Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Neurochem Res ; 2019 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-31713091

RÉSUMÉ

Neuronal hippocampal death can be induced by exacerbated levels of cortisol, a condition usually observed in patients with Major depressive disorder (MDD). Previous in vitro and in vivo studies showed that ursolic acid (UA) elicits antidepressant and neuroprotective properties. However, the protective effects of UA against glucocorticoid-induced cytotoxicity have never been addressed. Using an in vitro model of hippocampal cellular death induced by elevated levels of corticosterone, we investigated if UA prevents corticosterone-induced cytotoxicity in HT22 mouse hippocampal derived cells. Concentrations lower than 25 µM UA did not alter cell viability. Co-incubation with UA for 48 h was able to protect HT22 cells from the reduction on cell viability and from the increase in apoptotic cells induced by corticosterone. Inhibition of protein kinase A (PKA), protein kinase C (PKC) and, Ca2+/calmodulin-dependent protein kinase II (CaMKII), but not phosphoinositide 3-kinase(PI3K), by using the pharmacological the inhibitors: H-89, chelerythrine, KN-62, and LY294002, respectively totally abolished the cytoprotective effects of UA. Finally, UA abrogated the reduction in phospho-extracellular signal-regulated kinases 1 and 2 (ERK1/2) but not in phospho-c-Jun kinases induced by corticosterone. These results indicate that the protective effect of UA against the cytotoxicity induced by corticosterone in HT22 cells may involve PKA, PKC, CaMKII, and ERK1/2 activation. The cytoprotective potential of UA against corticosterone-induced cytotoxicity and its ability to modulate intracellular signaling pathways involved in cell proliferation and survival suggest that UA may be a relevant strategy to manage stress-related disorders such as MDD.

2.
Food Chem Toxicol ; 132: 110665, 2019 Oct.
Article de Anglais | MEDLINE | ID: mdl-31283947

RÉSUMÉ

In this study we evaluate the chemical composition and neuroprotective effects of alkaloid fractions of the Amaryllidaceae species Rhodophiala pratensis, Rhodolirium speciosum, Phycella australis and Phaedranassa lehmannii. Gas chromatography-mass spectrometry (GC/MS) enable the identification of 41 known alkaloids. Rhodolirium speciosum and Rhodophiala pratensis were the most active extracts against acetylcholinesterase (AChE), with IC50 values of 35.22 and 38.13 µg/mL, respectively. The protective effect of these extracts on human neuroblastoma cells (SH-SY5Y) subjected to mitochondrial oxidative stress with rotenone/oligomycin A (R/O) and toxicity promoted by okadaic acid (OA) was evaluated. Only Phycella australis and Rhodophiala pratensis at 0.75 and 1.5 µg/mL, tend to reverse the cell death induced by R/O by around 12%. In OA assay, alkaloid fractions of Phycella Australis and Phaedranassa lehmannii displayed a concentration-dependent (0.375-3.0 µg/mL) effect with a maximum neuroprotective response of 78% and 84%, respectively. Afterwards, neuroprotective effects of Phycella australis (3 and 6 µg/mL) in mouse hippocampal slices stressed with oxygen glucose deprivation/reoxygenation (OGD/R), shown a protection greater than 14%. Finally, Phycella Australis (6 µg/mL) reverted the cell viability from 65% to 90% in slices treated with OA, representing a protection of 25% attributable to the alkaloids of this species.


Sujet(s)
Alcaloïdes des Amaryllidaceae/pharmacologie , Amaryllidaceae/composition chimique , Hippocampe/effets des médicaments et des substances chimiques , Isoquinoléines/pharmacologie , Neuroblastome/métabolisme , Neuroprotecteurs/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Chromatographie gazeuse-spectrométrie de masse , Hippocampe/métabolisme , Humains , Techniques in vitro , Souris , Souris de lignée C57BL , Neuroblastome/anatomopathologie
3.
Mol Neurobiol ; 55(2): 1580-1589, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-28185129

