Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Mol Psychiatry ; 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704508

ABSTRACT

Sensory abnormalities are observed in ~90% of individuals with autism spectrum disorders (ASD), but the underlying mechanisms are poorly understood. GluN2B, an NMDA receptor subunit that regulates long-term depression and circuit refinement during brain development, has been strongly implicated in ASD, but whether GRIN2B mutations lead to sensory abnormalities remains unclear. Here, we report that Grin2b-mutant mice show behavioral sensory hypersensitivity and brain hyperconnectivity associated with the anterior cingulate cortex (ACC). Grin2b-mutant mice with a patient-derived C456Y mutation (Grin2bC456Y/+) show sensory hypersensitivity to mechanical, thermal, and electrical stimuli through supraspinal mechanisms. c-fos and functional magnetic resonance imaging indicate that the ACC is hyperactive and hyperconnected with other brain regions under baseline and stimulation conditions. ACC pyramidal neurons show increased excitatory synaptic transmission. Chemogenetic inhibition of ACC pyramidal neurons normalizes ACC hyperconnectivity and sensory hypersensitivity. These results suggest that GluN2B critically regulates ASD-related cortical connectivity and sensory brain functions.

2.
Cereb Cortex ; 32(12): 2508-2520, 2022 06 07.
Article in English | MEDLINE | ID: mdl-34607355

ABSTRACT

To understand how incoming cortical inputs are processed by different types of cortical projection neurons in the medial prefrontal cortex, we compared intrinsic physiological properties of and commissural excitatory/inhibitory influences on layer 5 intratelencephalic (IT), layer 5 pyramidal tract (PT), and layers 2/3 IT projection neurons. We found that intrinsic physiological properties and commissural synaptic transmission varied across the three types of projection neurons. The rank order of intrinsic excitability was layer 5 PT > layer 5 IT > layers 2/3 IT neurons. Commissural connectivity was higher in layers 2/3 than layer 5 projection neurons, but commissural excitatory influence was stronger on layer 5 than layers 2/3 pyramidal neurons. Paired-pulse ratio was also greater in PT than IT neurons. These results indicate that commissural inputs activate deep layer PT neurons most preferentially and superficial layer IT neurons least preferentially. Deep layer PT neurons might faithfully transmit cortical input signals to downstream subcortical structures for reliable control of behavior, whereas superficial layer IT neurons might integrate cortical input signals from diverse sources in support of higher-order cognitive functions.


Subject(s)
Neurons , Pyramidal Cells , Interneurons , Neurons/physiology , Pyramidal Cells/physiology , Pyramidal Tracts/physiology , Synaptic Transmission/physiology
3.
J Neurosci ; 41(34): 7162-7170, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34290082

ABSTRACT

Hyperphosphorylation of the microtubule associated protein tau (tau) is inextricably linked to several neurodegenerative diseases, collectively termed tauopathies, in which synapse dysfunction occurs through largely unidentified mechanisms. Our research aimed to uncover molecular mechanisms by which phosphorylation of tau (pTau) affects synapse function. Using combined molecular and electrophysiological analysis with in vitro genetic knock-in of phosphorylation mutant human tau in male rat CA1 hippocampal neurons, we show an interplay between tau and protein kinase C and casein kinase substrate in neurons protein 1 (PACSIN1) that regulates synapse function. pTau at serine residues 396/404 decreases tau:PACSIN1 binding and evokes PACSIN1-dependent functional and structural synapse weakening. Knock-down of tau or PACSIN1 increases AMPA receptor (AMPAR)-mediated current at extrasynaptic regions, supporting a role for these proteins in affecting AMPAR trafficking. The pTau-induced PACSIN1 dissociation may represent a pathophysiological regulator of synapse function that underlies tauopathy-associated synapse defects.SIGNIFICANCE STATEMENT Knowledge is still lacking for how hyperphosphorylation of tau and its effectors lead to synaptic and neuronal dysfunction. Our results provide crucial insight for this mechanistic understanding; we show that specific tau phosphorylation events modulate its protein interaction with PACSIN1 and thus elicits synapse weakening likely through PACSIN1-dependent regulation of AMPA receptor (AMPAR) trafficking. These findings develop our understanding of molecular events that may be relevant to cellular changes underpinning tauopathy-associated neurodegenerative diseases.


