Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Neuron ; 111(20): 3133-3135, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37857086

ABSTRACT

In this issue of Neuron, Essayan-Perez and Südhof1 demonstrate roles for γ-secretase in the regulation of synaptic functions in human neurons. Chronic attenuation of γ-secretase activity increases synapse formation but decreases neurotransmission (i.e., the probability of presynaptic release), likely due to impairment of cholesterol metabolism.


Subject(s)
Amyloid Precursor Protein Secretases , Neurons , Humans , Amyloid Precursor Protein Secretases/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Homeostasis , Cholesterol/metabolism , Synapses/metabolism
2.
Semin Cell Dev Biol ; 139: 3-12, 2023 04.
Article in English | MEDLINE | ID: mdl-35918217

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by the progressive deterioration of cognitive functions. Due to the extended global life expectancy, the prevalence of AD is increasing among aging populations worldwide. While AD is a multifactorial disease, synaptic dysfunction is one of the major neuropathological changes that occur early in AD, before clinical symptoms appear, and is associated with the progression of cognitive deterioration. However, the underlying pathological mechanisms leading to this synaptic dysfunction remains unclear. Recent large-scale genomic analyses have identified more than 40 genetic risk factors that are associated with AD. In this review, we discuss the functional roles of these genes in synaptogenesis and synaptic functions under physiological conditions, and how their functions are dysregulated in AD. This will provide insights into the contributions of these encoded proteins to synaptic dysfunction during AD pathogenesis.


Subject(s)
Alzheimer Disease , Cognition Disorders , Neurodegenerative Diseases , Humans , Alzheimer Disease/metabolism , Synapses/genetics , Synapses/metabolism , Neurodegenerative Diseases/metabolism , Cognition Disorders/pathology , Risk Factors
3.
Nat Aging ; 2(7): 616-634, 2022 07.
Article in English | MEDLINE | ID: mdl-37117777

ABSTRACT

Changes in the levels of circulating proteins are associated with Alzheimer's disease (AD), whereas their pathogenic roles in AD are unclear. Here, we identified soluble ST2 (sST2), a decoy receptor of interleukin-33-ST2 signaling, as a new disease-causing factor in AD. Increased circulating sST2 level is associated with more severe pathological changes in female individuals with AD. Genome-wide association analysis and CRISPR-Cas9 genome editing identified rs1921622 , a genetic variant in an enhancer element of IL1RL1, which downregulates gene and protein levels of sST2. Mendelian randomization analysis using genetic variants, including rs1921622 , demonstrated that decreased sST2 levels lower AD risk and related endophenotypes in females carrying the Apolipoprotein E (APOE)-ε4 genotype; the association is stronger in Chinese than in European-descent populations. Human and mouse transcriptome and immunohistochemical studies showed that rs1921622 /sST2 regulates amyloid-beta (Aß) pathology through the modulation of microglial activation and Aß clearance. These findings demonstrate how sST2 level is modulated by a genetic variation and plays a disease-causing role in females with AD.


Subject(s)
Alzheimer Disease , Humans , Female , Animals , Mice , Alzheimer Disease/genetics , Interleukin-1 Receptor-Like 1 Protein/genetics , Genome-Wide Association Study , Apolipoprotein E4/genetics , Amyloid beta-Peptides/genetics
4.
ACS Chem Neurosci ; 12(22): 4249-4256, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34738783

ABSTRACT

Alzheimer's disease (AD), the most common neurodegenerative disease, has limited treatment options. As such, extensive studies have been conducted to identify novel therapeutic approaches. We previously reported that rhynchophylline (Rhy), a small molecule EphA4 inhibitor, rescues impaired hippocampal synaptic plasticity and cognitive dysfunctions in APP/PS1 mice, an AD transgenic mouse model. To assess whether Rhy can be developed as an alternative treatment for AD, it is important to examine its pharmacokinetics and effects on other disease-associated pathologies. Here, we show that Rhy ameliorates amyloid plaque burden and reduces inflammation in APP/PS1 mice. Transcriptome analysis revealed that Rhy regulates various molecular pathways in APP/PS1 mouse brains associated with amyloid metabolism and inflammation, specifically the ubiquitin proteasome system, angiogenesis, and microglial functional states. These results show that Rhy, which is blood-brain barrier permeable, is beneficial to amyloid pathology and regulates multiple molecular pathways.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxindoles , Plaque, Amyloid/drug therapy , Presenilin-1/genetics
5.
Sci Rep ; 11(1): 4359, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33623128

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder with no disease-modifying treatment. AD progression is characterized by cognitive decline, neuroinflammation, and accumulation of amyloid-beta (Aß) and neurofibrillary tangles in the brain, leading to neuronal and glial dysfunctions. Neuropeptides govern diverse pathophysiological processes and represent key players in AD pathogenesis, regulating synaptic plasticity, glial cell functions and amyloid pathology. Activation of the pro-opiomelanocortin (POMC)-derived neuropeptide and its receptor from the melanocortin receptor (MCR) family have previously been shown to rescue the impairment in hippocampus-dependent synaptic plasticity in the APP/PS1 mouse model of AD. However, the functional roles of MCR signaling in AD conditions, particularly in glial functions, are largely unknown. In this study, we investigated the potential benefits of MCR activation in AD. In APP/PS1 transgenic mice, we demonstrate that MCR activation mediated by the central administration of its agonist D-Tyr MTII substantially reduces Aß accumulation, while alleviating global inflammation and astrocytic activation, particularly in the hippocampus. MCR activation prominently reduces the A1 subtype of reactive astrocytes, which is considered a key source of astrocytic neurotoxicity in AD. Concordantly, MCR activation suppresses microglial activation, while enhancing their association with amyloid plaques. The blunted activation of microglia may contribute to the reduction in the neurotoxic phenotypes of astrocytes. Importantly, transcriptome analysis reveals that MCR activation restores the impaired homeostatic processes and microglial reactivity in the hippocampus in APP/PS1 mice. Collectively, our findings demonstrate the potential of MCR signaling as therapeutic target for AD.


Subject(s)
Alzheimer Disease/drug therapy , Astrocytes/metabolism , Receptors, Melanocortin/agonists , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Inbred C57BL , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptides, Cyclic/chemistry , Receptors, Melanocortin/metabolism , Tyrosine/analogs & derivatives , alpha-MSH/analogs & derivatives , alpha-MSH/chemistry
6.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443211

ABSTRACT

Hippocampal synaptic plasticity is important for learning and memory formation. Homeostatic synaptic plasticity is a specific form of synaptic plasticity that is induced upon prolonged changes in neuronal activity to maintain network homeostasis. While astrocytes are important regulators of synaptic transmission and plasticity, it is largely unclear how they interact with neurons to regulate synaptic plasticity at the circuit level. Here, we show that neuronal activity blockade selectively increases the expression and secretion of IL-33 (interleukin-33) by astrocytes in the hippocampal cornu ammonis 1 (CA1) subregion. This IL-33 stimulates an increase in excitatory synapses and neurotransmission through the activation of neuronal IL-33 receptor complex and synaptic recruitment of the scaffold protein PSD-95. We found that acute administration of tetrodotoxin in hippocampal slices or inhibition of hippocampal CA1 excitatory neurons by optogenetic manipulation increases IL-33 expression in CA1 astrocytes. Furthermore, IL-33 administration in vivo promotes the formation of functional excitatory synapses in hippocampal CA1 neurons, whereas conditional knockout of IL-33 in CA1 astrocytes decreases the number of excitatory synapses therein. Importantly, blockade of IL-33 and its receptor signaling in vivo by intracerebroventricular administration of its decoy receptor inhibits homeostatic synaptic plasticity in CA1 pyramidal neurons and impairs spatial memory formation in mice. These results collectively reveal an important role of astrocytic IL-33 in mediating the negative-feedback signaling mechanism in homeostatic synaptic plasticity, providing insights into how astrocytes maintain hippocampal network homeostasis.


Subject(s)
Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , Interleukin-33/metabolism , Neuronal Plasticity , Signal Transduction/drug effects , Spatial Memory/drug effects , Animals , Astrocytes/drug effects , Disks Large Homolog 4 Protein/metabolism , Gene Knockout Techniques , Hippocampus/metabolism , Homeostasis , Interleukin-33/administration & dosage , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Synapses/drug effects , Synapses/genetics , Synapses/metabolism , Synaptic Transmission/drug effects , Tetrodotoxin/pharmacology
7.
Sci Rep ; 10(1): 18746, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127972

ABSTRACT

Dendrites, branched structures extending from neuronal cell soma, are specialized for processing information from other neurons. The morphogenesis of dendritic structures is spatiotemporally regulated by well-orchestrated signaling cascades. Dysregulation of these processes impacts the wiring of neuronal circuit and efficacy of neurotransmission, which contribute to the pathogeneses of neurological disorders. While Cdk5 (cyclin-dependent kinase 5) plays a critical role in neuronal dendritic development, its underlying molecular control is not fully understood. In this study, we show that p39, one of the two neuronal Cdk5 activators, is a key regulator of dendritic morphogenesis. Pyramidal neurons deficient in p39 exhibit aberrant dendritic morphology characterized by shorter length and reduced arborization, which is comparable to dendrites in Cdk5-deficient neurons. RNA sequencing analysis shows that the adaptor protein, WDFY1 (WD repeat and FYVE domain-containing 1), acts downstream of Cdk5/p39 to regulate dendritic morphogenesis. While WDFY1 is elevated in p39-deficient neurons, suppressing its expression rescues the impaired dendritic arborization. Further phosphoproteomic analysis suggests that Cdk5/p39 mediates dendritic morphogenesis by modulating various downstream signaling pathways, including PI3K/Akt-, cAMP-, or small GTPase-mediated signaling transduction pathways, thereby regulating cytoskeletal organization, protein synthesis, and protein trafficking.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Lipid-Linked Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cyclic AMP/metabolism , Cyclin-Dependent Kinase 5/genetics , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Lipid-Linked Proteins/genetics , Mass Spectrometry , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Nervous System/cytology , Nervous System/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
8.
Cell Rep ; 31(3): 107530, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320664

ABSTRACT

Impairment of microglial clearance activity contributes to beta-amyloid (Aß) pathology in Alzheimer's disease (AD). While the transcriptome profile of microglia directs microglial functions, how the microglial transcriptome can be regulated to alleviate AD pathology is largely unknown. Here, we show that injection of interleukin (IL)-33 in an AD transgenic mouse model ameliorates Aß pathology by reprogramming microglial epigenetic and transcriptomic profiles to induce a microglial subpopulation with enhanced phagocytic activity. These IL-33-responsive microglia (IL-33RMs) express a distinct transcriptome signature that is highlighted by increased major histocompatibility complex class II genes and restored homeostatic signature genes. IL-33-induced remodeling of chromatin accessibility and PU.1 transcription factor binding at the signature genes of IL-33RM control their transcriptome reprogramming. Specifically, disrupting PU.1-DNA interaction abolishes the microglial state transition and Aß clearance that is induced by IL-33. Thus, we define a PU.1-dependent transcriptional pathway that drives the IL-33-induced functional state transition of microglia, resulting in enhanced Aß clearance.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Interleukin-33/pharmacology , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Disease Models, Animal , Female , Humans , Interleukin-33/genetics , Male , Mice , Mice, Transgenic , Microglia/pathology , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/pharmacology , Trans-Activators/metabolism , Transcriptome/drug effects
9.
Sci Rep ; 9(1): 1190, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718786

ABSTRACT

Major depressive disorders are emerging health problems that affect millions of people worldwide. However, treatment options and targets for drug development are limited. Impaired adult hippocampal neurogenesis is emerging as a key contributor to the pathology of major depressive disorders. We previously demonstrated that increasing the expression of the multifunctional scaffold protein Axis inhibition protein (Axin) by administration of the small molecule XAV939 enhances embryonic neurogenesis and affects social interaction behaviors. This prompted us to examine whether increasing Axin protein level can enhance adult hippocampal neurogenesis and thus contribute to mood regulation. Here, we report that stabilizing Axin increases adult hippocampal neurogenesis and exerts an antidepressant effect. Specifically, treating adult mice with XAV939 increased the amplification of adult neural progenitor cells and neuron production in the hippocampus under both normal and chronic stress conditions. Furthermore, XAV939 injection in mice ameliorated depression-like behaviors induced by chronic restraint stress. Thus, our study demonstrates that Axin/XAV939 plays an important role in adult hippocampal neurogenesis and provides a potential therapeutic approach for mood-related disorders.


Subject(s)
Axin Protein/metabolism , Depression/metabolism , Neurogenesis/drug effects , Animals , Antidepressive Agents/pharmacology , Axin Protein/genetics , Brain/metabolism , Cell Differentiation/drug effects , Depression/pathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Disease Models, Animal , Heterocyclic Compounds, 3-Ring/pharmacology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Stress, Psychological
10.
ACS Chem Neurosci ; 10(2): 872-879, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30221933

ABSTRACT

Alzheimer's disease is a progressive neurodegenerative disease, and its incidence is expected to increase owing to the aging population worldwide. Current therapies merely provide symptomatic relief. Therefore, interventions for AD that delay the disease onset or progression are urgently required. Recent genomics and functional studies suggest that immune/inflammatory pathways are involved in the pathogenesis of AD. Although many anti-inflammatory drug candidates have undergone clinical trials, most have failed. This might be because of our limited understanding of the pathological mechanisms of neuroinflammation in AD. However, recent advances in the understanding of immune/inflammatory pathways in AD and their regulatory mechanisms could open up new avenues for drug development targeting neuroinflammation. In this Review, we discuss the mechanisms and status of different anti-inflammatory drug candidates for AD that have undergone or are undergoing clinical trials and explore new opportunities for targeting neuroinflammation in AD drug development.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Anti-Inflammatory Agents/administration & dosage , Drug Delivery Systems/trends , Drug Discovery/trends , Inflammation Mediators/metabolism , Alzheimer Disease/immunology , Animals , Drug Delivery Systems/methods , Drug Discovery/methods , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology
11.
Sci Rep ; 8(1): 7377, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29743517

ABSTRACT

The receptor tyrosine kinase, erythropoietin-producing hepatocellular A4 (EphA4), was recently identified as a molecular target for Alzheimer's disease (AD). We found that blockade of the interaction of the receptor and its ligands, ephrins, alleviates the disease phenotype in an AD transgenic mouse model, suggesting that targeting EphA4 is a potential approach for developing AD interventions. In this study, we identified five FDA-approved drugs-ergoloid, cyproheptadine, nilotinib, abiraterone, and retapamulin-as potential inhibitors of EphA4 by using an integrated approach combining virtual screening with biochemical and cellular assays. We initially screened a database of FDA-approved drugs using molecular docking against the ligand-binding domain of EphA4. Then, we selected 22 candidate drugs and examined their inhibitory activity towards EphA4. Among them, five drugs inhibited EphA4 clustering induced by ephrin-A in cultured primary neurons. Specifically, nilotinib, a kinase inhibitor, inhibited the binding of EphA4 and ephrin-A at micromolar scale in a dosage-dependent manner. Furthermore, nilotinib inhibited the activation of EphA4 and EphA4-dependent growth cone collapse in cultured hippocampal neurons, demonstrating that the drug exhibits EphA4 inhibitory activity in cellular context. As demonstrated in our combined computational and experimental approaches, repurposing of FDA-approved drugs to inhibit EphA4 may provide an alternative fast-track approach for identifying and developing new treatments for AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Evaluation, Preclinical , Molecular Docking Simulation , Pyrimidines/pharmacology , Receptor, EphA4/antagonists & inhibitors , Alzheimer Disease/metabolism , Androstenes/metabolism , Androstenes/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyproheptadine/metabolism , Cyproheptadine/pharmacology , Disease Models, Animal , Diterpenes/metabolism , Diterpenes/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Ligands , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Protein Binding , Protein Domains , Pyrimidines/metabolism , Receptor, EphA4/metabolism
12.
Proc Natl Acad Sci U S A ; 113(19): E2705-13, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27091974

ABSTRACT

Alzheimer's disease (AD) is a devastating condition with no known effective treatment. AD is characterized by memory loss as well as impaired locomotor ability, reasoning, and judgment. Emerging evidence suggests that the innate immune response plays a major role in the pathogenesis of AD. In AD, the accumulation of ß-amyloid (Aß) in the brain perturbs physiological functions of the brain, including synaptic and neuronal dysfunction, microglial activation, and neuronal loss. Serum levels of soluble ST2 (sST2), a decoy receptor for interleukin (IL)-33, increase in patients with mild cognitive impairment, suggesting that impaired IL-33/ST2 signaling may contribute to the pathogenesis of AD. Therefore, we investigated the potential therapeutic role of IL-33 in AD, using transgenic mouse models. Here we report that IL-33 administration reverses synaptic plasticity impairment and memory deficits in APP/PS1 mice. IL-33 administration reduces soluble Aß levels and amyloid plaque deposition by promoting the recruitment and Aß phagocytic activity of microglia; this is mediated by ST2/p38 signaling activation. Furthermore, IL-33 injection modulates the innate immune response by polarizing microglia/macrophages toward an antiinflammatory phenotype and reducing the expression of proinflammatory genes, including IL-1ß, IL-6, and NLRP3, in the cortices of APP/PS1 mice. Collectively, our results demonstrate a potential therapeutic role for IL-33 in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Brain/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Interleukin-33/administration & dosage , Alzheimer Disease/diagnosis , Animals , Brain/drug effects , Cognition Disorders/diagnosis , Cytokines/metabolism , Female , Male , Mice , Mice, Transgenic , Neuroprotective Agents/administration & dosage , Treatment Outcome
13.
Nat Commun ; 6: 8665, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26503494

ABSTRACT

Precise regulation of synaptic strength requires coordinated activity and functions of synaptic proteins, which is controlled by a variety of post-translational modification. Here we report that S-nitrosylation of p35, the activator of cyclin-dependent kinase 5 (Cdk5), by nitric oxide (NO) is important for the regulation of excitatory synaptic strength. While blockade of NO signalling results in structural and functional synaptic deficits as indicated by reduced mature dendritic spine density and surface expression of glutamate receptor subunits, phosphorylation of numerous synaptic substrates of Cdk5 and its activity are aberrantly upregulated following reduced NO production. The results show that the NO-induced reduction in Cdk5 activity is mediated by S-nitrosylation of p35, resulting in its ubiquitination and degradation by the E3 ligase PJA2. Silencing p35 protein in hippocampal neurons partially rescues the NO blockade-induced synaptic deficits. These findings collectively demonstrate that p35 S-nitrosylation by NO signalling is critical for regulating hippocampal synaptic strength.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Hippocampus/enzymology , Proteasome Endopeptidase Complex/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Hippocampus/metabolism , Mice , Neurons/enzymology , Neurons/metabolism , Nitric Oxide/metabolism , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Processing, Post-Translational , Proteolysis , Rats , Synaptic Transmission
14.
PLoS One ; 10(7): e0133115, 2015.
Article in English | MEDLINE | ID: mdl-26204446

ABSTRACT

During development, scaffold proteins serve as important platforms for orchestrating signaling complexes to transduce extracellular stimuli into intracellular responses that regulate dendritic spine morphology and function. Axin ("axis inhibitor") is a key scaffold protein in canonical Wnt signaling that interacts with specific synaptic proteins. However, the cellular functions of these protein-protein interactions in dendritic spine morphology and synaptic regulation are unclear. Here, we report that Axin protein is enriched in synaptic fractions, colocalizes with the postsynaptic marker PSD-95 in cultured hippocampal neurons, and interacts with a signaling protein Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in synaptosomal fractions. Axin depletion by shRNA in cultured neurons or intact hippocampal CA1 regions significantly reduced dendritic spine density. Intriguingly, the defective dendritic spine morphogenesis in Axin-knockdown neurons could be restored by overexpression of the small Rho-GTPase Cdc42, whose activity is regulated by CaMKII. Moreover, pharmacological stabilization of Axin resulted in increased dendritic spine number and spontaneous neurotransmission, while Axin stabilization in hippocampal neurons reduced the elimination of dendritic spines. Taken together, our findings suggest that Axin promotes dendritic spine stabilization through Cdc42-dependent cytoskeletal reorganization.


Subject(s)
Axin Protein/physiology , Dendritic Spines/ultrastructure , Signal Transduction/physiology , cdc42 GTP-Binding Protein/physiology , Animals , Axin Protein/genetics , CA1 Region, Hippocampal/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Nucleus/chemistry , Cells, Cultured , Cytosol/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Mice , Morphogenesis , Neurogenesis , Post-Synaptic Density/chemistry , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptosomes/metabolism
15.
Neuropsychopharmacology ; 40(8): 1877-87, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25649278

ABSTRACT

Compounds that have the ability to both strengthen synaptic function and facilitate neuroprotection are valuable cognitive enhancers that may improve health and quality of life, as well as retard age-related cognitive deterioration. Medicinal plants are an abundant source of potential cognitive enhancers. Here we report that anemoside A3 (AA3) isolated from Pulsatilla chinensis modulates synaptic connectivity in circuits central to memory enhancement. AA3 specifically modulates the function of AMPA-type glutamate receptors (AMPARs) by increasing serine phosphorylation within the GluA1 subunit, which is a modification required for the trafficking of GluA1-containing AMPARs to synapses. Furthermore, AA3 administration activates several synaptic signaling molecules and increases protein expressions of the neurotrophin brain-derived neurotrophic factor and monoamine neurotransmitters in the mouse hippocampus. In addition to acting through AMPARs, AA3 also acts as a non-competitive NMDA receptor (NMDAR) modulator with a neuroprotective capacity against ischemic brain injury and overexcitation in rats. These findings collectively suggest that AA3 possesses a unique ability to modulate the functions of both AMPARs and NMDARs. Concordantly, behavioral studies indicate that AA3 not only facilitates hippocampal long-term potentiation but also enhances spatial reference memory formation in mice. These multifaceted roles suggest that AA3 is an attractive candidate for further development as a cognitive enhancer capable of alleviating memory dysfunctions associated with aging and neurodegenerative diseases.


Subject(s)
Cognition/drug effects , Hippocampus , Neuroprotective Agents/pharmacology , Prefrontal Cortex/drug effects , Saponins/pharmacology , Synapses/drug effects , Triterpenes/pharmacology , Animals , Disease Models, Animal , Escape Reaction/drug effects , Excitatory Postsynaptic Potentials/drug effects , Exploratory Behavior/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , MAP Kinase Signaling System/drug effects , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , N-Methylaspartate/pharmacology , Nerve Net/drug effects , Rats , Rats, Sprague-Dawley , Spatial Navigation/drug effects
16.
PLoS One ; 9(10): e110584, 2014.
Article in English | MEDLINE | ID: mdl-25329792

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase, which plays critical roles in a wide spectrum of neuronal functions including neuronal survival, neurite outgrowth, and synapse development and plasticity. Cdk5 activity is controlled by its specific activators: p35 or p39. While knockout studies reveal that Cdk5/p35 is critical for neuronal migration during early brain development, functions of Cdk5/p35 have been unraveled through the identification of the interacting proteins of p35, most of which are Cdk5/p35 substrates. However, it remains unclear whether p35 can regulate neuronal functions independent of Cdk5 activity. Here, we report that a nuclear protein, nuclear hormone receptor coregulator (NRC)-interacting factor 1 (NIF-1), is a new interacting partner of p35. Interestingly, p35 regulates the functions of NIF-1 independent of Cdk5 activity. NIF-1 was initially discovered as a transcriptional regulator that enhances the transcriptional activity of nuclear hormone receptors. Our results show that p35 interacts with NIF-1 and regulates its nucleocytoplasmic trafficking via the nuclear export pathway. Furthermore, we identified a nuclear export signal on p35; mutation of this site or blockade of the CRM1/exportin-dependent nuclear export pathway resulted in the nuclear accumulation of p35. Intriguingly, blocking the nuclear export of p35 attenuated the nuclear accumulation of NIF-1. These findings reveal a new p35-dependent mechanism in transcriptional regulation that involves the nucleocytoplasmic shuttling of transcription regulators.


Subject(s)
Active Transport, Cell Nucleus/genetics , Intracellular Signaling Peptides and Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/physiology , Nuclear Proteins/biosynthesis , Animals , COS Cells , Chlorocebus aethiops , Cytoplasm/genetics , Cytoplasm/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Karyopherins/genetics , Mice , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors , Exportin 1 Protein
17.
Proc Natl Acad Sci U S A ; 111(27): 9959-64, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958880

ABSTRACT

Alzheimer's disease (AD), characterized by cognitive decline, has emerged as a disease of synaptic failure. The present study reveals an unanticipated role of erythropoietin-producing hepatocellular A4 (EphA4) in mediating hippocampal synaptic dysfunctions in AD and demonstrates that blockade of the ligand-binding domain of EphA4 reverses synaptic impairment in AD mouse models. Enhanced EphA4 signaling was observed in the hippocampus of amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD, whereas soluble amyloid-ß oligomers (Aß), which contribute to synaptic loss in AD, induced EphA4 activation in rat hippocampal slices. EphA4 depletion in the CA1 region or interference with EphA4 function reversed the suppression of hippocampal long-term potentiation in APP/PS1 transgenic mice, suggesting that the postsynaptic EphA4 is responsible for mediating synaptic plasticity impairment in AD. Importantly, we identified a small-molecule rhynchophylline as a novel EphA4 inhibitor based on molecular docking studies. Rhynchophylline effectively blocked the EphA4-dependent signaling in hippocampal neurons, and oral administration of rhynchophylline reduced the EphA4 activity effectively in the hippocampus of APP/PS1 transgenic mice. More importantly, rhynchophylline administration restored the impaired long-term potentiation in transgenic mouse models of AD. These findings reveal a previously unidentified role of EphA4 in mediating AD-associated synaptic dysfunctions, suggesting that it is a new therapeutic target for this disease.


Subject(s)
Alzheimer Disease/physiopathology , Disease Models, Animal , Hippocampus/physiopathology , Receptor, EphA4/metabolism , Synapses/physiology , Alzheimer Disease/metabolism , Animals , Hippocampus/metabolism , Mice , Mice, Transgenic , Receptor, EphA4/genetics , Synapses/metabolism
18.
J Neurosci ; 33(2): 464-72, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303927

ABSTRACT

Learning and memory require orchestrated regulation of both structural and functional synaptic plasticity in the hippocampus. While a neuropeptide alpha-melanocyte-stimulating hormone, α-MSH, has been implicated in memory acquisition and retention, the functional role of its cognate receptor, melanocortin-4 receptor (MC4R), in hippocampal-dependent synaptic plasticity has not been explored. In this study, we report that activation of MC4R enhances synaptic plasticity through the regulation of dendritic spine morphology and abundance of AMPA receptors. We show that activation of postsynaptic MC4R increases the number of mature dendritic spines and enhances surface expression of AMPA receptor subunit GluA1, resulting in synaptic accumulation of GluA1-containing AMPA receptors. Moreover, MC4R stimulates surface GluA1 trafficking through phosphorylation of GluA1 at Ser845 in a Gα(s)-cAMP/PKA-dependent manner. Blockade of protein kinase A (PKA) signaling abolishes the MC4R-mediated enhancement of neurotransmission and hippocampal long-term potentiation. Importantly, in vivo application of MC4R agonists increases LTP in the mouse hippocampal CA1 region. These findings reveal that MC4R in the hippocampus plays a critical role in the regulation of structural and functional plasticity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Hippocampus/physiology , Neuronal Plasticity/physiology , Receptor, Melanocortin, Type 4/physiology , Synapses/physiology , Animals , Biotinylation , Blotting, Western , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , DNA Primers , Electrophysiological Phenomena , HEK293 Cells , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Learning/physiology , Long-Term Potentiation/physiology , Memory/physiology , Mice , Real-Time Polymerase Chain Reaction , Receptors, AMPA/physiology , Stereotaxic Techniques , Synaptic Transmission/physiology
19.
Neurosignals ; 21(1-2): 55-60, 2013.
Article in English | MEDLINE | ID: mdl-22398430

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5), a member of the cyclin-dependent kinase family, is critical for regulating neural development and neuronal survival. Dysregulation of Cdk5 is associated with abnormal expression of cell cycle-related proteins during neuronal apoptosis. We have previously found that p35, a Cdk5 activator, interacts with mSds3, an integral component of the histone deacetylase complex in vitro, suggesting a functional role of Cdk5 in gene regulation through modulation of chromatin integrity. In this study, we further demonstrate that Cdk5-dependent phosphorylation of mSds3 at Ser228 occurs in mouse brain nuclei. The expression of mSds3 protein and its interaction with Cdk5 activators is developmentally regulated in the mouse brain. Importantly, our findings suggest that the ability of Cdk5 to regulate activity deprivation-induced apoptosis of cerebellar granule neurons is likely mediated by the regulation of histone acetylation. Suppression of Cdk5 not only attenuates the induction of histone H3 acetylation and the aberrant upregulation of cyclin proteins in neurons after activity deprivation, but also results in protection of neurons against apoptotic cell death. Taken together, our findings suggest that Cdk5 regulates neuronal survival by precise epigenetic control through modulation of histone acetylation.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Neurons/metabolism , Acetylation , Animals , Cell Death/physiology , Cells, Cultured , Cerebellum/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation/physiology
20.
J Neurosci ; 32(24): 8263-9, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22699907

ABSTRACT

Dendrites are the primary sites on neurons for receiving and integrating inputs from their presynaptic partners. Defects in dendrite development perturb the formation of neural circuitry and impair information processing in the brain. Extracellular cues are important for shaping the dendritic morphogenesis, but the underlying molecular mechanisms are not well understood. In this study, we examined the role of ARMS (ankyrin repeat-rich membrane spanning protein), also known as Kidins220 (kinase D-interacting substrate of 220 kDa), previously identified as a downstream target of neurotrophin and ephrin receptors, in dendrite development. We report here that knockdown of ARMS/Kidins220 by in utero electroporation impairs dendritic branching in mouse cerebral cortex, and silencing of ARMS/Kidins220 in primary rat hippocampal neurons results in a significant decrease in the length, number, and complexity of the dendritic arbors. Overexpression of cell surface receptor tyrosine kinases, including TrkB and EphB2, in ARMS/Kidins220-deficient neurons can partially rescue the defective dendritic phenotype. More importantly, we show that PI3K (phosphoinositide-3-kinase)- and Akt-mediated signaling pathway is crucial for ARMS/Kidins220-dependent dendrite development. Furthermore, loss of ARMS/Kidins220 significantly reduced the clustering of EphB2 receptor signaling complex in neurons. Our results collectively suggest that ARMS/Kidins220 is a key player in organizing the signaling complex to transduce the extracellular stimuli to cellular responses during dendrite development.


Subject(s)
Membrane Proteins/physiology , Phosphoproteins/physiology , Receptor, trkB/physiology , Receptors, Eph Family/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dendrites/metabolism , Dendrites/physiology , Female , Gene Knockdown Techniques/methods , Hippocampus/cytology , Hippocampus/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred ICR , Mice, Transgenic , Neurogenesis/physiology , Phosphoproteins/genetics , Primary Cell Culture , Rats , Receptor Protein-Tyrosine Kinases/biosynthesis , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...