Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Chin Med ; 51(7): 1823-1843, 2023.
Article in English | MEDLINE | ID: mdl-37650420

ABSTRACT

Traditional Chinese Medicine (TCM) prescriptions are organically composed of compatible herbs according to the TCM theory. The complex ingredients of TCM could act on multiple targets through various pathways simultaneously to exert pharmacological effects, making TCM an unrivaled gem in the medical world. However, due to a lack of comprehensive and standard study methods, the research of TCM products has been quite limited. A novel paradigm that could aid in the discovery of the material basis and fully clarify the mechanism of TCM prescriptions is urgently needed. In this study, a similarity analysis based on molecular fingerprints was adopted to explore the representative molecules of the Tiaoxin recipe, a Chinese patent formula approved by the National Medical Products Administration (NMPA) for the treatment of mild-to-moderate Alzheimer's disease (AD), and 38 out of 1047 chemicals were finally screened out. Next, we tried to define a new concept of a "functional molecule cluster" for chemicals with similar pharmacological effects to elucidate how the chemical mixture from TCMs produce their therapeutic effects. Four anti-AD functional molecule clusters from the Tiaoxin recipe were identified: an anti-inflammatory cluster, an anti-ROS cluster, an anti-AChE activity cluster, and an anti-A[Formula: see text] aggregation cluster. Furthermore, the chemicals from the anti-inflammatory cluster and anti-ROS cluster were proved to display their multi-target and multi-pathway roles partially or mainly through molecules of the TLR4-MYD88-NF-[Formula: see text]B and Keap1-Nrf2-ARE pathways. The functional molecule clusters may be vital to the explanation of the efficacy of the Tiaoxin recipe, which could give us a more profound understanding of TCM prescriptions. Our paradigm may open a novel path for TCM research.

2.
Int J Mol Sci ; 24(8)2023 Apr 16.
Article in English | MEDLINE | ID: mdl-37108518

ABSTRACT

The selectivity of drugs for G protein-coupled receptor (GPCR) signaling pathways is crucial for their therapeutic efficacy. Different agonists can cause receptors to recruit effector proteins at varying levels, thus inducing different signaling responses, called signaling bias. Although several GPCR-biased drugs are currently being developed, only a limited number of biased ligands have been identified regarding their signaling bias for the M1 muscarinic acetylcholine receptor (M1mAChR), and the mechanism is not yet well understood. In this study, we utilized bioluminescence resonance energy transfer (BRET) assays to compare the efficacy of six agonists in inducing Gαq and ß-arrestin2 binding to M1mAChR. Our findings reveal notable variations in agonist efficacy in the recruitment of Gαq and ß-arrestin2. Pilocarpine preferentially promoted the recruitment of ß-arrestin2 (∆∆RAi = -0.5), while McN-A-343 (∆∆RAi = 1.5), Xanomeline (∆∆RAi = 0.6), and Iperoxo (∆∆RAi = 0.3) exhibited a preference for the recruitment of Gαq. We also used commercial methods to verify the agonists and obtained consistent results. Molecular docking revealed that certain residues (e.g., Y404, located in TM7 of M1mAChR) could play crucial roles in Gαq signaling bias by interacting with McN-A-343, Xanomeline, and Iperoxo, whereas other residues (e.g., W378 and Y381, located in TM6) contributed to ß-arrestin recruitment by interacting with Pilocarpine. The preference of activated M1mAChR for different effectors may be due to significant conformational changes induced by biased agonists. By characterizing bias towards Gαq and ß-arrestin2 recruitment, our study provides insights into M1mAChR signaling bias.


Subject(s)
Acetylcholine , Receptor, Muscarinic M1 , Humans , beta-Arrestins/metabolism , Molecular Docking Simulation , Receptor, Muscarinic M1/metabolism , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride , Pilocarpine/pharmacology , GTP-Binding Proteins/metabolism , beta-Arrestin 2/metabolism , beta-Arrestin 1/metabolism , Energy Transfer , HEK293 Cells
3.
Transl Neurodegener ; 12(1): 1, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36624510

ABSTRACT

BACKGROUND: Ribosomal protein S6 kinase 1 (S6K1) is a serine-threonine kinase that has two main isoforms: p70S6K (70-kDa isoform) and p85S6K (85-kDa isoform). p70S6K, with its upstream mammalian target of rapamycin (mTOR), has been shown to be involved in learning and memory and participate in the pathophysiology of Alzheimer's disease (AD). However, the function of p85S6K has long been neglected due to its high similarity to p70S6k. The role of p85S6K in learning and memory is still largely unknown. METHODS: We fractionated the postsynaptic densities to illustrate the differential distribution of p85S6K and p70S6K. Coimmunoprecipitation was performed to unveil interactions between p85S6K and the GluA1 subunit of AMPA receptor. The roles of p85S6K in synaptic targeting of GluA1 and learning and memory were evaluated by specific knockdown or overexpression of p85S6K followed by a broad range of methodologies including immunofluorescence, Western blot, in situ proximity ligation assay, morphological staining and behavioral examination. Further, the expression level of p85S6K was measured in brains from AD patients and AD model mice. RESULTS: p85S6K, but not p70S6K, was enriched in the postsynaptic densities. Moreover, knockdown of p85S6K resulted in defective spatial and recognition memory. In addition, p85S6K could interact with the GluA1 subunit of AMPA receptor through synapse-associated protein 97 and A-kinase anchoring protein 79/150. Mechanistic studies demonstrated that p85S6K could directly phosphorylate GluA1 at Ser845 and increase the amount of GluA1 in synapses, thus sustaining synaptic function and spine densities. Moreover, p85S6K was found to be specifically decreased in the synaptosomal compartment in the brains of AD patients and AD mice. Overexpression of p85S6K ameliorated the synaptic deficits and cognitive impairment in transgenic AD model mice. CONCLUSIONS: These results strongly imply a significant role for p85S6K in maintaining synaptic and cognitive function by interacting with GluA1. The findings provide an insight into the rational targeting of p85S6K as a therapeutic potential for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Animals , Mice , Alzheimer Disease/genetics , Receptors, AMPA , Cognitive Dysfunction/genetics , Cognition , Mice, Transgenic , Mammals
4.
Front Chem ; 10: 842208, 2022.
Article in English | MEDLINE | ID: mdl-35646819

ABSTRACT

Background: A series of α-Mangostin (α-M) derivatives were designed and synthesized. α-M and four analogues were evaluated for their multifunctional anti-Alzheimer's disease (anti-AD) effects on fibrillogenesis, microglial uptake, microglial degradation, and anti-neurotoxicity of Aß, as well as LPS-induced neuroinflammation. The differences in bioactivities were analyzed to understand the structure-activity relationship for further modifications. Purpose: This study aims to investigate the anti-AD effects of α-M and elucidate its structure-activity relationship by comparing difference between α-M and several analogues. Methods: Aß fibrillogenesis was detected by Thioflavin T fluorometric assay. The levels of Aß1-42 and inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay. Neuron viability was examined by the CCK-8 assay. The morphology of ZO-1 of bEnd.3 cultured in BV-2-conditioned medium was evaluated by immunofluorescence staining. Results: Aß fibrillogenesis was significantly inhibited by co-incubation with α-M, Zcbd-2 or Zcbd-3. α-M, Zcbd-2, Zcbd-3, and Zcbd-4 decreased the levels of Aß1-42 and inflammatory cytokines, and promoted Aß uptake, degradation and anti-inflammation effects inflammation in microglia. α-M and Zcbd-3 protected neuron viability from Aß-induced neurotoxicity, and preserved tight junction integrity of bEnd.3 against LPS-induced neuroinflammation. Conclusion: Zcbd-3 acted as α-M almost in all effects. The structure-activity analysis indicated that the 3-methyl-2-butenyl group at C-8 is essential for the bioactivity of α-M, while modifying the double hydroxylation at the C-2 position may improve the multifunctional anti-AD effects.

5.
Transl Neurodegener ; 11(1): 18, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35300725

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease in the elderly worldwide. However, the complexity of AD pathogenesis leads to discrepancies in the understanding of this disease, and may be the main reason for the failure of AD drug development. Fortunately, many ongoing preclinical and clinical studies will continually open up avenues to unravel disease mechanisms and guide strategies for AD diagnosis and drug development. For example, immunotherapeutic strategies targeting amyloid-ß (Aß) and tau proteins were once deemed almost certainly effective in clinical treatment due to the excellent preclinical results. However, the repeated failures of clinical trials on vaccines and humanized anti-Aß and anti-tau monoclonal antibodies have resulted in doubts on this strategy. Recently, a new anti-Aß monoclonal antibody (Aducanumab) has been approved by the US Food and Drug Administration, which brings us back to the realization that immunotherapy strategies targeting Aß may be still promising. Meanwhile, immunotherapies based on other targets such as tau, microglia and gut-brain axis are also under development. Further research is still needed to clarify the forms and epitopes of targeted proteins to improve the accuracy and effectiveness of immunotherapeutic drugs. In this review, we focus on the immunotherapies based on Aß, tau and microglia and their mechanisms of action in AD. In addition, we present up-to-date advances and future perspectives on immunotherapeutic strategies for AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Aged , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Humans , Immunologic Factors , Immunotherapy/methods , tau Proteins/metabolism
6.
FASEB J ; 35(6): e21658, 2021 06.
Article in English | MEDLINE | ID: mdl-34010470

ABSTRACT

Alzheimer's disease (AD) is a complicated neurodegenerative disease and therefore addressing multiple targets simultaneously has been believed as a promising therapeutic strategy against AD. α7 nicotinic acetylcholine receptor (nAChR), which plays an important role in improving cognitive function and alleviating neuroinflammation in central nervous system (CNS), has been regarded as a potential target in the treatment of AD. However, the regulation of α7 nAChR at post-transcriptional level in mammalian brain remains largely speculated. Herein, we uncovered a novel post-transcriptional regulatory mechanism of α7 nAChR expression in AD and further demonstrated that miR-98-5p suppressed α7 nAChR expression through directly binding to the 3'UTR of mRNA. Knockdown of miR-98-5p activated Ca2+ signaling pathway and consequently reversed cognitive deficits and Aß burden in APP/PS1 mice. Furthermore, miR-98-5p downregulation increased α7 nAChR expression, and ameliorated neuroinflammation via inhibiting NF-κB pathway and upregulating Nrf2 target genes. Our findings illustrate a prominent regulatory role of miR-98-5p in targeting inflammation and cognition, and provide an insight into the potential of miR-98-5p/α7 nAChR axis as a novel therapeutic strategy for AD.


Subject(s)
Alzheimer Disease/complications , Cognitive Dysfunction/pathology , MicroRNAs/genetics , Neurodegenerative Diseases/pathology , RNA Processing, Post-Transcriptional , alpha7 Nicotinic Acetylcholine Receptor/genetics , Amyloid beta-Protein Precursor/physiology , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Presenilin-1/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
7.
Bioorg Med Chem Lett ; 30(24): 127632, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33132116

ABSTRACT

Muscarinic acetylcholine receptors (mAChRs) comprise five distinct subtypes denoted M1 to M5. The antagonism of M2 subtype could increase the release of acetylcholine from vesicles into the synaptic cleft and improve postsynaptic functions in the hippocampus via M1 receptor activation, displaying therapeutic potentials for Alzheimer's disease. However, drug development for M2 antagonists is still challenged among different receptor subtypes. In this study, by optimizing a scaffold from virtual screening, we synthesized two focused libraries and generated up to 50 derivatives. By measuring potency and binding selectivity, we discovered a novel M2 antagonist, ligand 47, featuring submicromolar IC50, high M2/M4 selectivity (~30-fold) and suitable lipophilicity (cLogP = 4.55). Further study with these compounds also illustrates the structure-activity relationship of this novel scaffold. Our study could not only provide novel lead structure, which was easy to synthesize, but also offer valuable information for further development of selective M2 ligands.


Subject(s)
Muscarinic Antagonists/chemistry , Muscarinic Antagonists/pharmacology , Receptor, Muscarinic M2/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Molecular Docking Simulation , Receptor, Muscarinic M2/metabolism , Structure-Activity Relationship
8.
Cell Mol Neurobiol ; 40(4): 547-554, 2020 May.
Article in English | MEDLINE | ID: mdl-31721013

ABSTRACT

M1 muscarinic acetylcholine receptors (M1 mAChRs) have long been an attractive target for the treatment of Alzheimer's disease (AD), the most common cause of dementia in the elderly. M1 mAChR agonists show desirably preclinical activities; however, most have not gone further into late clinical trials due to ineffectiveness or side effects. Thus, to understand the signaling pathways involved in M1 mAChR-mediated memory improvement may be important for design of biased agonists with on-target therapeutic effects. M1 mAChRs are classically coupled to Gαq or ectopically to Gαs to activate multiple kinases such as protein kinase C (PKC), Ras and protein kinase A (PKA). Our previous studies have found that M1 mAChRs could improve learning and memory through modulating AMPA receptor GluA1 subunit via PKA-PI3K-Akt signaling. Here, we further investigated whether PKC and Ras were involved in M1 mAChR-mediated modulation of GluA1. We demonstrated the role of PKC and Ras in the signaling pathway, as both PKC inhibitors Ro-31-8425 or Gö6983 and Ras inhibitor salirasib abolished the membrane insertion of GluA1 and enhancement of its phosphorylation at Ser845 induced by M1 mAChRs in the primary cultured neurons and hippocampus in vivo. We further showed that PKC and Ras modulated PKA-PI3K-Akt signaling since the increases of PKA, Akt and mTOR activities by M1 mAChR activation were blocked by PKC and Ras inhibitors. These data demonstrated the detailed mechanism underlying M1 mAChR-mediated modulation of GluA1 through Gαq/11 coupling, broadening the knowledge of the downstream signaling after M1 mAChR-Gαq/11 coupling.


Subject(s)
Protein Kinase C/metabolism , Receptor, Muscarinic M1/metabolism , Receptors, AMPA/metabolism , ras Proteins/metabolism , Animals , Animals, Newborn , Cells, Cultured , Male , Mice, Inbred C57BL , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction
9.
Curr Alzheimer Res ; 16(8): 753-763, 2019.
Article in English | MEDLINE | ID: mdl-31441725

ABSTRACT

BACKGROUND: APOE4 is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). LOAD patients carrying or not carrying APOE4 manifest distinct clinico-pathological characteristics. APOE4 has been shown to play a critical role in the pathogenesis of AD by affecting various aspects of pathological processes. However, the pathogenesis involved in LOAD not-carrying APOE4 remains elusive. OBJECTIVE: We aimed to identify the associated genes involved in LOAD not-carrying APOE4. METHODS: An integrated genomic analysis of datasets of genome-wide association study, genome-wide expression profiling and genome-wide linkage scan and protein-protein interaction network construction were applied to identify associated gene clusters in APOE4 non-carriers. The role of one of hub gene of an APOE4 non-carrier-associated gene cluster in tau phosphorylation was studied by knockdown and western blot. RESULTS: We identified 12 gene clusters associated with AD APOE4 non-carriers. The hub genes associated with AD in these clusters were MAPK8, POU2F1, XRCC1, PRKCG, EXOC6, VAMP4, SIRT1, MME, NOS1, ABCA1 and LDLR. The associated genes for APOE4 non-carriers were enriched in hereditary disorder, neurological disease and psychological disorders. Moreover, knockdown of PRKCG to reduce the expression of protein kinase Cγ isoform enhanced tau phosphorylation at Thr181 and Thr231 and the expression of glycogen synthase kinase 3ß and cyclin-dependent kinase 5 in the presence of APOE3 but not APOE4. CONCLUSION: The study provides new insight into the mechanism of distinct pathogenesis of LOAD not carrying APOE4 and prompts the functional exploration of identified genes based on APOE genotypes.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Apolipoprotein E4/genetics , Cells, Cultured , Genetic Predisposition to Disease , Genome-Wide Association Study , Heterozygote , Hippocampus/metabolism , Humans , Male , Mice, Inbred C57BL , Multigene Family , Neurons/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , tau Proteins/genetics , tau Proteins/metabolism
10.
Neuroscience ; 408: 239-247, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30981860

ABSTRACT

M1 muscarinic receptors have long been identified as a potential therapeutic target for the treatment of cognitive impairment in Alzheimer's disease (AD). Our previous study has shown that M1 receptors promote membrane insertion and synaptic delivery of AMPA receptor GluA1 subunit. In this study, we sought to determine whether activation of M1 receptor would rescue the cognitive impairment in AD model mice through modulation of GluA1 subunit. For the mice injected with aggregated ß-amyloid (Aß) fragments to impair learning and memory, activation of M1 receptors could rescue it by reducing the latency to find the platform and spending more time in the target quadrant in the probe test in the Morris water maze. However, such an effect was ablated in mice with Ser845 residue of GluA1 mutated to alanine. Furthermore, the activation of M1 receptors enhanced the expression of GluA1 and its phosphorylation at Ser845 and drove GluA1 to incorporate with PSD95, a postsynaptic marker, in the hippocampi from Aß-injected wild type mice but not from the mutant mice. Moreover, for 9-month-old APP/PS1 transgenic AD model mice, which may resemble the late AD, M1 receptor activation could not improve the cognitive impairment significantly. In addition, the enhancement of GluA1 expression and its phosphorylation at Ser845 were not observed in their hippocampi. Taken together, the study indicated that M1 receptor activation rescued the cognitive deficit through modulating the trafficking of GluA1-containing AMPA receptors and the therapeutics targeting M1 receptors should aim at mild AD or even pre-AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cognitive Dysfunction/drug therapy , Muscarinic Agonists/pharmacology , Peptide Fragments/pharmacology , Receptors, AMPA/metabolism , Spatial Learning/drug effects , Spatial Memory/drug effects , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Muscarinic Agonists/therapeutic use , Phosphorylation , Receptor, Muscarinic M1/metabolism
11.
FASEB J ; 33(5): 6622-6631, 2019 05.
Article in English | MEDLINE | ID: mdl-30794430

ABSTRACT

M1 muscarinic acetylcholine receptors are highly expressed in key areas that control cognition, such as the cortex and hippocampus, representing one potential therapeutic target for cognitive dysfunctions of Alzheimer's disease and schizophrenia. We have reported that M1 receptors facilitate cognition by promoting membrane insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor AMPA receptor subunit 1 (GluA1) through phosphorylation at Ser845. However, the signaling pathway is still unclear. Here we showed that adenylyl cyclase inhibitor 2',5'-dideoxyadenosine and PKA inhibitor KT5720 inhibited enhancement of phosphorylation of Ser845 and membrane insertion of GluA1 induced by M1 receptor activation. Furthermore, PI3K inhibitor LY294002 and protein kinase B (Akt) inhibitor IV blocked the effects of M1 receptors as well. Remarkably, the increase of the activity of PI3K-Akt signaling induced by M1 receptor activation could be abolished by cAMP-PKA inhibitors. Moreover, inhibiting the mammalian target of rapamycin (mTOR) complex 1, an important downstream effector of PI3K-Akt, by short-term application of rapamycin attenuated the effects of M1 receptors on GluA1. Furthermore, such effect was unrelated to possible protein synthesis promoted by mTOR. Taken together, these data demonstrate that M1 receptor activation induces membrane insertion of GluA1 via a signaling linking cAMP-PKA and PI3K-Akt-mTOR pathways but is irrelevant to protein synthesis.-Zhao, L.-X., Ge, Y.-H., Li, J.-B., Xiong, C.-H., Law, P.-Y., Xu, J.-R., Qiu, Y., Chen, H.-Z. M1 muscarinic receptors regulate the phosphorylation of AMPA receptor subunit GluA1 via a signaling pathway linking cAMP-PKA and PI3K-Akt.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Muscarinic M1/metabolism , Receptors, AMPA/metabolism , Second Messenger Systems/physiology , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Rats , Rats, Sprague-Dawley
12.
Neuropharmacology ; 146: 242-251, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30529302

ABSTRACT

Cognitive flexibility is an important aspect of executive function. The cholinergic system, an important component of cognition, has been shown to modulate cognitive flexibility mainly through the striatum and prefrontal cortex. The role of M1 muscarinic receptors (M1 mAChRs), an important therapeutic target in the cholinergic system, in hippocampus-dependent cognitive flexibility is unclarified. In the present study, we demonstrated that selective activation of M1 mAChRs promoted extinction of initial learned response and facilitated acquisition of reversal learning in the Morris water maze, a behavior test that is mainly dependent on the hippocampus. However, these effects were abolished in GluA2 mutant mice with deficiency in phosphorylation of Ser880 by protein kinase C (PKC). Further long-term depression (LTD) in the hippocampal CA1 area induced by M1 mAChR activation was shown to be dependent on AMPA receptor subunit GluA2 but not GluA1. M1 mAChRs increased GluA2 endocytosis through phosphorylation of Ser880 by PKC. Inhibition of PKC blocked M1 mAChR-mediated LTD, memory switching and reversal learning facilitation. Moreover, the slow memory extinction observed in GluA2 mutant mice and PKC inhibitor-treated mice appeared to affect the consolidation and retrieval of reversal learning. Thus, these results demonstrate that M1 mAChRs mainly facilitate acquisition in spatial reversal learning and further elucidate that such an effect is dependent on the phosphorylation of GluA2 by PKC. The study helps clarify the role of M1 mAChRs in cognitive flexibility and may prompt the earlier prevention of cognitive inflexibility.


Subject(s)
Receptor, Muscarinic M1/drug effects , Receptor, Muscarinic M1/metabolism , Receptors, AMPA/metabolism , Reversal Learning/drug effects , Animals , Behavior, Animal/drug effects , CA1 Region, Hippocampal/drug effects , Cognition/physiology , Hippocampus , Learning/physiology , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Piperidines , Protein Kinase C/antagonists & inhibitors , Quinolones , Receptor, Muscarinic M1/agonists , Receptors, AMPA/deficiency
13.
FASEB J ; 32(8): 4247-4257, 2018 08.
Article in English | MEDLINE | ID: mdl-29509512

ABSTRACT

M1 muscarinic acetylcholine receptors (M1 mAChRs) are the most abundant muscarinic receptors in the hippocampus and have been shown to have procognitive effects. AMPA receptors (AMPARs), an important subtype of ionotropic glutamate receptors, are key components in neurocognitive networks. However, the role of AMPARs in procognitive effects of M1 mAChRs and how M1 mAChRs affect the function of AMPARs remain poorly understood. Here, we found that basal expression of GluA1, a subunit of AMPARs, and its phosphorylation at Ser845 were maintained by M1 mAChR activity. Activation of M1 mAChRs promoted membrane insertion of GluA1, especially to postsynaptic densities. Impairment of hippocampus-dependent learning and memory by antagonism of M1 mAChRs paralleled the reduction of GluA1 expression, and improvement of learning and memory by activation of M1 mAChRs was accompanied by the synaptic insertion of GluA1 and its increased phosphorylation at Ser845. Furthermore, abrogation of phosphorylation of Ser845 residue of GluA1 ablated M1 mAChR-mediated improvement of learning and memory. Taken together, these results show a functional correlation of M1 mAChRs and GluA1 and the essential role of GluA1 in M1 mAChR-mediated cognitive improvement.-Zhao, L.-X., Ge, Y.-H., Xiong, C.-H., Tang, L., Yan, Y.-H., Law, P.-Y., Qiu, Y., Chen, H.-Z. M1 muscarinic receptor facilitates cognitive function by interplay with AMPA receptor GluA1 subunit.


Subject(s)
Cognition/physiology , Protein Subunits/metabolism , Receptor, Muscarinic M1/metabolism , Receptors, AMPA/metabolism , Animals , Chromosome Pairing/physiology , Hippocampus/metabolism , Learning/physiology , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic/metabolism , Phosphorylation/physiology , Receptors, Muscarinic/metabolism
14.
CNS Neurosci Ther ; 23(6): 526-534, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28429536

ABSTRACT

AIMS: ß-amyloid (Aß) aggregation and deposition play a central role in the pathogenic process of Alzheimer's disease (AD). α-Mangostin (α-M), a polyphenolic xanthone, have been shown to dissociate Aß oligomers. In this study, we further investigated the effect of α-M on Aß production and its molecular mechanism. METHODS: The Aß and soluble amyloid precursor protein α (sAPPα) in culture medium of cortical neurons were measured by ELISA. The activities of α-, ß-, and γ-secretases were assayed, and the interaction between α-M and ß- or γ-secretases was simulated by molecular docking. RESULTS: α-M significantly decreased Aß40 and Aß42 production. α-M did not affect the expression of enzymes involved in nonamyloidogenic and amyloidogenic pathways, but significantly decreased the activities of ß-secretase and likely γ-secretase with IC50 13.22 nmol·L-1 and 16.98 nmol·L-1 , respectively. Molecular docking demonstrated that α-M interacted with ß-site amyloid precursor protein cleaving enzyme 1 and presenilin 1 to interfere with their active sites. CONCLUSIONS: Our data demonstrate that α-M decreases Aß production through inhibiting activities of ß-secretase and likely γ-secretase in the amyloidogenic pathway. The current data together with previous study indicated that α-M could be a novel neuroprotective agent through intervention of multiple pathological processes of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , Xanthones/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Embryo, Mammalian , Molecular Docking Simulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Presenilin-1/metabolism , RNA, Messenger , Rats
15.
Neurosci Lett ; 566: 231-235, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24607931

ABSTRACT

Agonist-activated µ-opioid receptor (OPRM1) undergoes robust receptor phosphorylation by G protein-coupled receptor kinases and subsequent ß-arrestin recruitment, triggering receptor internalization and desensitization. Morphine, a widely prescribed opioid, induces receptor phosphorylation inefficiently. Previously we reported that FK506 binding protein 12 (FKBP12) specifically interacts with OPRM1 and such interaction attenuates receptor phosphorylation and facilitates morphine-induced recruitment and activation of protein kinase C. In the current study, we demonstrated that the association of FKBP12 with OPRM1 also affects morphine-induced receptor internalization and G protein-dependent adenylyl cyclase desensitization. Morphine induced faster receptor internalization and adenylyl cyclase desensitization in cells expressing OPRM1 with Pro(353) mutated to Ala (OPRM1P353A), which does not interact with FKBP12, or in the presence of FK506 which dissociates the receptor-FKBP12 interaction. Furthermore, knockdown of cellular FKBP12 level by siRNA accelerated morphine-induced receptor internalization and adenylyl cyclase desensitization. Our study further demonstrated that peptidyl prolyl cis-trans isomerase activity of FKBP12 probably plays a role in inhibition of receptor phosphorylation. In the view that internalized receptor recycles and thus counteracts the development of analgesic tolerance, receptor's association with FKBP12 could also contribute to the development of morphine tolerance through modulation of receptor trafficking.


Subject(s)
Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Tacrolimus Binding Protein 1A/metabolism , Adenylyl Cyclases/metabolism , HEK293 Cells , Humans , Mutation , Phosphorylation , RNA, Small Interfering/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/genetics
16.
Antimicrob Agents Chemother ; 58(4): 2344-55, 2014.
Article in English | MEDLINE | ID: mdl-24514088

ABSTRACT

Pterostilbene (PTE) is a stilbene-derived phytoalexin that originates from several natural plant sources. In this study, we evaluated the activity of PTE against Candida albicans biofilms and explored the underlying mechanisms. In 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) reduction assays, biofilm biomass measurement, confocal laser scanning microscopy, and scanning electron microscopy, we found that ≤16 µg/ml PTE had a significant effect against C. albicans biofilms in vitro, while it had no fungicidal effect on planktonic C. albicans cells, which suggested a unique antibiofilm effect of PTE. Then we found that PTE could inhibit biofilm formation and destroy the maintenance of mature biofilms. At 4 µg/ml, PTE decreased cellular surface hydrophobicity (CSH) and suppressed hyphal formation. Gene expression microarrays and real-time reverse transcription-PCR showed that exposure of C. albicans to 16 µg/ml PTE altered the expression of genes that function in morphological transition, ergosterol biosynthesis, oxidoreductase activity, and cell surface and protein unfolding processes (heat shock proteins). Filamentation-related genes, especially those regulated by the Ras/cyclic AMP (cAMP) pathway, including ECE1, ALS3, HWP1, HGC1, and RAS1 itself, were downregulated upon PTE treatment, indicating that the antibiofilm effect of PTE was related to the Ras/cAMP pathway. Then, we found that the addition of exogenous cAMP reverted the PTE-induced filamentous growth defect. Finally, with a rat central venous catheter infection model, we confirmed the in vivo activity of PTE against C. albicans biofilms. Collectively, PTE had strong activities against C. albicans biofilms both in vitro and in vivo, and these activities were associated with the Ras/cAMP pathway.


Subject(s)
Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Biofilms/drug effects , Candida albicans/drug effects , Candida albicans/physiology , Stilbenes/pharmacology , Stilbenes/therapeutic use , Animals , Candida albicans/metabolism , Female , Fungal Proteins/metabolism , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Rats , Rats, Sprague-Dawley
17.
PLoS One ; 8(11): e79671, 2013.
Article in English | MEDLINE | ID: mdl-24260276

ABSTRACT

Candida albicans is the most common human fungal pathogen and has a high propensity to develop biofilms that are resistant to traditional antifungal agents. In this study, we investigated the effect of tetrandrine (TET) on growth, biofilm formation and yeast-to-hypha transition of C. albicans. We characterized the inhibitory effect of TET on hyphal growth and addressed its possible mechanism of action. Treatment of TET at a low concentration without affecting fungal growth inhibited hyphal growth in both liquid and solid Spider media. Real-time RT-PCR revealed that TET down-regulated the expression of hypha-specific genes ECE1, ALS3 and HWP1, and abrogated the induction of EFG1 and RAS1, regulators of hyphal growth. Addition of cAMP restored the normal phenotype of the SC5314 strain. These results indicate that TET may inhibit hyphal growth through the Ras1p-cAMP-PKA pathway. In vivo, at a range of concentrations from 4 mg/L to 32 mg/L, TET prolonged the survival of C. albicans-infected Caenorhabditis elegans significantly. This study provides useful information for the development of new strategies to reduce the incidence of C. albicans biofilm-associated infections.


Subject(s)
Antifungal Agents/pharmacology , Benzylisoquinolines/pharmacology , Biofilms/drug effects , Candida albicans/drug effects , Drugs, Chinese Herbal/pharmacology , Candida albicans/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism
18.
Antimicrob Agents Chemother ; 57(12): 6016-27, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24060867

ABSTRACT

It was found in our previous study that berberine (BBR) and fluconazole (FLC) used concomitantly exhibited a synergism against FLC-resistant Candida albicans in vitro. The aim of the present study was to clarify how BBR and FLC worked synergistically and the underlying mechanism. Antifungal time-kill curves indicated that the synergistic effect of the two drugs was BBR dose dependent rather than FLC dose dependent. In addition, we found that BBR accumulated in C. albicans cells, especially in the nucleus, and resulted in cell cycle arrest and significant change in the transcription of cell cycle-related genes. Besides BBR, other DNA intercalators, including methylene blue, sanguinarine, and acridine orange, were all found to synergize with FLC against FLC-resistant C. albicans. Detection of intracellular BBR accumulation by fluorescence measurement showed that FLC played a role in increasing intracellular BBR concentration, probably due to its effect in disrupting the fungal cell membrane. Similar to the case with FLC, other antifungal agents acting on the cell membrane were able to synergize with BBR. Interestingly, we found that the efflux of intracellular BBR was FLC independent but strongly glucose dependent and associated with the drug efflux pump Cdr2p. These results suggest that BBR plays a major antifungal role in the synergism of FLC and BBR, while FLC plays a role in increasing the intracellular BBR concentration.


Subject(s)
Antifungal Agents/pharmacology , Berberine/pharmacology , Candida albicans/drug effects , Fluconazole/pharmacology , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Acridine Orange/pharmacology , Benzophenanthridines/pharmacology , Biological Transport , Candida albicans/genetics , Candida albicans/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Fungal/genetics , Drug Synergism , Fungal Proteins/metabolism , Intercalating Agents/pharmacology , Isoquinolines/pharmacology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methylene Blue/pharmacology , Microbial Sensitivity Tests
19.
Biol Pharm Bull ; 36(9): 1482-7, 2013.
Article in English | MEDLINE | ID: mdl-23995660

ABSTRACT

Candida albicans is the most common fungal pathogen. Galleria mellonella is widely used as an infection model host. Nevertheless, the G. mellonella-C. albicans infection model had not been optimized for drug evaluation before this study. In this work, we revealed that 5 × 10(5) colony forming unit (CFU)/larva was a suitable inoculum to optimize the G. mellonella-C. albicans infection model in order to evaluate antifungal agents. Using our optimized model, the antifungal effect of fluconazole, amphotericin B and flucytosine, and the synergy between amphotericin B and flucytosine were successfully verified. Thus, this study provides a rapid, inexpensive and reliable way to evaluate antifungals in vivo.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Disease Models, Animal , Moths/microbiology , Amphotericin B/pharmacology , Animals , Candida albicans/pathogenicity , Fluconazole/pharmacology , Flucytosine/pharmacology , Larva/microbiology
20.
FEBS J ; 280(11): 2633-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23517286

ABSTRACT

Candida albicans is the most common opportunistic fungal pathogen and its apoptosis is inducible by environmental stress. Based on our previous finding that transcription factor Cap1p was involved in baicalein-induced apoptosis, the present study aimed to further clarify the role of Cap1p in apoptosis by observing the impact of CAP1 deletion on cell fate. It was found that apoptotic stimulation with amphotericin B, acetic acid and hydrogen peroxide increased the number of apoptotic and necrotic cells, caspase activity and the accumulation of reactive oxygen species, whereas it decreased the mitochondrial membrane potential and intracellular ATP level in the cap1Δ/Δ mutant. The cell fate was, at least partly, caused by glutathione depletion and attenuation of the expression of the glutathione reductase gene in the cap1Δ/Δ mutant. Collectively, our data suggest that Cap1p participated in the apoptosis of C. albicans by regulating the expression of the glutathione reductase gene and glutathione content.


Subject(s)
Apoptosis/physiology , Candida albicans/cytology , Candida albicans/metabolism , Cell Cycle Proteins/physiology , Fungal Proteins/physiology , Transcription Factors/physiology , Amphotericin B/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Basic-Leucine Zipper Transcription Factors , Candida albicans/genetics , Cell Cycle Proteins/genetics , Fungal Proteins/genetics , Gene Deletion , Gene Expression Regulation, Fungal/drug effects , Genes, Fungal , Glutathione/metabolism , Glutathione Reductase/genetics , Glutathione Reductase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...