Subject(s)
Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , tau Proteins/metabolism , Action Potentials , Animals , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Excitatory Postsynaptic Potentials , Gene Knockdown Techniques , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mutation, Missense , Patch-Clamp Techniques , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Receptors, Glutamate/physiology , Recombinant Proteins/metabolism , tau Proteins/antagonists & inhibitors , tau Proteins/genetics
4.
Biochem Biophys Res Commun ; 637: 100-107, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36395691

ABSTRACT

Recently, we reported that auditory fear conditioning leads to the presynaptic potentiation at lateral amygdala to basal amygdala (LA-BA) synapses that shares the mechanism with high-frequency stimulation (HFS)-induced long-term potentiation (LTP) ex vivo. In the present study, we further examined the molecular mechanisms underlying the HFS-induced presynaptic LTP. We found that a presynaptic elevation of Ca2+ was required for the LTP induction. Interestingly, the blockade of presynaptic but not postsynaptic HCN channels with ZD7288 completely abolished LTP induction. While ZD7288 did not alter basal synaptic transmission, the blocker fully reversed previously established LTP, indicating that HCN channels are also required for the maintenance of LTP. Indeed, HCN3 and HCN4 channels were preferentially localized in the presynaptic boutons of LA afferents. Furthermore, an inhibition of either GABAB receptors or GIRK channels eliminated the inhibitory effect of HCN blockade on the LTP induction. Collectively, we suggest that activation of presynaptic HCN channels may counteract membrane hyperpolarization during tetanic stimulation, and thereby contributes to the presynaptic LTP at LA-BA synapses.


Subject(s)
Basolateral Nuclear Complex , Long-Term Potentiation , Synapses , Synaptic Transmission , Presynaptic Terminals
5.
Biochem Biophys Res Commun ; 584: 39-45, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34768080

ABSTRACT

The lateral amygdala (LA) is a main sensory input site from the cortical and thalamic regions. In turn, LA glutamatergic pyramidal neurons strongly project to the basal amygdala (BA). Although it is well known that auditory fear conditioning involves synaptic potentiation in the LA, it is not clear whether the LA-BA synaptic transmission is modified upon auditory fear conditioning. Here we found that high-frequency stimulation ex vivo resulted in long-term potentiation (LTP) with a concomitant enhancement of neurotransmitter release at LA-BA synapses. Auditory fear conditioning also led to the presynaptic facilitation at LA-BA synapses. Meanwhile, AMPA/NMDA current ratio was not changed upon fear conditioning, excluding the involvement of postsynaptic mechanism. Notably, fear conditioning occluded electrically induced ex vivo LTP in the LA-BA pathway, indicating that the conditioning and electrically induced LTP share common mechanisms. Our findings suggest that the presynaptic potentiation of LA-BA synapses may be involved in fear conditioning.


Subject(s)
Basolateral Nuclear Complex/physiology , Conditioning, Classical/physiology , Fear/physiology , Neurotransmitter Agents/metabolism , Synapses/physiology , Acoustic Stimulation , Amygdala/metabolism , Amygdala/physiology , Animals , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/metabolism , Long-Term Potentiation/physiology , Male , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, AMPA/physiology , Synapses/metabolism , Synaptic Transmission/physiology
6.
Int J Mol Sci ; 21(10)2020 May 13.
Article in English | MEDLINE | ID: mdl-32414166

ABSTRACT

The aim of this study was to examine whether rubrofusarin, an active ingredient of the Cassia species, has an antidepressive effect in chronic restraint stress (CRS) mouse model. Although acute treatment using rubrofusarin failed, chronic treatment using rubrofusarin ameliorated CRS-induced depressive symptoms. Rubrofusarin treatment significantly reduced the number of Fluoro-Jade B-positive cells and caspase-3 activation within the hippocampus of CRS-treated mice. Moreover, rubrofusarin treatment significantly increased the number of newborn neurons in the hippocampus of CRS-treated mice. CRS induced activation of glycogen synthase kinase-3ß and regulated development and DNA damage responses, and reductions in the extracellular-signal-regulated kinase pathway activity were also reversed by rubrofusarin treatment. Microglial activation and inflammasome markers, including nod-like receptor family pyrin domain containing 3 and adaptor protein apoptosis-associated speck-like protein containing CARD, which were induced by CRS, were ameliorated by rubrofusarin. Synaptic plasticity dysfunction within the hippocampus was also rescued by rubrofusarin treatment. Within in vitro experiments, rubrofusarin blocked corticosterone-induced long-term potentiation impairments. These were blocked by LY294002, which is an Akt inhibitor. Finally, we found that the antidepressant effects of rubrofusarin were blocked by an intracerebroventricular injection of LY294002. These results suggest that rubrofusarin ameliorated CRS-induced depressive symptoms through PI3K/Akt signaling.


Subject(s)
Depression/drug therapy , Neurons/drug effects , Pyrones/pharmacology , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/pharmacology , Depression/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Neurons/pathology , Restraint, Physical/psychology , Signal Transduction/drug effects , Stress, Psychological/pathology
7.
Int J Mol Sci ; 21(24)2020 Dec 13.
Article in English | MEDLINE | ID: mdl-33322202

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by neurological dysfunction, including memory impairment, attributed to the accumulation of amyloid ß (Aß) in the brain. Although several studies reported possible mechanisms involved in Aß pathology, much remains unknown. Previous findings suggested that a protein regulated in development and DNA damage response 1 (REDD1), a stress-coping regulator, is an Aß-responsive gene involved in Aß cytotoxicity. However, we still do not know how Aß increases the level of REDD1 and whether REDD1 mediates Aß-induced synaptic dysfunction. To elucidate this, we examined the effect of Aß on REDD1-expression using acute hippocampal slices from mice, and the effect of REDD1 short hairpin RNA (shRNA) on Aß-induced synaptic dysfunction. Lastly, we observed the effect of REDD1 shRNA on memory deficit in an AD-like mouse model. Through the experiments, we found that Aß-incubated acute hippocampal slices showed increased REDD1 levels. Moreover, Aß injection into the lateral ventricle increased REDD1 levels in the hippocampus. Anisomycin, but not actinomycin D, blocked Aß-induced increase in REDD1 levels in the acute hippocampal slices, suggesting that Aß may increase REDD1 translation rather than transcription. Aß activated Fyn/ERK/S6 cascade, and inhibitors for Fyn/ERK/S6 or mGluR5 blocked Aß-induced REDD1 upregulation. REDD1 inducer, a transcriptional activator, and Aß blocked synaptic plasticity in the acute hippocampal slices. REDD1 inducer inhibited mTOR/Akt signaling. REDD1 shRNA blocked Aß-induced synaptic deficits. REDD1 shRNA also blocked Aß-induced memory deficits in passive-avoidance and object-recognition tests. Collectively, these results demonstrate that REDD1 participates in Aß pathology and could be a target for AD therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Memory Disorders/metabolism , Synapses/metabolism , Transcription Factors/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Anisomycin/pharmacology , Dactinomycin/pharmacology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Memory Disorders/genetics , Memory Disorders/pathology , Memory and Learning Tests , Mice , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , RNA, Small Interfering , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/metabolism , Synapses/drug effects , Synapses/genetics , Synapses/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Up-Regulation
8.
Pharmacol Res ; 121: 42-51, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28416463

ABSTRACT

Evidence suggests that the stress hormones glucocorticoids (GCs) can cause cognitive deficits and neurodegeneration. Previous studies have found GCs facilitate physiological synapse weakening, termed long-term depression (LTD), though the precise mechanisms underlying this are poorly understood. Here we show that GCs activate glycogen synthase kinase-3 (GSK-3), a kinase crucial to synapse weakening signals. Critically, this ultimately leads to phosphorylation of the microtubule associated protein tau, specifically at the serine 396 residue, and this is a causal factor in the GC-mediated impairment of synaptic function. These findings reveal the link between GCs and synapse weakening signals, and the potential for stress-induced priming of neurodegeneration. This could have important implications for our understanding of how stress can lead to neurodegenerative disease.


Subject(s)
Glucocorticoids/metabolism , Hippocampus/physiology , Long-Term Potentiation , Synapses/physiology , tau Proteins/metabolism , Animals , Glycogen Synthase Kinase 3/metabolism , Phosphorylation , Rats , Signal Transduction
9.
J Neurosci ; 35(12): 4804-12, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25810511

ABSTRACT

Tau is required for the induction of long-term depression (LTD) of synaptic transmission in the hippocampus. Here we probe the role of tau in LTD, finding that an AMPA receptor internalization mechanism is impaired in tau KO mice, and that LTD causes specific phosphorylation at the serine 396 and 404 residues of tau. Surprisingly, we find that phosphorylation at serine 396, specifically, is critical for LTD but has no role in LTP. Finally, we show that tau KO mice exhibit deficits in spatial reversal learning. These findings underscore the physiological role for tau at the synapse and identify a behavioral correlate of its role in LTD.


Subject(s)
Hippocampus/physiology , Long-Term Synaptic Depression/physiology , tau Proteins/physiology , Animals , Endocytosis/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Knockout , Phosphorylation , Rats , Receptors, AMPA/metabolism , Reversal Learning/physiology , Serine/metabolism , tau Proteins/genetics
10.
Pharmacol Res ; 104: 97-107, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687096

ABSTRACT

This study describes a fundamental functional difference between the two main polymorphisms of the pro-form of brain-derived neurotrophic factor (proBDNF), providing an explanation as to why these forms have such different age-related neurological outcomes. Healthy young carriers of the Met66 form (present in ∼30% Caucasians) have reduced hippocampal volume and impaired hippocampal-dependent memory function, yet the same polymorphic population shows enhanced cognitive recovery after traumatic brain injury, delayed cognitive dysfunction during aging, and lower risk of late-onset Alzheimer's disease (AD) compared to those with the more common Val66 polymorphism. To examine the differences between the protein polymorphisms in structure, kinetics of binding to proBDNF receptors and in vitro function, we generated purified cleavage-resistant human variants. Intriguingly, we found no statistical differences in those characteristics. As anticipated, exogenous application of proBDNF Val66 to rat hippocampal slices dysregulated synaptic plasticity, inhibiting long-term potentiation (LTP) and facilitating long-term depression (LTD). We subsequently observed that this occurred via the glycogen synthase kinase 3ß (GSK3ß) activation pathway. However, surprisingly, we found that Met66 had no such effects on either LTP or LTD. These novel findings suggest that, unlike Val66, the Met66 variant does not facilitate synapse weakening signaling, perhaps accounting for its protective effects with aging.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Protein Precursors/genetics , Synapses/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/physiology , Humans , L-Lactate Dehydrogenase/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Polymorphism, Genetic , Protein Precursors/metabolism , Rats, Wistar , Recombinant Proteins/pharmacology , Synapses/drug effects , tau Proteins/metabolism
11.
BMC Neurosci ; 15: 65, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24884833

ABSTRACT

BACKGROUND: Glucocorticoid released by stressful stimuli elicits various stress responses. Acute treatment with a single dose of corticosterone (CORT; predominant glucocorticoid of rats) alone has previously been shown to trigger anxiety behavior and robust dendritic hypertrophy of neurons in the basolateral amygdala (BLA). Neurons in the medial prefrontal cortex (mPFC) are also known to be highly sensitive to stress and regulate anxiety-like behaviors. Nevertheless, we know less about acute CORT-induced structural changes of other brain regions and their behavioral outcomes. In addition, the temporal profile of acute CORT effects remains to be examined. The current study investigates time course changes of dendritic architectures in the stress vulnerable brain areas, the BLA and mPFC, and their behavioral consequences after acute treatment with a single dose of CORT. RESULTS: Acute CORT treatment produced delayed onset of dendritic remodeling in the opposite direction in the BLA and mPFC with different time courses. Acute CORT induced dendritic hypertrophy of BLA spiny neurons, which was paralleled by heightened anxiety, both peaked 12 days after the treatment. Meanwhile, CORT-induced dendritic atrophy of mPFC pyramidal neurons peaked on day 6, concomitantly with impaired working memory. Both changed dendritic morphologies and altered behavioral outcomes were fully recovered. CONCLUSION: Our results suggest that stress-induced heightened anxiety appears to be a functional consequence of dendritic remodeling of BLA neurons but not that of mPFC. Instead, stress-induced dendritic atrophy of mPFC neurons is relevant to working memory deficit. Therefore, structural changes in the BLA and the mPFC might be specifically associated with distinct behavioral symptoms observed in stress-related mental disorders. Remarkably, stress-induced dendritic remodeling in the BLA as well as mPFC is readily reversible. The related behavioral outcomes also follow the similar time course in a reversible manner. Therefore, further studies on the cellular mechanism for the plasticity of dendrites architecture might provide new insight into the etiological factors for stress-related mental illness such as posttraumatic stress disorder (PTSD).


Subject(s)
Anxiety/chemically induced , Anxiety/physiopathology , Brain/pathology , Brain/physiopathology , Corticosterone/toxicity , Dendrites/drug effects , Dendrites/pathology , Animals , Anxiety/pathology , Behavior, Animal/drug effects , Brain/drug effects , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
12.
Biomed Pharmacother ; 161: 114511, 2023 May.
Article in English | MEDLINE | ID: mdl-36913892

ABSTRACT

Alzheimer's disease (AD) is a well-known neurodegenerative brain disease, and no curative treatment has yet been developed. The main symptoms include various brain lesions, caused by amyloid ß (Aß) aggregation, and cognitive decline. Therefore, it is believed that substances that control Aß will inhibit the onset of Alzheimer's disease and slow its progression. In this study, the effect of phyllodulcin, a major component of hydrangea, on Aß aggregation and brain pathology in an animal model of AD was studied. Phyllodulcin inhibited the aggregation of Aß and decomposed the pre-aggregated Aß in a concentration-dependent manner. In addition, it inhibited the cytotoxicity of Aß aggregates. Oral administration of phyllodulcin improved Aß-induced memory impairments in normal mice, reduced Aß deposition in the hippocampus, inhibited the activation of microglia and astrocytes, and improved synaptic plasticity in 5XFAD mice. These results suggest that phyllodulcin may be a candidate for the treatment of AD.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Long-Term Potentiation , Hippocampus , Mice, Transgenic , Disease Models, Animal
13.
Life Sci ; 328: 121901, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37391067

ABSTRACT

Stress is an inevitable part of life and, simultaneously, a stimulus that can trigger various neuropsychiatric disorders. Therefore, proper stress management is essential for maintaining a healthy life. In this study, we investigated the suppression of stress-induced cognitive deficit by controlling changes in synaptic plasticity caused by stress and confirmed that ethyl pyruvate (EP) has such an effect. Corticosterone, a stress hormone, suppresses long-term potentiation (LTP) in mouse acute hippocampal slices. EP blocked the LTP inhibitory effect of corticosterone by regulating GSK-3ß function. Restraint stress for 2 weeks increased the anxiety levels and caused the cognitive decline in the experimental animals. Administration of EP for 14 days did not affect the increase in anxiety caused by stress but improved cognitive decline caused by stress. In addition, the decrease in neurogenesis and synaptic function deficits in the hippocampus, which cause of cognitive decline due to stress, were improved by EP administration. These effects appear via regulation of Akt/GSK-3ß signaling, as in in vitro studies. These results suggest that EP prevents stress-induced cognitive decline through the modulation of Akt/GSK-3ß-mediated synaptic regulation.


Subject(s)
Cognitive Dysfunction , Proto-Oncogene Proteins c-akt , Mice , Animals , Glycogen Synthase Kinase 3 beta , Proto-Oncogene Proteins c-akt/metabolism , Corticosterone , Long-Term Potentiation , Hippocampus/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control
14.
Biochim Biophys Acta ; 1812(4): 536-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20832469

ABSTRACT

Hearing loss is a common communication disorder caused by various environmental and genetic factors. Hereditary hearing loss is very heterogeneous, and most of such cases involve sensorineural defects in the auditory pathway. There are currently 57 known autosomal dominant non-syndromic hearing loss (DFNA) loci, and the causative genes have been identified at 22 of these loci. In the present study, we performed a genome-wide linkage analysis in a Korean family segregating autosomal dominant hearing loss. We observed linkage on chromosome 1p34, and at this locus, we detected a novel mutation consisting of an 18 nucleotide deletion in exon 4 of the KCNQ4 gene, which encodes a voltage-gated potassium channel. We carried out a functional in vitro study to analyze the effects of this mutation (c.664_681del) along with two previously described KCNQ4 mutations, p.W276S and p.G285C. Although the c.664_681del mutation is located in the intercellular loop and the two previously described mutations, p.W276S and p.G285C, are located in the pore region, all mutants inhibit normal channel function by a dominant negative effect. Our analysis indicates that the intercellular loop is as significant as the pore region as a potential site of pathogenic effects on KCNQ4 channel function.


Subject(s)
Amino Acid Sequence , Hearing Loss/genetics , KCNQ Potassium Channels/genetics , Sequence Deletion , Cell Line , Genes, Dominant , Genetic Linkage , Haplotypes , Molecular Sequence Data , Pedigree
15.
Eur J Pharmacol ; 931: 175188, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35948162

ABSTRACT

Alzheimer's disease (AD) is the most common degenerative disease and is indicative of dementia. The cerebral accumulation of amyloid ß (Aß), a crucial factor in AD, initiates synaptic and cognitive dysfunction. Therefore, the elevation of synaptic and cognitive functions may help manage dementia in AD. In this study, we suggest hyperoside as a synaptic function- and memory-enhancing agent. Hyperoside enhanced learning and memory in passive avoidance and object recognition tasks. Hyperoside facilitated synaptic long-term potentiation (LTP) in acute hippocampal slices. IEM-1460, a calcium-permeable amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (CP-AMPAR) antagonist, blocked the facilitation effect of hyperoside. Hyperoside also induced N-methyl-d-aspartate receptor (NMDAR)-independent LTP, which was blocked by IEM-1460, suggesting the involvement of CP-AMPARs in the synaptic effects of hyperoside-mediated LTP. PKI (a PKA inhibitor) or SQ22536 (adenylyl cyclase, an AC inhibitor) blocked hyperoside-facilitated LTP and hyperoside-induced NMDAR-independent LTP. Hyperoside-enhanced learning and memory were blocked by IEM-1460, suggesting the involvement of CP-AMPARs in the effect of hyperoside on learning and memory. Finally, hyperoside ameliorated Aß-induced memory impairments in an AD mouse model. These results suggest that hyperoside enhances learning and memory, and this may be due to the effect of CP-AMPARs.


Subject(s)
Alzheimer Disease , Receptors, AMPA , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Animals , Calcium/metabolism , Hippocampus , Long-Term Potentiation , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Quercetin/analogs & derivatives , Receptors, AMPA/metabolism , Synapses
16.
Biochem Biophys Res Commun ; 404(4): 952-7, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21185265

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed in the central nervous system and play a regulatory role in neuronal excitability. In the present study, we examined a physiological role of HCN channels in the rat basolateral amygdala (BLA). In vitro electrophysiological studies showed that ZD7288 decreased spontaneous inhibitory postsynaptic current (sIPSC) without changing miniature IPSC (mIPSC). HCN channel blockade also attenuated feedback inhibitions in BLA principal neurons. However, blockade of HCN channel had little effects on spontaneous excitatory postsynaptic current (sEPSC) and mEPSC. Therefore, HCN channel appeared to decrease BLA excitability by increasing the action potential-dependent inhibitory control over the BLA principal neurons. Anxiety is reported to be influenced by neuronal excitability in the BLA and inhibitory synaptic transmission is thought to play a pivotal role in regulating overall excitability of the amygdala. As expected, blockade of HCN channels by targeted injection of ZD7288 to the BLA increased anxiety-like behavior under elevated plus maze test. Our results suggest that HCN channel activity can modulate the GABAergic synaptic transmission in the BLA, which in turn control the amygdala-related emotional behaviors such as anxiety.


Subject(s)
Amygdala/physiology , Anxiety/physiopathology , Cyclic Nucleotide-Gated Cation Channels/physiology , Potassium Channels/physiology , Synaptic Transmission/physiology , Amygdala/drug effects , Animals , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Maze Learning , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
17.
Brain Commun ; 2(2): fcaa058, 2020.
Article in English | MEDLINE | ID: mdl-32766549

ABSTRACT

Aggregation of amyloid beta and loss of cholinergic innervation in the brain are predominant components of Alzheimer's disease pathology and likely underlie cognitive impairment. Acetylcholinesterase inhibitors are one of the few treatment options for Alzheimer's disease, where levels of available acetylcholine are enhanced to counteract the cholinergic loss. However, these inhibitors show limited clinical efficacy. One potential explanation for this is a concomitant dysregulation of cholinergic receptors themselves as a consequence of the amyloid beta pathology. We tested this hypothesis by examining levels of M1 muscarinic acetylcholine receptors in the temporal cortex from seven Alzheimer's disease and seven non-disease age-matched control brain tissue samples (control: 85 ± 2.63 years old, moderate Alzheimer's disease: 84 ± 2.32 years old, P-value = 0.721; eight female and six male patients). The samples were categorized into two groups: 'control' (Consortium to Establish a Registry for Alzheimer's Disease diagnosis of 'No Alzheimer's disease', and Braak staging pathology of I-II) and 'moderate Alzheimer's disease' (Consortium to Establish a Registry for Alzheimer's Disease diagnosis of 'possible/probable Alzheimer's disease', and Braak staging pathology of IV). We find that in comparison to age-matched controls, there is a loss of M1 muscarinic acetylcholine receptors in moderate Alzheimer's disease tissue (control: 2.17 ± 0.27 arbitrary units, n = 7, Mod-AD: 0.83 ± 0.16 arbitrary units, n = 7, two-tailed t-test, t = 4.248, P = 0.00113). Using a functional rat cortical brain slice model, we find that postsynaptic muscarinic acetylcholine receptor function is dysregulated by aberrant amyloid beta-mediated activation of metabotropic glutamate receptor 5. Crucially, blocking metabotropic glutamate receptor 5 restores muscarinic acetylcholine receptor function and object recognition memory in 5XFAD transgenic mice. This indicates that the amyloid beta-mediated activation of metabotropic glutamate receptor 5 negatively regulates muscarinic acetylcholine receptor and illustrates the importance of muscarinic acetylcholine receptors as a potential disease-modifying target in the moderate pathological stages of Alzheimer's disease.

18.
Commun Biol ; 3(1): 79, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32071389

ABSTRACT

The molecular events causing memory loss and neuronal cell death in Alzheimer's disease (AD) over time are still unknown. Here we found that picomolar concentrations of soluble oligomers of synthetic beta amyloid (Aß42) aggregates incubated with BV2 cells or rat astrocytes caused a sensitised response of Toll-like receptor 4 (TLR4) with time, leading to increased production of TNF-α. Aß aggregates caused long term potentiation (LTP) deficit in hippocampal slices and predominantly neuronal cell death in co-cultures of astrocytes and neurons, which was blocked by TLR4 antagonists. Soluble Aß aggregates cause LTP deficit and neuronal death via an autocrine/paracrine mechanism due to TLR4 signalling. These findings suggest that the TLR4-mediated inflammatory response may be a key pathophysiological process in AD.


Subject(s)
Amyloid beta-Peptides/physiology , Neurons/physiology , Protein Aggregates/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Embryo, Mammalian , Long-Term Potentiation/drug effects , Male , Mice , Neurons/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/physiopathology , Protein Aggregation, Pathological/psychology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
19.
Food Chem Toxicol ; 136: 111011, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31790773

ABSTRACT

Neurite outgrowth is important process in synaptic formation and neuronal development. Many previous studies reported that natural compounds as well as neurotrophins induce neurite outgrowth through various signaling pathways. In this study, we tested the effect of cryptotanshinone (CPT), a constituent of Salvia miltiorrhiza Bunge, on neurite outgrowth using neuro2a cell line, a mouse neuroblastoma cell line. And then, we examined the effect of CPT on learning and memory. We first found that CPT facilitated neurite outgrowth in a concentration-dependent manner. Although CPT induced MTT reduction, CPT did not induce LDH release. Moreover, CPT suppressed cell proliferation. CPT increased ERK1/2 phosphorylation and ERK1/2 inhibitor blocked CPT-facilitated neurite outgrowth. CPT also enhanced learning and memory without affecting basal sensory conditions and increased ERK1/2 phosphorylation in the hippocampus in a dose-dependent manner. These results demonstrate that CPT facilitates neurite outgrowth and enhances learning and memory, which may be mediated by facilitating ERK1/2 signal.


Subject(s)
MAP Kinase Signaling System/drug effects , Memory/drug effects , Neuronal Outgrowth/drug effects , Phenanthrenes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Hippocampus/metabolism , Learning/drug effects , Male , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects
20.
Cell Death Dis ; 10(12): 943, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819047

ABSTRACT

In the current study we present evidence suggesting that PARP-1 regulates neurogenesis and its deficiency may result in schizophrenia-like behavioral deficits in mice. PARP-1 knockout neural stem cells exhibited a marked upregulation of embryonic stem cell phosphatase that can suppress the proliferative signaling of PI3K-Akt and ERK. The suppressed activity of Akt and ERK in the absence of PARP-1 results in the elevation of FOXO1 activity and its downstream target genes p21 and p27, leading to the inhibition of neural stem cell proliferation. Moreover, expression of neurogenic factors and neuronal differentiation were decreased in the PARP-1 knockout neural stem cells whereas glial differentiation was increased. In accordance with the in vitro data, PARP-1 knockout mice exhibited reduced brain weight with enlarged ventricle as well as decreased adult neurogenesis in the hippocampus. Interestingly, PARP-1 knockout mice exhibited schizophrenia-like symptoms such as anxiety, depression, social interaction deficits, cognitive impairments, and prepulse inhibition deficits. Taken together, our results suggest that PARP-1 regulates neurogenesis during development and in adult and its absence may lead to the schizophrenia-like behavioral abnormality in mice.


Subject(s)
Behavior, Animal , Neurogenesis/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Schizophrenia/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Hippocampus/metabolism , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL