Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.996
1.
Neurobiol Aging ; 139: 20-29, 2024 Jul.
Article En | MEDLINE | ID: mdl-38583392

Brazilian green propolis (propolis) is a chemically complex resinous substance that is a potentially viable therapeutic agent for Alzheimer's disease. Herein, propolis induced a transient increase in intracellular Ca2+ concentration ([Ca2+]i) in Neuro-2A cells; moreover, propolis-induced [Ca2+]i elevations were suppressed prior to 24-h pretreatment with amyloid-ß. To reveal the effect of [Ca2+]i elevation on impaired cognition, we performed memory-related behavioral tasks in APP-KI mice relative to WT mice at 4 and 12 months of age. Propolis, at 300-1000 mg/kg/d for 8 wk, significantly ameliorated cognitive deficits in APP-KI mice at 4 months, but not at 12 months of age. Consistent with behavioral observations, injured hippocampal long-term potentiation was markedly ameliorated in APP-KI mice at 4 months of age following repeated propolis administration. In addition, repeated administration of propolis significantly activated intracellular calcium signaling pathway in the CA1 region of APP-KI mice. These results suggest a preventive effect of propolis on cognitive decline through the activation of intracellular calcium signaling pathways in CA1 region of AD mice model.


Alzheimer Disease , Calcium , Cognitive Dysfunction , Disease Models, Animal , Propolis , Animals , Propolis/therapeutic use , Propolis/administration & dosage , Propolis/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Alzheimer Disease/etiology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/drug therapy , Calcium/metabolism , Mice, Transgenic , Calcium Signaling/drug effects , Long-Term Potentiation/drug effects , Male , Amyloid beta-Peptides/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Mice
2.
Behav Brain Res ; 466: 114974, 2024 May 28.
Article En | MEDLINE | ID: mdl-38554850

Polygala tenuifolia Wild is an ancient traditional Chinese medicine. Its main component, tenuifolin (TEN), has been proven to improve cognitive impairment caused by neurodegenerative diseases and ovariectomy. However, there was hardly any pharmacological research about TEN and its potential gender differences. Considering the reduction of TEN on learning and memory dysfunction in ovariectomized animals, therefore, we focused on the impact of TEN in different mice genders in the current study. Spontaneous alternation behavior (SAB), light-dark discrimination, and Morris water maze (MWM) tests were used to evaluate the mice's learning and memory abilities. The field excitatory postsynaptic potential (fEPSP) of the hippocampal CA1 region was recorded using an electrophysiological method, and the morphology of the dendritic structure was examined using Golgi staining. In the behavioral experiments, TEN improved the correct rate in female mice in the SAB test, the correct rate in the light-dark discrimination test, and the number of crossing platforms in the MWM test. Additionally, TEN reduced the latency of female mice rather than male mice in light-dark discrimination and MWM tests. Moreover, TEN could significantly increase the slope of fEPSP in hippocampal Schaffer-CA1 and enhance the total length and the number of intersections of dendrites in the hippocampal CA1 area in female mice but not in male mice. Collectively, the results of the current study showed that TEN improved learning and memory by regulating long-term potentiation (LTP) and dendritic structure of hippocampal CA1 area in female mice but not in males. These findings would help to explore the improvement mechanism of TEN on cognition and expand the knowledge of the potential therapeutic value of TEN in the treatment of cognitive impairment.


CA1 Region, Hippocampal , Dendrites , Diterpenes, Kaurane , Long-Term Potentiation , Animals , Female , Male , CA1 Region, Hippocampal/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Mice , Dendrites/drug effects , Memory/drug effects , Sex Factors , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Maze Learning/drug effects , Maze Learning/physiology
3.
Neuroscience ; 545: 148-157, 2024 May 03.
Article En | MEDLINE | ID: mdl-38513764

In this study, the electrophysiological and biochemical consequences of repeated exposure to morphine in male rats on glutamatergic synaptic transmission, synaptic plasticity, the expression of GABA receptors and glutamate receptors at the temporoammonic-CA1 synapse along the longitudinal axis of the hippocampus (dorsal, intermediate, ventral, DH, IH, VH, respectively) were investigated. Slice electrophysiological methods, qRT-PCR, and western blotting techniques were used to characterize synaptic plasticity properties. We showed that repeated morphine exposure (RME) reduced excitatory synaptic transmission and ability for long-term potentiation (LTP) in the VH as well as eliminated the dorsoventral difference in paired-pulse responses. A decreased expression of NR2B subunit in the VH and an increased expression GABAA receptor of α1 and α5 subunits in the DH were observed following RME. Furthermore, RME did not affect the expression of NR2A, AMPA receptor subunits, and γ2GABAA and GABAB receptors in either segment of the hippocampus. In sum, the impact of morphine may differ depending on the region of the hippocampus studied. A distinct change in the short- and long-term synaptic plasticity along the hippocampus long axis due to repeated morphine exposure, partially mediated by a change in the expression profile of glutamatergic receptor subunits. These findings can be useful in further understanding the cellular mechanism underlying deficits in information storage and, more generally, cognitive processes resulting from chronic opioid abuse.


Morphine , Neuronal Plasticity , Rats, Sprague-Dawley , Animals , Male , Morphine/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Rats , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Narcotics/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Receptors, GABA/metabolism , Receptors, GABA/drug effects
4.
J Alzheimers Dis ; 92(4): 1413-1426, 2023.
Article En | MEDLINE | ID: mdl-36911940

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by amyloid-ß peptide (Aß) deposition. Aß accumulation induces oxidative stress, leading to mitochondrial dysfunction, apoptosis, and so forth. Octadecaneuropeptide (ODN), a diazepam-binding inhibitor (DBI)-derived peptide, has been reported to have antioxidant properties. However, it is unclear whether ODN has neuroprotective effects in AD. OBJECTIVE: To profile the potential effects of ODN on AD. METHODS: We established a mouse model of AD via microinjection of Aß in the lateral ventricle. Utilizing a combination of western blotting assays, electrophysiological recordings, and behavioral tests, we investigated the neuroprotective effects of ODN on AD. RESULTS: DBI expression was decreased in AD model mice and cells. Meanwhile, ODN decreased Aß generation by downregulating amyloidogenic AßPP processing in HEK-293 cells stably expressing human Swedish mutant APP695 and BACE1 (2EB2). Moreover, ODN could inhibit Aß-induced oxidative stress in primary cultured cells and mice, as reflected by a dramatic increase in antioxidants and a decrease in pro-oxidants. We also found that ODN could reduce oxidative stress-induced apoptosis by restoring mitochondrial membrane potential, intracellular Ca2+ and cleaved caspase-3 levels in Aß-treated primary cultured cells and mice. More importantly, intracerebroventricular injection of ODN attenuated cognitive impairments as well as long-term potentiation in Aß-treated mice. CONCLUSION: These results suggest that ODN may exert a potent neuroprotective effect against Aß-induced neurotoxicity and memory decline via its antioxidant effects, indicating that ODN may be a potential therapeutic agent for AD.


Alzheimer Disease , Brain , Cognitive Dysfunction , Diazepam Binding Inhibitor , Neuropeptides , Neuroprotective Agents , Oxidative Stress , Peptide Fragments , Animals , Humans , Mice , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , CA1 Region, Hippocampal/drug effects , Cells, Cultured , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Diazepam Binding Inhibitor/pharmacology , Diazepam Binding Inhibitor/therapeutic use , Disease Models, Animal , HEK293 Cells , Long-Term Potentiation/drug effects , Membrane Potential, Mitochondrial/drug effects , Memory/drug effects , Mice, Inbred C57BL , Neurons/drug effects , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use
5.
Cells ; 11(2)2022 01 13.
Article En | MEDLINE | ID: mdl-35053378

Nicotine addiction develops predominantly during human adolescence through smoking. Self-administration experiments in rodents verify this biological preponderance to adolescence, suggesting evolutionary-conserved and age-defined mechanisms which influence the susceptibility to nicotine addiction. The hippocampus, a brain region linked to drug-related memory storage, undergoes major morpho-functional restructuring during adolescence and is strongly affected by nicotine stimulation. However, the signaling mechanisms shaping the effects of nicotine in young vs. adult brains remain unclear. MicroRNAs (miRNAs) emerged recently as modulators of brain neuroplasticity, learning and memory, and addiction. Nevertheless, the age-dependent interplay between miRNAs regulation and hippocampal nicotinergic signaling remains poorly explored. We here combined biophysical and pharmacological methods to examine the impact of miRNA-132/212 gene-deletion (miRNA-132/212-/-) and nicotine stimulation on synaptic functions in adolescent and mature adult mice at two hippocampal synaptic circuits: the medial perforant pathway (MPP) to dentate yrus (DG) synapses (MPP-DG) and CA3 Schaffer collaterals to CA1 synapses (CA3-CA1). Basal synaptic transmission and short-term (paired-pulse-induced) synaptic plasticity was unaltered in adolescent and adult miRNA-132/212-/- mice hippocampi, compared with wild-type controls. However, nicotine stimulation promoted CA3-CA1 synaptic potentiation in mature adult (not adolescent) wild-type and suppressed MPP-DG synaptic potentiation in miRNA-132/212-/- mice. Altered levels of CREB, Phospho-CREB, and acetylcholinesterase (AChE) expression were further detected in adult miRNA-132/212-/- mice hippocampi. These observations propose miRNAs as age-sensitive bimodal regulators of hippocampal nicotinergic signaling and, given the relevance of the hippocampus for drug-related memory storage, encourage further research on the influence of miRNAs 132 and 212 in nicotine addiction in the young and the adult brain.