RÉSUMÉ

Folic acid (folate) is a vitamin of the B-complex group crucial for neurological function. Considering that excitotoxicity and cell death induced by glutamate are involved in many disorders, the potential protective effect of folic acid on glutamate-induced cell damage in rat hippocampal slices and the possible intracellular signaling pathway involved in such effect were investigated. The treatment of hippocampal slices with folic acid (100 µM) significantly abrogated glutamate (1 mM)-induced reduction of cell viability measured by MTT reduction assay and inhibited glutamate-induced D-[3H]-aspartate release. To investigate the putative intracellular signaling pathways implicated in the protective effect of folic acid, we used a PI3K inhibitor, LY294002, which abolished the protective effects of folic acid against glutamate-induced cell damage and D-[3H] aspartate release. Moreover, hippocampal slices incubated with folic acid alone for 30 min presented increased phosphorylation of GSK-3ß at Ser9, indicating an inhibition of the activity of this enzyme. Furthermore, folic acid in the presence of glutamate insult in hippocampal slices maintained for an additional period of 6 h in fresh culture medium without glutamate and/or folic acid induced phosphorylation of GSK-3ß and ß-catenin expression. In addition, glutamate-treated hippocampal slices showed increased iNOS expression that was reversed by folic acid. In conclusion, the results of this study show that the protective effect of folic acid against glutamate-induced excitotoxicity may involve the modulation of PI3K/GSK-3ß/ß-catenin pathway and iNOS inhibition.


Sujet(s)
Acide folique/pharmacologie , Acide glutamique/pharmacologie , Glycogen synthase kinase 3 beta/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Nitric oxide synthase type II/métabolisme , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Mâle , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques
4.
Redox Biol ; 14: 285-294, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-28987867

RÉSUMÉ

Human erythropoietin is mainly recognized for its hematopoietic function; however, by binding to its receptor (EpoR), it can activate different signaling pathways as STAT, PI3K, MAPK and RAS to increase cellular differentiation or provide neuroprotective effects, among others. A recombinant human erythropoietin variant with low glycosylation and without hematopoietic effect (EpoL) was purified from skimmed goat milk. Recombinant human erythropoietin (Epo) was obtained from CHO cell line and used as control to compare EpoL effects. Neuroprotection studies were performed in PC12 cells and rat hippocampal slices. Cells were pretreated during 1h with EpoL or Epo and exposed to oxidative agents (H2O2 or FCCP); cell viability was assayed at the end of the experiment by the MTT method. Hippocampal slices were exposed to 15min of oxygen and glucose deprivation (OGD) and the neuroprotective drugs EpoL or Epo were incubated for 2h post-OGD in re-oxygenated medium. Cell cultures stressed with oxidative agents, and pretreated with EpoL, showed neuroprotective effects of 30% at a concentration 10 times lower than that of Epo. Moreover, similar differences were observed in OGD ex vivo assays. Neuroprotection elicited by EpoL was lost when an antibody against EpoR was present, indicating that its effect is EpoR-dependent. In conclusion, our results suggest that EpoL has a more potent neuroprotective profile than Epo against oxidative stress, mediated by activation of EpoR, thus EpoL represents an important target to develop a potential biopharmaceutical to treat different central nervous system pathologies related to oxidative stress such as stroke or neurodegenerative diseases.


Sujet(s)
Érythropoïétine/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Cellules CHO , Survie cellulaire/effets des médicaments et des substances chimiques , Cricetulus , Érythropoïétine/génétique , Humains , Neurones/cytologie , Neurones/métabolisme , Neuroprotecteurs/métabolisme , Cellules PC12 , Rats , Récepteur érythropoïétine/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/pharmacologie
5.
Amino Acids ; 47(4): 795-811, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25555469

RÉSUMÉ

The modulation of N-methyl-D-aspartate receptor (NMDAR) and L-arginine/nitric oxide (NO) pathway is a therapeutic strategy for treating depression and neurologic disorders that involves excitotoxicity. Literature data have reported that creatine exhibits antidepressant and neuroprotective effects, but the implication of NMDAR and L-arginine/nitric oxide (NO) pathway in these effects is not established. This study evaluated the influence of pharmacological agents that modulate NMDAR/L-arginine-NO pathway in the anti-immobility effect of creatine in the tail suspension test (TST) in mice. The NOx levels and cellular viability in hippocampal and cerebrocortical slices of creatine-treated mice were also evaluated. The anti-immobility effect of creatine (10 mg/kg, po) in the TST was abolished by NMDA (0.1 pmol/mouse, icv), D-serine (30 µg/mouse, icv, glycine-site NMDAR agonist), arcaine (1 mg/kg, ip, polyamine site NMDAR antagonist), L-arginine (750 mg/kg, ip, NO precursor), SNAP (25 µg/mouse, icv, NO donor), L-NAME (175 mg/kg, ip, non-selective NOS inhibitor) or 7-nitroindazole (50 mg/kg, ip, neuronal NOS inhibitor), but not by DNQX (2.5 µg/mouse, icv, AMPA receptor antagonist). The combined administration of sub-effective doses of creatine (0.01 mg/kg, po) and NMDAR antagonists MK-801 (0.001 mg/kg, po) or ketamine (0.1 mg/kg, ip) reduced immobility time in the TST. Creatine (10 mg/kg, po) increased cellular viability in hippocampal and cerebrocortical slices and enhanced hippocampal and cerebrocortical NO x levels, an effect potentiated by L-arginine or SNAP and abolished by 7-nitroindazole or L-NAME. In conclusion, the anti-immobility effect of creatine in the TST involves NMDAR inhibition and enhancement of NO levels accompanied by an increase in neural viability.


Sujet(s)
Antidépresseurs/pharmacologie , Arginine/métabolisme , Créatine/pharmacologie , Dépression/métabolisme , Monoxyde d'azote/métabolisme , Récepteurs du N-méthyl-D-aspartate/métabolisme , Animaux , Dépression/traitement médicamenteux , Dépression/génétique , Femelle , Suspension des membres postérieurs , Humains , Souris , Rats , Rat Wistar , Récepteurs du N-méthyl-D-aspartate/génétique , Transduction du signal
6.
J Neurochem ; 126(4): 437-50, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23713463

RÉSUMÉ

Guanosine (GUO) is an endogenous modulator of glutamatergic excitotoxicity and has been shown to promote neuroprotection in in vivo and in vitro models of neurotoxicity. This study was designed to understand the neuroprotective mechanism of GUO against oxidative damage promoted by oxygen/glucose deprivation and reoxygenation (OGD). GUO (100 µM) reduced reactive oxygen species production and prevented mitochondrial membrane depolarization induced by OGD. GUO also exhibited anti-inflammatory actions as inhibition of nuclear factor kappa B activation and reduction of inducible nitric oxide synthase induction induced by OGD. These GUO neuroprotective effects were mediated by adenosine A1 receptor, phosphatidylinositol-3 kinase and MAPK/ERK. Furthermore, GUO recovered the impairment of glutamate uptake caused by OGD, an effect that occurred via a Pertussis toxin-sensitive G-protein-coupled signaling, blockade of adenosine A2A receptors (A2A R), but not via A1 receptor. The modulation of glutamate uptake by GUO also involved MAPK/ERK activation. In conclusion, GUO, by modulating adenosine receptor function and activating MAPK/ERK, affords neuroprotection of hippocampal slices subjected to OGD by a mechanism that implicates the following: (i) prevention of mitochondrial membrane depolarization, (ii) reduction of oxidative stress, (iii) regulation of inflammation by inhibition of nuclear factor kappa B and inducible nitric oxide synthase, and (iv) promoting glutamate uptake.


Sujet(s)
Encéphalite , Guanosine/métabolisme , Hippocampe/immunologie , Hippocampe/métabolisme , Hypoxie cérébrale , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Encéphalite/traitement médicamenteux , Encéphalite/immunologie , Encéphalite/métabolisme , Sous-unités alpha Gi-Go des protéines G/métabolisme , Glucose/pharmacologie , Acide glutamique/pharmacocinétique , Guanosine/pharmacologie , Hippocampe/cytologie , Hypoxie cérébrale/traitement médicamenteux , Hypoxie cérébrale/immunologie , Hypoxie cérébrale/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/immunologie , Mâle , Potentiel de membrane mitochondriale/physiologie , Facteur de transcription NF-kappa B/métabolisme , Protéines de tissu nerveux/métabolisme , Neuroprotecteurs/pharmacologie , Nitric oxide synthase type II/métabolisme , Techniques de culture d'organes , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/physiologie , Oxygène/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Rats , Rat Wistar , Espèces réactives de l'oxygène/métabolisme , Récepteur A1 à l'adénosine/métabolisme , Synaptotagmines , Tritium
7.
Neurochem Int ; 62(6): 836-42, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23454192

RÉSUMÉ

The excitotoxicity induced by excessive activation of the glutamatergic neurotransmission pathway is involved in several neuropathologies. In this sense, molecules that prevent the release of glutamate or the excessive activation of its receptors can be useful in preventing the neuronal cell death observed in these diseases. Lectins are proteins capable of reversible binding to the carbohydrates in glycoconjugates, and some have been used in the study and purification of glutamate receptors. ConBr is a mannose/glucose-binding lectin purified from Canavalia brasiliensis seeds. In the present study, we aimed to evaluate the neuroprotective activity of ConBr against glutamate-induced excitotoxicity. Hippocampal slices were isolated from adult male mice and incubated for 6h in Krebs saline/DMEM buffer alone (control), in the presence of glutamate or glutamate plus ConBr. The phosphorylation of Akt and mitogen activated protein kinases (MAPKs) such as ERK1/2, p38(MAPK) and JNK1/2/3 was evaluated with western blotting. The results indicate that glutamate provoked a reduction in the hippocampal slice viability (-25%), diminished the phosphorylation of Akt and augmented p38(MAPK) and ERK1 phosphorylation. No changes were observed in the phosphorylation of JNK1/2/3 or ERK2. Notably, ConBr, through a mechanism dependent on carbohydrate interaction, prevented the reduction of cell viability and Akt phosphorylation induced by glutamate. Furthermore, in the presence of the PI3K inhibitor LY294002, ConBr was unable to reverse glutamate neurotoxicity. Taken together, our data suggest that the neuroprotective effect of ConBr against glutamate neurotoxicity requires oligosaccharide interaction and is dependent on the PI3K/Akt pathway.


Sujet(s)
Canavalia/composition chimique , Antagonistes des acides aminés excitateurs , Acide glutamique/toxicité , Hippocampe/effets des médicaments et des substances chimiques , Neuroprotecteurs , Protéine oncogène v-akt/physiologie , Phosphatidylinositol 3-kinases/physiologie , Lectines végétales/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Technique de Western , Métabolisme glucidique/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , 4H-1-Benzopyran-4-ones/pharmacologie , Antienzymes/pharmacologie , Hippocampe/anatomopathologie , Techniques in vitro , Mâle , Souris , Mitogen-Activated Protein Kinases/physiologie , Morpholines/pharmacologie , Phosphorylation , Lectines végétales/composition chimique , Lectines végétales/isolement et purification , Sels de tétrazolium , Thiazoles
8.
Neurochem Int ; 61(3): 397-404, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22683349

RÉSUMÉ

Mitochondrial perturbation and oxidative stress are key factors in neuronal vulnerability in several neurodegenerative diseases or during brain ischemia. Here we have investigated the protective mechanism of action of guanosine, the guanine nucleoside, in a human neuroblastoma cell line, SH-SY5Y, subjected to mitochondrial oxidative stress. Blockade of mitochondrial complexes I and V with rotenone plus oligomycin (Rot/oligo) caused a significant decrease in cell viability and an increase in ROS production. Guanosine that the protective effect of guanosine incubated concomitantly with Rot/oligo abolished Rot/oligo-induced cell death and ROS production in a concentration dependent manner; maximum protection was achieved at the concentration of 1mM. The cytoprotective effect afforded by guanosine was abolished by adenosine A(1) or A(2A) receptor antagonists (DPCPX or ZM241385, respectively), or by a large (big) conductance Ca(2+)-activated K(+) channel (BK) blocker (charybdotoxin). Evaluation of signaling pathways showed that the protective effect of guanosine was not abolished by a MEK inhibitor (PD98059), by a p38(MAPK) inhibitor (SB203580), or by a PKC inhibitor (cheleritrine). However, when blocking the PI3K/Akt pathway with LY294002, the neuroprotective effect of guanosine was abolished. Guanosine increased Akt and p-Ser-9-GSK-3ß phosphorylation confirming this pathway plays a key role in guanosine's neuroprotective effect. Guanosine induced the antioxidant enzyme heme oxygenase-1 (HO-1) expression. The protective effects of guanosine were prevented by heme oxygenase-1 inhibitor, SnPP. Moreover, bilirubin, an antioxidant and physiologic product of HO-1, is protective against mitochondrial oxidative stress. In conclusion, our results show that guanosine can afford protection against mitochondrial oxidative stress by a signaling pathway that implicates PI3K/Akt/GSK-3ß proteins and induction of the antioxidant enzyme HO-1.


Sujet(s)
Guanosine/pharmacologie , Heme oxygenase-1/biosynthèse , Mitochondries/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Technique de Western , Lignée cellulaire tumorale , Induction enzymatique , Glycogen Synthase Kinase 3/métabolisme , Glycogen synthase kinase 3 beta , Humains , Mitochondries/enzymologie , Mitochondries/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Espèces réactives de l'oxygène/métabolisme
9.
J Psychopharmacol ; 26(5): 714-23, 2012 May.
Article de Anglais | MEDLINE | ID: mdl-22037925

RÉSUMÉ

Preclinical and clinical studies indicate that deficiency in folic acid plays a role in the pathophysiology of depression. Considering that alterations in the signaling pathways that regulate neuroplasticity and cellular survival are implicated in depressive disorders, the present study investigated the involvement of the phosphoinositide 3-kinase (PI3K), glycogen synthase kinase-3 (GSK-3ß), and peroxisome proliferator-activated receptor-γ (PPARγ) in the antidepressant-like effect of folic acid in the forced swimming test (FST). The intracerebroventricular (i.c.v.) pre-treatment of mice with LY294002 (10 nmol/site, a PI3K inhibitor) or GW-9662 (1 µg/site, a PPARγ antagonist) prevented the antidepressant-like effect of folic acid (50 mg/kg, p.o.) in the FST. In addition, the administration of subeffective doses of the selective GSK-3ß inhibitor, AR-A014418 (3 mg/kg, i.p.), a non-selective GSK-3ß inhibitor, lithium chloride (10 mg/kg, p.o) or a PPARγ agonist, rosiglitazone (1 µg/site, i.c.v.) in combination with a subeffective dose of folic acid (10 mg/kg, p.o.) significantly reduced the immobility time in the FST as compared with either drug alone, without altering the locomotor activity. These results indicate that the antidepressant-like effect of folic acid in the FST might be dependent on inhibition of GSK-3ß and activation of PPARγ, reinforcing the notion that these are important targets for antidepressant activity.


Sujet(s)
Antidépresseurs/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Dépression/traitement médicamenteux , Acide folique/pharmacologie , Glycogen Synthase Kinase 3/métabolisme , Récepteur PPAR gamma/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Animaux , Dépression/métabolisme , Glycogen synthase kinase 3 beta , Mâle , Souris , Activité motrice/effets des médicaments et des substances chimiques , Natation
10.
J Pain ; 12(3): 315-22, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-20705523

RÉSUMÉ

UNLABELLED: We investigated the antinociceptive effects of AR-A014418, a selective inhibitor of glycogen synthase kinase-3ß (GSK-3ß) in mice. A 30-minute pretreatment with AR-A014418 (.1 and 1 mg/kg, intraperitoneal [ip]) inhibited nociception induced by an ip injection of acetic acid. AR-A014418 pretreatment (.1 and .3 mg/kg, ip) also decreased the late (inflammatory) phase of formalin-induced licking, without affecting responses of the first (neurogenic) phase. In a different set of experiments, AR-A014418 (.1-10 µg/site) coinjected intraplantarly (ipl) with formalin inhibited the late phase of formalin-induced nociception. Furthermore, AR-A014418 administration (1 and 10 ng/site, intrathecal [it]) inhibited both phases of formalin-induced licking. In addition, AR-A014418 coinjection (10 ng/site, it) inhibited nociception induced by glutamate, N-methyl-D-aspartate (NMDA), (±)-1-aminocyclopentane-trans-1,3-dicarboxylic acid (trans-ACPD), tumor necrosis factor-alpha (TNF-α), and interleukin-1beta (IL-1ß) by 47 ± 12%, 48 ± 11%, 31 ± 8%, 46 ± 13%, and 44 ± 11%, respectively. In addition, a 30-minute pretreatment with NP031115 (3 and 10 mg/kg, ip), a different GSK-3 ß inhibitor, also attenuated the late phase of formalin-induced nociception. Collectively, these results provide convincing evidence that AR-A014418, given by local, systemic, and central routes, produces antinociception in several mouse models of nociception. The AR-A014418-dependent antinociceptive effects were induced by modulation of the glutamatergic system through metabotropic and ionotropic (NMDA) receptors and the inhibition of the cytokine (TNF-α and IL-1ß) signaling. PERSPECTIVE: These results suggest that GSK-3ß may be a novel pharmacological target for the treatment of pain.


Sujet(s)
Douleur abdominale/prévention et contrôle , Analgésiques/administration et posologie , Glycogen Synthase Kinase 3/antagonistes et inhibiteurs , Thiazoles/administration et posologie , Urée/analogues et dérivés , Douleur abdominale/induit chimiquement , Agressivité/effets des médicaments et des substances chimiques , Analyse de variance , Animaux , Anti-inflammatoires non stéroïdiens , Azotures/administration et posologie , Cytokines/administration et posologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Voies d'administration de substances chimiques et des médicaments , Formaldéhyde/effets indésirables , Acide glutamique/effets indésirables , Inflammation/induit chimiquement , Inflammation/traitement médicamenteux , Mâle , Souris , N-Méthyl-aspartate/administration et posologie , Mesure de la douleur/méthodes , Oses acides/effets indésirables , Urée/administration et posologie , Xylose/effets indésirables , Xylose/analogues et dérivés
11.
Behav Brain Res ; 168(2): 318-22, 2006 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-16387371

RÉSUMÉ

This study investigated the involvement of the L-arginine-nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) pathway in the antidepressant-like effect of an acute administration of memantine in the forced swimming test (FST) in mice, since this signaling pathway is supposed to play a significant role in depression. The antidepressant-like effect of memantine (3 mg/kg, i.p.) was prevented by pretreatment with L-arginine (750 mg/kg, i.p.) or S-nitroso-N-acetyl-penicillamine (SNAP, 25 microg/site, i.c.v.), but not with d-arginine (750 mg/kg, i.p.).The treatment of mice with NG-nitro-L-arginine (L-NNA, 0.03 and 0.3 mg/kg, i.p.) potentiated the effect of a subeffective dose of memantine (0.3 mg/kg, i.p.) in the FST. Moreover, the pretreatment of mice with (1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one) (ODQ, 30 pmol/site, i.c.v.) produced a synergistic antidepressant-like effect with subeffective doses of memantine (0.3 and 1 mg/kg, i.p.) in the FST. Furthermore, the reduction in the immobility time elicited by memantine (3 mg/kg, i.p.) in the FST was prevented by pretreatment with sildenafil (5 mg/kg, i.p.). Taken together, the results demonstrate that memantine produced an antidepressant-like effect in the FST that seems to be mediated through an interaction with the L-arginine-NO-cGMP pathway.


Sujet(s)
Antidépresseurs/administration et posologie , Arginine/physiologie , GMP cyclique/physiologie , Dépression/traitement médicamenteux , Mémantine/administration et posologie , Monoxyde d'azote/physiologie , Analyse de variance , Animaux , GMP cyclique/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Interactions médicamenteuses , Antienzymes/pharmacologie , Femelle , Réaction d'immobilité tonique/effets des médicaments et des substances chimiques , Mâle , Souris , Donneur d'oxyde nitrique/pharmacologie , Nitroarginine/pharmacologie , Pénicillamine/analogues et dérivés , Pénicillamine/pharmacologie , Pipérazines/pharmacologie , Purines , Citrate de sildénafil , Sulfones , Natation
12.
Neurosci Lett ; 395(2): 93-7, 2006 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-16289784

RÉSUMÉ

This study investigated the cellular signaling pathways involved in the acute antidepressant-like action of memantine in the forced swimming test (FST) in mice. The immobility time in the FST was reduced by memantine (3-10 mg/kg, i.p.). The anti-immobility effect of memantine (3 mg/kg, i.p.) was prevented by pretreatment with H-89 (1 microg/site, i.c.v., an inhibitor of PKA), PD098059 (5 microg/site, i.c.v., an inhibitor of MAPK/ERK), KN-62 (1 microg/site, i.c.v., an inhibitor of CaMKII), but not with chelerythrine (1 microg/site, i.c.v., an inhibitor of PKC). Taken together, these results firstly demonstrate that the acute antidepressant-like effect of memantine seems to be dependent on the cellular signaling modulated by PKA, CaMKII and MAPK/ERK, but not by PKC.


Sujet(s)
Antidépresseurs/pharmacologie , Antagonistes des acides aminés excitateurs/pharmacologie , Mémantine/pharmacologie , Protein kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/métabolisme , Cyclic AMP-Dependent Protein Kinases , Antienzymes/pharmacologie , Femelle , Mâle , Souris , Mitogen-Activated Protein Kinases/métabolisme , Activité motrice/effets des médicaments et des substances chimiques , Protéine kinase C/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Transduction du signal/physiologie , Stress psychologique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE