Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 13: 552787, 2020.
Article in English | MEDLINE | ID: mdl-33192290

ABSTRACT

Although numerous studies have indicated that chronic stress causes cognitive dysfunction with the impairment of synaptic structures and functions, the relationship between cognitive deficits induced by repeated restraint stress and the level of NMDA receptors in the subregion of the hippocampus has been relatively unknown until now. In this study, 3-week-old male Sprague-Dawley rats were exposed to repeated restraint stress for seven consecutive days, their cognitive functions were evaluated through behavioral tests, and then they were sacrificed for electrophysiological, morphological, and biochemical assays. Chronic repeated restraint stress led to cognitive and electrophysiological impairments, with a reduced density of dendritic spines. We also found that the protein level of NMDA receptors only increased in the hippocampal CA3 region. Nevertheless, repeated restraint stress-induced cognitive and synaptic dysfunction were effectively reversed by Ro25-6981, an inhibitor of the GluN2B receptor. These findings suggest that repeated restraint stress-induced synaptic and cognitive deficits are probably mediated through NMDA receptors.

2.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1477-1489, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30826466

ABSTRACT

BACKGROUND: Maternal immune activation (MIA) is an independent risk factor for psychiatric disorders including depression spectrum in the offsprings, but the molecular mechanism is unclear. Recent studies show that interferon-stimulated gene-15 (ISG15) is involved in inflammation and neuronal dendrite development; here we studied the role of ISG15 in MIA-induced depression and the underlying mechanisms. METHODS: By vena caudalis injecting polyinosinic: polycytidylic acid (poly I:C) into the pregnant rats to mimic MIA, we used AAV or lentivirus to introduce or silence ISG15 expression. Synaptic plasticity was detected by confocal microscope and Golgi staining. Cognitive performances of the offspring were measured by Open field, Forced swimming and Sucrose preference test. RESULTS: We found that MIA induced depression-like behaviors with dendrite impairments in the offspring with ISG15 level increased in the offsprings' brain. Overexpressing ISG15 in the prefrontal cortex of neonatal cubs (P0) could mimic dendritic pathology and depressive like behaviors, while downregulating ISG15 rescued these abnormalities in the offsprings. Further studies demonstrated that MIA-induced upregulation of inflammatory cytokines promoted ISG15 expression in the offspring' brain which suppressed Rap2A ubiquitination via NEDD4 and thus induced Rap2A accumulation, while upregulating NEDD4 abolished ISG15-induced dendrite impairments. CONCLUSIONS: These data reveal that MIA impedes offsprings' dendrite development and causes depressive like behaviors by upregulating ISG15 and suppressing NEDD4/Rap2A signaling. The current findings suggest that inhibiting ISG15 may be a promising intervention of MIA-induced psychiatric disorders in the offsprings.


Subject(s)
Cytokines/genetics , Dendrites/metabolism , GTP-Binding Proteins/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Prenatal Exposure Delayed Effects/genetics , Ubiquitins/genetics , Animals , Behavior Rating Scale , Cytokines/antagonists & inhibitors , Cytokines/immunology , Dendrites/immunology , Dendrites/pathology , Depression , Disease Models, Animal , Female , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/immunology , Gene Expression Regulation , Immunity, Innate/drug effects , Inflammation , Injections, Intravenous , Nedd4 Ubiquitin Protein Ligases/antagonists & inhibitors , Nedd4 Ubiquitin Protein Ligases/immunology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurogenesis/immunology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/immunology , Poly I-C/administration & dosage , Prefrontal Cortex/immunology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Ubiquitins/antagonists & inhibitors , Ubiquitins/immunology
3.
J Alzheimers Dis ; 63(4): 1537-1546, 2018.
Article in English | MEDLINE | ID: mdl-29782322

ABSTRACT

There is accumulating evidence that decreased histone acetylation is involved in normal aging and neurodegenerative diseases. Recently, we found that ANP32A, a key component of INHAT (inhibitor of acetyltransferases) that suppresses histone acetylation, increased in aged and cognitively impaired C57 mice and expressing wild-type human full length tau (htau) transgenic mice. Downregulating ANP32A restored cognitive function and synaptic plasticity through upregulation of the expressions of synaptic-related proteins via increasing histone acetylation. However, there is no direct evidence that ANP32A can induce neurodegeneration and memory deficits. In the present study, we overexpressed ANP32A in the hippocampal CA3 region of C57 mice and found that ANP32A overexpression induced cognitive abilities and synaptic plasticity deficits, with decreased synaptic-related protein expression and histone acetylation. Combined with our recent studies, our findings reveal that upregulated ANP32A induced-suppressing histone acetylation may underlie the cognitive decline in neurodegenerative disease, and suppression of ANP32A may represent a promising therapeutic approach for neurodegenerative diseases including Alzheimer's disease.


Subject(s)
Histones/metabolism , Memory Disorders/enzymology , Memory Disorders/genetics , Nuclear Proteins/metabolism , Up-Regulation/genetics , Acetylation , Age Factors , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Dependovirus/genetics , Disease Models, Animal , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Maze Learning/physiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , RNA-Binding Proteins , Receptors, Glutamate/metabolism , Synapsins/metabolism , Synaptophysin/metabolism , Transduction, Genetic
4.
J Huazhong Univ Sci Technolog Med Sci ; 37(3): 307-312, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28585125

ABSTRACT

As a major microtubule-associated protein, tau plays an important role in promoting microtubule assembly and stabilizing microtubules. In Alzheimer's disease (AD) and other tauopathies, the abnormally hyperphosphorylated tau proteins are aggregated into paired helical filaments and accumulated in the neurons with the form of neurofibrillary tangles. An imbalanced regulation in protein kinases and protein phosphatases is the direct cause of tau hyperphosphorylation. Among various kinases and phosphatases, glycogen synthase kinase-3ß (GSK-3ß) and protein phosphatase 2A (PP2A) are the most implicated. Accumulation of the hyperphosphorylated tau induces synaptic toxicity and cognitive impairments. Here, we review the upstream factors or pathways that can regulate GSK-3ß or PP2A activity mainly based on our recent findings. We will also discuss the mechanisms that may underlie tau-induced synaptic toxicity.


Subject(s)
Alzheimer Disease/metabolism , Cognitive Dysfunction/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Nerve Degeneration/metabolism , Protein Phosphatase 2/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/genetics , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Phosphatase 2/genetics , Signal Transduction , Synapses/metabolism , Synapses/pathology , tau Proteins/genetics
5.
Front Aging Neurosci ; 9: 104, 2017.
Article in English | MEDLINE | ID: mdl-28473768

ABSTRACT

Aging is a cause of cognitive decline in the elderly and the major risk factor for Alzheimer's disease, however, aging people are not all destined to develop into cognitive deficits, the molecular mechanisms underlying this difference in cognition of aging people are obscure. Epigenetic modifications, particularly histone acetylation in the nervous system, play a critical role in regulation of gene expression for learning and memory. An inhibitor of acetyltransferases (INHAT) is reported to suppress histone acetylation via a histone-masking mechanism, and pp32 is a key component of INHAT complex. In the present study, we divided ~18 m-old aged mice into the cognitive-normal and the cognitive-impaired group by Morris water maze, and found that pp32 level was significantly increased in the hippocampus of cognitive-impaired aged mice. The mRNA and protein levels of synaptic-associated proteins decreased with reduced dendrite complexity and histone acetylation. Knockdown of pp32 rescued cognitive decline in cognitive-impaired aged mice with restoration of synaptic-associated proteins, the increase of spine density and elevation of histone acetylation. Our study reveals a novel mechanism underlying the aging-associated cognitive disturbance, indicating that suppression of pp32 might represent a promising therapeutic approach for learning and memory impairments.

6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-238364

ABSTRACT

As a major microtubule-associated protein,tau plays an important role in promoting microtubule assembly and stabilizing microtubules.In Alzheimer's disease (AD) and other tauopathies,the abnormally hyperphosphorylated tau proteins are aggregated into paired helical filaments and accumulated in the neurons with the form of neurofibrillary tangles.An imbalanced regulation in protein kinases and protein phosphatases is the direct cause of tau hyperphosphorylation.Among various kinases and phosphatases,glycogen synthase kinase-3β (GSK-3β) and protein phosphatase 2A (PP2A) are the most implicated.Accumulation of the hyperphosphorylated tau induces synaptic toxicity and cognitive impairments.Here,we review the upstream factors or pathways that can regulate GSK-3β or PP2A activity mainly based on our recent findings.We will also discuss the mechanisms that may underlie tau-induced synaptic toxicity.

7.
Neurobiol Aging ; 45: 64-75, 2016 09.
Article in English | MEDLINE | ID: mdl-27459927

ABSTRACT

Neurogenesis plays a role in hippocampus-dependent learning and impaired neurogenesis may correlate with cognitive deficits in Alzheimer's disease. Spatial training influences the production and fate of newborn cells in hippocampus of normal animals, whereas the effects on neurogenesis in Alzheimer-like animal are not reported until now. Here, for the first time, we investigated the effect of Morris water maze training on proliferation, survival, apoptosis, migration, and differentiation of newborn cells in ß-amyloid-treated Alzheimer-like rats. We found that spatial training could preserve a short-term survival of newborn cells generated before training, during the early phase, and the late phase of training. However, the training had no effect on the long-term survival of mature newborn cells generated at previously mentioned 3 different phases. We also demonstrated that spatial training promoted newborn cell differentiation preferentially to the neuron direction. These findings suggest a time-independent neurogenesis induced by spatial training, which may be indicative for the cognitive stimulation in Alzheimer's disease therapy.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/administration & dosage , Cell Differentiation , Cognition/physiology , Hippocampus/cytology , Maze Learning/physiology , Neurogenesis/physiology , Peptide Fragments/administration & dosage , Spatial Navigation/physiology , Animals , Cell Survival , Disease Models, Animal , Hippocampus/physiology , Male , Rats, Sprague-Dawley , Time Factors
8.
Neurosci Bull ; 30(6): 923-935, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25391447

ABSTRACT

Amyloid ß-peptide (Aß) has been implicated as a key molecule in the neurodegenerative cascades of Alzheimer's disease (AD). Humanin (HN) is a secretory peptide that inhibits the neurotoxicity of Aß. However, the mechanism(s) by which HN exerts its neuroprotection against Aß-induced AD-like pathological changes and memory deficits are yet to be completely defined. In the present study, we provided evidence that treatment of rats with HN increases the number of dendritic branches and the density of dendritic spines, and upregulates pre- and post-synaptic protein levels; these effects lead to enhanced long-term potentiation and amelioration of the memory deficits induced by Aß(1-42). HN also attenuated Aß(1-42)-induced tau hyperphosphorylation, apparently by inhibiting the phosphorylation of Tyr307 on the inhibitory protein phosphatase-2A (PP2A) catalytic subunit and thereby activating PP2A. HN also inhibited apoptosis and reduced the oxidative stress induced by Aß(1-42). These findings provide novel mechanisms of action for the ability of HN to protect against Aß(1-42)-induced AD-like pathological changes and memory deficits.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Cognition Disorders/drug therapy , Dendrites/drug effects , Intracellular Signaling Peptides and Proteins/therapeutic use , Maze Learning/drug effects , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Brain/pathology , Cognition/drug effects , Cognition Disorders/pathology , Cognition Disorders/psychology , Dendrites/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Rats , Rats, Wistar
9.
Front Aging Neurosci ; 6: 123, 2014.
Article in English | MEDLINE | ID: mdl-24987368

ABSTRACT

Increase of inhibitor-2 of protein phosphatase-2A [Formula: see text] is associated with protein phosphatase-2A (PP2A) inhibition and tau hyperphosphorylation in Alzheimer's disease (AD). Down-regulating [Formula: see text] attenuated amyloidogenesis and improved the cognitive functions in transgenic mice expressing amyloid precursor protein (tg2576). Here, we found that silencing [Formula: see text] by hippocampal infusion of [Formula: see text] down-regulated [Formula: see text] (~45%) with reduction of tau phosphorylation/accumulation, improvement of memory deficits, and dendritic plasticity in 12-month-old human tau transgenic mice. Silencing [Formula: see text] not only restored PP2A activity but also inhibited glycogen synthase kinase-3ß (GSK-3ß) with a significant activation of protein kinase A (PKA) and Akt. In HEK293/tau and N2a/tau cells, silencing [Formula: see text] by [Formula: see text] also significantly reduced tau hyperphosphorylation with restoration of PP2A activity and inhibition of GSK-3ß, demonstrated by the decreased GSK-3ß total protein and mRNA levels, and the increased inhibitory phosphorylation of GSK-3ß at serine-9. Furthermore, activation of PKA but not Akt mediated the inhibition of GSK-3ß by [Formula: see text] silencing. We conclude that targeting [Formula: see text] can improve tau pathologies and memory deficits in human tau transgenic mice, and activation of PKA contributes to GSK-3ß inhibition induced by silencing [Formula: see text]in vitro, suggesting that [Formula: see text] is a promising multiple target of AD.

SELECTION OF CITATIONS
SEARCH DETAIL
...