Aging/genetics , Hippocampus/physiology , MicroRNAs/metabolism , Neuronal Plasticity/genetics , Nicotine/pharmacology , Acetylcholinesterase/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/physiology , Gene Expression Regulation/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Neuronal Plasticity/drug effects , Phosphorylation/drug effects , Synaptic Transmission/drug effects
6.
Biomed Pharmacother ; 147: 112663, 2022 Mar.
Article En | MEDLINE | ID: mdl-35093759

Memory-enhancing agents have long been required for various reasons such as for obtaining a good score in a test in the young and for retaining memory in the aged. Although many studies have found that several natural products may be good candidates for memory enhancement, there is still a need for better agents. The present study investigated whether rubrofusarin, an active ingredient in Cassiae semen, enhances learning and memory in normal mice. Passive avoidance and Morris water maze tests were performed to determine the memory-enhancing ability of rubrofusarin. To investigate synaptic function, hippocampal long-term potentiation (LTP) was measured. Western blotting was performed to determine protein levels. To investigate neurite outgrowth, DCX immunohistochemistry and cell culture were utilised. Rubrofusarin (1, 3, 10, 30 mg/kg) enhanced memory in passive avoidance and Morris water maze tests. Moreover, rubrofusarin ameliorated scopolamine-induced memory impairment. In the rubrofusarin-treated group, high-frequency stimulation induced higher LTP in the hippocampal Schaffer-collateral pathway compared to the control group. The rubrofusarin-treated group showed a higher number of DCX-positive immature neurons with an increase in the length of dendrites compared to the control group in the hippocampal dentate gyrus region. In vitro experiments showed that rubrofusarin facilitated neurite outgrowth in neuro2a cells through extracellular signal-regulated kinase (ERK). Finally, we found that extracellular signal-regulated kinase (ERK) is required for rubrofusarin-induced enhancement of neurite outgrowth, learning and memory. These results demonstrate that rubrofusarin enhances learning and memory and neurite outgrowth, and these might need activation of ERK pathway.


Cognition/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Neuronal Outgrowth/drug effects , Pyrones/pharmacology , Animals , Cell Culture Techniques , Dose-Response Relationship, Drug , Hippocampus/drug effects , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Pyrones/administration & dosage
7.
Life Sci ; 293: 120349, 2022 Mar 15.
Article En | MEDLINE | ID: mdl-35065162

AIMS: Propofol, the most commonly used intravenous anesthetic, is known for its protective effect in various human and animal disease models such as post-traumatic stress disease (PTSD). However, it still needs efforts to clarify the effect of propofol on fear memory extinction and the relevant mechanisms. METHODS: Fear memory extinction was examined in PTSD mice model. Thirty-six mice were randomly divided into three groups: a shock + propofol group (sh + Pro), shock + normal saline group (sh + NS), and sham group. The mice were treated with propofol (150 mg/kg) or normal saline (of the same volume) intraperitoneally 30 min after the conditioning. These mice's behavior was analysed with contextual test, sucrose preference test (SPT) and Morris water maze (MWM). Additionally, the synaptic plasticity of the hippocampus was examined by long-term potentiation (LTP) and long-term depression (LTD). KEY FINDINGS: Compared with the sham group, the sh + NS group showed increased freezing time and depressive behavior, meanwhile the sh + Pro group showed minor behavioral changes. What's more, we found that propofol rescued the impaired long-term potentiation (LTP) and long-term depression (LTD) in hippocampus of PTSD mice. All these suggest that propofol can accelerate fear memory extinction and change synaptic plasticity of PTSD mice. SIGNIFICANCE: The study proved that propofol can protect the mice from PTSD by reserving synaptic plasticity and brought a new insight into PTSD treatment indicating that propofol maybe a potential cure for PTSD.


Hippocampus/drug effects , Maze Learning/drug effects , Memory/drug effects , Neuronal Plasticity/drug effects , Propofol/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Animals , Hippocampus/physiology , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/therapeutic use , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Propofol/pharmacology , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology
8.
Exp Neurol ; 347: 113892, 2022 01.
Article En | MEDLINE | ID: mdl-34634309

Intrapleural injections of cholera toxin B conjugated to saporin (CTB-SAP) selectively eliminates respiratory (e.g., phrenic) motor neurons, and mimics motor neuron death and respiratory deficits observed in rat models of neuromuscular diseases. Additionally, microglial density increases in the phrenic motor nucleus following CTB-SAP. This CTB-SAP rodent model allows us to study the impact of motor neuron death on the output of surviving phrenic motor neurons, and the underlying mechanisms that contribute to enhancing or constraining their output at 7 days (d) or 28d post-CTB-SAP injection. 7d CTB-SAP rats elicit enhanced phrenic long-term facilitation (pLTF) through the Gs-pathway (inflammation-resistant in naïve rats), while pLTF is elicited though the Gq-pathway (inflammation-sensitive in naïve rats) in control and 28d CTB-SAP rats. In 7d and 28d male CTB-SAP rats and controls, we evaluated the effect of cyclooxygenase-1/2 enzymes on pLTF by delivery of the nonsteroidal anti-inflammatory drug, ketoprofen (IP), and we hypothesized that pLTF would be unaffected by ketoprofen in 7d CTB-SAP rats, but pLTF would be enhanced in 28d CTB-SAP rats. In anesthetized, paralyzed and ventilated rats, pLTF was surprisingly attenuated in 7d CTB-SAP rats and enhanced in 28d CTB-SAP rats (both p < 0.05) following ketoprofen delivery. Additionally in CTB-SAP rats: 1) microglia were more amoeboid in the phrenic motor nucleus; and 2) cervical spinal inflammatory-associated factor expression (TNF-α, BDNF, and IL-10) was increased vs. controls in the absence of ketoprofen (p < 0.05). Following ketoprofen delivery, TNF-α and IL-10 expression was decreased back to control levels, while BDNF expression was differentially affected over the course of motor neuron death in CTB-SAP rats. This study furthers our understanding of factors (e.g., cyclooxygenase-1/2-induced inflammation) that contribute to enhancing or constraining pLTF and its implications for breathing following respiratory motor neuron death.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ketoprofen/pharmacology , Long-Term Potentiation/drug effects , Motor Neurons/drug effects , Phrenic Nerve/drug effects , Animals , Cell Death/drug effects , Cholera Toxin/toxicity , Male , Microglia/metabolism , Motor Neurons/pathology , Neuromuscular Diseases/chemically induced , Neuromuscular Diseases/pathology , Neuromuscular Diseases/physiopathology , Phrenic Nerve/pathology , Rats , Rats, Sprague-Dawley , Saporins/toxicity
9.
Neuropharmacology ; 202: 108846, 2022 01 01.
Article En | MEDLINE | ID: mdl-34687710

Drugs that block N-methyl-d-aspartate receptors (NMDARs) suppress hippocampus-dependent memory formation; they also block long-term potentiation (LTP), a cellular model of learning and memory. However, the fractional block that is required to achieve these effects is unknown. Here, we measured the dose-dependent suppression of contextual memory in vivo by systemic administration of the competitive antagonist (R,S)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP); in parallel, we measured the concentration-dependent block by CPP of NMDAR-mediated synapses and LTP of excitatory synapses in hippocampal brain slices in vitro. We found that the dose of CPP that suppresses contextual memory in vivo (EC50 = 2.3 mg/kg) corresponds to a free concentration of 53 nM. Surprisingly, applying this concentration of CPP to hippocampal brain slices had no effect on the NMDAR component of evoked field excitatory postsynaptic potentials (fEPSPNMDA), or on LTP. Rather, the IC50 for blocking the fEPSPNMDA was 434 nM, and for blocking LTP was 361 nM - both nearly an order of magnitude higher. We conclude that memory impairment produced by systemically administered CPP is not due primarily to its blockade of NMDARs on hippocampal pyramidal neurons. Rather, systemic CPP suppresses memory formation by actions elsewhere in the memory-encoding circuitry.


CA1 Region, Hippocampal/physiology , Learning/drug effects , Long-Term Potentiation/drug effects , Memory/drug effects , Pyramidal Cells/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Female , In Vitro Techniques , Male , Mice, Inbred C57BL
10.
Neuropharmacology ; 202: 108840, 2022 01 01.
Article En | MEDLINE | ID: mdl-34678377

Different types of memory are thought to rely on different types of synaptic plasticity, many of which depend on the activation of the N-Methyl-D Aspartate (NMDA) subtype of glutamate receptors. Accordingly, there is considerable interest in the possibility of using positive allosteric modulators (PAMs) of NMDA receptors (NMDARs) as cognitive enhancers. Here we firstly review the evidence that NMDA receptor-dependent forms of synaptic plasticity: short-term potentiation (STP), long-term potentiation (LTP) and long-term depression (LTD) can be pharmacologically differentiated by using NMDAR ligands. These observations suggest that PAMs of NMDAR function, depending on their subtype selectivity, might differentially regulate STP, LTP and LTD. To test this hypothesis, we secondly performed experiments in rodent hippocampal slices with UBP714 (a GluN2A/2B preferring PAM), CIQ (a GluN2C/D selective PAM) and UBP709 (a pan-PAM that potentiates all GluN2 subunits). We report here, for the first time, that: (i) UBP714 potentiates sub-maximal LTP and reduces LTD; (ii) CIQ potentiates STP without affecting LTP; (iii) UBP709 enhances LTD and decreases LTP. We conclude that PAMs can differentially regulate distinct forms of NMDAR-dependent synaptic plasticity due to their subtype selectivity.


Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Allosteric Regulation , Animals , Benzimidazoles/pharmacology , Hippocampus , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
11.
J Neurosci ; 42(3): 350-361, 2022 01 19.
Article En | MEDLINE | ID: mdl-34815314

Highly correlated firing of primary afferent inputs and lamina I projection neurons evokes synaptic long-term potentiation (LTP), a mechanism by which ascending nociceptive transmission can be amplified at the level of the spinal dorsal horn. However, the degree to which neuromodulatory signaling shapes the temporal window governing spike-timing-dependent plasticity (STDP) at sensory synapses onto projection neurons remains unclear. The present study demonstrates that activation of spinal D1/D5 dopamine receptors (D1/D5Rs) creates a highly permissive environment for the production of LTP in male and female adult mouse spinoparabrachial neurons by promoting non-Hebbian plasticity. Bath application of the mixed D1/D5R agonist SKF82958 unmasked LTP at STDP pairing intervals that normally fail to alter synaptic efficacy. Furthermore, during D1/D5R signaling, action potential discharge in projection neurons became dispensable for LTP generation, and primary afferent stimulation alone was sufficient to induce strengthening of sensory synapses. This non-Hebbian LTP was blocked by the D1/D5R antagonist SCH 39166 or genetic deletion of D5R, and required activation of mGluR5 and intracellular Ca2+ release but was independent of NMDAR activation. D1/D5R-enabled non-Hebbian plasticity was observed across multiple neuronal subpopulations in the superficial dorsal horn but was more prevalent in spinoparabrachial neurons than interneurons. Interestingly, the ability of neonatal tissue damage to promote non-Hebbian LTP in adult projection neurons was not observed in D5R knock-out mice. Collectively, these findings suggest that joint spinal D1/D5R and mGluR5 activation can allow unfettered potentiation of sensory synapses onto the output neurons responsible for conveying pain and itch information to the brain.SIGNIFICANCE STATEMENT Synaptic LTP in spinal projection neurons has been implicated in the generation of chronic pain. Under normal conditions, plasticity at sensory synapses onto adult mouse spinoparabrachial neurons follows strict Hebbian learning rules, requiring coincident presynaptic and postsynaptic firing. Here, we demonstrate that the activation of spinal D1/D5Rs promotes a switch from Hebbian to non-Hebbian LTP so that primary afferent stimulation alone is sufficient to evoke LTP in the absence of action potential discharge in projection neurons, which required joint activation of mGluR5 and intracellular Ca2+ release but not NMDARs. These results suggest that D1/D5Rs cooperate with mGluR5 receptors in the spinal dorsal horn to powerfully influence the amplification of ascending nociceptive transmission to the brain.


Long-Term Potentiation/drug effects , Neurons/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D5/agonists , Spinal Cord Dorsal Horn/drug effects , Synapses/drug effects , Action Potentials/drug effects , Animals , Benzazepines/pharmacology , Calcium/metabolism , Dopamine Agonists/pharmacology , Female , Male , Mice , Mice, Knockout , Neurons/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism , Spinal Cord Dorsal Horn/metabolism , Synapses/metabolism
12.
J Alzheimers Dis ; 85(1): 343-357, 2022.
Article En | MEDLINE | ID: mdl-34806605

BACKGROUND: Cognitive deficit is mainly clinical characteristic of Alzheimer's disease (AD). Recent reports showed adiponectin and its analogues could reverse cognitive impairments, lower amyloid-ß protein (Aß) deposition, and exert anti-inflammatory effects in different APP/PS1 AD model mice mainly exhibiting amyloid plaque pathology. However, the potential in vivo electrophysiological mechanism of adiponectin protecting against cognitive deficits in AD and the neuroprotective effects of adiponectin on 3xTg-AD mice including both plaque and tangle pathology are still unclear. OBJECTIVE: To observe the effects of adiponectin treatment on cognitive deficits in 3xTg-AD mice, investigate its potential in vivo electrophysiological mechanism, and testify its anti-inflammatory effects. METHODS: Barnes maze test, Morris water maze test, and fear conditioning test were used to evaluate the memory-ameliorating effects of adiponectin on 3xTg-AD mice. In vivo hippocampal electrophysiological recording was used to observe the change of basic synaptic transmission, long-term potentiation, and long-term depression. Immunohistochemistry staining and western blot were used to observe the activation of microglia and astroglia, and the expression levels of proinflammatory factors and anti-inflammtory factor IL-10. RESULTS: Adiponectin treatment could alleviate spatial memory and conditioned fear memory deficits observed in 3xTg-AD mice, improve in vivo LTP depression and LTD facilitation, inhibit overactivation of microglia and astroglia, decrease the expression of proinflammatory factors NF- κB and IL-1ß, and increase the expression level of IL-10 in the hippocampus of 3xTg-AD mice. CONCLUSION: Adiponectin could ameliorate cognitive deficits in 3xTg-AD mice through improving in vivo synaptic plasticity impairments and alleviating neuroinflammation in the hippocampus of 3xTg-AD mice.


Adiponectin/pharmacology , Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Behavior, Animal/drug effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Mice, Transgenic , Spatial Memory/drug effects , Synaptic Transmission/drug effects
13.
J Med Chem ; 65(1): 217-233, 2022 01 13.
Article En | MEDLINE | ID: mdl-34962802

Cognitive impairment and learning ability of the brain are directly linked to synaptic plasticity as measured in changes of long-term potentiation (LTP) and long-term depression (LTD) in animal models of brain diseases. LTD reflects a sustained reduction of the synaptic AMPA receptor content based on targeted clathrin-mediated endocytosis. AMPA receptor endocytosis is initiated by dephosphorylation of Tyr876 on the C-terminus of the AMPAR subunit GluA2. The brain-specific striatal-enriched protein tyrosine phosphatase (STEP) is responsible for this process. To identify new, highly effective inhibitors of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) internalization, we performed structure-based design of peptides able to inhibit STEP-GluA2-CT complex formation. Two short peptide derivatives were found as efficient in vitro inhibitors. Our in vivo experiments evidenced that both peptides restore the memory deficits and display anxiolytic and antidepressant effects in a scopolamine-treated rat model. The interference peptides identified and characterized here represent promising lead compounds for novel cognitive enhancers and/or behavioral modulators.


Cognition/drug effects , Long-Term Potentiation/drug effects , Peptide Fragments/pharmacology , Protein Interaction Domains and Motifs/drug effects , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Animals , Endocytosis , Hippocampus/drug effects , Male , Mice , Neuronal Plasticity , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Synapses/drug effects
14.
Exp Neurol ; 350: 113929, 2022 04.
Article En | MEDLINE | ID: mdl-34813840

Obstructive sleep apnea-hypopnea syndrome (OSAHS) is widely known for its multiple systems damage, especially neurocognitive deficits in children. Since their discovery, adenosine A2A receptors (A2ARs) have been considered as key elements in signaling pathways mediating neurodegenerative diseases such as Huntington's and Alzheimer's, as well as cognitive function regulation. Herein, we investigated A2AR role in cognitive impairment induced by chronic intermittent hypoxia (CIH). Mice were exposed to CIH 7 h every day for 4 weeks, and intraperitoneally injected with A2AR agonist CGS21680 or A2AR antagonist SCH58261 half an hour before IH exposure daily. The 8-arm radial arm maze was utilized to assess spatial memory after CIH exposures.To validate findings using pharmacology, the impact of intermittent hypoxia was investigated in A2AR knockout mice. CIH-induced memory dysfunction was manifested by increased error rates in the radial arm maze test. The behavioral changes were associated with hippocampal pathology, neuronal apoptosis, and synaptic plasticity impairment. The stimulation of adenosine A2AR exacerbated memory impairment with more serious neuropathological damage, attenuated long-term potentiation (LTP), syntaxin down-regulation, and increased BDNF protein. Moreover, apoptosis-promoting protein cleaved caspase-3 was upregulated while anti-apoptotic protein Bcl-2 was downregulated. Consistent with these findings, A2AR inhibition with SCH58261 and A2AR deletion exhibited the opposite result. Overall, these findings suggest that A2AR plays a critical role in CIH-induced impairment of learning and memory by accelerating hippocampal neuronal apoptosis and reducing synaptic plasticity. Blockade of adenosine A2A receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice.


Adenosine A2 Receptor Antagonists/therapeutic use , Cognition Disorders/prevention & control , Hypoxia, Brain/drug therapy , Hypoxia, Brain/psychology , Receptor, Adenosine A2A/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , Chronic Disease , Cognition Disorders/chemically induced , Cognitive Dysfunction , Hippocampus/pathology , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Psychomotor Performance/drug effects , Pyrimidines/therapeutic use , Receptor, Adenosine A2A/genetics , Triazoles/therapeutic use
15.
Int J Mol Sci ; 22(24)2021 Dec 12.
Article En | MEDLINE | ID: mdl-34948152

Status epilepticus (SE) causes persistent abnormalities in the functioning of neuronal networks, often resulting in worsening epileptic seizures. Many details of cellular and molecular mechanisms of seizure-induced changes are still unknown. The lithium-pilocarpine model of epilepsy in rats reproduces many features of human temporal lobe epilepsy. In this work, using the lithium-pilocarpine model in three-week-old rats, we examined the morphological and electrophysiological changes in the hippocampus within a week following pilocarpine-induced seizures. We found that almost a third of the neurons in the hippocampus and dentate gyrus died on the first day, but this was not accompanied by impaired synaptic plasticity at that time. A diminished long-term potentiation (LTP) was observed following three days, and the negative effect of SE on plasticity increased one week later, being accompanied by astrogliosis. The attenuation of LTP was caused by the weakening of N-methyl-D-aspartate receptor (NMDAR)-dependent signaling. NMDAR-current was more than two-fold weaker during high-frequency stimulation in the post-SE rats than in the control group. Application of glial transmitter D-serine, a coagonist of NMDARs, allows the enhancement of the NMDAR-dependent current and the restoration of LTP. These results suggest that the disorder of neuron-astrocyte interactions plays a critical role in the impairment of synaptic plasticity.


Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Lithium/adverse effects , Long-Term Potentiation/drug effects , Pilocarpine/adverse effects , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Lithium/pharmacology , Male , Pilocarpine/pharmacology , Rats , Rats, Wistar
16.
Molecules ; 26(20)2021 Oct 10.
Article En | MEDLINE | ID: mdl-34684684

Memory deterioration in Alzheimer's disease (AD) is thought to be underpinned by aberrant amyloid ß (Aß) accumulation, which contributes to synaptic plasticity impairment. Avenanthramide-C (Avn-C), a polyphenol compound found predominantly in oats, has a range of biological properties. Herein, we performed methanolic extraction of the Avns-rich fraction (Fr. 2) from germinated oats using column chromatography, and examined the effects of Avn-C on synaptic correlates of memory in a mouse model of AD. Avn-C was identified in Fr. 2 based on 1H-NMR analysis. Electrophysiological recordings were performed to examine the effects of Avn-C on the hippocampal long-term potentiation (LTP) in a Tg2576 mouse model of AD. Avn-C from germinated oats restored impaired LTP in Tg2576 mouse hippocampal slices. Furthermore, Avn-C-facilitated LTP was associated with changes in the protein levels of phospho-glycogen synthase kinase-3ß (p-GSK3ß-S9) and cleaved caspase 3, which are involved in Aß-induced synaptic impairment. Our findings suggest that the Avn-C extract from germinated oats may be beneficial for AD-related synaptic plasticity impairment and memory decline.


Alzheimer Disease/drug therapy , Hippocampus/drug effects , Long-Term Potentiation/drug effects , ortho-Aminobenzoates/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Avena/chemistry , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Male , Mice , Mice, Transgenic , Neuronal Plasticity , Plant Extracts/pharmacology
17.
Cell Rep ; 37(1): 109786, 2021 10 05.
Article En | MEDLINE | ID: mdl-34610314

Regulated insertion and removal of postsynaptic AMPA glutamate receptors (AMPARs) mediates hippocampal long-term potentiation (LTP) and long-term depression (LTD) synaptic plasticity underlying learning and memory. In Alzheimer's disease ß-amyloid (Aß) oligomers may impair learning and memory by altering AMPAR trafficking and LTP/LTD balance. Importantly, Ca2+-permeable AMPARs (CP-AMPARs) assembled from GluA1 subunits are excluded from hippocampal synapses basally but can be recruited rapidly during LTP and LTD to modify synaptic strength and signaling. By employing mouse knockin mutations that disrupt anchoring of the kinase PKA or phosphatase Calcineurin (CaN) to the postsynaptic scaffold protein AKAP150, we find that local AKAP-PKA signaling is required for CP-AMPAR recruitment, which can facilitate LTP but also, paradoxically, prime synapses for Aß impairment of LTP mediated by local AKAP-CaN LTD signaling that promotes subsequent CP-AMPAR removal. These findings highlight the importance of PKA/CaN signaling balance and CP-AMPARs in normal plasticity and aberrant plasticity linked to disease.


A Kinase Anchor Proteins/genetics , Amyloid beta-Peptides/pharmacology , Calcineurin/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Receptors, AMPA/metabolism , A Kinase Anchor Proteins/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Calcineurin/metabolism , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Excitatory Postsynaptic Potentials/drug effects , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, AMPA/antagonists & inhibitors , Receptors, Glutamate/chemistry , Receptors, Glutamate/metabolism , Signal Transduction/drug effects , Spermine/analogs & derivatives , Spermine/pharmacology , Synapses/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
18.
Biomed Pharmacother ; 144: 112266, 2021 Dec.
Article En | MEDLINE | ID: mdl-34634555

BACKGROUND: In the present study, we aimed to investigate the effects of probucol on aging-related hippocampus-dependent cognitive impairment and explore the potential mechanisms. METHODS: D-galactose (100 mg/kg, once daily for 6 weeks) was subcutaneously injected to induce aging in mice. Then the mice were administered with probucol or vehicle once a day for 2 weeks. The hippocampus-related cognition was evaluated with Morris water maze test, novel object recognition test, and contextual fear conditioning test. Moreover, synaptic plasticity was assessed, and RNA-sequencing was applied to further explore the molecular mechanisms. RESULTS: Aging mice induced by D-galactose showed conspicuous learning and memory impairment, which was significantly ameliorated by probucol. Meanwhile, probucol enhanced the spine density and dendritic branches, improved long-term potentiation, and increased the expression of PSD95 of aging mice. Probucol regulated 70 differentially expressed genes compared to D-galactose group, of which 38 genes were upregulated and 32 genes were downregulated. At last, RNA-sequencing results were verified by quantitative reverse transcription-polymerase chain reaction. CONCLUSIONS: Probucol improved learning and memory in aging mice through enhancing synaptic plasticity and regulating gene expression, indicating the potential application of probucol to prevent and treat aging-related disorders.


Behavior, Animal/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Hippocampus/drug effects , Neurons/drug effects , Nootropic Agents/pharmacology , Probucol/pharmacology , Age Factors , Animals , Cellular Senescence/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Fear/drug effects , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation/drug effects , Male , Mice, Inbred C57BL , Morris Water Maze Test/drug effects , Neurons/metabolism , Neurons/pathology , Open Field Test/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Pak J Pharm Sci ; 34(3): 909-914, 2021 May.
Article En | MEDLINE | ID: mdl-34602413

N-Methyl-D-aspartate receptor (NMDAR)-induced antioxidation is a significant cause of neuronal injury after ischemic stroke. In a previous work, we verified the neuroprotective roles of geniposide during tMCAO in vivo. However, it remains unknown whether geniposide ameliorates injury to hippocampal neurons during Ischemic Long Term Potentiation (iLTP) induction in vitro. After induction of cells oxygen-glucose deprivation or hydrogen peroxide, the protection of geniposide evaluated by MTT assay and electrophysiological tests. In this study, we suggested neuronal cell apoptosis was attenuated by geniposide. Furthermore, field excitatory postsynaptic potentials (fEPSCs) following postischemic LTP were assessed by electrophysiological tests. Finally, we determined that medium and high doses of geniposide attenuated oxidative stress insult and improved iLTP. Importantly, these effects were abolished by cotreatment with geniposide and the GluN2A antagonist NVP. In contrast, the GluN2B inhibitor ifenprodil failed to have an effect. In conclusion, we suggest for the first time that treatment with geniposide can attenuate postischemic LTP induction in a concentration-dependent manner. We infer that GluN2A-containing NMDARs are involved in the neuroprotection induced by geniposide treatment in ischemia.


Excitatory Postsynaptic Potentials/drug effects , Hypoxia-Ischemia, Brain/metabolism , Iridoids/pharmacology , Long-Term Potentiation/drug effects , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Apoptosis/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiopathology , Hydrogen Peroxide/pharmacology , Hypoxia-Ischemia, Brain/physiopathology , In Vitro Techniques , Infarction, Middle Cerebral Artery/physiopathology , Neurons/metabolism , Oxidants/pharmacology , PC12 Cells , Piperidines/pharmacology , Quinoxalines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
20.
J Alzheimers Dis ; 84(1): 239-248, 2021.
Article En | MEDLINE | ID: mdl-34511503

BACKGROUND: Tauopathies are a group of neurodegenerative disorders, including Alzheimer's disease (AD) and frontotemporal lobar degeneration with tau pathology. Hyperphosphorylation modification promotes tau protein misfolding and aggregation into neurofibrillary tangles, leading to impairments of synaptic plasticity and learning and memory. However, very limited therapeutic strategies are available. OBJECTIVE: In the present study, we wanted to investigate the potential effects of Dihydroartemisinin (DHA) on tauopathies. METHODS: We constructed adeno-associated virus carrying hTau cDNA (AAVhTau) to establish a mouse model of tauopathy through intrahippocampal microinjection. Using a combination of behavioral test, electrophysiological recording, and western blotting assay, we examined the neuroprotective effects of DHA on learning and memory deficits in mice with tauopathy. RESULTS: DHA improved learning and memory and increased hippocampal CA1 long-term potentiation (LTP) in mice overexpressed human tau (hTau) in the hippocampus. More importantly, further study revealed that DHA could induce protein O-GlcNAcylation modification and reduce protein phosphorylation. O-GlcNAc transferase inhibitor alloxan could suppress DHA-induced protein O-GlcNAcylation, and subsequently prevent therapeutic effect of DHA on the deficits of learning and memory as well as synaptic plasticity in hTau mice. CONCLUSION: These results indicate that DHA may exert neuroprotective role in tauopathy through a crosstalk between O-GlcNAcylation and phosphorylation, suggesting a potential therapeutic for learning and memory deficits associated with tau pathology.


Antimalarials/therapeutic use , Artemisinins/therapeutic use , Cognition/drug effects , Long-Term Potentiation/drug effects , Neuroprotective Agents/pharmacology , Tauopathies/drug therapy , Alzheimer Disease/pathology , Animals , Brain/drug effects , Disease Models, Animal , Humans , Learning/drug effects , Mice , Phosphorylation
...