Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Int J Neuropsychopharmacol ; 22(10): 665-674, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31325908

RESUMO

BACKGROUND: Although recent studies provide insight into the molecular mechanisms of the effects of ketamine, the antidepressant mechanism of ketamine enantiomers and their metabolites is not fully understood. In view of the involvement of mechanisms other than the N-methyl-D-aspartate receptor in ketamine's action, we investigated the effects of (R)-ketamine, (S)-ketamine, (R)-norketamine [(R)-NK], (S)-NK, (2R,6R)-hydroxynorketamine [(2R,6R)-HNK], and (2S,6S)-HNK on monoaminergic neurotransmission in the prefrontal cortex of mice. METHODS: The extracellular monoamine levels in the prefrontal cortex were measured by in vivo microdialysis. RESULTS: (R)-Ketamine and (S)-ketamine acutely increased serotonin release in a dose-dependent manner, and the effect of (R)-ketamine was greater than that of (S)-ketamine. In contrast, (S)-ketamine caused a robust increase in dopamine release compared with (R)-ketamine. Both ketamine enantiomers increased noradrenaline release, but these effects did not differ. (2R,6R)-HNK caused a slight but significant increase in serotonin and noradrenaline but not dopamine release. (S)-NK increased dopamine and noradrenaline but not serotonin release. Differential effects between (R)-ketamine and (S)-ketamine were also observed in a lipopolysaccharide-induced model of depression. An α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor antagonist, 2,3-dioxo-6-nitro-1,2,3,4- tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX), attenuated (S)-ketamine-induced, but not (R)-ketamine-induced serotonin release, whereas NBQX blocked dopamine release induced by both enantiomers. Local application of (R)-ketamine into the prefrontal cortex caused a greater increase in prefrontal serotonin release than that of (S)-ketamine. CONCLUSIONS: (R)-Ketamine strongly activates the prefrontal serotonergic system through an AMPA receptor-independent mechanism. (S)-Ketamine-induced serotonin and dopamine release was AMPA receptor-dependent. These findings provide a neurochemical basis for the underlying pharmacological differences between ketamine enantiomers and their metabolites.


Assuntos
Ketamina/análogos & derivados , Ketamina/farmacologia , Córtex Pré-Frontal/metabolismo , Serotonina/metabolismo , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Ketamina/administração & dosagem , Ketamina/antagonistas & inibidores , Lipopolissacarídeos , Masculino , Camundongos , Microdiálise , Microinjeções , Norepinefrina/metabolismo , Quinoxalinas/farmacologia , Receptores de AMPA/metabolismo , Estereoisomerismo
2.
Gen Physiol Biophys ; 38(5): 427-434, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31411575

RESUMO

The objective of this study is to investigate the effects of nimesulide on ketamine-induced ovarian and uterine toxicity by biochemical and histopathological examinations. Ketamine is an anesthetic agent whose use leads to overproduction of catecholamines. Nimesulide is a cyclooxygenase-2 inhibitor, which has also been reported to exert a significant antioxidant effect. Wistar albino female rats were randomly divided into three groups as follows: ketamine group (60 mg/kg), ketamine (60 mg/kg) + nimesulide (50 mg/kg) group, and a healthy control group. Then, the biochemical levels and histopathological findings in the ovaries and uteri of the rats were examined for malondialdehyde, myeloperoxidase, total glutathione and superoxide dismutase. The study demonstrated that, in the uterine and ovarian tissues of rats that have been administered ketamine, there was a decrease in the levels of total glutathione and superoxide dismutase, while malondialdehyde and myeloperoxidase was increased: however it was observed that these ratios were reversed in the ketamine+nimesulide group. It was also proved that the negative effects of ketamine can be corrected with nimesulide when the myometrial and endometrial thicknesses are compared. Antioxidants such as nimesulide may protect against the damage caused by ketamine to the genital organs in young women.


Assuntos
Antioxidantes/farmacologia , Ketamina/efeitos adversos , Ketamina/antagonistas & inibidores , Ovário/efeitos dos fármacos , Sulfonamidas/farmacologia , Útero/efeitos dos fármacos , Animais , Feminino , Glutationa/metabolismo , Malondialdeído/metabolismo , Ovário/enzimologia , Ovário/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Útero/enzimologia , Útero/metabolismo
3.
Biochem Biophys Res Commun ; 489(4): 420-425, 2017 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-28577999

RESUMO

Ketamine shows promise as a therapeutic agent for the treatment of depression. The increased expression of brain-derived neurotrophic factor (BDNF) has been associated with the antidepressant-like effects of ketamine, but the mechanism of BDNF induction is not well understood. In the current study, we demonstrate that the treatment of rats with ketamine results in the dose-dependent rapid upregulation of Bdnf promoter IV activity and expression of Bdnf exon IV mRNAs in rat hippocampal neurons. Transfection of histone deacetylase 5 (HDAC5) into rat hippocampal neurons similarly induces Bdnf mRNA expression in response to ketamine, whereas transfection of a HDAC5 phosphorylation-defective mutant (Ser259 and Ser498 replaced by Ala259 and Ala498), results in the suppression of ketamine-mediated BDNF promoter IV transcriptional activity. Viral-mediated hippocampal knockdown of HDAC5 induces Bdnf mRNA and protein expression, and blocks the enhancing effects of ketamine on BDNF expression in both unstressed and stressed rats, and thereby providing evidence for the role of HDAC5 in the regulation of Bdnf expression. Taken together, our findings implicate HDAC5 in the ketamine-induced transcriptional regulation of Bdnf, and suggest that the phosphorylation of HDAC5 regulates the therapeutic actions of ketamine.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Hipocampo/citologia , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Ketamina/farmacologia , Neurônios/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Ketamina/antagonistas & inibidores , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Relação Estrutura-Atividade
4.
Metab Brain Dis ; 32(5): 1475-1489, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28508340

RESUMO

Effects of sertraline, haloperidol or olanzapine administration on ketamine-induced behaviours in mice were examined. The aim was to ascertain the degree of reversal of such behaviours by sertraline, and compare its effectiveness to haloperidol and olanzapine. Ten-week old mice (N = 120) were equally divided into main groups; 1 (open-field, radial-arm maze and elevated plus maze {EPM} tests), and 2 (social interaction test). Mice in each main group were assigned into six groups of ten (n = 10) each. Group 1 received intraperitoneal (i.p) injection of vehicle, while groups 2-6 received i.p ketamine at 15 mg/kg daily for 10 days. From day 11 to 24, mice in group 1 (vehicle) were given distilled water (i.p at 2 ml/kg and oral at 10 ml/kg), group 2 (ketamine control) received daily i.p ketamine and oral distilled water; while animals in groups 3-6 received daily i.p. ketamine and oral haloperidol (4 mg/kg), olanzapine (2 mg/kg), or one of two doses of sertraline (SERT) (2.5 or 5 mg/kg), respectively. Treatments were administered daily, and behaviours assessed on days 11 and 24. Results showed that repeated ketamine administration caused hyperlocomotion, increased self-grooming, memory loss and social withdrawal. Administration of sertraline (both doses), haloperidol, and olanzapine reversed ketamine-induced behavioural changes. However, in the EPM, sertraline and olanzapine were anxiolytic, while haloperidol was anxiogenic. Sertraline's effect on behaviours tested was comparable to olanzapine and better than haloperidol. In conclusion, this study shows that sertraline's ability to counteract ketamine-induced behavioural changes in mice is comparable to known antipsychotics.


Assuntos
Antidepressivos/farmacologia , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Benzodiazepinas/farmacologia , Antagonistas de Aminoácidos Excitatórios/toxicidade , Haloperidol/farmacologia , Ketamina/antagonistas & inibidores , Sertralina/farmacologia , Animais , Ansiedade/psicologia , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Asseio Animal/efeitos dos fármacos , Relações Interpessoais , Ketamina/toxicidade , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/psicologia , Memória de Curto Prazo/efeitos dos fármacos , Camundongos , Atividade Motora/efeitos dos fármacos , Olanzapina
5.
Anesthesiology ; 125(2): 333-45, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27341276

RESUMO

BACKGROUND: Calabadion 2 is a new drug-encapsulating agent. In this study, the authors aim to assess its utility as an agent to reverse general anesthesia with etomidate and ketamine and facilitate recovery. METHODS: To evaluate the effect of calabadion 2 on anesthesia recovery, the authors studied the response of rats to calabadion 2 after continuous and bolus intravenous etomidate or ketamine and bolus intramuscular ketamine administration. The authors measured electroencephalographic predictors of depth of anesthesia (burst suppression ratio and total electroencephalographic power), functional mobility impairment, blood pressure, and toxicity. RESULTS: Calabadion 2 dose-dependently reverses the effects of ketamine and etomidate on electroencephalographic predictors of depth of anesthesia, as well as drug-induced hypotension, and shortens the time to recovery of righting reflex and functional mobility. Calabadion 2 displayed low cytotoxicity in MTS-3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium-based cell viability and adenylate kinase release cell necrosis assays, did not inhibit the human ether-à-go-go-related channel, and was not mutagenic (Ames test). On the basis of maximum tolerable dose and acceleration of righting reflex recovery, the authors calculated the therapeutic index of calabadion 2 in recovery as 16:1 (95% CI, 10 to 26:1) for the reversal of ketamine and 3:1 (95% CI, 2 to 5:1) for the reversal of etomidate. CONCLUSIONS: Calabadion 2 reverses etomidate and ketamine anesthesia in rats by chemical encapsulation at nontoxic concentrations.


Assuntos
Anestesia Geral/métodos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Ácidos Sulfônicos/farmacologia , Anestésicos Dissociativos/toxicidade , Anestésicos Intravenosos/toxicidade , Animais , Pressão Sanguínea/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Etomidato/antagonistas & inibidores , Etomidato/toxicidade , Ketamina/antagonistas & inibidores , Ketamina/toxicidade , Masculino , Mutagênicos/toxicidade , Necrose/prevenção & controle , Equilíbrio Postural/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reflexo/efeitos dos fármacos
6.
Int J Neuropsychopharmacol ; 17(8): 1321-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24852262

RESUMO

Several lines of evidence indicate that ketamine has a rapid antidepressant-like effect in rodents and humans, but underlying mechanisms are unclear. In the present study, we investigated the effect of ketamine on serotonin (5-HT) release in the rat prefrontal cortex by in vivo microdialysis. A subcutaneous administration of ketamine (5 and 25 mg/kg) significantly increased the prefrontal 5-HT level in a dose-dependent manner, which was attenuated by local injection of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) antagonists into the dorsal raphe nucleus (DRN). Direct stimulation of AMPARs in the DRN significantly increased prefrontal 5-HT level, while intra-DRN injection of ketamine (36.5 nmol) had no effect. Furthermore, intra-DRN injection of an α 4 ß 2-nicotinic acetylcholine receptor (nAChR) antagonist, dihydro-ß-erythroidine (10 nmol), significantly attenuated the subcutaneous ketamine-induced increase in prefrontal 5-HT levels. These results suggest that AMPARs and α 4 ß 2-nAChRs in the DRN play a key role in the ketamine-induced 5-HT release in the prefrontal cortex.


Assuntos
Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/metabolismo , Ketamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores Nicotínicos/metabolismo , Serotonina/metabolismo , Animais , Antidepressivos/administração & dosagem , Antidepressivos/farmacologia , Benzotiadiazinas/administração & dosagem , Benzotiadiazinas/farmacologia , Di-Hidro-beta-Eritroidina/administração & dosagem , Di-Hidro-beta-Eritroidina/farmacologia , Relação Dose-Resposta a Droga , