Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.601
1.
CNS Neurol Disord Drug Targets ; 21(4): 292-301, 2022.
Article En | MEDLINE | ID: mdl-34477538

BACKGROUND: Regulation of glutamate release is crucial for maintaining normal brain function, but excess glutamate release is implicated in many neuropathological conditions. Therefore, the minimum glutamate release from presynaptic nerve terminals is an important neuroprotective mechanism. OBJECTIVE: In this mini-review, we analyze the three B vitamins, namely vitamin B2 (riboflavin), vitamin B6 (pyridoxine), and vitamin B12 (cyanocobalamin), that affect the 4-aminopyridine (4- AP)-evoked glutamate release from presynaptic nerve terminal in rat and discuss their neuroprotective role. METHODS: In this study, the measurements include glutamate release, DiSC3(5), and Fura-2. RESULTS: The riboflavin, pyridoxine, and cyanocobalamin produced significant inhibitory effects on 4-aminopyridine-evoked glutamate release from rat cerebrocortical nerve terminals (synaptosomes) in a dose-dependent relationship. These presynaptic inhibitory actions of glutamate release are attributed to inhibition of physiologic Ca2+-dependent vesicular exocytosis but not Ca2+-independent nonvesicular release. These effects also did not affect membrane excitability, while diminished cytosolic (Ca2+)c through a reduction of direct Ca2+ influx via Cav2.2 (N-type) and Cav2.1 (P/Q-type) Ca2+ channels, rather than through indirect Ca2+induced Ca2+ release from ryanodine-sensitive intracellular stores. Furthermore, their effects were attenuated by GF109203X and Ro318220, two protein kinase C (PKC) inhibitors, suggesting suppression of PKC activity. Taken together, these results suggest that riboflavin, pyridoxine, and cyanocobalamin inhibit presynaptic vesicular glutamate release from rat cerebrocortical synaptosomes, through the depression Ca2+ influx via voltage- dependent Cav2.2 (N-type) and Cav2.1 (P/Q-type) Ca2+ channels, and PKC signaling cascade. CONCLUSION: Therefore, these B vitamins may reduce the strength of glutamatergic synaptic transmission and is of considerable importance as potential targets for therapeutic agents in glutamate- induced excitation-related diseases.


Glutamic Acid/metabolism , Synaptic Transmission/drug effects , Vitamin B Complex/metabolism , 4-Aminopyridine , Animals , Calcium/metabolism , Calcium Channels, N-Type , Cerebral Cortex/metabolism , Male , Membrane Potentials/drug effects , Presynaptic Terminals/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synaptosomes/drug effects
2.
Biomolecules ; 11(10)2021 10 18.
Article En | MEDLINE | ID: mdl-34680170

Reduction in glutamate release is a key mechanism for neuroprotection and we investigated the effect of isoliquiritigenin (ISL), an active ingredient of Glycyrrhiza with neuroprotective activities, on glutamate release in rat cerebrocortical nerve terminals (synaptosomes). ISL produced a concentration-dependent inhibition of glutamate release and reduced the intraterminal [Ca2+] increase. The inhibition of glutamate release by ISL was prevented after removing extracellular Ca2+ or blocking P/Q-type Ca2+ channels. This inhibition was mediated through the γ-aminobutyric acid type B (GABAB) receptors because ISL was unable to inhibit glutamate release in the presence of baclofen (an GABAB agonist) or CGP3548 (an GABAB antagonist) and docking data revealed that ISL interacted with GABAB receptors. Furthermore, the ISL inhibition of glutamate release was abolished through the inhibition of Gi/o-mediated responses or Gßγ subunits, but not by 8-bromoadenosine 3',5'-cyclic monophosphate or adenylate cyclase inhibition. The ISL inhibition of glutamate release was also abolished through the inhibition of protein kinase C (PKC), and ISL decreased the phosphorylation of PKC. Thus, we inferred that ISL, through GABAB receptor activation and Gßγ-coupled inhibition of P/Q-type Ca2+ channels, suppressed the PKC phosphorylation to cause a decrease in evoked glutamate release at rat cerebrocortical nerve terminals.


Chalcones/pharmacology , Glycyrrhiza/chemistry , Receptors, GABA-B/genetics , Synaptosomes/drug effects , Animals , Baclofen/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Calcium/metabolism , Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Chalcones/chemistry , GABA-B Receptor Antagonists/pharmacology , Glutamic Acid/biosynthesis , Humans , Rats , Synaptosomes/metabolism
3.
Biomolecules ; 11(9)2021 08 25.
Article En | MEDLINE | ID: mdl-34572487

BACKGROUND: In the brain, polyamines are mainly synthesized in neurons, but preferentially accumulated in astrocytes, and are proposed to be involved in neurodegenerative/neuroinflammatory disorders and neuron injury. A transgenic mouse overexpressing spermine oxidase (SMOX, which specifically oxidizes spermine) in the neocortex neurons (Dach-SMOX mouse) was proved to be a model of increased susceptibility to excitotoxic injury. METHODS: To investigate possible alterations in synapse functioning in Dach-SMOX mouse, both cerebrocortical nerve terminals (synaptosomes) and astrocytic processes (gliosomes) were analysed by assessing polyamine levels, ezrin and vimentin content, glutamate AMPA receptor activation, calcium influx, and catalase activity. RESULTS: The main findings are as follows: (i) the presence of functional calcium-permeable AMPA receptors in synaptosomes from both control and Dach-SMOX mice, and in gliosomes from Dach-SMOX mice only; (ii) reduced content of spermine in gliosomes from Dach-SMOX mice; and (iii) down-regulation and up-regulation of catalase activity in synaptosomes and gliosomes, respectively, from Dach-SMOX mice. CONCLUSIONS: Chronic activation of SMOX in neurons leads to major changes in the astrocyte processes including reduced spermine levels, increased calcium influx through calcium-permeable AMPA receptors, and stimulation of catalase activity. Astrocytosis and the astrocyte process alterations, depending on chronic activation of polyamine catabolism, result in synapse dysregulation and neuronal suffering.


Gliosis/metabolism , Gliosis/pathology , Polyamines/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Calcium/metabolism , Catalase/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Nerve Endings/drug effects , Nerve Endings/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Receptors, AMPA/metabolism , Spermine/analogs & derivatives , Spermine/metabolism , Spermine/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , Vimentin/metabolism
4.
EMBO J ; 40(21): e107915, 2021 11 02.
Article En | MEDLINE | ID: mdl-34585770

Synaptic refinement is a critical physiological process that removes excess synapses to establish and maintain functional neuronal circuits. Recent studies have shown that focal exposure of phosphatidylserine (PS) on synapses acts as an "eat me" signal to mediate synaptic pruning. However, the molecular mechanism underlying PS externalization at synapses remains elusive. Here, we find that murine CDC50A, a chaperone of phospholipid flippases, localizes to synapses, and that its expression depends on neuronal activity. Cdc50a knockdown leads to phosphatidylserine exposure at synapses and subsequent erroneous synapse removal by microglia partly via the GPR56 pathway. Taken together, our data support that CDC50A safeguards synapse maintenance by regulating focal phosphatidylserine exposure at synapses.


Membrane Proteins/genetics , Microglia/drug effects , Neurons/drug effects , Phosphatidylserines/pharmacology , Receptors, G-Protein-Coupled/genetics , Synapses/drug effects , Animals , Gene Expression Regulation , Genes, Reporter , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/metabolism , Neuronal Plasticity , Neurons/cytology , Neurons/metabolism , Phosphatidylserines/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/genetics , Synapses/metabolism , Synaptic Transmission , Synaptosomes/drug effects , Synaptosomes/metabolism , Vesicular Glutamate Transport Protein 2
5.
Biomolecules ; 11(7)2021 07 15.
Article En | MEDLINE | ID: mdl-34356653

Rosmarinic acid, a major component of rosemary, is a polyphenolic compound with potential neuroprotective effects. Asreducing the synaptic release of glutamate is crucial to achieving neuroprotectant's pharmacotherapeutic effects, the effect of rosmarinic acid on glutamate release was investigated in rat cerebrocortical nerve terminals (synaptosomes). Rosmarinic acid depressed the 4-aminopyridine (4-AP)-induced glutamate release in a concentration-dependent manner. The removal of extracellular calcium and the blockade of vesicular transporters prevented the inhibition of glutamate release by rosmarinic acid. Rosmarinic acid reduced 4-AP-induced intrasynaptosomal Ca2+ elevation. The inhibition of N-, P/Q-type Ca2+ channels and the calcium/calmodulin-dependent kinase II (CaMKII) prevented rosmarinic acid from having effects on glutamate release. Rosmarinic acid also reduced the 4-AP-induced activation of CaMKII and the subsequent phosphorylation of synapsin I, the main presynaptic target of CaMKII. In addition, immunocytochemistry confirmed the presence of GABAA receptors. GABAA receptor agonist and antagonist blocked the inhibitory effect of rosmarinic acid on 4-AP-evoked glutamate release. Docking data also revealed that rosmarinic acid formed a hydrogen bond with the amino acid residues of GABAA receptor. These results suggested that rosmarinic acid activates GABAA receptors in cerebrocortical synaptosomes to decrease Ca2+ influx and CaMKII/synapsin I pathway to inhibit the evoked glutamate release.


Cinnamates/pharmacology , Depsides/pharmacology , Glutamic Acid/metabolism , Synaptosomes/drug effects , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cinnamates/chemistry , Depsides/chemistry , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Male , Membrane Potentials/drug effects , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Synaptosomes/metabolism
6.
J Neurochem ; 159(1): 156-171, 2021 10.
Article En | MEDLINE | ID: mdl-34309872

The regulation of the serotonin transporter (SERT) by guanine nucleotide-binding protein alpha (Gα) q was investigated using Gαq knockout mice. In the absence of Gαq, SERT-mediated uptake of 5-hydroxytryptamine (5HT) was enhanced in midbrain and frontal cortex synaptosomes, but only in female mice. The mechanisms underlying this sexual dimorphism were investigated using quantitative western blot analysis revealing brain region-specific differences. In the frontal cortex, SERT protein expression was decreased in male knockout mice, seemingly explaining the sex-dependent variation in SERT activity. The differential expression of Gαi1 in female mice contributes to the sex differences in the midbrain. In fact, Gαi1 levels inversely correlate with 5HT uptake rates across both sexes and genotypes. Likely due to differential SERT regulation as well as sex differences in the expression of tryptophan hydroxylase 2, Gαq knockout mice also displayed sex- and genotype-dependent alterations in total 5HT tissue levels as determined by high-performance liquid chromatography. Gαq inhibitors, YM-254890 and BIM-46187, differentially affected SERT activity in both, synaptosomes and cultured cells. YM-254890 treatment mimicked the effect of Gαq knockout in the frontal cortex. BIM-46187, which promotes the nucleotide-free form of Gα proteins, substantially inhibited 5HT uptake, prompting us to hypothesise that Gαq interacts with SERT similarly as with G-protein-coupled receptors and inhibits SERT activity by modulating transport-associated conformational changes. Taken together, our findings reveal a novel mechanism of SERT regulation and impact our understanding of sex differences in diseases associated with dysregulation of serotonin transmission, such as depression and anxiety.


Brain/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , Serotonin Plasma Membrane Transport Proteins/metabolism , Sex Characteristics , Synaptosomes/metabolism , Animals , Brain/drug effects , Female , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Peptides, Cyclic/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Synaptosomes/drug effects
7.
Exp Neurol ; 345: 113811, 2021 11.
Article En | MEDLINE | ID: mdl-34298012

Methamphetamine (METH) is a highly addictive and powerful central nervous system psychostimulant with no FDA-approved pharmacotherapy. Parkin is a neuroprotective protein and its loss of function contributes to Parkinson's disease. This study used 3-month-old homozygous parkin knockout (PKO) rats to determine whether loss of parkin protein potentiates neurotoxicity of chronic METH to the nigrostriatal dopamine pathway. PKO rats were chronically treated with 10 mg/kg METH for 10 consecutive days and assessed for neurotoxicity markers in the striatum on the 5th and 10th day of withdrawal from METH. The PKO rats showed higher METH-induced hyperthermia; however, they did not display augmented deficits in dopaminergic and serotonergic neurotoxicity markers, astrocyte activation or decreased mitochondrial enzyme levels as compared to wild-type (WT) rats. Interestingly, saline-treated PKO rats had lower levels of dopamine (DA) as well as mitochondrial complex I and II levels while having increased basal levels of glial fibrillary acidic protein (GFAP), a marker of gliosis. These results indicate PKO display a certain resistance to METH neurotoxicity, possibly mediated by lowered DA levels and downregulated mitochondria.


Central Nervous System Stimulants/toxicity , Dopamine/metabolism , Locomotion/drug effects , Methamphetamine/toxicity , Ubiquitin-Protein Ligases/deficiency , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Central Nervous System Stimulants/administration & dosage , Dopamine/genetics , Drug Administration Schedule , Hyperthermia, Induced/adverse effects , Hyperthermia, Induced/methods , Locomotion/physiology , Male , Methamphetamine/administration & dosage , Rats , Rats, Long-Evans , Rats, Transgenic , Synaptosomes/drug effects , Synaptosomes/metabolism , Ubiquitin-Protein Ligases/genetics
8.
J Neurochem ; 159(1): 116-127, 2021 10.
Article En | MEDLINE | ID: mdl-34320222

Methcathinone (MCAT) is a psychostimulant of abuse that can cause both persistent striatal dopaminergic and serotonergic, as well as hippocampal serotonergic, deficits. Evidence suggests that the rapid effects of stimulants that are structurally and mechanistically similar to MCAT on monoamine transporter function may contribute to the abuse liability and/or persistent monoaminergic deficits caused by these agents. Thus, effects of MCAT on 1) striatal dopamine (DA) transporter (DAT); and 2) striatal and hippocampal serotonin transporter (SERT) function, as determined in tissues from adult male rats, were assessed. As reported previously, a single administration of MCAT rapidly (within 1 hr) decreases striatal [3 H]DA uptake. Similarly, incubation of rat synaptosomes with MCAT at 37℃ (but not 4˚C) decreased striatal [3 H]DA uptake. Incubation with MCAT likewise decreased [3 H]5HT but not vesicular [3 H]DA uptake. MCAT incubation in vitro was without effect on [3 H]DA uptake in striatal synaptosomes prepared from MCAT-treated rats. The decrease in [3 H]DA uptake caused by MCAT incubation: (a) reflected a decrease in Vmax , with minimal change in Km , and (b) was attenuated by co-incubation with the cell-permeable calcium chelator, N,N'-[1,2-ethanediylbis(oxy-2,1-phenylene)]bis[N-[2-[(acetyloxy)methoxy]-2-oxoethyl]-1,1'-bis[(acetyloxy)methyl] ester-glycine (BAPTA-AM), as well as the non-selective protein kinase-C (PKC) inhibitors bisindolylmaleimide-1 (BIM-1) and 2-[1-3(Aminopropyl)indol-3-yl]-3(1-methyl-1H-indol-3-yl)maleimide (or Bisindolylmaleimide VIII; Ro-31-7549). Taken together, these results suggest that in vitro MCAT incubation may model important aspects of MCAT administration in vivo, and that calcium and PKC contribute to the in vitro effects of MCAT on DAT.


Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/physiology , Propiophenones/pharmacology , Protein Kinase C/physiology , Animals , Corpus Striatum/drug effects , Corpus Striatum/physiology , Male , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/physiology
9.
Neurochem Int ; 148: 105100, 2021 09.
Article En | MEDLINE | ID: mdl-34139299

Nitric oxide generation is related to the activity of certain proteins located at synaptic sites. Previous findings show that NOS activity, nNOS protein expression, respiratory parameters and mitochondrial complex activities are altered in rat cerebral cortex by administration of levocabastine, an antagonist of histamine H1 and neurotensin NTS2 receptors. ATP provision by mitochondria may play an important role in the functional interaction between synaptic proteins NMDA receptor and PSD-95 with NO synthesis. In this context, our purpose was to evaluate the effect of levocabastine administration on protein expression of PSD-95, GluN2B and iNOS, as well as on mitochondrial ATP production. Male Wistar rats received a single (i.p.) dose of levocabastine (50 µg/kg) or saline solution (controls) and were decapitated 18 h later. Mitochondrial and synaptosomal membrane fractions were isolated from cerebral cortex by differential and sucrose gradient centrifugation. Expression of synaptic proteins was evaluated by Western blot assays in synaptosomal membrane fractions. Oxygen consumption, mitochondrial membrane potential and ATP production rate were determined in fresh crude mitochondrial fractions. After levocabastine treatment, protein expression of PSD-95, GluN2B and ß-actin decreased 97, 45 and 55%, respectively, whereas that of iNOS enhanced 3.5-fold versus controls. In crude mitochondrial fractions levocabastine administration reduced roughly 15% respiratory control rate as assayed with malate-glutamate or succinate as substrates, decreased mitochondrial membrane potential (21%), and ATP production rates (57%). Results suggested that levocabastine administration induces alterations in synaptic proteins of the protein complex PSD-95/NMDA receptor/nNOS and in neuron cytoskeleton. Mitochondrial bioenergetics impairment may play a role in the functional link between synaptic proteins and NO synthesis.


Disks Large Homolog 4 Protein/metabolism , Histamine H1 Antagonists/pharmacology , Nitric Oxide Synthase Type II/metabolism , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Nitric Oxide Synthase Type II/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects , Synaptosomes/drug effects
10.
Front Immunol ; 12: 586521, 2021.
Article En | MEDLINE | ID: mdl-33717067

Antibodies recognizing the amino-terminal domain of receptor subunit proteins modify the receptor efficiency to controlling transmitter release in isolated nerve endings (e.g., synaptosomes) indirectly confirming their presence in these particles but also allowing to speculate on their subunit composition. Western blot analysis and confocal microscopy unveiled the presence of the GluA1, GluA2, GluA3, and GluA4 receptor subunits in cortical synaptosomes. Functional studies confirmed the presence of presynaptic release-regulating AMPA autoreceptors in these terminals, whose activation releases [3H]D-aspartate ([3H]D-Asp, here used as a marker of glutamate) in a NBQX-dependent manner. The AMPA autoreceptors traffic in a constitutive manner, since entrapping synaptosomes with the pep2-SVKI peptide (which interferes with the GluA2-GRIP1/PICK1 interaction) amplified the AMPA-evoked releasing activity, while the inactive pep2-SVKE peptide was devoid of activity. Incubation of synaptosomes with antibodies recognizing the NH2 terminus of the GluA2 and the GluA3 subunits increased, although to a different extent, the GluA2 and 3 densities in synaptosomal membranes, also amplifying the AMPA-evoked glutamate release in a NBQX-dependent fashion. We then analyzed the releasing activity of complement (1:300) from both treated and untreated synaptosomes and found that the complement-induced overflow occurred in a DL-t-BOA-sensitive, NBQX-insensitive fashion. We hypothesized that anti-GluA/GluA complexes in neuronal membranes could trigger the classic pathway of activation of the complement, modifying its releasing activity. Accordingly, the complement-evoked release of [3H]D-Asp from antiGluA2 and anti-GluA3 antibody treated synaptosomes was significantly increased when compared to untreated terminals and facilitation was prevented by omitting the C1q component of the immunocomplex. Antibodies recognizing the NH2 terminus of the GluA1 or the GluA4 subunits failed to affect both the AMPA and the complement-evoked tritium overflow. Our results suggest the presence of GluA2/GluA3-containing release-regulating AMPA autoreceptors in cortical synaptosomes. Incubation of synaptosomes with commercial anti-GluA2 or anti-GluA3 antibodies amplifies the AMPA-evoked exocytosis of glutamate through a complement-independent pathway, involving an excessive insertion of AMPA autoreceptors in plasma membranes but also affects the complement-dependent releasing activity, by promoting the classic pathway of activation of the immunocomplex. Both events could be relevant to the development of autoimmune diseases typified by an overproduction of anti-GluA subunits.


Antibodies/pharmacology , Protein Interaction Domains and Motifs/drug effects , Protein Subunits/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Complement C1q/immunology , Fluorescent Antibody Technique , Male , Mice , Receptors, AMPA/chemistry , Synaptosomes/drug effects , Synaptosomes/metabolism
11.
Chem Res Toxicol ; 34(5): 1286-1295, 2021 05 17.
Article En | MEDLINE | ID: mdl-33621091

Glutamate is the major excitatory neurotransmitter in the brain and is involved in many brain functions. In this study, we investigated whether typhaneoside, a flavonoid from Typhae angustifolia pollen, affects endogenous glutamate release from rat cortical synaptosomes. Using a one-line enzyme-coupled fluorometric assay, glutamate release stimulated by the K+ channel blocker 4-aminopyridine was monitored to explore the possible underlying mechanisms. The vesicular transporter inhibitor bafilomycin A1 and chelation of extracellular Ca2+ ions with EGTA suppressed the effect of typhaneoside on the induced glutamate release. Nevertheless, the typhaneoside activity has not been affected by the glutamate transporter inhibitor dl-threo-beta-benzyloxyaspartate. The synaptosomal plasma membrane potential was assayed using a membrane potential-sensitive dye DiSC3(5), and cytosolic Ca2+ concentrations ([Ca2+]C) was monitored by a Ca2+ indicator Fura-2. Results showed that typhaneoside did not alter the synaptosomal membrane potential but lowered 4-aminopyridine-induced increases in [Ca2+]C. Furthermore, the Cav2.2 (N-type) channel blocker ω-conotoxin GVIA blocked Ca2+ entry and inhibited the effect of typhaneoside on 4-aminopyridine-induced glutamate release. However, the inhibitor of intracellular Ca2+ release dantrolene and the mitochondrial Na+/Ca2+ exchanger blocker 7-chloro-5-(2-chloropheny)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one have no effect on the suppression of glutamate release mediated by typhaneoside. Moreover, inhibition of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) prevented the inhibitory effect of typhaneoside on induced glutamate release. Typhaneoside reduced 4-aminopyridine-induced phosphorylation of ERK1/2 and the major presynaptic ERK target synapsin I, which is a synaptic vesicle-associated protein. In conclusion, these findings suggest a role for typhaneoside in modulating glutamate release by suppressing voltage-dependent Ca2+ channel mediated presynaptic Ca2+ influx and the MAPK/ERK/synapsin I signaling cascade.


Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Cerebral Cortex/drug effects , Glutamic Acid/metabolism , Glycosides/pharmacology , Animals , Cerebral Cortex/metabolism , Male , Membrane Potentials/drug effects , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism
12.
Eur J Pharmacol ; 898: 173986, 2021 May 05.
Article En | MEDLINE | ID: mdl-33640406

The high-affinity choline transporter CHT1 mediates choline uptake, the rate-limiting and regulatory step in acetylcholine synthesis at cholinergic presynaptic terminals. CHT1-medated choline uptake is specifically inhibited by hemicholinium-3, which is a type of choline analog that acts as a competitive inhibitor. Although the substrate choline and the inhibitor hemicholinium-3 are well-established ligands of CHT1, few potent ligands other than choline analogs have been reported. Here we show that tetrahydropyrimidine anthelmintics, known as nicotinic acetylcholine receptor agonists, act as competitive inhibitors of CHT1. A ligand-dependent trafficking assay in cell lines expressing human CHT1 was designed to search for CHT1 ligands from a collection of biologically active compounds. We found that morantel as well as other tetrahydropyrimidines, pyrantel and oxantel, potently inhibits the high-affinity choline uptake activity of CHT1 in a competitive manner similar to the inhibitor hemicholinium-3. They also inhibit the high-affinity choline transporter from the nematode Caenorhabditis elegans. Finally, tetrahydropyrimidines potently inhibit the high-affinity choline uptake in rat brain synaptosomes at a low micromolar level, resulting in the inhibition of acetylcholine synthesis. The rank order of potency in synaptosomes is as follows: morantel > pyarantel > oxantel (Ki = 1.3, 5.7, and 8.3 µM, respectively). Our results reveal that tetrahydropyrimidine anthelmintics are novel CHT1 ligands that inhibit the high-affinity choline uptake for acetylcholine synthesis in cholinergic neurons.


Anthelmintics/pharmacology , Brain/drug effects , Cation Transport Proteins/antagonists & inhibitors , Choline/metabolism , Pyrimidines/pharmacology , Symporters/antagonists & inhibitors , Animals , Anthelmintics/metabolism , Binding, Competitive , Biological Transport , Brain/metabolism , Cation Transport Proteins/metabolism , Female , HEK293 Cells , Humans , Ligands , Mice , Morantel/metabolism , Morantel/pharmacology , Protein Binding , Protein Transport , Pyrantel/analogs & derivatives , Pyrantel/metabolism , Pyrantel/pharmacology , Pyrimidines/metabolism , Symporters/genetics , Symporters/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
13.
Neurochem Res ; 46(4): 804-818, 2021 Apr.
Article En | MEDLINE | ID: mdl-33428094

In this study, we were aimed to investigate the neuroprotective effects of bexarotene and nicotinamide in synaptosomes incubated with amyloid-beta (Aß). Our study consists of 2 parts, in vivo and in vitro. In the in vivo section, twenty-four Wistar albino male rats were divided into 4 groups (control, dimethyl sulfoxide (DMSO), nicotinamide and bexarotene) with six animals in each group. DMSO(1%), nicotinamide(100 mg/kg) and bexarotene(0.1 mg/kg) were administered intraperitoneally to animals in the experimental groups for seven days. In the in vitro part of our study, three different isolation methods were used to obtain the synaptosomes from the brain tissue. Total antioxidant capacity(TAS), total oxidant capacity(TOS), cleaved caspase 3(CASP3), cytochrome c(Cyt c), sirtuin 1(SIRT1), peroxisome proliferator-activated receptor gamma(PPARγ) and poly(ADP-ribose) polymerase-1(PARP-1) levels in the synaptosomes incubated with a concentration of 10 µM Aß(1-42) were measured by enzyme-linked immunosorbent assay method. Biochemical analysis and histopathological examinations in serum and brain samples showed that DMSO, nicotinamide and bexarotene treatments did not cause any damage to the rat brain tissue. We found that in vitro Aß(1-42) administration decreased TAS, SIRT1 and PPARγ levels in synaptosomes while increasing TOS, CASP3, Cyt c, and PARP1 levels. Nicotinamide treatment suppressed oxidative stress and apoptosis by supporting antioxidant capacity and increased PPARγ through SIRT1 activation, causing PARP1 to decrease. On the other hand, bexarotene caused a moderate increase in SIRT1 levels with PPARγ activation. Consequently, we found that nicotinamide can be more effective than bexarotene in AD pathogenesis by regulating mitochondrial functions in synaptosomes.


Bexarotene/pharmacology , Neuroprotective Agents/pharmacology , Niacinamide/pharmacology , Synaptosomes/drug effects , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Male , Oxidative Stress/drug effects , PPAR gamma/metabolism , Peptide Fragments , Poly (ADP-Ribose) Polymerase-1/metabolism , Rats, Wistar , Signal Transduction/drug effects , Sirtuin 1/metabolism , Synaptosomes/metabolism
14.
Article En | MEDLINE | ID: mdl-33400944

Ketamine produces a rapid antidepressant effect, but its use can be associated with serious side effects. Hence, other therapeutic options that will allow us to obtain a quick and safe antidepressant effect by modulating glutamatergic transmission are needed. Antagonists of mGlu2/3 receptors, which share some mechanisms of action with ketamine, may be good candidates to obtain this effect. Here, we show that the metabotropic glutamate (mGlu) 2/3 receptor antagonist LY341495 induced a dose-dependent antidepressant-like effect in the chronic unpredictable mild stress (CUMS) model of depression in C57BL/6J mice after both single and subchronic (three-day) administration. Furthermore, a noneffective dose of LY341495 (0.3 mg/kg) given jointly with a noneffective dose of ketamine (3 mg/kg) reversed the CUMS-induced behavioral effects, indicating that coadministration of ketamine with an mGlu2/3 receptor antagonist might allow its therapeutically effective dose to be lowered. Western blot results indicate that mTOR pathway activation might be involved in the mechanism of action of this drug combination. Moreover, the combined doses of both substances did not produce undesirable behavioral effects characteristic of a higher dose of ketamine (10 mg/kg) commonly used in rodent studies to induce antidepressant effects. Coadministration of low doses of ketamine and LY341495 did not induce the hyperactivity typical of NMDA channel blockers, did not disturb short-term memory in the novel object recognition (NOR) test, and did not disturb motor coordination in the rotarod test. Our research not only confirmed the earlier data on the rapid antidepressant effect of mGlu2/3 receptor antagonists but also indicated that such compounds can safely lower the effective dose of ketamine.


Amino Acids/therapeutic use , Antidepressive Agents/therapeutic use , Depression/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Ketamine/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Xanthenes/therapeutic use , Amino Acids/pharmacology , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Ketamine/pharmacology , Male , Mice , Mice, Inbred C57BL , Motor Skills/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Xanthenes/pharmacology
15.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article En | MEDLINE | ID: mdl-35008791

Methamphetamine (METH) is a highly abused psychostimulant that is neurotoxic to dopaminergic (DAergic) nerve terminals in the striatum and increases the risk of developing Parkinson's disease (PD). In vivo, METH-mediated DA release, followed by DA-mediated oxidative stress and mitochondrial dysfunction in pre- and postsynaptic neurons, mediates METH neurotoxicity. METH-triggered oxidative stress damages parkin, a neuroprotective protein involved in PD etiology via its involvement in the maintenance of mitochondria. It is not known whether METH itself contributes to mitochondrial dysfunction and whether parkin regulates complex I, an enzymatic complex downregulated in PD. To determine this, we separately assessed the effects of METH or DA alone on electron transport chain (ETC) complexes and the protein parkin in isolated striatal mitochondria. We show that METH decreases the levels of selected complex I, II, and III subunits (NDUFS3, SDHA, and UQCRC2, respectively), whereas DA decreases the levels only of the NDUFS3 subunit in our preparations. We also show that the selected subunits are not decreased in synaptosomal mitochondria under similar experimental conditions. Finally, we found that parkin overexpression does not influence the levels of the NDUFS3 subunit in rat striatum. The presented results indicate that METH itself is a factor promoting dysfunction of striatal mitochondria; therefore, it is a potential drug target against METH neurotoxicity. The observed decreases in ETC complex subunits suggest that DA and METH decrease activities of the ETC complexes via oxidative damage to their subunits and that synaptosomal mitochondria may be somewhat "resistant" to DA- and METH-induced disruption in mitochondrial ETC complexes than perikaryal mitochondria. The results also suggest that parkin does not regulate NDUFS3 turnover in rat striatum.


Corpus Striatum/metabolism , Dopamine/pharmacology , Methamphetamine/toxicity , Neurotoxins/toxicity , Ubiquitin-Protein Ligases/metabolism , Animals , Buffers , Corpus Striatum/drug effects , Electron Transport/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , NADH Dehydrogenase/metabolism , Protein Subunits/metabolism , Rats , Synaptosomes/drug effects , Synaptosomes/metabolism
16.
Neurochem Res ; 46(1): 100-107, 2021 Jan.
Article En | MEDLINE | ID: mdl-32130629

Reductions in the activities of mitochondrial electron transport chain (ETC) enzymes have been implicated in the pathogenesis of numerous chronic neurodegenerative disorders. Maintenance of the mitochondrial membrane potential (Δψm) is a primary function of these enzyme complexes, and is essential for ATP production and neuronal survival. We examined the effects of inhibition of mitochondrial ETC complexes I, II/III, III and IV activities by titrations of respective inhibitors on Δψm in synaptosomal mitochondria. Small perturbations in the activity of complex I, brought about by low concentrations of rotenone (1-50 nM), caused depolarisation of Δψm. Small decreases in complex I activity caused an immediate and partial Δψm depolarisation, whereas inhibition of complex II/III activity by more than 70% with antimycin A was required to affect Δψm. A similarly high threshold of inhibition was found when complex III was inhibited with myxothiazol, and inhibition of complex IV by more than 90% with KCN was required. The plasma membrane potential (Δψp) had a complex I inhibition threshold of 40% whereas complex III and IV had to be inhibited by more than 90% before changes in Δψp were registered. These data indicate that in synaptosomes, both Δψm and Δψp are more susceptible to reductions in complex I activity than reductions in the other ETC complexes. These findings may be of relevance to the mechanism of neuronal cell death in Parkinson's disease in particular, where such reductions in complex I activity are present.


Electron Transport Complex I/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Animals , Antimycin A/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Enzyme Assays , Enzyme Inhibitors/pharmacology , Female , Membrane Potential, Mitochondrial/drug effects , Methacrylates/pharmacology , Mitochondria/drug effects , Potassium Cyanide/pharmacology , Rats, Wistar , Rotenone/pharmacology , Synaptosomes/drug effects , Thiazoles/pharmacology
17.
Bioorg Chem ; 107: 104529, 2021 02.
Article En | MEDLINE | ID: mdl-33339665

In our screening program for new biologically active secondary metabolites, nine new polycyclic polyprenyled acylphloroglucinols, hyperscabins D-L, together with three known compounds, were obtained from the aerial parts of Hypericum scabrum. The chemical structures of 1-9 were characterized by extensive spectroscopic analyses, nuclear magnetic resonance calculation with DP4+ probability analysis, and the electronic circular dichroism spectra were calculated. Compound 1 was an unusual prenylated acylphloroglucinol decorated with a 5-oxaspiro [4,5] deca-1,9-dione skeleton. Compound 2 was a newly identified spirocyclic polyprenylated acylphloroglucinol possessing a rare 5,5-spiroketal segment. Compounds 3, 8, and 10 (10 µM) exhibited pronounced hepatoprotective activity against d-galactosamine-induced WB-F344 cell damage in vitro assays. All test compounds (1, 3, and 7-12) demonstrated potential inhibitory effects at 10 µM against noradrenalinet ([3H]-NE) reuptake in rat brain synaptosome.


Antidepressive Agents/pharmacology , Hemiterpenes/pharmacology , Hypericum/chemistry , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Protective Agents/pharmacology , Animals , Antidepressive Agents/chemical synthesis , Antidepressive Agents/isolation & purification , Cell Line , Hemiterpenes/chemical synthesis , Hemiterpenes/isolation & purification , Neurotransmitter Uptake Inhibitors/chemical synthesis , Neurotransmitter Uptake Inhibitors/isolation & purification , Neurotransmitter Uptake Inhibitors/pharmacology , Norepinephrine/metabolism , Phloroglucinol/isolation & purification , Plant Components, Aerial/chemistry , Protective Agents/chemical synthesis , Protective Agents/isolation & purification , Rats , Synaptosomes/drug effects , Synaptosomes/metabolism
18.
Food Funct ; 11(11): 9858-9867, 2020 Nov 18.
Article En | MEDLINE | ID: mdl-33089839

3ß,7ß,25-Trihydroxycucurbita-5,23(E)-dien-19-al (TCD) is a triterpenoid isolated from wild bitter gourd that is a common tropical vegetable with neuroprotective effects. Because excessive glutamate release is a major cause of neuronal damage in various neurological disorders, the aims of this study were to examine the effect of TCD on glutamate release in vitro and to examine the effect of TCD in vivo. In rat cerebrocortical synaptosomes, TCD reduced 4-aminopyridine (4-AP)-stimulated glutamate release and Ca2+ concentration elevation, but had no effect on plasma membrane potential. TCD-mediated inhibition of 4-AP-induced glutamate release was dependent on the presence of extracellular calcium; persisted in the presence of the glutamate transporter inhibitor dl-TBOA, P/Q-type Ca2+ channel blocker ω-agatoxin IVA, and intracellular Ca2+-releasing inhibitors dantrolene and CGP37157; and was blocked by the vesicular transporter inhibitor bafilomycin A1 and the N-type Ca2+ channel blocker ω-conotoxin GVIA. Molecular docking studies have demonstrated that TCD binds to N-type Ca2+ channels. TCD-mediated inhibition of 4-AP-induced glutamate release was abolished by the Ca2+-dependent protein kinase C (PKC) inhibitor Go6976, but was unaffected by the Ca2+-independent PKC inhibitor rottlerin. Furthermore, TCD considerably reduced the phosphorylation of PKC, PKCα, and myristoylated alanine-rich C kinase substrate, a major presynaptic substrate for PKC. In a rat model of kainic acid (KA)-induced excitotoxicity, TCD pretreatment substantially attenuated KA-induced neuronal death in the CA3 hippocampal region. These results suggest that TCD inhibits synaptosomal glutamate release by suppressing N-type Ca2+ channels and PKC activity and exerts protective effects against KA-induced excitotoxicity in vivo.


Glutamic Acid/metabolism , Kainic Acid/adverse effects , Momordica charantia/chemistry , Nervous System Diseases/drug therapy , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Synaptosomes/drug effects , Triterpenes/administration & dosage , Animals , Calcium/metabolism , Calcium Channel Blockers/administration & dosage , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Death/drug effects , Humans , Male , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism
19.
Brain Res Bull ; 165: 263-271, 2020 12.
Article En | MEDLINE | ID: mdl-33049353

Synaptic dopamine (DA) concentrations are largely determined by the activities of presynaptic D2 and D3 autoreceptors (D2R and D3R) and DA transporter (DAT). Furthermore, the activity of DAT is regulated by phosphorylation events and protein interactions that affect its surface expression. Because DA autoreceptors and DAT coordinately maintain synaptic DA homeostasis, we hypothesized that D3R might crosstalk with DAT to fine-tune synaptic DA concentrations. To test this hypothesis, we established [3H]DA uptake and DAT surface expression assays in hD3/rDAT-double-transfected HEK-293 cells or limbic forebrain synaptosomal preparations. Ropinirole, a preferential D3R agonist, reduced [3H]DA uptake in HEK-hD3/rDAT cells in a dose-dependent manner, an effect which could be blocked by the D2R/D3R antagonist, raclopride. Furthermore, ropinirole also reduced DAT surface expression in limbic forebrain synaptosomes, and this effect could be blocked by raclopride or the internalization inhibitor, concanavalin A. To identify potential mediators of this apparent D3R-DAT crosstalk, DAT-associated proteins were co-immunoprecipitated from limbic forebrain synaptosomes after D3R activation and identified by MALDI-TOF. From this analysis, the Hsc70 chaperone was identified as a DAT-associated protein. Interestingly, ropinirole induced the association of Hsc70/Hsp70 with DAT, and the Hsc70/Hsp70 inhibitor, apoptozole, prevented the ropinirole-induced reduction of DAT surface expression. Together, these results suggest that D3R negatively regulates DAT activity by promoting the association of DAT and Hsc70/Hsp70.


Dopamine Plasma Membrane Transport Proteins/metabolism , HSC70 Heat-Shock Proteins/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Receptors, Dopamine D3/metabolism , Animals , Dopamine Agonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , HEK293 Cells , HSC70 Heat-Shock Proteins/genetics , Humans , Indoles/pharmacology , Mice , Neurons/drug effects , Phosphorylation/drug effects , Prosencephalon/drug effects , Receptors, Dopamine D3/agonists , Synaptosomes/drug effects , Synaptosomes/metabolism
20.
Front Immunol ; 11: 2187, 2020.
Article En | MEDLINE | ID: mdl-33013920

Microvilli are finger-like membrane protrusions, supported by the actin cytoskeleton, and found on almost all cell types. A growing body of evidence suggests that the dynamic lymphocyte microvilli, with their highly curved membranes, play an important role in signal transduction leading to immune responses. Nevertheless, challenges in modulating local membrane curvature and monitoring the high dynamicity of microvilli hampered the investigation of the curvature-generation mechanism and its functional consequences in signaling. These technical barriers have been partially overcome by recent advancements in adapted super-resolution microscopy. Here, we review the up-to-date progress in understanding the mechanisms and functional consequences of microvillus formation in T cell signaling. We discuss how the deformation of local membranes could potentially affect the organization of signaling proteins and their biochemical activities. We propose that curved membranes, together with the underlying cytoskeleton, shape microvilli into a unique compartment that sense and process signals leading to lymphocyte activation.


Cell Membrane/immunology , Lymphocyte Activation/physiology , Microvilli/immunology , Signal Transduction/immunology , T-Lymphocytes/ultrastructure , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Carrier Proteins/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Shape , Cyclodextrins/pharmacology , Cytokines/physiology , Glycocalyx/drug effects , Glycocalyx/immunology , Humans , Lymphocyte Activation/drug effects , Membrane Lipids/immunology , Membrane Proteins/immunology , Mice , Microfilament Proteins/pharmacology , Microscopy, Electron, Scanning , Microvilli/drug effects , Microvilli/ultrastructure , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Stress, Mechanical , Surface Properties , Synaptosomes/drug effects , Synaptosomes/immunology , Synaptosomes/ultrastructure , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
...