Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Int J Mol Sci ; 25(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39201534

ABSTRACT

The purpose of this study was to investigate whether and how albiflorin, a natural monoterpene glycoside, affects the release of glutamate, one of the most important neurotransmitters involved in neurotoxicity, from cerebrocortical nerve terminals (synaptosomes) in rats. The results showed that albiflorin reduced 4-aminopyridine (4-AP)-elicited glutamate release from synaptosomes, which was abrogated in the absence of extracellular Ca2+ or in the presence of the vesicular glutamate transporter inhibitor or a P/Q-type Ca2+ channel inhibitor, indicating a mechanism of action involving Ca2+-dependent depression of vesicular exocytotic glutamate release. Albiflorin failed to alter the increase in the fluorescence intensity of 3,3-diethylthiacarbocyanine iodide (DiSC3(5)), a membrane-potential-sensitive dye. In addition, the suppression of protein kinase A (PKA) abolished the effect of albiflorin on glutamate release. Albiflorin also reduced the phosphorylation of PKA and synaptosomal-associated protein of 25 kDa (SNAP-25) and synapsin I at PKA-specific residues, which correlated with decreased available synaptic vesicles. The results of transmission electron microscopy (TEM) also observed that albiflorin reduces the release competence of synaptic vesicles evoked by 4-AP in synaptosomes. In conclusion, by studying synaptosomally released glutamate, we suggested that albiflorin reduces vesicular exocytotic glutamate release by decreasing extracellular Ca2+ entry via P/Q-type Ca2+ channels and reducing PKA-mediated synapsin I and SNAP-25 phosphorylation.


Subject(s)
Cerebral Cortex , Cyclic AMP-Dependent Protein Kinases , Glutamic Acid , Synaptosomes , Animals , Glutamic Acid/metabolism , Synaptosomes/metabolism , Synaptosomes/drug effects , Rats , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Male , Cyclic AMP-Dependent Protein Kinases/metabolism , Calcium Channels, Q-Type/metabolism , Rats, Sprague-Dawley , Calcium Channels, P-Type/metabolism , Bridged-Ring Compounds/pharmacology , Calcium/metabolism , Phosphorylation/drug effects , Synapsins/metabolism
2.
Molecules ; 28(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36770979

ABSTRACT

Inhibiting the excessive release of glutamate in the brain is emerging as a promising therapeutic option and is efficient for treating neurodegenerative disorders. The aim of this study is to investigate the effect and mechanism of plantainoside D (PD), a phenylenthanoid glycoside isolated from Plantago asiatica L., on glutamate release in rat cerebral cortical nerve terminals (synaptosomes). We observed that PD inhibited the potassium channel blocker 4-aminopyridine (4-AP)-evoked release of glutamate and elevated concentration of cytosolic Ca2+. Using bafilomycin A1 to block glutamate uptake into synaptic vesicles and EDTA to chelate extracellular Ca2+, the inhibitory effect of PD on 4-AP-evoked glutamate release was prevented. In contrast, the action of PD on the 4-AP-evoked release of glutamate in the presence of dl-TBOA, a potent nontransportable inhibitor of glutamate transporters, was unaffected. PD does not alter the 4-AP-mediated depolarization of the synaptosomal membrane potential, suggesting that the inhibitory effect of PD on glutamate release is associated with voltage-dependent Ca2+ channels (VDCCs) but not the modulation of plasma membrane potential. Pretreatment with the Ca2+ channel blocker (N-type) ω-conotoxin GVIA abolished the inhibitory effect of PD on the evoked glutamate release, as did pretreatment with the protein kinase C inhibitor GF109203x. However, the PD-mediated inhibition of glutamate release was eliminated by applying the mitochondrial Na+/Ca2+ exchanger inhibitor CGP37157 or dantrolene, which inhibits Ca2+ release through ryanodine receptor channels. These data suggest that PD mediates the inhibition of evoked glutamate release from synaptosomes primarily by reducing the influx of Ca2+ through N-type Ca2+ channels, subsequently reducing the protein kinase C cascade.


Subject(s)
4-Aminopyridine , Glutamic Acid , Rats , Animals , Glutamic Acid/metabolism , Rats, Sprague-Dawley , 4-Aminopyridine/pharmacology , Synaptosomes/metabolism , Calcium Signaling , Protein Kinase C/metabolism , Cerebral Cortex/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology
3.
Int J Mol Sci ; 23(5)2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35269784

ABSTRACT

The neurotransmitter glutamate plays an essential role in excitatory neurotransmission; however, excessive amounts of glutamate lead to excitotoxicity, which is the most common pathogenic feature of numerous brain disorders. This study aimed to investigate the role of butyl 2-[2-(2-fluorophenyl)acetamido]benzoate (HFP034), a synthesized anthranilate derivative, in the central glutamatergic system. We used rat cerebro-cortical synaptosomes to examine the effect of HFP034 on glutamate release. In addition, we used a rat model of kainic acid (KA)-induced glutamate excitotoxicity to evaluate the neuroprotective potential of HFP034. We showed that HFP034 inhibits 4-aminopyridine (4-AP)-induced glutamate release from synaptosomes, and this inhibition was absent in the absence of extracellular calcium. HFP034-mediated inhibition of glutamate release was associated with decreased 4-AP-evoked Ca2+ level elevation and had no effect on synaptosomal membrane potential. The inhibitory effect of HFP034 on evoked glutamate release was suppressed by blocking P/Q-type Ca2+ channels and protein kinase C (PKC). Furthermore, HFP034 inhibited the phosphorylation of PKC and its substrate, myristoylated alanine-rich C kinase substrate (MARCKS) in synaptosomes. We also observed that HFP034 pretreatment reduced neuronal death, glutamate concentration, glial activation, and the levels of endoplasmic reticulum stress-related proteins, calpains, glucose-regulated protein 78 (GRP 78), C/EBP homologous protein (CHOP), and caspase-12 in the hippocampus of KA-injected rats. We conclude that HFP034 is a neuroprotective agent that prevents glutamate excitotoxicity, and we suggest that this effect involves inhibition of presynaptic glutamate release through the suppression of P/Q-type Ca2+ channels and PKC/MARCKS pathways.


Subject(s)
Glutamic Acid , Synaptosomes , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Kainic Acid/pharmacology , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism , ortho-Aminobenzoates
4.
Int J Mol Sci ; 23(8)2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35456948

ABSTRACT

Current anti-seizure drugs fail to control approximately 30% of epilepsies. Therefore, there is a need to develop more effective anti-seizure drugs, and medicinal plants provide an attractive source for new compounds. This study aimed to evaluate the possible anti-seizure and neuroprotective effects of neferine, an alkaloid from the lotus seed embryos of Nelumbo nucifera, in a kainic acid (KA)-induced seizure rat model and its underlying mechanisms. Rats were intraperitoneally (i.p.) administrated neferine (10 and 50 mg/kg) 30 min before KA injection (15 mg/kg, i.p.). Neferine pretreatment increased seizure latency and reduced seizure scores, prevented glutamate elevation and neuronal loss, and increased presynaptic protein synaptophysin and postsynaptic density protein 95 expression in the hippocampi of rats with KA. Neferine pretreatment also decreased glial cell activation and proinflammatory cytokine (interleukin-1ß, interleukin-6, tumor necrosis factor-α) expression in the hippocampi of rats with KA. In addition, NOD-like receptor 3 (NLRP3) inflammasome, caspase-1, and interleukin-18 expression levels were decreased in the hippocampi of seizure rats pretreated with neferine. These results indicated that neferine reduced seizure severity, exerted neuroprotective effects, and ameliorated neuroinflammation in the hippocampi of KA-treated rats, possibly by inhibiting NLRP3 inflammasome activation and decreasing inflammatory cytokine secretion. Our findings highlight the potential of neferine as a therapeutic option in the treatment of epilepsy.


Subject(s)
Alkaloids , Antineoplastic Agents , Benzylisoquinolines , Neuroprotective Agents , Alkaloids/pharmacology , Alkaloids/therapeutic use , Animals , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Cytokines/metabolism , Inflammasomes/metabolism , Kainic Acid/adverse effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Seeds/metabolism , Seizures/chemically induced , Seizures/drug therapy
5.
Int J Mol Sci ; 23(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36362193

ABSTRACT

The inhibition of synaptic glutamate release to maintain glutamate homeostasis contributes to the alleviation of neuronal cell injury, and accumulating evidence suggests that natural products can repress glutamate levels and associated excitotoxicity. In this study, we investigated whether eupatilin, a constituent of Artemisia argyi, affected glutamate release in rat cortical nerve terminals (synaptosomes). Additionally, we evaluated the effect of eupatilin in an animal model of kainic acid (KA) excitotoxicity, particularly on the levels of glutamate and N-methyl-D-aspartate (NMDA) receptor subunits (GluN2A and GluN2B). We found that eupatilin decreased depolarization-evoked glutamate release from rat cortical synaptosomes and that this effect was accompanied by a reduction in cytosolic Ca2+ elevation, inhibition of P/Q-type Ca2+ channels, decreased synapsin I Ca2+-dependent phosphorylation and no detectable effect on the membrane potential. In a KA-induced glutamate excitotoxicity rat model, the administration of eupatilin before KA administration prevented neuronal cell degeneration, glutamate elevation, glutamate-generating enzyme glutaminase increase, excitatory amino acid transporter (EAAT) decrease, GluN2A protein decrease and GluN2B protein increase in the rat cortex. Taken together, the results suggest that eupatilin depresses glutamate exocytosis from cerebrocortical synaptosomes by decreasing P/Q-type Ca2+ channels and synapsin I phosphorylation and alleviates glutamate excitotoxicity caused by KA by preventing glutamatergic alterations in the rat cortex. Thus, this study suggests that eupatilin can be considered a potential therapeutic agent in the treatment of brain impairment associated with glutamate excitotoxicity.


Subject(s)
Artemisia , Neurotoxicity Syndromes , Rats , Animals , Glutamic Acid/metabolism , Synapsins/metabolism , Artemisia/metabolism , 4-Aminopyridine/pharmacology , Rats, Sprague-Dawley , Cerebral Cortex/metabolism , Calcium/metabolism , Synaptosomes/metabolism , Exocytosis , Kainic Acid/pharmacology , Neurotoxicity Syndromes/metabolism
6.
Int J Mol Sci ; 23(15)2022 Aug 06.
Article in English | MEDLINE | ID: mdl-35955884

ABSTRACT

Excessive glutamate release is known to be involved in the pathogenesis of neurological diseases, and suppression of glutamate release from nerve terminals is considered to be a treatment strategy. In this study, we investigated whether isosaponarin, a flavone glycoside isolated from wasabi leaves, could affect glutamate release in rat cerebral cortex nerve terminals (synaptosomes). The release of glutamate was evoked by the K+ channel blocker 4-aminopyridine (4-AP) and measured by an online enzyme-coupled fluorimetric assay. Isosaponarin produced a concentration-dependent inhibition of 4-AP-evoked glutamate release with a half-maximum inhibition of release value of 22 µM. The inhibition caused by isosaponarin was prevented by eliminating extracellular Ca2+ or by using bafilomycin A1, an inhibitor of synaptic vesicle exocytosis. Isosaponarin decreased intrasynaptosomal rises in Ca2+ levels that were induced by 4-AP, without affecting the synaptosomal membrane potential. The isosaponarin-induced inhibition of glutamate release was significantly prevented in synaptosomes that were pretreated with a combination of the calcium channel blockers ω-conotoxin GVIA (N-type) and ω-agatoxin IVA (P/Q-types). The protein kinase C (PKC) pan-inhibitor GF109203X and the Ca2+-dependent PKC inhibitor Go6976 abolished the inhibition of glutamate release by isosaponarin, while the Ca2+-independent PKC inhibitor rottlerin did not show any effect. The results from immunoblotting assays also showed that isosaponarin lowered PKC, PKCα, synaptosomal-associated protein of 25 kDa (SNAP-25), and myristoylated alanine-rich C-kinase substrate (MARCKS) phosphorylation induced by 4-AP. In addition, FM1-43-labeled synaptic vesicles in synaptosomes showed that treatment with isosaponarin resulted in an attenuation of the 4-AP-induced decrease in fluorescence intensity that is consistent with glutamate release. Transmission electron microscopy of synaptosomes also provided evidence that isosaponarin altered the number of synaptic vesicles. These results indicate that isosaponarin suppresses the Ca2+-dependent PKC/SNAP-25 and MARCKS pathways in synaptosomes, causing a decrease in the number of available synaptic vesicles, which inhibits vesicular glutamate release from synaptosomes.


Subject(s)
Glutamic Acid , Synaptosomes , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Membrane Potentials , Nerve Endings/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism
7.
Molecules ; 27(9)2022 Apr 20.
Article in English | MEDLINE | ID: mdl-35565989

ABSTRACT

The neuroprotective properties of piperine, the major alkaloid extracted from black pepper, have been under investigation, but its mechanism of action in excitotoxicity is still poorly understood. This study aimed to evaluate the protective effects of piperine with a focus on nerve growth factor (NGF) signalling in a kainic acid (KA) rat model of excitotoxicity. Rats were administered intraperitoneally (i.p.) piperine (10 or 50 mg/kg) before KA injection (15 mg/kg, i.p.). Our results show that KA exposure in rats caused seizure behaviour, intrinsic neuronal hyperactivity, glutamate elevation, hippocampal neuronal damage, and cognitive impairment. These KA-induced alterations could be restored to the normal state by piperine treatment. In addition, piperine decreased the expression of the NGF precursor proNGF and NGF-degrading protease matrix metalloproteinase 9, whereas it increased the expression of proNGF processing enzyme matrix metalloproteinase 7, NGF, and NGF-activated receptor TrkA in the hippocampus of KA-treated rats. Furthermore, KA decreased phosphorylation of the protein kinase B (Akt) and glycogen synthase kinase 3ß (GSK3ß) in the hippocampus, and piperine reversed these changes. Our data suggest that piperine protects hippocampal neurons against KA-induced excitotoxicity by upregulating the NGF/TrkA/Akt/GSK3ß signalling pathways.


Subject(s)
Alkaloids , Neuroprotective Agents , Neurotoxicity Syndromes , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Benzodioxoles , Excitatory Amino Acid Agonists/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Kainic Acid/toxicity , Nerve Growth Factor/metabolism , Neuroprotection , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/metabolism , Piperidines , Polyunsaturated Alkamides , Proto-Oncogene Proteins c-akt/metabolism , Rats
8.
Molecules ; 27(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35209129

ABSTRACT

Excess synaptic glutamate release has pathological consequences, and the inhibition of glutamate release is crucial for neuroprotection. Kaempferol 3-rhamnoside (KR) is a flavonoid isolated from Schima superba with neuroprotective properties, and its effecton the release of glutamate from rat cerebrocortical nerve terminals was investigated. KR produced a concentration-dependent inhibition of 4-aminopyridine (4-AP)-evoked glutamate release with half-maximal inhibitory concentration value of 17 µM. The inhibition of glutamate release by KR was completely abolished by the omission of external Ca2+ or the depletion of glutamate in synaptic vesicles, and it was unaffected by blocking carrier-mediated release. In addition, KR reduced the 4-AP-evoked increase in Ca2+ concentration, while it did not affect 4-AP-evoked membrane potential depolarization. The application of selective antagonists of voltage-dependent Ca2+ channels revealed that the KR-mediated inhibition of glutamate release involved the suppression of P/Q-type Ca2+ channel activity. Furthermore, the inhibition of release was abolished by the calmodulin antagonist, W7, and Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor, KN62, but not by the protein kinase A (PKA) inhibitor, H89, or the protein kinase C (PKC) inhibitor, GF109203X. We also found that KR reduced the 4-AP-induced increase in phosphorylation of CaMKII and its substrate synapsin I. Thus, the effect of KR on evoked glutamate release is likely linked to a decrease in P/Q-type Ca2+ channel activity, as well as to the consequent reduction in the CaMKII/synapsin I pathway.


Subject(s)
Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Kaempferols/pharmacology , Synapses/drug effects , Synapses/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Kaempferols/chemistry , Membrane Potentials/drug effects , Molecular Structure , Phosphorylation , Rats , Signal Transduction/drug effects , Synapsins/metabolism
9.
Molecules ; 27(5)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35268651

ABSTRACT

Gold has always been regarded as a symbol of nobility, and its shiny golden appearance has always attracted the attention of many people. Gold has good ductility, molecular recognition properties, and good biocompatibility. At present, gold is being used in many fields. When gold particles are as small as several nanometers, their physical and chemical properties vary with their size in nanometers. The surface area of a nano-sized gold surface has a special effect. Therefore, gold nanoparticles can, directly and indirectly, give rise to different biological activities. For example, if the surface of the gold is sulfided. Various substances have a strong chemical reactivity and are easy to combine with sulfhydryl groups; hence, nanogold is often used in biomedical testing, disease diagnosis, and gene detection. Nanogold is easy to bind to proteins, such as antibodies, enzymes, or cytokines. In fact, scientists use nanogold to bind special antibodies, as a tool for targeting cancer cells. Gold nanoparticles are also directly cytotoxic to cancer cells. For diseases caused by inflammation and oxidative damage, gold nanoparticles also have antioxidant and anti-inflammatory effects. Based on these unique properties, gold nanoparticles have become the most widely studied metal nanomaterials. Many recent studies have further demonstrated that gold nanoparticles are beneficial for humans, due to their functional pharmacological properties in a variety of diseases. The content of this review will be the application of gold nanoparticles in treating or diagnosing pressing diseases, such as cancers, retinopathy, neurological diseases, skin disorders, bowel diseases, bone cartilage disorders, cardiovascular diseases, infections, and metabolic syndrome. Gold nanoparticles have shown very obvious therapeutic and application potential.


Subject(s)
Gold
10.
Molecules ; 27(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35164225

ABSTRACT

Excessive release of glutamate induces excitotoxicity and causes neuronal damage in several neurodegenerative diseases. Natural products have emerged as potential neuroprotective agents for preventing and treating neurological disorders. Dehydrocorydaline (DHC), an active alkaloid compound isolated from Corydalis yanhusuo, possesses neuroprotective capacity. The present study investigated the effect of DHC on glutamate release using a rat brain cortical synaptosome model. Our results indicate that DHC inhibited 4-aminopyridine (4-AP)-evoked glutamate release and elevated intrasynaptosomal calcium levels. The inhibitory effect of DHC on 4-AP-evoked glutamate release was prevented in the presence of the vesicular transporter inhibitor bafilomycin A1 and the N- and P/Q-type Ca2+ channel blocker ω-conotoxin MVIIC but not the intracellular inhibitor of Ca2+ release dantrolene or the mitochondrial Na+/Ca2+ exchanger inhibitor CGP37157. Moreover, the inhibitory effect of DHC on evoked glutamate release was prevented by the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) inhibitor PD98059. Western blotting data in synaptosomes also showed that DHC significantly decreased the level of ERK1/2 phosphorylation and synaptic vesicle-associated protein synapsin I, the main presynaptic target of ERK. Together, these results suggest that DHC inhibits presynaptic glutamate release from cerebrocortical synaptosomes by suppressing presynaptic voltage-dependent Ca2+ entry and the MAPK/ERK/synapsin I signaling pathway.


Subject(s)
Alkaloids/pharmacology , Calcium/metabolism , Cerebral Cortex/drug effects , Corydalis/chemistry , Glutamic Acid/metabolism , Nerve Tissue/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Cerebral Cortex/metabolism , Male , Nerve Tissue/metabolism , Rats , Rats, Sprague-Dawley
11.
Chem Res Toxicol ; 34(5): 1286-1295, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33621091

ABSTRACT

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.


Subject(s)
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.
Can J Physiol Pharmacol ; 99(7): 737-743, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33175564

ABSTRACT

Oxycodone, a semisynthetic opioid analgesic with actions similar to morphine, is extensively prescribed for treatment of moderate to severe acute pain. Given that glutamate plays a crucial role in mediating pain transmission, the purpose of this study was to investigate the effect of oxycodone on glutamatergic synaptic transmission in rat hippocampal CA3 area, which is associated with the modulation of nociceptive perception. Whole-cell patch-clamp recordings revealed that oxycodone effectively reduced presynaptic glutamate release, as detected by decreased frequencies of spontaneous excitatory postsynaptic currents (sEPSCs) and miniature EPSCs (mEPSCs), without eliciting significant changes in the amplitudes of sEPSCs and mEPSCs and glutamate-evoked inward currents. The inhibitory effect of oxycodone on the frequency of sEPSCs was blocked by the nonselective opioid receptor antagonist naloxone. In addition, oxycodone suppressed burst firing induced by 4-aminopyridine and tonic repetitive firing evoked by the applied depolarizing current. These results suggest that oxycodone inhibits spontaneous presynaptic glutamate release possibly by activating opioid receptors and consequently suppressing the neuronal excitability of hippocampal CA3 neurons.


Subject(s)
Neurons , Animals , Excitatory Postsynaptic Potentials , Oxycodone , Rats
13.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34768876

ABSTRACT

The glutamatergic neurotransmitter system has received substantial attention in research on the pathophysiology and treatment of neurological disorders. The study investigated the effect of the polyphenolic compound chlorogenic acid (CGA) on glutamate release in rat cerebrocortical nerve terminals (synaptosomes). CGA inhibited 4-aminopyridine (4-AP)-induced glutamate release from synaptosomes. This inhibition was prevented in the absence of extracellular Ca2+ and was associated with the inhibition of 4-AP-induced elevation of Ca2+ but was not attributed to changes in synaptosomal membrane potential. In line with evidence observed through molecular docking, CGA did not inhibit glutamate release in the presence of P/Q-type Ca2+ channel inhibitors; therefore, CGA-induced inhibition of glutamate release may be mediated by P/Q-type Ca2+ channels. CGA-induced inhibition of glutamate release was also diminished by the calmodulin and Ca2+/calmodilin-dependent kinase II (CaMKII) inhibitors, and CGA reduced the phosphorylation of CaMKII and its substrate, synapsin I. Furthermore, pretreatment with intraperitoneal CGA injection attenuated the glutamate increment and neuronal damage in the rat cortex that were induced by kainic acid administration. These results indicate that CGA inhibits glutamate release from cortical synaptosomes by suppressing P/Q-type Ca2+ channels and CaMKII/synapsin I pathways, thereby preventing excitotoxic damage to cortical neurons.


Subject(s)
Calcium Channels/metabolism , Chlorogenic Acid/pharmacology , Glutamic Acid/metabolism , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Chlorogenic Acid/metabolism , Excitatory Amino Acid Agents , Glutamic Acid/drug effects , Kainic Acid/metabolism , Male , Membrane Potentials/drug effects , Molecular Docking Simulation , Neurons/drug effects , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synaptic Vesicles/metabolism , Synaptosomes/metabolism
14.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884781

ABSTRACT

This study investigated the effects of enmein, an active constituent of Isodon japonicus Hara, on glutamate release in rat cerebrocortical nerve terminals (synaptosomes) and evaluated its neuroprotective potential in a rat model of kainic acid (KA)-induced glutamate excitotoxicity. Enmein inhibited depolarization-induced glutamate release, FM1-43 release, and Ca2+ elevation in cortical nerve terminals but had no effect on the membrane potential. Removing extracellular Ca2+ and blocking vesicular glutamate transporters, N- and P/Q-type Ca2+ channels, or protein kinase C (PKC) prevented the inhibition of glutamate release by enmein. Enmein also decreased the phosphorylation of PKC, PKC-α, and myristoylated alanine-rich C kinase substrates in synaptosomes. In the KA rat model, intraperitoneal administration of enmein 30 min before intraperitoneal injection of KA reduced neuronal cell death, glial cell activation, and glutamate elevation in the hippocampus. Furthermore, in the hippocampi of KA rats, enmein increased the expression of synaptic markers (synaptophysin and postsynaptic density protein 95) and excitatory amino acid transporters 2 and 3, which are responsible for glutamate clearance, whereas enmein decreased the expression of glial fibrillary acidic protein (GFAP) and CD11b. These results indicate that enmein not only inhibited glutamate release from cortical synaptosomes by suppressing Ca2+ influx and PKC but also increased KA-induced hippocampal neuronal death by suppressing gliosis and decreasing glutamate levels by increasing glutamate uptake.


Subject(s)
Apoptosis/drug effects , Brain Injuries/prevention & control , Diterpenes/pharmacology , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Synaptosomes/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Brain Injuries/chemically induced , CD11b Antigen/metabolism , Calcium/metabolism , Disks Large Homolog 4 Protein/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Membrane Potentials/drug effects , Neuroglia/metabolism , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synaptophysin/metabolism
15.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681651

ABSTRACT

Mast cells play a very important role in skin allergy and inflammation, including atopic dermatitis and psoriasis. In the past, it was found that neferine has anti-inflammatory and anti-aging effects on the skin, but its effect on mast cells has not yet been studied in detail. In this study, we used mast cells (RBL-2H3 cells) and mouse models to study the anti-allergic and inflammatory effects of neferine. First, we found that neferine inhibits the degranulation of mast cells and the expression of cytokines. In addition, we observed that when mast cells were stimulated by A23187/phorbol 12-myristate-13-acetate (PMA), the elevation of intracellular calcium was inhibited by neferine. The phosphorylation of the MAPK/NF-κB pathway is also reduced by pretreatment of neferine. The results of in vivo studies show that neferine can improve the appearance of dermatitis and mast cell infiltration caused by dinitrochlorobenzene (DNCB). Moreover, the expressions of barrier proteins in the skin are also restored. Finally, it was found that neferine can reduce the scratching behavior caused by compound 48/80. Taken together, our results indicate that neferine is a very good anti-allergic and anti-inflammatory natural product. Its effect on mast cells contributes to its pharmacological mechanism.


Subject(s)
Anti-Allergic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzylisoquinolines/pharmacology , Mast Cells/drug effects , Animals , Anti-Allergic Agents/therapeutic use , Benzylisoquinolines/therapeutic use , Calcimycin/pharmacology , Calcium/metabolism , Cell Line , Cell Movement/drug effects , Cytokines/genetics , Cytokines/metabolism , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Dinitrochlorobenzene/pharmacology , Disease Models, Animal , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
16.
J Neuroinflammation ; 17(1): 177, 2020 Jun 06.
Article in English | MEDLINE | ID: mdl-32505192

ABSTRACT

BACKGROUND: Upregulation of matrix metalloproteinase-9 (MMP-9) has been indicated as one of the inflammatory biomarkers. In the central nervous system (CNS), the MMP-9 is induced by several proinflammatory mediators and participates in the CNS disorders, including inflammation and neurodegeneration. In addition, protein kinase Cs (PKCs) has been shown to be involved in regulation of various inflammatory factors like MMP-9 by several stimuli in many cell types. Several phytochemicals are believed to reduce the risk of several inflammatory disorders including the CNS diseases. The rottlerin, a principal phenolic compound of the Kamala plant Mallotus philippinensis, has been shown to possess an array of medicinal properties, including anti-PKC-δ, antitumor, anti-oxidative, and anti-inflammatory activities. METHODS: Herein, we used rat brain astrocytes (RBA) to demonstrate the signaling mechanisms of phorbol 12-myristate 13-acetate (PMA)-induced MMP-9 expression by zymographic, RT-PCR, subcellular isolation, Western blot, ROS detection, and promoter reporter analyses. Then, we evaluate the effects of rottlerin on PMA-induced MMP-9 expression in RBA and its influencing mechanism. RESULTS: We first demonstrated that PMA stimulated activation of various types of PKC, including PKC-δ in RBA. Subsequently, PMA induced MMP-9 expression via PKCδ-mediated reactive oxygen species (ROS) generation, extracellular signal-regulated kinase 1/2 (ERK1/2) activation, and then induced c-Fos/AP-1 signaling pathway. Finally, upregulation of MMP-9 by PMA via the pathway may promote astrocytic migration, and the event could be attenuated by rottlerin. CONCLUSIONS: These data indicated that rottlerin may have anti-inflammatory activity by reducing these related pathways of PKC-δ-dependent ROS-mediated MMP-9 expression in brain astrocytes.


Subject(s)
Acetophenones/pharmacology , Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Benzopyrans/pharmacology , Brain/drug effects , Signal Transduction/drug effects , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Line , Cell Movement/drug effects , Matrix Metalloproteinase 9/metabolism , Protein Kinase C-delta/metabolism , Rats , Reactive Oxygen Species/metabolism , Up-Regulation
17.
Cell Commun Signal ; 18(1): 185, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33228717

ABSTRACT

BACKGROUND: The matrix metalloproteinase-9 (MMP-9) is up-regulated by several proinflammatory mediators in the central nervous system (CNS) diseases. Increasing reports show that MMP-9 expression is an inflammatory biomarker of several CNS disorders, including the CNS inflammation and neurodegeneration. Bradykinin (BK) is a common proinflammatory mediator and elevated in several brain injury and inflammatory disorders. The raised BK may be detrimental effects on the CNS that may aggravate brain inflammation through MMP-9 up-regulation or cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) production in brain astrocytes. However, the relationship between BK-induced MMP-9 expression and COX-2-derived PGE2 release in brain astrocytes remains unclear. METHODS: Herein we used rat brain astrocytes (RBA) to investigate the role of the COX-2/PGE2 system in BK-induced MMP-9 expression. We used zymographic, RT-PCR, EIA, and Western blotting analyses to confirm that BK induces MMP-9 expression via a COX-2/PGE2-dependent pathway. RESULTS: Our results show activation of native COX-2 by BK led to PGE2 production and release. Subsequently, PGE2 induced MMP-9 expression via PGE2 receptor (EP)-mediated c-Src, Jak2, ERK1/2, and then activated signal transducer and activator of transcription 3 (STAT3) signaling pathway. Finally, up-regulation of MMP-9 by BK via the pathway may promote astrocytic migration. CONCLUSION: These results demonstrated that a novel autocrine pathway for BK-induced MMP-9 protein expression is mediated through activation of STAT3 by native COX-2/PGE2-mediated c-Src/Jak2/ERK cascades in brain astrocytes. Video Abstract.


Subject(s)
Astrocytes/cytology , Astrocytes/enzymology , Autocrine Communication , Bradykinin/pharmacology , Cell Movement/drug effects , Dinoprostone/metabolism , Matrix Metalloproteinase 9/metabolism , STAT3 Transcription Factor/metabolism , Animals , Astrocytes/drug effects , Autocrine Communication/drug effects , Celecoxib/pharmacology , Cell Line , Janus Kinase 2/metabolism , MAP Kinase Signaling System/drug effects , Rats , Receptors, Prostaglandin E/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , src-Family Kinases/metabolism
18.
Planta Med ; 86(6): 434-441, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32097973

ABSTRACT

Excessive glutamate concentration induces neuronal death in acute brain injuries and chronic neurodegenerative diseases. Natural compounds from medicinal plants have attracted considerable attention for their use in the prevention and treatment of neurological disorders. 11-Keto-ß-boswellic acid, a triterpenoid found in the medicinal plant Boswellia serrata, has neuroprotective potential. The present study investigated the effect of 11-keto-ß-boswellic acid on glutamate release in vitro and kainic acid-induced glutamate excitotoxicity in vivo in the rat hippocampus. In rat hippocampal nerve terminals (synaptosomes), 11-keto-ß-boswellic acid dose-dependently inhibited 4-aminopyridine-stimulated glutamate release. This effect was dependent on extracellular calcium, persisted in the presence of the glutamate transporter inhibitor DL-threo-ß-benzyloxyaspartate, and was blocked by the vesicular transporter inhibitor bafilomycin A1. In addition, 11-keto-ß-boswellic acid reduced the 4-aminopyridine-induced increase in intrasynaptosomal Ca2+ levels. The N- and P/Q-type channel blocker ω-conotoxin MVIIC and the protein kinase A inhibitor H89 significantly suppressed the 11-keto-ß-boswellic acid-mediated inhibition of glutamate release, whereas the intracellular Ca2+-releasing inhibitors dantrolene, CGP37157, and xestospongin C, mitogen-activated protein kinase inhibitor PD98059, as well as protein kinase C inhibitor calphostin C had no effect. In a rat model of excitotoxicity induced by intraperitoneal kainic acid injection (15 mg/kg), intraperitoneal 11-keto-ß-boswellic acid administration (10 or 50 mg/kg) 30 min before kainic acid injection considerably ameliorated kainic acid-induced glutamate concentration elevation and CA3 neuronal death. These data suggested that 11-keto-ß-boswellic acid inhibits glutamate release from the rat hippocampal synaptosomes by suppressing N- and P/Q-type Ca2+ channels and protein kinase A activity, as well as exerts protective effects against kainic acid-induced excitotoxicity in vivo.


Subject(s)
Glutamic Acid , Triterpenes , Animals , Calcium , Cerebral Cortex , Hippocampus , Kainic Acid , Membrane Potentials , Rats , Rats, Sprague-Dawley
19.
Pharmacology ; 105(7-8): 445-453, 2020.
Article in English | MEDLINE | ID: mdl-31825942

ABSTRACT

BACKGROUND: Tapentadol, a centrally acting oral analgesic, activates µ-opioid receptor and inhibits norepinephrine reuptake. Given that glutamate plays a crucial role in mediating pain, this study investigated the influence of tapentadol on spontaneous glutamatergic synaptic transmission and evoked neuronal excitability in rat hippocampal CA3 pyramidal neurons, which has been suggested to be involved in nociceptive perception. METHODS: We used electrophysiological technique to determine the effect of tapentadol on spontaneous excitatory postsynaptic currents (sEPSC), glutamate-activated currents, and neuronal excitability in CA3 pyramidal neurons in rat hippocampal slices. We also used isolated nerve terminals (synaptosomes) prepared from the rat hippocampus to examine the effect of tapentadol on glutamate release. RESULTS: Whole-cell patch clamp recordings revealed that tapentadol effectively decreased the frequencies of sEPSCs and miniature EPSCs (mEPSCs) without changing their amplitudes in hippocampal CA3 pyramidal neurons. However, glutamate-evoked inward currents were not affected by tapentadol. Further, tapentadol decreased 4-aminopyridine-induced glutamate release from hippocampal synaptosomes, and this effect was prevented by chelating the extracellular Ca2+ ions and blocking the N- and P/Q-type Ca2+ channels. In addition, burst firing induced by 4-aminopyridine and tonic repetitive firing induced by depolarizing pulses were attenuated by tapentadol. CONCLUSIONS: We conclude that tapentadol inhibits glutamatergic synaptic transmission, without modifying postsynaptic receptor sensitivity, and that this decline of excitation consequently suppresses neuronal hyperexcitability in the hippocampal CA3 area.


Subject(s)
Analgesics, Opioid/pharmacology , CA3 Region, Hippocampal/drug effects , Glutamic Acid/metabolism , Pyramidal Cells/drug effects , Tapentadol/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/drug effects , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
20.
Chem Res Toxicol ; 32(8): 1591-1598, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31264847

ABSTRACT

The effect of cycloheterophyllin, a prenylflavone isolated from Artocarpus heteophyllus, on glutamate release was studied in the rat hippocampus using synaptosome and slice preparations. In rat hippocampal synaptosomes, cycloheterophyllin inhibited 4-aminopyridine (4-AP)-evoked glutamate release and elevation of intrasynaptosomal calcium levels. The inhibitory effect of cycloheterophyllin on 4-AP-evoked glutamate release was prevented in the presence of the vesicular transporter inhibitor, the N- and P/Q-type calcium channel blocker, and the protein kinase C (PKC) inhibitor but was insensitive to the intracellular Ca2+ release inhibitors, the protein kinase A inhibitor, and the mitogen-activated/extracellular signal-regulated kinase inhibitor. Western blotting data in synaptosomes also showed that cycloheterophyllin significantly decreased the level of phosphorylation of PKC. In addition, cycloheterophyllin decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) without influencing the amplitude of sEPSCs and glutamate-activated currents in hippocampal slices, supporting a presynaptic action. Together, these results suggest that cycloheterophyllin inhibits presynaptic glutamate release by suppressing N- and P/Q-type calcium channel and PKC activity in the rat hippocampus.


Subject(s)
Flavonoids/pharmacology , Glutamic Acid/metabolism , Hippocampus/drug effects , Neurons/drug effects , Animals , Artocarpus/chemistry , Calcium/analysis , Flavonoids/chemistry , Flavonoids/isolation & purification , Glutamic Acid/analysis , Hippocampus/metabolism , Male , Molecular Structure , Neurons/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL