Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.291
Filter
1.
Cell Mol Life Sci ; 81(1): 224, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769196

ABSTRACT

Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.


Subject(s)
Brain Ischemia , Brain , Cystatin C , Extracellular Vesicles , Mice, Inbred C57BL , Synapses , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Cystatin C/metabolism , Synapses/metabolism , Mice , Male , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Brain/pathology , Proteomics/methods , Synaptosomes/metabolism , Neurons/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Cells, Cultured , Disease Models, Animal
2.
Methods Mol Biol ; 2799: 139-150, 2024.
Article in English | MEDLINE | ID: mdl-38727906

ABSTRACT

Epilepsy is one of the most represented neurological diseases worldwide. However, in many cases, the precise molecular mechanisms of epileptogenesis and ictiogenesis are unknown. Because of their important role in synaptic function and neuronal excitability, NMDA receptors are implicated in various epileptogenic mechanisms. Most of these are subunit specific and require a precise analysis of the subunit composition of the NMDARs implicated. Here, we describe an express electrophysiological method to analyze the contribution of NMDAR subunits to spontaneous postsynaptic activity in identified cells in brain slices using patch clamp whole cell recordings.


Subject(s)
Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate , Synapses , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Patch-Clamp Techniques/methods , Synapses/metabolism , Synapses/physiology , Brain/metabolism , Brain/cytology , Neurons/metabolism , Mice , Rats , Protein Subunits/metabolism
3.
Methods Mol Biol ; 2799: 257-267, 2024.
Article in English | MEDLINE | ID: mdl-38727912

ABSTRACT

The NMDAR is a heterotetramer composed of two GluN1 subunits and two GluN2 and/or GluN3 subunits, with the GluN2 subunits exhibiting significant diversity in their structure and function. Recent studies have highlighted the importance of characterizing the specific roles of each GluN2 subunit across central nervous system regions and developmental stages, as well as their unique contributions to NMDAR-mediated signaling and plasticity. Understanding the distinct functions of GluN2 subunits is critical for the development of targeted therapeutic strategies for NMDAR-related disorders. However, measuring the functional contribution of individual GluN2 subtypes in ex vivo slices is challenging. Conventionally, pharmacological or genetic approaches are used, but, in many cases, this is not possible or is restricted to population-level NMDAR responses. Here, we describe a technique for using biophysical properties of miniature synaptic NMDAR responses as a proxy to measure the functional contribution of specific GluN2-NMDAR subunits to individual synapses within a neuron.


Subject(s)
Protein Subunits , Receptors, N-Methyl-D-Aspartate , Synapses , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Synapses/metabolism , Protein Subunits/metabolism , Mice , Neurons/metabolism , Rats , Patch-Clamp Techniques/methods , Synaptic Transmission
4.
Article in English | MEDLINE | ID: mdl-38697654

ABSTRACT

A coordinated and complex interplay of signals between motor neurons, skeletal muscle cells, and Schwann cells controls the formation and maintenance of neuromuscular synapses. Deficits in the signaling pathway for building synapses, caused by mutations in critical genes or autoantibodies against key proteins, are responsible for several neuromuscular diseases, which cause muscle weakness and fatigue. Here, we describe the role that four key genes, Agrin, Lrp4, MuSK, and Dok7, play in this signaling pathway, how an understanding of their mechanisms of action has led to an understanding of several neuromuscular diseases, and how this knowledge has contributed to emerging therapies for treating neuromuscular diseases.


Subject(s)
Neuromuscular Junction , Signal Transduction , Humans , Animals , Agrin/metabolism , LDL-Receptor Related Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Muscle Proteins/metabolism , Neuromuscular Diseases , Receptors, Cholinergic/metabolism , Synapses/physiology , Synapses/metabolism , Motor Neurons/physiology , Motor Neurons/metabolism
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.
Cereb Cortex ; 34(13): 121-128, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38696601

ABSTRACT

Previous studies in autism spectrum disorder demonstrated an increased number of excitatory pyramidal cells and a decreased number of inhibitory parvalbumin+ chandelier interneurons in the prefrontal cortex of postmortem brains. How these changes in cellular composition affect the overall abundance of excitatory and inhibitory synapses in the cortex is not known. Herein, we quantified the number of excitatory and inhibitory synapses in the prefrontal cortex of 10 postmortem autism spectrum disorder brains and 10 control cases. To identify excitatory synapses, we used VGlut1 as a marker of the presynaptic component and postsynaptic density protein-95 as marker of the postsynaptic component. To identify inhibitory synapses, we used the vesicular gamma-aminobutyric acid transporter as a marker of the presynaptic component and gephyrin as a marker of the postsynaptic component. We used Puncta Analyzer to quantify the number of co-localized pre- and postsynaptic synaptic components in each area of interest. We found an increase in the number of excitatory synapses in upper cortical layers and a decrease in inhibitory synapses in all cortical layers in autism spectrum disorder brains compared with control cases. The alteration in the number of excitatory and inhibitory synapses could lead to neuronal dysfunction and disturbed network connectivity in the prefrontal cortex in autism spectrum disorder.


Subject(s)
Membrane Proteins , Prefrontal Cortex , Synapses , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Humans , Male , Female , Synapses/pathology , Synapses/metabolism , Adult , Middle Aged , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Young Adult , Adolescent , Child , Autistic Disorder/metabolism , Autistic Disorder/pathology , Neural Inhibition/physiology , Vesicular Glutamate Transport Protein 1/metabolism
7.
Sci Adv ; 10(21): eadl2882, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781346

ABSTRACT

Neuromorphic sensors, designed to emulate natural sensory systems, hold the promise of revolutionizing data extraction by facilitating rapid and energy-efficient analysis of extensive datasets. However, a challenge lies in accurately distinguishing specific analytes within mixtures of chemically similar compounds using existing neuromorphic chemical sensors. In this study, we present an artificial olfactory system (AOS), developed through the integration of human olfactory receptors (hORs) and artificial synapses. This AOS is engineered by interfacing an hOR-functionalized extended gate with an organic synaptic device. The AOS generates distinct patterns for odorants and mixtures thereof, at the molecular chain length level, attributed to specific hOR-odorant binding affinities. This approach enables precise pattern recognition via training and inference simulations. These findings establish a foundation for the development of high-performance sensor platforms and artificial sensory systems, which are ideal for applications in wearable and implantable devices.


Subject(s)
Odorants , Receptors, Odorant , Humans , Receptors, Odorant/metabolism , Odorants/analysis , Smell/physiology , Synapses/metabolism , Pattern Recognition, Automated/methods , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/physiology , Biosensing Techniques/methods
8.
Science ; 384(6698): eadg5136, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781388

ABSTRACT

The complexity and heterogeneity of schizophrenia have hindered mechanistic elucidation and the development of more effective therapies. Here, we performed single-cell dissection of schizophrenia-associated transcriptomic changes in the human prefrontal cortex across 140 individuals in two independent cohorts. Excitatory neurons were the most affected cell group, with transcriptional changes converging on neurodevelopment and synapse-related molecular pathways. Transcriptional alterations included known genetic risk factors, suggesting convergence of rare and common genomic variants on neuronal population-specific alterations in schizophrenia. Based on the magnitude of schizophrenia-associated transcriptional change, we identified two populations of individuals with schizophrenia marked by expression of specific excitatory and inhibitory neuronal cell states. This single-cell atlas links transcriptomic changes to etiological genetic risk factors, contextualizing established knowledge within the human cortical cytoarchitecture and facilitating mechanistic understanding of schizophrenia pathophysiology and heterogeneity.


Subject(s)
Neurons , Prefrontal Cortex , Schizophrenia , Single-Cell Analysis , Transcriptome , Schizophrenia/genetics , Humans , Prefrontal Cortex/metabolism , Neurons/metabolism , Cohort Studies , Male , Female , Genetic Predisposition to Disease , Adult , Synapses/metabolism , Risk Factors
9.
Biomolecules ; 14(5)2024 May 16.
Article in English | MEDLINE | ID: mdl-38785996

ABSTRACT

Excitotoxicity is a common pathological process in neurological diseases caused by excess glutamate. The purpose of this study was to evaluate the effect of gypenoside XVII (GP-17), a gypenoside monomer, on the glutamatergic system. In vitro, in rat cortical nerve terminals (synaptosomes), GP-17 dose-dependently decreased glutamate release with an IC50 value of 16 µM. The removal of extracellular Ca2+ or blockade of N-and P/Q-type Ca2+ channels and protein kinase A (PKA) abolished the inhibitory effect of GP-17 on glutamate release from cortical synaptosomes. GP-17 also significantly reduced the phosphorylation of PKA, SNAP-25, and synapsin I in cortical synaptosomes. In an in vivo rat model of glutamate excitotoxicity induced by kainic acid (KA), GP-17 pretreatment significantly prevented seizures and rescued neuronal cell injury and glutamate elevation in the cortex. GP-17 pretreatment decreased the expression levels of sodium-coupled neutral amino acid transporter 1, glutamate synthesis enzyme glutaminase and vesicular glutamate transporter 1 but increased the expression level of glutamate metabolism enzyme glutamate dehydrogenase in the cortex of KA-treated rats. In addition, the KA-induced alterations in the N-methyl-D-aspartate receptor subunits GluN2A and GluN2B in the cortex were prevented by GP-17 pretreatment. GP-17 also prevented the KA-induced decrease in cerebral blood flow and arginase II expression. These results suggest that (i) GP-17, through the suppression of N- and P/Q-type Ca2+ channels and consequent PKA-mediated SNAP-25 and synapsin I phosphorylation, reduces glutamate exocytosis from cortical synaptosomes; and (ii) GP-17 has a neuroprotective effect on KA-induced glutamate excitotoxicity in rats through regulating synaptic glutamate release and cerebral blood flow.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Glutamic Acid , Gynostemma , Animals , Glutamic Acid/metabolism , Rats , Male , Gynostemma/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Rats, Sprague-Dawley , Synaptosomes/metabolism , Synaptosomes/drug effects , Neuroprotective Agents/pharmacology , Kainic Acid/toxicity , Seizures/chemically induced , Seizures/metabolism , Seizures/drug therapy , Seizures/prevention & control , Synapses/drug effects , Synapses/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synapsins/metabolism , Phosphorylation/drug effects , Calcium/metabolism , Plant Extracts
10.
Cells ; 13(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38786059

ABSTRACT

In recent decades, there has been a dramatic rise in the rates of children being born after in utero exposure to drugs of abuse, particularly opioids. Opioids have been shown to have detrimental effects on neurons and glia in the central nervous system (CNS), but the impact of prenatal opioid exposure (POE) on still-developing synaptic circuitry is largely unknown. Astrocytes exert a powerful influence on synaptic development, secreting factors to either promote or inhibit synapse formation and neuronal maturation in the developing CNS. Here, we investigated the effects of the partial µ-opioid receptor agonist buprenorphine on astrocyte synaptogenic signaling and morphological development in cortical cell culture. Acute buprenorphine treatment had no effect on the excitatory synapse number in astrocyte-free neuron cultures. In conditions where neurons shared culture media with astrocytes, buprenorphine attenuated the synaptogenic capabilities of astrocyte-secreted factors. Neurons cultured from drug-naïve mice showed no change in synapses when treated with factors secreted by astrocytes from POE mice. However, this same treatment was synaptogenic when applied to neurons from POE mice, indicating a complex neuroadaptive response in the event of impaired astrocyte signaling. In addition to promoting morphological and connectivity changes in neurons, POE exerted a strong influence on astrocyte development, disrupting their structural maturation and promoting the accumulation of lipid droplets (LDs), suggestive of a maladaptive stress response in the developing CNS.


Subject(s)
Analgesics, Opioid , Astrocytes , Neurons , Prenatal Exposure Delayed Effects , Signal Transduction , Synapses , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Animals , Synapses/metabolism , Synapses/drug effects , Female , Pregnancy , Mice , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/metabolism , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Signal Transduction/drug effects , Buprenorphine/pharmacology , Cells, Cultured , Mice, Inbred C57BL
11.
Hear Res ; 447: 109022, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705005

ABSTRACT

The disruption of ribbon synapses in the cochlea impairs the transmission of auditory signals from the cochlear sensory receptor cells to the auditory cortex. Although cisplatin-induced loss of ribbon synapses is well-documented, and studies have reported nitration of cochlear proteins after cisplatin treatment, yet the underlying mechanism of cochlear synaptopathy is not fully understood. This study tests the hypothesis that cisplatin treatment alters the abundance of cochlear synaptosomal proteins, and selective targeting of nitrative stress prevents the associated synaptic dysfunction. Auditory brainstem responses of mice treated with cisplatin showed a reduction in amplitude and an increase in latency of wave I, indicating cisplatin-induced synaptic dysfunction. The mass spectrometry analysis of cochlear synaptosomal proteins identified 102 proteins that decreased in abundance and 249 that increased in abundance after cisplatin treatment. Pathway analysis suggested that the dysregulated proteins were involved in calcium binding, calcium ion regulation, synapses, and endocytosis pathways. Inhibition of nitrative stress by co-treatment with MnTBAP, a peroxynitrite scavenger, attenuated cisplatin-induced changes in the abundance of 27 proteins. Furthermore, MnTBAP co-treatment prevented the cisplatin-induced decrease in the amplitude and increase in the latency of wave I. Together, these findings suggest a potential role of oxidative/nitrative stress in cisplatin-induced cochlear synaptic dysfunction.


Subject(s)
Cisplatin , Cochlea , Evoked Potentials, Auditory, Brain Stem , Proteomics , Synapses , Synaptosomes , Cisplatin/toxicity , Cisplatin/pharmacology , Animals , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/drug effects , Oxidative Stress/drug effects , Mice, Inbred CBA , Male , Ototoxicity/metabolism , Ototoxicity/physiopathology , Mice
12.
ACS Nano ; 18(20): 13277-13285, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38728175

ABSTRACT

Synapses in the brain utilize two distinct communication mechanisms: chemical and electrical. For a comprehensive investigation of neural circuitry, neural interfaces should be capable of both monitoring and stimulating these types of physiological interactions. However, previously developed interfaces for neurotransmitter monitoring have been limited in interaction modality due to constraints in device size, fabrication techniques, and the usage of flexible materials. To address this obstacle, we propose a multifunctional and flexible fiber probe fabricated through the microwire codrawing thermal drawing process, which enables the high-density integration of functional components with various materials such as polymers, metals, and carbon fibers. The fiber enables real-time monitoring of transient dopamine release in vivo, real-time stimulation of cell-specific neuronal populations via optogenetic stimulation, single-unit electrophysiology of individual neurons localized to the tip of the neural probe, and chemical stimulation via drug delivery. This fiber will improve the accessibility and functionality of bidirectional interrogation of neurochemical mechanisms in implantable neural probes.


Subject(s)
Brain , Neurons , Synapses , Animals , Brain/metabolism , Synapses/metabolism , Synapses/chemistry , Neurons/metabolism , Optogenetics , Dopamine/metabolism , Mice , Temperature
14.
Sci Adv ; 10(18): eadm7039, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701209

ABSTRACT

Long-range glutamatergic inputs originating from the cortex and thalamus are indispensable for striatal development, providing the foundation for motor and cognitive functions. Despite their significance, transcriptional regulation governing these inputs remains largely unknown. We investigated the role of a transcription factor encoded by a high-risk autism-associated gene, FOXP1, in sculpting glutamatergic inputs onto spiny projection neurons (SPNs) within the striatum. We find a neuron subtype-specific role of FOXP1 in strengthening and maturing glutamatergic inputs onto dopamine receptor 2-expressing SPNs (D2 SPNs). We also find that FOXP1 promotes synaptically driven excitability in these neurons. Using single-nuclei RNA sequencing, we identify candidate genes that mediate these cell-autonomous processes through postnatal FOXP1 function at the post-synapse. Last, we demonstrate that postnatal FOXP1 reinstatement rescues electrophysiological deficits, cell type-specific gene expression changes, and behavioral phenotypes. Together, this study enhances our understanding of striatal circuit development and provides proof of concept for a therapeutic approach for FOXP1 syndrome and other neurodevelopmental disorders.


Subject(s)
Corpus Striatum , Forkhead Transcription Factors , Neurons , Receptors, Dopamine D2 , Repressor Proteins , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Corpus Striatum/metabolism , Corpus Striatum/cytology , Mice , Neurons/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Phenotype , Synapses/metabolism , Synapses/physiology , Male
15.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731870

ABSTRACT

Transcranial magneto-acoustic stimulation (TMAS), which is characterized by high spatiotemporal resolution and high penetrability, is a non-invasive neuromodulation technology based on the magnetic-acoustic coupling effect. To reveal the effects of TMAS treatment on amyloid-beta (Aß) plaque and synaptic plasticity in Alzheimer's disease, we conducted a comparative analysis of TMAS and transcranial ultrasound stimulation (TUS) based on acoustic effects in 5xFAD mice and BV2 microglia cells. We found that the TMAS-TUS treatment effectively reduced amyloid plaque loads and plaque-associated neurotoxicity. Additionally, TMAS-TUS treatment ameliorated impairments in long-term memory formation and long-term potentiation. Moreover, TMAS-TUS treatment stimulated microglial proliferation and migration while enhancing the phagocytosis and clearance of Aß. In 5xFAD mice with induced microglial exhaustion, TMAS-TUS treatment-mediated Aß plaque reduction, synaptic rehabilitation improvement, and the increase in phospho-AKT levels were diminished. Overall, our study highlights that stimulation of hippocampal microglia by TMAS treatment can induce anti-cognitive impairment effects via PI3K-AKT signaling, providing hope for the development of new strategies for an adjuvant therapy for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Microglia , Plaque, Amyloid , Animals , Microglia/metabolism , Mice , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Transcranial Magnetic Stimulation/methods , Acoustic Stimulation , Mice, Transgenic , Disease Models, Animal , Synapses/metabolism , Hippocampus/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Neuronal Plasticity , Long-Term Potentiation , Signal Transduction
16.
PLoS Biol ; 22(5): e3002596, 2024 May.
Article in English | MEDLINE | ID: mdl-38718086

ABSTRACT

Autism spectrum disorders (ASD) frequently accompany macrocephaly, which often involves hydrocephalic enlargement of brain ventricles. Katnal2 is a microtubule-regulatory protein strongly linked to ASD, but it remains unclear whether Katnal2 knockout (KO) in mice leads to microtubule- and ASD-related molecular, synaptic, brain, and behavioral phenotypes. We found that Katnal2-KO mice display ASD-like social communication deficits and age-dependent progressive ventricular enlargements. The latter involves increased length and beating frequency of motile cilia on ependymal cells lining ventricles. Katnal2-KO hippocampal neurons surrounded by enlarged lateral ventricles show progressive synaptic deficits that correlate with ASD-like transcriptomic changes involving synaptic gene down-regulation. Importantly, early postnatal Katnal2 re-expression prevents ciliary, ventricular, and behavioral phenotypes in Katnal2-KO adults, suggesting a causal relationship and a potential treatment. Therefore, Katnal2 negatively regulates ependymal ciliary function and its deletion in mice leads to ependymal ciliary hyperfunction and hydrocephalus accompanying ASD-related behavioral, synaptic, and transcriptomic changes.


Subject(s)
Autism Spectrum Disorder , Cilia , Ependyma , Mice, Knockout , Phenotype , Animals , Cilia/metabolism , Ependyma/metabolism , Mice , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Hydrocephalus/physiopathology , Hippocampus/metabolism , Male , Neurons/metabolism , Mice, Inbred C57BL , Synapses/metabolism , Behavior, Animal , Katanin/metabolism , Katanin/genetics , Transcriptome/genetics , Disease Models, Animal
17.
Exp Brain Res ; 242(6): 1507-1515, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719948

ABSTRACT

Alzheimer's disease is a progressive neurodegenerative disorder characterized by impairments in synaptic plasticity and cognitive performance. Current treatments are unable to achieve satisfactory therapeutic effects or reverse the progression of the disease. Calcineurin has been implicated as part of a critical signaling pathway for learning and memory, and neuronal calcineurin may be hyperactivated in AD. To investigate the effects and underlying mechanisms of FK506, a calcineurin inhibitor, on Alzheimer-like behavior and synaptic dysfunction in the 3 × Tg-AD transgenic mouse model of Alzheimer's disease, we investigated the effect of FK506 on cognitive function and synaptic plasticity in the 3 × Tg-AD transgenic mouse model of Alzheimer's disease. The results showed that FK506 treatment ameliorated cognitive deficits, as indicated by the decreased latency in the water maze, and attenuated tau hyperphosphorylation in 3 × Tg-AD mice. Treatment with FK506 also reduced the levels of certain markers of postsynaptic deficits, including PSD-95 and NR2B, and reversed the long-term potentiation deficiency and dendritic spine impairments in 3 × Tg-AD mice. These findings suggest that treatment with calcineurin inhibitors such as FK506 can be an effective therapeutic strategy to rescue synaptic deficit and cognitive impairment in familial Alzheimer's disease and related tauopathies.


Subject(s)
Alzheimer Disease , Calcineurin Inhibitors , Disease Models, Animal , Mice, Transgenic , Tacrolimus , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Tacrolimus/pharmacology , Calcineurin Inhibitors/pharmacology , Mice , Maze Learning/drug effects , Maze Learning/physiology , Calcineurin/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , tau Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Male , Synapses/drug effects , Synapses/metabolism , Disks Large Homolog 4 Protein/metabolism
18.
CNS Neurosci Ther ; 30(5): e14744, 2024 05.
Article in English | MEDLINE | ID: mdl-38727249

ABSTRACT

BACKGROUND: Stroke is an acute cerebrovascular disease in which brain tissue is damaged due to sudden obstruction of blood flow to the brain or the rupture of blood vessels in the brain, which can prompt ischemic or hemorrhagic stroke. After stroke onset, ischemia, hypoxia, infiltration of blood components into the brain parenchyma, and lysed cell fragments, among other factors, invariably increase blood-brain barrier (BBB) permeability, the inflammatory response, and brain edema. These changes lead to neuronal cell death and synaptic dysfunction, the latter of which poses a significant challenge to stroke treatment. RESULTS: Synaptic dysfunction occurs in various ways after stroke and includes the following: damage to neuronal structures, accumulation of pathologic proteins in the cell body, decreased fluidity and release of synaptic vesicles, disruption of mitochondrial transport in synapses, activation of synaptic phagocytosis by microglia/macrophages and astrocytes, and a reduction in synapse formation. CONCLUSIONS: This review summarizes the cellular and molecular mechanisms related to synapses and the protective effects of drugs or compounds and rehabilitation therapy on synapses in stroke according to recent research. Such an exploration will help to elucidate the relationship between stroke and synaptic damage and provide new insights into protecting synapses and restoring neurologic function.


Subject(s)
Stroke , Synapses , Humans , Animals , Synapses/pathology , Synapses/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/complications , Stroke/physiopathology
19.
Cell ; 187(10): 2574-2594.e23, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38729112

ABSTRACT

High-resolution electron microscopy of nervous systems has enabled the reconstruction of synaptic connectomes. However, we do not know the synaptic sign for each connection (i.e., whether a connection is excitatory or inhibitory), which is implied by the released transmitter. We demonstrate that artificial neural networks can predict transmitter types for presynapses from electron micrographs: a network trained to predict six transmitters (acetylcholine, glutamate, GABA, serotonin, dopamine, octopamine) achieves an accuracy of 87% for individual synapses, 94% for neurons, and 91% for known cell types across a D. melanogaster whole brain. We visualize the ultrastructural features used for prediction, discovering subtle but significant differences between transmitter phenotypes. We also analyze transmitter distributions across the brain and find that neurons that develop together largely express only one fast-acting transmitter (acetylcholine, glutamate, or GABA). We hope that our publicly available predictions act as an accelerant for neuroscientific hypothesis generation for the fly.


Subject(s)
Brain , Drosophila melanogaster , Microscopy, Electron , Neurons , Neurotransmitter Agents , Synapses , Animals , Drosophila melanogaster/ultrastructure , Drosophila melanogaster/metabolism , Neurotransmitter Agents/metabolism , Synapses/ultrastructure , Synapses/metabolism , Microscopy, Electron/methods , Brain/ultrastructure , Brain/metabolism , Neurons/metabolism , Neurons/ultrastructure , Neural Networks, Computer , Connectome , gamma-Aminobutyric Acid/metabolism
20.
Nature ; 629(8012): 639-645, 2024 May.
Article in English | MEDLINE | ID: mdl-38693264

ABSTRACT

Sleep is a nearly universal behaviour with unclear functions1. The synaptic homeostasis hypothesis proposes that sleep is required to renormalize the increases in synaptic number and strength that occur during wakefulness2. Some studies examining either large neuronal populations3 or small patches of dendrites4 have found evidence consistent with the synaptic homeostasis hypothesis, but whether sleep merely functions as a permissive state or actively promotes synaptic downregulation at the scale of whole neurons is unclear. Here, by repeatedly imaging all excitatory synapses on single neurons across sleep-wake states of zebrafish larvae, we show that synapses are gained during periods of wake (either spontaneous or forced) and lost during sleep in a neuron-subtype-dependent manner. However, synapse loss is greatest during sleep associated with high sleep pressure after prolonged wakefulness, and lowest in the latter half of an undisrupted night. Conversely, sleep induced pharmacologically during periods of low sleep pressure is insufficient to trigger synapse loss unless adenosine levels are boosted while noradrenergic tone is inhibited. We conclude that sleep-dependent synapse loss is regulated by sleep pressure at the level of the single neuron and that not all sleep periods are equally capable of fulfilling the functions of synaptic homeostasis.


Subject(s)
Homeostasis , Larva , Neurons , Sleep , Synapses , Wakefulness , Zebrafish , Animals , Zebrafish/physiology , Synapses/metabolism , Synapses/physiology , Sleep/physiology , Neurons/physiology , Neurons/metabolism , Wakefulness/physiology , Larva/physiology , Adenosine/metabolism , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...