Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.010
Filter
1.
Trials ; 25(1): 441, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38956594

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a leading cause of disability worldwide across domains of health and cognition, affecting overall quality of life. Approximately one third of individuals with depression do not fully respond to treatments (e.g., conventional antidepressants, psychotherapy) and alternative strategies are needed. Recent early phase trials suggest psilocybin may be a safe and efficacious intervention with rapid-acting antidepressant properties. Psilocybin is thought to exert therapeutic benefits by altering brain network connectivity and inducing neuroplastic changes that endure for weeks post-treatment. Although early clinical results are encouraging, psilocybin's acute neurobiological effects on neuroplasticity have not been fully investigated. We aim to examine for the first time how psilocybin acutely (intraday) and subacutely (weeks) alters functional brain networks implicated in depression. METHODS: Fifty participants diagnosed with MDD or persistent depressive disorder (PDD) will be recruited from a tertiary mood disorders clinic and undergo 1:1 randomization into either an experimental or control arm. Participants will be given either 25 mg psilocybin or 25 mg microcrystalline cellulose (MCC) placebo for the first treatment. Three weeks later, those in the control arm will transition to receiving 25 mg psilocybin. We will investigate whether treatments are associated with changes in arterial spin labelling and blood oxygenation level-dependent contrast neuroimaging assessments at acute and subacute timepoints. Primary outcomes include testing whether psilocybin demonstrates acute changes in (1) cerebral blood flow and (2) functional brain activity in networks associated with mood regulation and depression when compared to placebo, along with changes in MADRS score over time compared to placebo. Secondary outcomes include changes across complementary clinical psychiatric, cognitive, and functional scales from baseline to final follow-up. Serum peripheral neurotrophic and inflammatory biomarkers will be collected at baseline and follow-up to examine relationships with clinical response, and neuroimaging measures. DISCUSSION: This study will investigate the acute and additive subacute neuroplastic effects of psilocybin on brain networks affected by depression using advanced serial neuroimaging methods. Results will improve our understanding of psilocybin's antidepressant mechanisms versus placebo response and whether biological measures of brain function can provide early predictors of treatment response. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT06072898. Registered on 6 October 2023.


Subject(s)
Affect , Brain , Depressive Disorder, Major , Psilocybin , Randomized Controlled Trials as Topic , Humans , Psilocybin/therapeutic use , Psilocybin/adverse effects , Psilocybin/administration & dosage , Psilocybin/pharmacology , Affect/drug effects , Brain/diagnostic imaging , Brain/drug effects , Brain/physiopathology , Depressive Disorder, Major/drug therapy , Magnetic Resonance Imaging , Time Factors , Treatment Outcome , Adult , Neuronal Plasticity/drug effects , Young Adult , Male , Antidepressive Agents/therapeutic use , Female , Middle Aged
2.
Sci Adv ; 10(26): eadm6951, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941461

ABSTRACT

Functional deficits in basal ganglia (BG) circuits contribute to cognitive and motor dysfunctions in alcohol use disorder. Chronic alcohol exposure alters synaptic function and neuronal excitability in the dorsal striatum, but it remains unclear how it affects BG output that is mediated by the substantia nigra pars reticulata (SNr). Here, we describe a neuronal subpopulation-specific synaptic organization of striatal and subthalamic (STN) inputs to the medial and lateral SNr. Chronic alcohol exposure (CIE) potentiated dorsolateral striatum (DLS) inputs but did not change dorsomedial striatum and STN inputs to the SNr. Chemogenetic inhibition of DLS direct pathway neurons revealed an enhanced role for DLS direct pathway neurons in execution of an instrumental lever-pressing task. Overall, we reveal a subregion-specific organization of striatal and subthalamic inputs onto the medial and lateral SNr and find that potentiated DLS-SNr inputs are accompanied by altered BG control of action execution following CIE.


Subject(s)
Basal Ganglia , Corpus Striatum , Ethanol , Neuronal Plasticity , Substantia Nigra , Animals , Neuronal Plasticity/drug effects , Basal Ganglia/physiology , Basal Ganglia/physiopathology , Substantia Nigra/drug effects , Substantia Nigra/physiology , Ethanol/pharmacology , Corpus Striatum/physiology , Male , Mice , Neurons/physiology , Neurons/drug effects , Alcoholism/physiopathology , Neural Pathways
3.
PLoS One ; 19(6): e0304481, 2024.
Article in English | MEDLINE | ID: mdl-38875235

ABSTRACT

Pro-inflammatory changes contribute to multiple neuropsychiatric illnesses. Understanding how these changes are involved in illnesses and identifying strategies to alter inflammatory responses offer paths to potentially novel treatments. We previously found that acute pro-inflammatory stimulation with high (µg/ml) lipopolysaccharide (LPS) for 10-15 min dampens long-term potentiation (LTP) in the hippocampus and impairs learning. Effects of LPS involved non-canonical inflammasome signaling but were independent of toll-like receptor 4 (TLR4), a known LPS receptor. Low (ng/ml) LPS also inhibits LTP when administered for 2-4 h, and here we report that this LPS exposure requires TLR4. We also found that effects of low LPS on LTP involve the oxysterol, 25-hydroxycholesterol, akin to high LPS. Effects of high LPS on LTP are blocked by inhibiting synthesis of 5α-reduced neurosteroids, indicating that neurosteroids mediate LTP inhibition. 5α-Neurosteroids also have anti-inflammatory effects, and we found that exogenous allopregnanolone (AlloP), a key 5α-reduced steroid, prevented effects of low but not high LPS on LTP. We also found that activation of TLR2, TLR3 and TLR7 inhibited LTP and that AlloP prevented the effects of TLR2 and TLR7, but not TLR3. The enantiomer of AlloP, a steroid that has anti-inflammatory actions but low activity at GABAA receptors, prevented LTP inhibition by TLR2, TLR3 and TLR7. In vivo, both AlloP enantiomers prevented LPS-induced learning defects. These studies indicate that neurosteroids play complex roles in network effects of acute neuroinflammation and have potential importance for development of AlloP analogues as therapeutic agents.


Subject(s)
Hippocampus , Lipopolysaccharides , Long-Term Potentiation , Neurosteroids , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Lipopolysaccharides/pharmacology , Long-Term Potentiation/drug effects , Male , Neurosteroids/metabolism , Toll-Like Receptors/metabolism , Learning/drug effects , Mice , Neuronal Plasticity/drug effects , Toll-Like Receptor 4/metabolism , Inflammation/metabolism , Mice, Inbred C57BL , Hydroxycholesterols/pharmacology , Hydroxycholesterols/metabolism , Pregnanolone/pharmacology , Pregnanolone/metabolism
4.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928248

ABSTRACT

Fluoxetine, a commonly prescribed medication for depression, has been studied in Alzheimer's disease (AD) patients for its effectiveness on cognitive symptoms. The aim of this systematic review is to investigate the therapeutic potential of fluoxetine in cognitive decline in AD, focusing on its anti-degenerative mechanisms of action and clinical implications. According to PRISMA, we searched MEDLINE, up to 1 April 2024, for animal and human studies examining the efficacy of fluoxetine with regard to the recovery of cognitive function in AD. Methodological quality was evaluated using the ARRIVE tool for animal AD studies and the Cochrane tool for clinical trials. In total, 22 studies were analyzed (19 animal AD studies and 3 clinical studies). Fluoxetine promoted neurogenesis and enhanced synaptic plasticity in preclinical models of AD, through a decrease in Aß pathology and increase in BDNF, by activating diverse pathways (such as the DAF-16-mediated, TGF-beta1, ILK-AKT-GSK3beta, and CREB/p-CREB/BDNF). In addition, fluoxetine has anti-inflammatory properties/antioxidant effects via targeting antioxidant Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome. Only three clinical studies showed that fluoxetine ameliorated the cognitive performance of people with AD; however, several methodological issues limited the generalizability of these results. Overall, the high-quality preclinical evidence suggests that fluoxetine may have neuroprotective, antioxidant, and anti-inflammatory effects in AD animal models. While more high-quality clinical research is needed to fully understand the mechanisms underlying these effects, fluoxetine is a promising potential treatment for AD patients. If future clinical trials confirm its anti-degenerative and neuroprotective effects, fluoxetine could offer a new therapeutic approach for slowing down the progression of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Fluoxetine , Fluoxetine/therapeutic use , Fluoxetine/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal , Neurogenesis/drug effects , Neuronal Plasticity/drug effects
5.
Int J Biol Macromol ; 273(Pt 2): 132854, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838879

ABSTRACT

Depression is a neuropsychiatric disorder characterized by persistent pleasure loss and behavioral despair. However, the potential mechanisms and therapeutic targets for depression treatment remain unclear. Therefore, identifying the underlying pathogenesis of depression would promote the development of novel treatment and provide effective targets for antidepressant drugs. In this study, proteomics analysis showed that the expression level of phosphatase and actin regulator 4 (Phactr4) was significantly increased in the CA1 hippocampus of depressed rats. The upregulated Phactr4 might induce dysfunction of the synaptic structure via suppressing the p-LIMK/p-Cofilin signaling pathway, and promote neuroinflammation via activating the NF-κB/NLRP3 pathway, which ultimately contributes to the pathogenesis of depression. In contrast, the downregulation of Phactr4 in hippocampal CA1 of depressed rats alleviated depression-like behaviors, along with reducing neuroinflammation and improving synaptic plasticity. In conclusion, these findings provide evidence that Phactr4 plays an important role in regulating neuroinflammatory response and impairment of synaptic plasticity, effects seem to involve in the pathogenesis of depression, and Phactr4 may serve as a potential target for antidepressant treatment.


Subject(s)
Depression , Neuroinflammatory Diseases , Neuronal Plasticity , Stress, Psychological , Animals , Neuronal Plasticity/drug effects , Rats , Depression/drug therapy , Depression/metabolism , Depression/etiology , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Stress, Psychological/complications , Stress, Psychological/metabolism , Microfilament Proteins/metabolism , Rats, Sprague-Dawley , Behavior, Animal/drug effects , Signal Transduction/drug effects , Disease Models, Animal , Antidepressive Agents/pharmacology , Hippocampus/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism
6.
Neurologia (Engl Ed) ; 39(5): 408-416, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38830720

ABSTRACT

Ataxias are characterized by aberrant movement patterns closely related to cerebellar dysfunction. Purkinje cell axons are the sole outputs from the cerebellar cortex, and dysfunctional activity of Purkinje cells has been associated with ataxic movements. However, the synaptic characteristics of Purkinje cells in cases of ataxia are not yet well understood. The nicotinamide antagonist 3-acethylpyridine (3-AP) selectively destroys inferior olivary nucleus neurons so it is widely used to induce cerebellar ataxia. Five days after 3-AP treatment (65mg/kg) in adult male Sprague-Dawley rats, motor incoordination was revealed through BBB and Rotarod testing. In addition, in Purkinje cells from lobules V-VII of the cerebellar vermis studied by the Golgi method, the density of dendritic spines decreased, especially the thin and mushroom types. Western blot analysis showed a decrease in AMPA and PSD-95 content with an increase of the α-catenin protein, while GAD-67 and synaptophysin were unchanged. Findings suggest a limited capacity of Purkinje cells to acquire and consolidate afferent excitatory inputs and an aberrant, rigid profile in the movement-related output patterns of Purkinje neurons that likely contributes to the motor-related impairments characteristic of cerebellar ataxias.


Subject(s)
Cerebellum , Purkinje Cells , Rats, Sprague-Dawley , Animals , Purkinje Cells/drug effects , Purkinje Cells/pathology , Male , Rats , Cerebellum/drug effects , Cerebellar Ataxia/chemically induced , Pyridines/pharmacology , Neuronal Plasticity/drug effects
7.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230221, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853554

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Adiponectin , Dentate Gyrus , Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neuronal Plasticity , Animals , Fragile X Syndrome/physiopathology , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/drug effects , Mice , Neuronal Plasticity/drug effects , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Adiponectin/metabolism , Long-Term Potentiation/drug effects , Male , Receptors, AMPA/metabolism
8.
Phytomedicine ; 131: 155802, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852473

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a serious neurodegenerative disease and brings a serious burden to society and families. Due to lack of effective drugs for the treatment of AD, it's urgent to develop new and effective drug for the treatment of AD. PURPOSE: The study aimed to investigate the potential of Zexieyin formula (ZXYF), a Chinese medicine formula, for the treatment of AD and its potential mechanism of action. METHODS: We used chronic scopolamine (SCOP) induction mice model and APP/PS1 mice to reveal and confirm ZXYF for the treatment of AD with donepezil (DON) as a positive reference. The learning and memory function were detected by morris water maze test (MWM) and y-maze test. Moreover, western blot and immunofluorescence were used to detect the molecular mechanism of ZXYF for the alleviation of AD in hippocampus. Lastly, pharmacological technology was applied to evaluate AMPA receptor involved in the role of ZXYF in the treatment of AD. RESULTS: The results showed that ZXYF could improve memory and learning deficits both in two AD models including scopolamine (SCOP)-induced mice model and APP/PS1mice. Moreover, ZXYF or not DON increased expressions of BrdU/DCX and Ki67 positive cells in dentate gyrus (DG), up-regulated the levels of AMPA subunit type (GluA1) and PKA in hippocampus in SCOP-induced mice model, although ZXYF and DON activated CaMKII, CaMKII-phosphorylation, CREB, CREB-phosphorylation and PSD95 in hippocampus in SCOP-induced mice model. ZXYF also activated CaMKII, CaMKII-phosphorylation and GluA1 in HT22 cells. Furthermore, transient inhibiting AMPA receptor was capable of blocking the effects of ZXYF to treat AD in MWM and suppressing the number of BrdU/DCX positive cells increased by ZXYF in DG in SCOP-induced mice model, but had no effect on the alteration of Ki67 positive cells. CONCLUSION: ZXYF had the therapeutic effects on AD-treatment, which activated CaMKII to promote AMPA receptor (GluA1) and subsequently up-regulated PKA/CREB signaling to facilitate neurogenesis to achieve enhanced postsynaptic protein.


Subject(s)
Alzheimer Disease , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Disease Models, Animal , Drugs, Chinese Herbal , Hippocampus , Neurogenesis , Neuronal Plasticity , Receptors, AMPA , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Receptors, AMPA/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Drugs, Chinese Herbal/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Neurogenesis/drug effects , Mice , Male , Neuronal Plasticity/drug effects , Scopolamine , Mice, Transgenic , Maze Learning/drug effects , Donepezil/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Memory/drug effects , Mice, Inbred C57BL
9.
Nature ; 630(8017): 677-685, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839962

ABSTRACT

All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.


Subject(s)
Dopaminergic Neurons , GABAergic Neurons , Morphine , Myelin Sheath , Neuronal Plasticity , Nucleus Accumbens , Oligodendroglia , Optogenetics , Reward , Ventral Tegmental Area , Ventral Tegmental Area/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Mice , Myelin Sheath/metabolism , Morphine/pharmacology , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Nucleus Accumbens/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Analgesics, Opioid/pharmacology , Dopamine/metabolism , Female , Mice, Inbred C57BL
10.
Dev Psychobiol ; 66(6): e22518, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38924086

ABSTRACT

All terrestrial vertebrate life must transition from aquatic gas exchange in the embryonic environment to aerial or pulmonary respiration at birth. In addition to being able to breathe air, neonates must possess functional sensory feedback systems for maintaining acid-base balance. Respiratory neurons in the brainstem act as pH sensors that can adjust breathing to regulate systemic pH. The central pH sensitivity of breathing-related motor output develops over the embryonic period in the zebra finch (Taeniopygia guttata). Due to the key role of chloride ions in electrochemical stability and developmental plasticity, we tested chloride's role in the development of central pH sensitivity. We blocked gamma-aminobutyric acid-A receptors and cation-chloride cotransport that subtly modulated the low-pH effects on early breathing biorhythms. Further, chloride-free artificial cerebrospinal fluid altered the pattern and timing of breathing biorhythms and blocked the stimulating effect of acidosis in E12-14 brainstems. Early and middle stage embryos exhibited rebound plasticity in brainstem motor outputs during low-pH treatment, which was eliminated by chloride-free solution. Results show that chloride modulates low-pH sensitivity and rebound plasticity in the zebra finch embryonic brainstem, but work is needed to determine the cellular and circuit mechanisms that control functional chloride balance during acid-base disturbances.


Subject(s)
Brain Stem , Chlorides , Finches , Neuronal Plasticity , Respiration , Animals , Hydrogen-Ion Concentration , Finches/physiology , Chlorides/metabolism , Chlorides/pharmacology , Brain Stem/physiology , Brain Stem/drug effects , Respiration/drug effects , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Embryo, Nonmammalian/physiology
11.
Mol Biol Rep ; 51(1): 782, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918289

ABSTRACT

Alcohol consumption is known to have detrimental effects on memory function, with various studies implicating ethanol in the impairment of cognitive processes related to memory retention and retrieval. This review aims to elucidate the complex neurobiological mechanisms underlying ethanol-induced memory impairment. Through a thorough search of existing literature using electronic databases, relevant articles focusing on the neurobiological mechanisms of ethanol on memory were identified and critically evaluated. This review focuses on the molecular and neural pathways through which ethanol exerts its effects on memory formation, consolidation, and recall processes. Key findings from the included studies shed light on the impact of ethanol on neurotransmitter systems, synaptic plasticity, and neuroinflammation in relation to memory impairment. This review contributes to a better understanding of the intricate mechanisms by which alcohol impairs memory function, offering insights for future research directions and the development of targeted interventions to alleviate these cognitive impairments.


Subject(s)
Brain , Ethanol , Memory Disorders , Neuronal Plasticity , Humans , Ethanol/adverse effects , Memory Disorders/metabolism , Memory Disorders/physiopathology , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Animals , Neuronal Plasticity/drug effects , Memory/drug effects , Alcohol Drinking/adverse effects , Neuroinflammatory Diseases , Neurotransmitter Agents/metabolism
12.
Eur J Pharmacol ; 977: 176726, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38852700

ABSTRACT

Short-chain fatty acids, such as butyric acid, derived from the intestinal fermentation of dietary fiber, have been proposed as a treatment for certain pathologies of the central nervous system. Our research group has shown that tributyrin (TB), a butyric acid prodrug, reverses deficits in spatial memory and modulates hippocampal synaptic plasticity. In the present work, diets enriched in either saturated (SOLF; Saturated OiL-enriched Food) or unsaturated (UOLF; Unsaturated OiL-enriched Food) fat were supplied during either 2 h or 8 weeks to 5-week-old male and female mice undergoing a treatment schedule with TB. After the dietary treatment, spatial learning and memory (SLM) was assessed in both the Y-maze and the eight-arm radial maze (RAM). Hippocampal expression of genes involved in glutamatergic transmission as well as synaptic plasticity (long-term potentiation -LTP- and long-term depression -LTD-) were also analyzed. Our results show that 2 h of SOLF intake impaired LTP as well as the performance in the Y-Maze in juvenile male mice whereas no effect was found in females. Moreover, TB reversed both effects in SLM and LTP in males. In the case of chronic intake, both SOLF and UOLF deteriorated SLM measured in the RAM in both sexes whereas TB only reversed LTP impairment induced by SOLF in male mice. These results suggest that TB may have a potentially beneficial influence on learning and memory processes, contingent upon the type of diet and the sex of the individuals.


Subject(s)
Hippocampus , Memory, Short-Term , Neuronal Plasticity , Triglycerides , Animals , Male , Female , Mice , Neuronal Plasticity/drug effects , Triglycerides/metabolism , Memory, Short-Term/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Maze Learning/drug effects , Sex Characteristics , Dietary Fats/adverse effects , Fatty Acids/metabolism , Long-Term Potentiation/drug effects
13.
Sci Adv ; 10(26): eadl2675, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941473

ABSTRACT

Declined memory is a hallmark of Alzheimer's disease (AD). Experiments in rodents and human postmortem studies suggest that serotonin (5-hydroxytryptamine, 5-HT) plays a role in memory, but the underlying mechanisms are unknown. Here, we investigate the role of 5-HT 2C receptor (5-HT2CR) in regulating memory. Transgenic mice expressing a humanized HTR2C mutation exhibit impaired plasticity of hippocampal ventral CA1 (vCA1) neurons and reduced memory. Further, 5-HT neurons project to and synapse onto vCA1 neurons. Disruption of 5-HT synthesis in vCA1-projecting neurons or deletion of 5-HT2CRs in the vCA1 impairs neural plasticity and memory. We show that a selective 5-HT2CR agonist, lorcaserin, improves synaptic plasticity and memory in an AD mouse model. Cumulatively, we demonstrate that hippocampal 5-HT2CR signaling regulates memory, which may inform the use of 5-HT2CR agonists in the treatment of dementia.


Subject(s)
Alzheimer Disease , Memory , Mice, Transgenic , Neuronal Plasticity , Receptor, Serotonin, 5-HT2C , Animals , Humans , Receptor, Serotonin, 5-HT2C/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Memory/drug effects , Memory/physiology , Mice , Neuronal Plasticity/drug effects , Alzheimer Disease/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Serotonin/metabolism , Disease Models, Animal , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Neurons/metabolism , Neurons/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology
14.
Glia ; 72(8): 1484-1500, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780213

ABSTRACT

Microglia are innate immune cells in the brain and show exceptional heterogeneity. They are key players in brain physiological development regulating synaptic plasticity and shaping neuronal networks. In pathological disease states, microglia-induced synaptic pruning mediates synaptic loss and targeting microglia was proposed as a promising therapeutic strategy. However, the effect of microglia depletion and subsequent repopulation on dendritic spine density and neuronal function in the adult brain is largely unknown. In this study, we investigated whether pharmacological microglia depletion affects dendritic spine density after long-term permanent microglia depletion and after short-term microglia depletion with subsequent repopulation. Long-term microglia depletion using colony-stimulating-factor-1 receptor (CSF1-R) inhibitor PLX5622 resulted in increased overall spine density, especially of mushroom spines, and increased excitatory postsynaptic current amplitudes. Short-term PLX5622 treatment with subsequent repopulation of microglia had an opposite effect resulting in activated microglia with increased synaptic phagocytosis and consequently decreased spine density and reduced excitatory neurotransmission, while Barnes maze and elevated plus maze testing was unaffected. Moreover, RNA sequencing data of isolated repopulated microglia showed an activated and proinflammatory phenotype. Long-term microglia depletion might be a promising therapeutic strategy in neurological diseases with pathological microglial activation, synaptic pruning, and synapse loss. However, repopulation after depletion induces activated microglia and results in a decrease of dendritic spines possibly limiting the therapeutic application of microglia depletion. Instead, persistent modulation of pathological microglia activity might be beneficial in controlling synaptic damage.


Subject(s)
Brain , Dendritic Spines , Mice, Inbred C57BL , Microglia , Animals , Microglia/drug effects , Microglia/metabolism , Dendritic Spines/drug effects , Male , Mice , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Phagocytosis/physiology , Phagocytosis/drug effects , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Mice, Transgenic , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Organic Chemicals
15.
Science ; 384(6699): eadd6260, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38815015

ABSTRACT

Abnormal calcium signaling is a central pathological component of Alzheimer's disease (AD). Here, we describe the identification of a class of compounds called ReS19-T, which are able to restore calcium homeostasis in cell-based models of tau pathology. Aberrant tau accumulation leads to uncontrolled activation of store-operated calcium channels (SOCCs) by remodeling septin filaments at the cell cortex. Binding of ReS19-T to septins restores filament assembly in the disease state and restrains calcium entry through SOCCs. In amyloid-ß and tau-driven mouse models of disease, ReS19-T agents restored synaptic plasticity, normalized brain network activity, and attenuated the development of both amyloid-ß and tau pathology. Our findings identify the septin cytoskeleton as a potential therapeutic target for the development of disease-modifying AD treatments.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Calcium , Homeostasis , Neuroprotective Agents , Septins , tau Proteins , Animals , Humans , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cytoskeleton/metabolism , Cytoskeleton/drug effects , Disease Models, Animal , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Septins/metabolism , tau Proteins/metabolism
16.
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
17.
Neuropharmacology ; 254: 109972, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38710443

ABSTRACT

Opioid use disorder (OUD) is a chronic condition associated with long-lasting molecular and behavioral changes. Animals with prolonged access to opioids develop behaviors similar to human OUD. Identifying associated molecular changes can provide insight to underpinnings that lead to or maintain OUD. In pilot studies, we identified several miRNA targets that are altered by the administration of oxycodone. We selected mir182 for follow up as it was recently shown to be dysregulated in plasma of men administered oxycodone. In addition, mir182 is increased in reward-related brain regions of male rats following exposure to various addictive substances. The present study utilizes a long-access oxycodone self-administration paradigm to examine changes in mir182 and its mRNA targets associated with neuroplasticity, which may be involved in the maintenance of OUD-like phenotype in rats. Male rats were trained to self-administer oxycodone (0.1 mg/kg/infusion, i. v.) for 6 h daily sessions for 12 days. Each animal had a yoked saline control that received matched saline infusions. Animals were then tested on a progressive ratio schedule to measure motivation to obtain a single infusion of oxycodone. Drug seeking was measured following 28 days of forced abstinence using a 90-min cued/test. RTqPCR was utilized to measure mir182 and mRNA targets related to neuroplasticity (wnt3, plppr4, pou3f3, tle4, cacna2d, and bdnf) from the nucleus accumbens. Data revealed that animals responded on a continuum for oxycodone. When divided into two groups termed high- and low responders, animals diverged during self-administration acquisition and maintained differences in behavior and gene expression throughout the study. mir182 was upregulated in the nucleus accumbens of both high and low responders and negatively correlated with tle4, which showed a strong negative correlation with reinstatement behavior. mRNA target levels were correlated with behaviors associated with increased severity of OUD behavior in male rats.


Subject(s)
MicroRNAs , Neuronal Plasticity , Oxycodone , Self Administration , Animals , Male , Oxycodone/administration & dosage , Oxycodone/pharmacology , Neuronal Plasticity/drug effects , Rats , MicroRNAs/metabolism , MicroRNAs/genetics , Individuality , Rats, Sprague-Dawley , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Opioid-Related Disorders/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics
18.
Eur J Pharmacol ; 975: 176638, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38734297

ABSTRACT

The underlying mechanisms of macamide's neuroprotective effects in Alzheimer's disease (AD) were investigated in the paper. Macamides are considered as unique ingredients in maca. Improvement effects and mechanisms of macamide on cognitive impairment have not been revealed. In this study, Vina 1.1.2 was used for docking to evaluate the binding abilities of 12 main macamides to acetylcholinesterase (AChE). N-benzyl-(9Z,12Z)-octadecadienamide (M 18:2) was selected to study the following experiments because it can stably bind to AChE with a strong binding energy. The animal experiments showed that M 18:2 prevented the scopolamine (SCP)-induced cognitive impairment and neurotransmitter disorders, increased the positive rates of Nrf2 and HO-1 in hippocampal CA1, improved the synaptic plasticity by maintaining synaptic morphology and increasing the synapse density. Moreover, the contents of IL-1ß, IL-6, and TNF-α in the hippocampus, serum, and colon were reduced by M 18:2. Furthermore, M 18:2 promoted colonic epithelial integrity and partially restored the composition of the gut microbiota to normal, including decreased genera Clostridiales_unclassified and Lachnospiraceae_unclassified, as well as increased genera Muribaculaceae_unclassified, Muribaculum, Alistipes, and Bacteroides, which may be the possible biomarkers of cognitive aging. In summary, M 18:2 exerted neuroprotective effects on SCP-induced AD mice possibly via activating the Nrf2/HO-1 signaling pathway and modulating the gut microbiota.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Gastrointestinal Microbiome , NF-E2-Related Factor 2 , Neuroprotective Agents , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Gastrointestinal Microbiome/drug effects , Signal Transduction/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Mice , Male , Acetylcholinesterase/metabolism , Scopolamine , Neuronal Plasticity/drug effects , Molecular Docking Simulation , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology
19.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791517

ABSTRACT

Maternal immune activation (MIA) is a risk factor for multiple neurodevelopmental disorders; however, animal models developed to explore MIA mechanisms are sensitive to experimental factors, which has led to complexity in previous reports of the MIA phenotype. We sought to characterize an MIA protocol throughout development to understand how prenatal immune insult alters the trajectory of important neurodevelopmental processes, including the microglial regulation of synaptic spines and complement signaling. We used polyinosinic:polycytidylic acid (polyI:C) to induce MIA on gestational day 9.5 in CD-1 mice, and measured their synaptic spine density, microglial synaptic pruning, and complement protein expression. We found reduced dendritic spine density in the somatosensory cortex starting at 3-weeks-of-age with requisite increases in microglial synaptic pruning and phagocytosis, suggesting spine density loss was caused by increased microglial synaptic pruning. Additionally, we showed dysregulation in complement protein expression persisting into adulthood. Our findings highlight disruptions in the prenatal environment leading to alterations in multiple dynamic processes through to postnatal development. This could potentially suggest developmental time points during which synaptic processes could be measured as risk factors or targeted with therapeutics for neurodevelopmental disorders.


Subject(s)
Complement System Proteins , Dendritic Spines , Microglia , Poly I-C , Animals , Microglia/metabolism , Microglia/drug effects , Microglia/immunology , Mice , Female , Pregnancy , Dendritic Spines/metabolism , Poly I-C/pharmacology , Complement System Proteins/metabolism , Complement System Proteins/immunology , Prenatal Exposure Delayed Effects , Phagocytosis , Disease Models, Animal , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Synapses/metabolism , Synapses/drug effects , Neuronal Plasticity/drug effects
20.
Pharmacol Biochem Behav ; 240: 173788, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734150

ABSTRACT

Autism is a complex neurodevelopmental disorder with no effective treatment available currently. Repetitive transcranial magnetic stimulation (rTMS) is emerging as a promising neuromodulation technique to treat autism. However, the mechanism how rTMS works remains unclear, which restrict the clinical application of magnetic stimulation in the autism treatment. In this study, we investigated the effect of low-frequency rTMS on the autistic-like symptoms and explored if this neuroprotective effect was associated with synaptic plasticity and neuroinflammation in the hippocampus. A rat model of autism was established by intraperitoneal injection of valproic acid (VPA) in pregnant rats and male offspring were treated with 1 Hz rTMS daily for two weeks continuously. Behavior tests were performed to identify behavioral abnormality. Synaptic plasticity was measured by in vivo electrophysiological recording and Golgi-Cox staining. Synapse and inflammation associated proteins were detected by immunofluorescence and Western blot analyses. Results showed prenatal VPA-exposed rats exhibited autistic-like and anxiety-like behaviors, and cognitive impairment. Synaptic plasticity deficits and the abnormality expression of synapse-associated proteins were found in the hippocampus of prenatal VPA-exposed rats. Prenatal VPA exposure increased the level of inflammation cytokines and promoted the excessive activation of microglia. rTMS significantly alleviated the prenatal VPA-induced abnormalities including behavioral and synaptic plasticity deficits, and excessive neuroinflammation. TMS maybe a potential strategy for autism therapy via rescuing synaptic plasticity and inhibiting neuroinflammation.


Subject(s)
Autistic Disorder , Disease Models, Animal , Hippocampus , Neuronal Plasticity , Prenatal Exposure Delayed Effects , Transcranial Magnetic Stimulation , Valproic Acid , Animals , Valproic Acid/pharmacology , Neuronal Plasticity/drug effects , Rats , Autistic Disorder/therapy , Autistic Disorder/chemically induced , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Sprague-Dawley , Neuroinflammatory Diseases/therapy , Behavior, Animal/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...