher than that in the TUS group ( 0.560±0.114 %) and sham group ( 0.570±0.088 %). TMAS significantly enhanced the synchronization of theta and gamma oscillations as well as theta-gamma cross-frequency coupling. Whereas, TUS did not show relative enhancements. TMAS provides enhanced effect for modulating the synaptic plasticity and rhythmic oscillations in hippocampus.


Subject(s)
Acoustic Stimulation , Hippocampus , Mice, Inbred C57BL , Transcranial Magnetic Stimulation , Animals , Mice , Transcranial Magnetic Stimulation/methods , Male , Hippocampus/physiology , Neuronal Plasticity/physiology , Cognition/physiology , Long-Term Potentiation/physiology , Ultrasonic Waves , Theta Rhythm/physiology
18.
Sci Rep ; 14(1): 10494, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714660

ABSTRACT

Binocular visual plasticity can be initiated via either bottom-up or top-down mechanisms, but it is unknown if these two forms of adult plasticity can be independently combined. In seven participants with normal binocular vision, sensory eye dominance was assessed using a binocular rivalry task, before and after a period of monocular deprivation and with and without selective attention directed towards one eye. On each trial, participants reported the dominant monocular target and the inter-ocular contrast difference between the stimuli was systematically altered to obtain estimates of ocular dominance. We found that both monocular light- and pattern-deprivation shifted dominance in favour of the deprived eye. However, this shift was completely counteracted if the non-deprived eye's stimulus was selectively attended. These results reveal that shifts in ocular dominance, driven by bottom-up and top-down selection, appear to act independently to regulate the relative contrast gain between the two eyes.


Subject(s)
Dominance, Ocular , Vision, Binocular , Humans , Vision, Binocular/physiology , Dominance, Ocular/physiology , Adult , Male , Female , Young Adult , Neuronal Plasticity/physiology , Photic Stimulation , Vision, Monocular/physiology , Visual Perception/physiology , Attention/physiology
19.
Transl Psychiatry ; 14(1): 218, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806461

ABSTRACT

Recent research shows that videogame training enhances neuronal plasticity and cognitive improvements in healthy individuals. As patients with schizophrenia exhibit reduced neuronal plasticity linked to cognitive deficits and symptoms, we investigated whether videogame-related cognitive improvements and plasticity changes extend to this population. In a training study, patients with schizophrenia and healthy controls were randomly assigned to 3D or 2D platformer videogame training or E-book reading (active control) for 8 weeks, 30 min daily. After training, both videogame conditions showed significant increases in sustained attention compared to the control condition, correlated with increased functional connectivity in a hippocampal-prefrontal network. Notably, patients trained with videogames mostly improved in negative symptoms, general psychopathology, and perceived mental health recovery. Videogames, incorporating initiative, goal setting and gratification, offer a training approach closer to real life than current psychiatric treatments. Our results provide initial evidence that they may represent a possible adjunct therapeutic intervention for complex mental disorders.


Subject(s)
Attention , Hippocampus , Magnetic Resonance Imaging , Neuronal Plasticity , Prefrontal Cortex , Schizophrenia , Video Games , Humans , Schizophrenia/physiopathology , Schizophrenia/rehabilitation , Hippocampus/physiopathology , Male , Female , Adult , Prefrontal Cortex/physiopathology , Attention/physiology , Neuronal Plasticity/physiology , Middle Aged , Schizophrenic Psychology
20.
Cell Rep ; 43(5): 114188, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38713584

ABSTRACT

Detecting novelty is ethologically useful for an organism's survival. Recent experiments characterize how different types of novelty over timescales from seconds to weeks are reflected in the activity of excitatory and inhibitory neuron types. Here, we introduce a learning mechanism, familiarity-modulated synapses (FMSs), consisting of multiplicative modulations dependent on presynaptic or pre/postsynaptic neuron activity. With FMSs, network responses that encode novelty emerge under unsupervised continual learning and minimal connectivity constraints. Implementing FMSs within an experimentally constrained model of a visual cortical circuit, we demonstrate the generalizability of FMSs by simultaneously fitting absolute, contextual, and omission novelty effects. Our model also reproduces functional diversity within cell subpopulations, leading to experimentally testable predictions about connectivity and synaptic dynamics that can produce both population-level novelty responses and heterogeneous individual neuron signals. Altogether, our findings demonstrate how simple plasticity mechanisms within a cortical circuit structure can produce qualitatively distinct and complex novelty responses.


Subject(s)
Models, Neurological , Neurons , Synapses , Synapses/physiology , Synapses/metabolism , Animals , Neurons/physiology , Neurons/metabolism , Neuronal Plasticity/physiology , Visual Cortex/physiology , Learning/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...