Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293.800
Filter
1.
Elife ; 132024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007235

ABSTRACT

The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor long form (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determined the role of glutamatergic neurotransmission from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces luteinizing hormone (LH) release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LeprCre (LepRb-Cre) mice. We collected blood sequentially before and for 1 hr after intravenous clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of Fos immunoreactive neurons in the PMv. Next, females with deletion of Slc17a6 (Vglut2) in LepRb neurons (LeprΔVGlut2) showed delayed age of puberty, disrupted estrous cycles, increased gonadotropin-releasing hormone (GnRH) concentration in the axon terminals, and disrupted LH secretion, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LeprloxTB) with concomitant deletion of Slc17a6 (Vglut2flox) mice. Rescue of Lepr and deletion of Slc17a6 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LeprloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation, and became pregnant, while LeprloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic neurotransmission from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.


Subject(s)
Glutamic Acid , Receptors, Leptin , Sexual Maturation , Synaptic Transmission , Animals , Female , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Mice , Glutamic Acid/metabolism , Neurons/metabolism , Neurons/physiology , Reproduction , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism
2.
Adv Neurobiol ; 38: 29-43, 2024.
Article in English | MEDLINE | ID: mdl-39008009

ABSTRACT

One of the most fascinating aspects of the brain is its ability to acquire new information from experience and retain it over time as memory. The search for physical correlates of memory, the memory engram, has been a longstanding priority in modern neurobiology. Advanced genetic approaches have led to the localization of engram cells in a few brain regions, including the hippocampus and cortex. Additionally, engram cells exhibit learning-induced, persistent modifications and have at least two states, active and silent. However, it has been hypothesized that engrams for a specific memory are distributed among multiple brain regions that are functionally connected, referred to as a unified engram complex. Recent tissue-clearing techniques have permitted high-throughput analyses of intact brain samples, which have been used to obtain a map of the engram complex for a contextual fear memory. Careful examination of these engram complex maps has revealed a potentially underappreciated contribution of subcortical regions, specifically thalamic nuclei, to memory function. These more holistic studies support the unified engram complex hypothesis for memory storage and have important implications for understanding dysfunctional engrams in the context of human disease.


Subject(s)
Brain , Hippocampus , Memory , Humans , Memory/physiology , Animals , Brain/physiology , Neurons/metabolism , Fear/physiology
3.
Transl Psychiatry ; 14(1): 289, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39009558

ABSTRACT

Prenatal exposure to infections is a risk factor for neurodevelopmental disorders in offspring, and alterations in mitochondrial function are discussed as a potential underlying factor. Here, using a mouse model of viral-like maternal immune activation (MIA) based on poly(I:C) (POL) treatment at gestational day (GD) 12, we show that adult offspring exhibit behavioral deficits, such as reduced levels of social interaction. In addition, we found increased nicotinamidadenindinucleotid (NADH)- and succinate-linked mitochondrial respiration and maximal electron transfer capacity in the prefrontal cortex (PFC) and in the amygdala (AMY) of males and females. The increase in respiratory capacity resulted from an increase in mitochondrial mass in neurons (as measured by complex IV activity and transcript expression), presumably to compensate for a reduction in mitochondrion-specific respiration. Moreover, in the PFC of control (CON) male offspring a higher excess capacity compared to females was observed, which was significantly reduced in the POL-exposed male offspring, and, along with a higher leak respiration, resulted in a lower mitochondrial coupling efficiency. Transcript expression of the uncoupling proteins (UCP4 and UCP5) showed a reduction in the PFC of POL male mice, suggesting mitochondrial dysfunction. In addition, in the PFC of CON females, a higher expression of the antioxidant enzyme superoxide dismutase (SOD1) was observed, suggesting a higher antioxidant capacity as compared to males. Finally, transcripts analysis of genes involved in mitochondrial biogenesis and dynamics showed reduced expression of fission/fusion transcripts in PFC of POL offspring of both sexes. In conclusion, we show that MIA causes alterations in neuronal mitochondrial function and mass in the PFC and AMY of adult offspring with some effects differing between males and females.


Subject(s)
Mitochondria , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Animals , Female , Prenatal Exposure Delayed Effects/immunology , Pregnancy , Mitochondria/metabolism , Mice , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/immunology , Poly I-C/pharmacology , Disease Models, Animal , Brain/immunology , Brain/metabolism , Amygdala/metabolism , Amygdala/immunology , Behavior, Animal , Mice, Inbred C57BL , Neurons/metabolism , Neurons/immunology
4.
Elife ; 122024 Jul 16.
Article in English | MEDLINE | ID: mdl-39010741

ABSTRACT

Multicellular organisms are composed of specialized cell types with distinct proteomes. While recent advances in single-cell transcriptome analyses have revealed differential expression of mRNAs, cellular diversity in translational profiles remains underinvestigated. By performing RNA-seq and Ribo-seq in genetically defined cells in the Drosophila brain, we here revealed substantial post-transcriptional regulations that augment the cell-type distinctions at the level of protein expression. Specifically, we found that translational efficiency of proteins fundamental to neuronal functions, such as ion channels and neurotransmitter receptors, was maintained low in glia, leading to their preferential translation in neurons. Notably, distribution of ribosome footprints on these mRNAs exhibited a remarkable bias toward the 5' leaders in glia. Using transgenic reporter strains, we provide evidence that the small upstream open-reading frames in the 5' leader confer selective translational suppression in glia. Overall, these findings underscore the profound impact of translational regulation in shaping the proteomics for cell-type distinction and provide new insights into the molecular mechanisms driving cell-type diversity.


Subject(s)
Neuroglia , Protein Biosynthesis , Animals , Neuroglia/metabolism , Neurons/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Gene Expression Regulation , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Brain/metabolism , Brain/cytology , Ribosomes/metabolism , Drosophila/genetics
5.
Braz J Med Biol Res ; 57: e13736, 2024.
Article in English | MEDLINE | ID: mdl-38985082

ABSTRACT

The present study utilized the spared nerve injury (SNI) to create a mouse model of depression to investigate the impact of esketamine on depressive-like behaviors, on the expression of PSD-95 and CRMP2 proteins, and on changes in neuronal dendritic spine plasticity in the prefrontal cortex (PFC). Depressive-like behavioral tests were performed 1 h after esketamine treatment, and the PFC tissues were obtained on the fourth day after completing the behavioral tests. Then, dendritic spine density and morphology in the PFC were measured using Golgi staining, and CRMP2 and PSD-95 proteins were obtained from PFC tissue by western blotting. The results of this study showed that esketamine significantly increased the immobility time in the forced swimming test and tail suspension test. In the open field test, esketamine increased the time spent in the open arms, the time spent in the central area, and the total distance covered. It also increased the protein expression levels of CRMP2 and PSD-95 in addition to the total and mature dendritic spine density of the PFC in SNI-depressed mice. Esketamine can significantly improve depression-like behaviors in SNI-depressed mice and promote an increase in dendritic spine density and maturation in the PFC. These effects may be associated with changes in CRMP2 and PSD-95 expression.


Subject(s)
Dendritic Spines , Depression , Disease Models, Animal , Ketamine , Neuronal Plasticity , Prefrontal Cortex , Animals , Prefrontal Cortex/drug effects , Ketamine/pharmacology , Neuronal Plasticity/drug effects , Male , Dendritic Spines/drug effects , Mice , Depression/drug therapy , Nerve Tissue Proteins/metabolism , Disks Large Homolog 4 Protein/metabolism , Intercellular Signaling Peptides and Proteins , Neurons/drug effects , Behavior, Animal/drug effects , Blotting, Western
6.
Elife ; 122024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985568

ABSTRACT

Accurate tracking of the same neurons across multiple days is crucial for studying changes in neuronal activity during learning and adaptation. Advances in high-density extracellular electrophysiology recording probes, such as Neuropixels, provide a promising avenue to accomplish this goal. Identifying the same neurons in multiple recordings is, however, complicated by non-rigid movement of the tissue relative to the recording sites (drift) and loss of signal from some neurons. Here, we propose a neuron tracking method that can identify the same cells independent of firing statistics, that are used by most existing methods. Our method is based on between-day non-rigid alignment of spike-sorted clusters. We verified the same cell identity in mice using measured visual receptive fields. This method succeeds on datasets separated from 1 to 47 days, with an 84% average recovery rate.


Subject(s)
Neurons , Animals , Neurons/physiology , Mice , Electrophysiology/methods , Electrophysiological Phenomena , Action Potentials/physiology , Cell Tracking/methods
7.
Nat Commun ; 15(1): 5861, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997274

ABSTRACT

Electrical stimulation is a key tool in neuroscience, both in brain mapping studies and in many therapeutic applications such as cochlear, vestibular, and retinal neural implants. Due to safety considerations, stimulation is restricted to short biphasic pulses. Despite decades of research and development, neural implants lead to varying restoration of function in patients. In this study, we use computational modeling to provide an explanation for how pulsatile stimulation affects axonal channels and therefore leads to variability in restoration of neural responses. The phenomenological explanation is transformed into equations that predict induced firing rate as a function of pulse rate, pulse amplitude, and spontaneous firing rate. We show that these equations predict simulated responses to pulsatile stimulation with a variety of parameters as well as several features of experimentally recorded primate vestibular afferent responses to pulsatile stimulation. We then discuss the implications of these effects for improving clinical stimulation paradigms and electrical stimulation-based experiments.


Subject(s)
Electric Stimulation , Animals , Electric Stimulation/methods , Models, Neurological , Macaca mulatta , Action Potentials/physiology , Neurons/physiology , Computer Simulation , Humans , Vestibule, Labyrinth/physiology
8.
Sci Rep ; 14(1): 16242, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39004628

ABSTRACT

Chemotherapy-induced neuropathic pain (CINP), a condition with unmet treatment needs, affects over half of cancer patients treated with chemotherapeutics. Researchers have recently focused on the endocannabinoid system because of its critical role in regulating our bodies' most important functions, including pain. We used in vitro and in vivo methods to determine the toxicity profile of a synthetic cannabinoid, JWH-182, and whether it could be potentially effective for CINP alleviation. In vitro, we evaluated JWH-182 general toxicity, measuring fibroblast viability treated with various concentrations of compound, and its neuroprotection on dorsal root ganglion neurons treated with paclitaxel. In vivo, we performed an evaluation of acute and 28-day repeated dose toxicity in mice, with monitoring of health status and a complete histopathological examination. Finally, we evaluated the efficacy of JWH-182 on a CINP model in mice using specific pain assessment tests. JWH-182 has an acceptable toxicity profile, in both, in vitro and in vivo studies and it was able to significantly reduce pain perception in a CINP model in mice. However, the translation of these results to the clinic needs further investigation.


Subject(s)
Cannabinoids , Neuralgia , Animals , Neuralgia/drug therapy , Neuralgia/chemically induced , Mice , Cannabinoids/pharmacology , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Male , Humans , Paclitaxel/adverse effects , Paclitaxel/pharmacology , Neurons/drug effects , Neurons/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism
9.
Commun Biol ; 7(1): 861, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39004677

ABSTRACT

Oxidative stress is a key contributor to AD pathology. However, the earliest role of pre-plaque neuronal oxidative stress, remains elusive. Using laser microdissected hippocampal neurons extracted from McGill-R-Thy1-APP transgenic rats we found that intraneuronal amyloid beta (iAß)-burdened neurons had increased expression of genes related to oxidative stress and DNA damage responses including Ercc2, Fancc, Sod2, Gsr, and Idh1. DNA damage was further evidenced by increased neuronal levels of XPD (Ercc2) and γH2AX foci, indicative of DNA double stranded breaks (DSBs), and by increased expression of Ercc6, Rad51, and Fen1, and decreased Sirt6 in hippocampal homogenates. We also found increased expression of synaptic plasticity genes (Grin2b (NR2B), CamkIIα, Bdnf, c-fos, and Homer1A) and increased protein levels of TOP2ß. Our findings indicate that early accumulation of iAß, prior to Aß plaques, is accompanied by incipient oxidative stress and DSBs that may arise directly from oxidative stress or from maladaptive synaptic plasticity.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , DNA Damage , Disease Models, Animal , Hippocampus , Neurons , Oxidative Stress , Rats, Transgenic , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Neurons/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Rats , Male , Neuronal Plasticity
10.
CNS Neurosci Ther ; 30(7): e14835, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39004783

ABSTRACT

AIMS: Necroptosis is one of programmed death that may aggravate spinal cord injury (SCI). We aimed to investigate the effect and mechanism of exendin-4 (EX-4) on the recovery of motor function and necroptosis after SCI. METHODS: The SD rats with left hemisection in the T10 spinal cord as SCI model were used. The behavior tests were measured within 4 weeks. The effects of EX-4 on necroptosis-associated proteins and autophagy flux were explored. In addition, the SHSY5Y cell model was introduced to explore the direct effect of EX-4 on neurons. The effect of lysosome was explored using mTOR activator and AO staining. RESULTS: EX-4 could improve motor function and limb strength, promote the recovery of autophagy flux, and accelerate the degradation of necroptosis-related protein at 3 d after injury in rats. EX-4 reduced lysosome membrane permeability, promoted the recovery of lysosome function and autophagy flux, and accelerated the degradation of necroptosis-related proteins by inhibiting the phosphorylation level of mTOR in the SHSY5Y cell model. CONCLUSION: Our results demonstrated that EX-4 may improve motor function after SCI via inhibiting mTOR phosphorylation level and accelerating the degradation of necroptosis-related proteins in neurons. Our findings may provide new therapeutic targets for clinical treatment after SCI.


Subject(s)
Autophagy , Exenatide , Necroptosis , Neurons , Rats, Sprague-Dawley , Spinal Cord Injuries , Animals , Autophagy/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Rats , Neurons/drug effects , Neurons/metabolism , Exenatide/pharmacology , Exenatide/therapeutic use , Necroptosis/drug effects , Humans , Recovery of Function/drug effects , Recovery of Function/physiology , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Neuroprotective Agents/pharmacology , Male
11.
J Clin Invest ; 134(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007270

ABSTRACT

Copy number variation (CNV) at 7q11.23 causes Williams-Beuren syndrome (WBS) and 7q microduplication syndrome (7Dup), neurodevelopmental disorders (NDDs) featuring intellectual disability accompanied by symmetrically opposite neurocognitive features. Although significant progress has been made in understanding the molecular mechanisms underlying 7q11.23-related pathophysiology, the propagation of CNV dosage across gene expression layers and their interplay remains elusive. Here we uncovered 7q11.23 dosage-dependent symmetrically opposite dynamics in neuronal differentiation and intrinsic excitability. By integrating transcriptomics, translatomics, and proteomics of patient-derived and isogenic induced neurons, we found that genes related to neuronal transmission follow 7q11.23 dosage and are transcriptionally controlled, while translational factors and ribosomal genes are posttranscriptionally buffered. Consistently, we found phosphorylated RPS6 (p-RPS6) downregulated in WBS and upregulated in 7Dup. Surprisingly, p-4EBP was changed in the opposite direction, reflecting dosage-specific changes in total 4EBP levels. This highlights different dosage-sensitive dyregulations of the mTOR pathway as well as distinct roles of p-RPS6 and p-4EBP during neurogenesis. Our work demonstrates the importance of multiscale disease modeling across molecular and functional layers, uncovers the pathophysiological relevance of ribosomal biogenesis in a paradigmatic pair of NDDs, and uncouples the roles of p-RPS6 and p-4EBP as mechanistically actionable relays in NDDs.


Subject(s)
Chromosomes, Human, Pair 7 , DNA Copy Number Variations , Neurons , Humans , Neurons/metabolism , Neurons/pathology , Chromosomes, Human, Pair 7/genetics , Ribosomes/metabolism , Ribosomes/genetics , Neurogenesis/genetics , Williams Syndrome/genetics , Williams Syndrome/metabolism , Williams Syndrome/pathology , Williams Syndrome/physiopathology , Ribosomal Protein S6/metabolism , Ribosomal Protein S6/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Male , Cell Differentiation , Female
12.
Adv Neurobiol ; 38: 237-257, 2024.
Article in English | MEDLINE | ID: mdl-39008019

ABSTRACT

Memory engrams in mice brains are potentially related to groups of concept cells in human brains. A single concept cell in human hippocampus responds, for example, not only to different images of the same object or person but also to its name written down in characters. Importantly, a single mental concept (object or person) is represented by several concept cells and each concept cell can respond to more than one concept. Computational work shows how mental concepts can be embedded in recurrent artificial neural networks as memory engrams and how neurons that are shared between different engrams can lead to associations between concepts. Therefore, observations at the level of neurons can be linked to cognitive notions of memory recall and association chains between memory items.


Subject(s)
Hippocampus , Memory , Neural Networks, Computer , Animals , Humans , Mice , Brain/physiology , Hippocampus/physiology , Memory/physiology , Mental Recall/physiology , Models, Neurological , Neurons/physiology
13.
Proc Natl Acad Sci U S A ; 121(30): e2319958121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39008673

ABSTRACT

Neuropeptides (NPs) and their cognate receptors are critical effectors of diverse physiological processes and behaviors. We recently reported of a noncanonical function of the Drosophila Glucose-6-Phosphatase (G6P) gene in a subset of neurosecretory cells in the central nervous system that governs systemic glucose homeostasis in food-deprived flies. Here, we show that G6P-expressing neurons define six groups of NP-secreting cells, four in the brain and two in the thoracic ganglion. Using the glucose homeostasis phenotype as a screening tool, we find that neurons located in the thoracic ganglion expressing FMRFamide NPs (FMRFaG6P neurons) are necessary and sufficient to maintain systemic glucose homeostasis in starved flies. We further show that G6P is essential in FMRFaG6P neurons for attaining a prominent Golgi apparatus and secreting NPs efficiently. Finally, we establish that G6P-dependent FMRFa signaling is essential for the build-up of glycogen stores in the jump muscle which expresses the receptor for FMRFamides. We propose a general model in which the main role of G6P is to counteract glycolysis in peptidergic neurons for the purpose of optimizing the intracellular environment best suited for the expansion of the Golgi apparatus, boosting release of NPs and enhancing signaling to respective target tissues expressing cognate receptors.


Subject(s)
Drosophila melanogaster , FMRFamide , Glucose-6-Phosphatase , Glycogen , Neurons , Neuropeptides , Signal Transduction , Animals , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , FMRFamide/metabolism , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Glucose-6-Phosphatase/genetics , Glycogen/metabolism , Golgi Apparatus/metabolism , Homeostasis , Muscles/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Neuropeptides/genetics
14.
Proc Natl Acad Sci U S A ; 121(30): e2404164121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39012823

ABSTRACT

The development of advanced neural modulation techniques is crucial to neuroscience research and neuroengineering applications. Recently, optical-based, nongenetic modulation approaches have been actively investigated to remotely interrogate the nervous system with high precision. Here, we show that a thin-film, silicon (Si)-based diode device is capable to bidirectionally regulate in vitro and in vivo neural activities upon adjusted illumination. When exposed to high-power and short-pulsed light, the Si diode generates photothermal effects, evoking neuron depolarization and enhancing intracellular calcium dynamics. Conversely, low-power and long-pulsed light on the Si diode hyperpolarizes neurons and reduces calcium activities. Furthermore, the Si diode film mounted on the brain of living mice can activate or suppress cortical activities under varied irradiation conditions. The presented material and device strategies reveal an innovated optoelectronic interface for precise neural modulations.


Subject(s)
Neurons , Optogenetics , Silicon , Animals , Silicon/chemistry , Neurons/physiology , Mice , Optogenetics/methods , Calcium/metabolism , Light , Brain/physiology
16.
J Mol Neurosci ; 74(3): 69, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39017898

ABSTRACT

The objective of this investigation was to examine the impact of multiple exposures to general anesthesia (GA) with sevoflurane on the offspring of pregnant mice, as well as to elucidate the underlying mechanism. Neurodevelopmental assessments, including various reflexes and behavioral tests, were conducted on the offspring in the GA group to evaluate neuronal cell development. Furthermore, neonatal mouse neuronal cells were isolated and transfected with a high-expression CREB vector (pcDNA3.1-CREB), followed by treatment with sevoflurane (0.72 mol/L), ZD7288 (50 µmol/L), and KN-62 (10 µmol/L), or a combination of these compounds. The expression of relevant genes was then analyzed using qRT-PCR and western blot techniques. In comparison to the sham group, neonatal mice in the GA group exhibited significantly prolonged latencies in surface righting reflex, geotaxis test, and air righting reflex. Furthermore, there was a notable deceleration in the development of body weight and tail in the GA group. These mice also displayed impairments in social ability, reduced reciprocal social interaction behaviors, diminished learning capacity, and heightened levels of anxious behaviors. Additionally, synaptic trigger malfunction was observed, along with decreased production of c-Fos and neurotrophic factors. Sevoflurane was found to notably decrease cellular c-Fos and neurotrophic factor production, as well as the expression of HCN2 and CaMKII/CREB-related proteins. The inhibitory effects of sevoflurane on HCN2 or CaMKII channels were similar to those observed with ZD7288 or KN-62 inhibition. However, overexpression of CREB mitigated the impact of sevoflurane on neuronal cells. Repetitive exposure to sevoflurane general anesthesia while pregnant suppresses the CaMKII/CREB pathway, leading to the development of autism-like characteristics in offspring mice through the reduction of HCN2 expression.


Subject(s)
Anesthetics, Inhalation , Autistic Disorder , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Down-Regulation , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Prenatal Exposure Delayed Effects , Sevoflurane , Animals , Sevoflurane/pharmacology , Sevoflurane/toxicity , Mice , Pregnancy , Female , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/adverse effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Autistic Disorder/genetics , Autistic Disorder/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Potassium Channels/metabolism , Potassium Channels/genetics , Cells, Cultured , Neurons/metabolism , Neurons/drug effects , Male , Mice, Inbred C57BL
17.
Biomed Phys Eng Express ; 10(5)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38959871

ABSTRACT

Biocompatible polymer-based scaffolds hold great promise for neural repair, especially when they are coupled with electrostimulation to induce neural differentiation. In this study, a combination of polyacrylonitrile/polyaniline (PAN/PANI) and Carbon Nanotubes (CNTs) were used to fabricate three different biomimetic electrospun scaffolds (samples 1, 2 and 3 containing 0.26 wt%, 1 wt% and 2 wt% of CNTs, respectively). These scaffolds underwent thorough characterization for assessing electroconductivity, tensile strength, wettability, degradability, swelling, XRD, and FTIR data. Notably, scanning electron microscopy (SEM) images revealed a three-dimensional scaffold morphology with aligned fibers ranging from 60 nm to 292 nm in diameter. To comprehensively investigate the impact of electrical stimulation on the nervous differentiation of the stem cells seeded on these scaffolds, cell morphology and adhesion were assessed based on SEM images. Additionally, scaffold biocompatibility was studied through MTT assay. Importantly, Real-Time PCR results indicated the expression of neural markers-Nestin,ß-tubulin III, and MAP2-by the cells cultured on these samples. In comparison with the control group, samples 1 and 2 exhibited significant increases in Nestin marker expression, indicating early stages of neuronal differentiation, whileß-tubulin III expression was significantly reduced and MAP2 expression remained statistically unchanged. In contrast, sample 3 did not display a statistically significant upturn in Nestin maker expression, while showcasing remarkable increases in the expression of both MAP2 andß-tubulin III, as markers of the end stages of differentiation, leading to postmitotic neurons. These results could be attributed to the higher electroconductivity of S3 compared to other samples. Our findings highlight the biomimetic potential of the prepared scaffolds for neural repair, illustrating their effectiveness in guiding stem cell differentiation toward a neural lineage.


Subject(s)
Acrylic Resins , Aniline Compounds , Cell Differentiation , Nanotubes, Carbon , Nerve Regeneration , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Nanotubes, Carbon/chemistry , Aniline Compounds/chemistry , Acrylic Resins/chemistry , Tissue Engineering/methods , Biocompatible Materials/chemistry , Electric Stimulation , Humans , Cell Adhesion , Microscopy, Electron, Scanning , Stem Cells/cytology , Tensile Strength , Neurons/metabolism , Neurons/cytology , Animals , Nestin/metabolism
18.
Neuromolecular Med ; 26(1): 29, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014255

ABSTRACT

Vascular dementia (VaD) is a cognitive disorder characterized by a decline in cognitive function resulting from cerebrovascular disease. The hippocampus is particularly susceptible to ischemic insults, leading to memory deficits in VaD. Astaxanthin (AST) has shown potential therapeutic effects in neurodegenerative diseases. However, the mechanisms underlying its protective effects in VaD and against hippocampal neuronal death remain unclear. In this study, We used the bilateral common carotid artery occlusion (BCCAO) method to establish a chronic cerebral hypoperfusion (CCH) rat model of VaD and administered a gastric infusion of AST at 25 mg/kg per day for 4 weeks to explore its therapeutic effects. Memory impairments were assessed using Y-maze and Morris water maze tests. We also performed biochemical analyses to evaluate levels of hippocampal neuronal death and apoptosis-related proteins, as well as the impact of astaxanthin on the PI3K/Akt/mTOR pathway and oxidative stress. Our results demonstrated that AST significantly rescued memory impairments in VaD rats. Furthermore, astaxanthin treatment protected against hippocampal neuronal death and attenuated apoptosis. We also observed that AST modulated the PI3K/Akt/mTOR pathway, suggesting its involvement in promoting neuronal survival and synaptic plasticity. Additionally, AST exhibited antioxidant properties, mitigating oxidative stress in the hippocampus. These findings provide valuable insights into the potential therapeutic effects of AST in VaD. By elucidating the mechanisms underlying the actions of AST, this study highlights the importance of protecting hippocampal neurons and suggests potential targets for intervention in VaD. There are still some unanswered questions include long-term effects and optimal dosage of the use in human. Further research is warranted to fully understand the therapeutic potential of AST and its application in the clinical treatment of VaD.


Subject(s)
Apoptosis , Dementia, Vascular , Hippocampus , Memory Disorders , Neurons , Neuroprotective Agents , Oxidative Stress , Rats, Sprague-Dawley , Xanthophylls , Animals , Xanthophylls/therapeutic use , Xanthophylls/pharmacology , Hippocampus/drug effects , Dementia, Vascular/drug therapy , Rats , Male , Memory Disorders/drug therapy , Memory Disorders/etiology , Oxidative Stress/drug effects , Neurons/drug effects , Apoptosis/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Maze Learning/drug effects , Disease Models, Animal , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Cell Death/drug effects , Antioxidants/therapeutic use , Antioxidants/pharmacology , Morris Water Maze Test/drug effects
19.
PLoS Comput Biol ; 20(7): e1012220, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38950068

ABSTRACT

Evidence for metastable dynamics and its role in brain function is emerging at a fast pace and is changing our understanding of neural coding by putting an emphasis on hidden states of transient activity. Clustered networks of spiking neurons have enhanced synaptic connections among groups of neurons forming structures called cell assemblies; such networks are capable of producing metastable dynamics that is in agreement with many experimental results. However, it is unclear how a clustered network structure producing metastable dynamics may emerge from a fully local plasticity rule, i.e., a plasticity rule where each synapse has only access to the activity of the neurons it connects (as opposed to the activity of other neurons or other synapses). Here, we propose a local plasticity rule producing ongoing metastable dynamics in a deterministic, recurrent network of spiking neurons. The metastable dynamics co-exists with ongoing plasticity and is the consequence of a self-tuning mechanism that keeps the synaptic weights close to the instability line where memories are spontaneously reactivated. In turn, the synaptic structure is stable to ongoing dynamics and random perturbations, yet it remains sufficiently plastic to remap sensory representations to encode new sets of stimuli. Both the plasticity rule and the metastable dynamics scale well with network size, with synaptic stability increasing with the number of neurons. Overall, our results show that it is possible to generate metastable dynamics over meaningful hidden states using a simple but biologically plausible plasticity rule which co-exists with ongoing neural dynamics.


Subject(s)
Action Potentials , Models, Neurological , Nerve Net , Neuronal Plasticity , Neurons , Synapses , Neuronal Plasticity/physiology , Nerve Net/physiology , Action Potentials/physiology , Neurons/physiology , Synapses/physiology , Animals , Cerebral Cortex/physiology , Computational Biology , Humans , Computer Simulation
20.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38960706

ABSTRACT

The cerebellum is a conserved structure of the vertebrate brain involved in the timing and calibration of movements. Its function is supported by the convergence of fibers from granule cells (GCs) and inferior olive neurons (IONs) onto Purkinje cells (PCs). Theories of cerebellar function postulate that IONs convey error signals to PCs that, paired with the contextual information provided by GCs, can instruct motor learning. Here, we use the larval zebrafish to investigate (1) how sensory representations of the same stimulus vary across GCs and IONs and (2) how PC activity reflects these two different input streams. We use population calcium imaging to measure ION and GC responses to flashes of diverse luminance and duration. First, we observe that GCs show tonic and graded responses, as opposed to IONs, whose activity peaks mostly at luminance transitions, consistently with the notion that GCs and IONs encode context and error information, respectively. Second, we show that GC activity is patterned over time: some neurons exhibit sustained responses for the entire duration of the stimulus, while in others activity ramps up with slow time constants. This activity could provide a substrate for time representation in the cerebellum. Together, our observations give support to the notion of an error signal coming from IONs and provide the first experimental evidence for a temporal patterning of GC activity over many seconds.


Subject(s)
Cerebellum , Photic Stimulation , Zebrafish , Animals , Zebrafish/physiology , Cerebellum/physiology , Photic Stimulation/methods , Purkinje Cells/physiology , Neurons/physiology , Visual Perception/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...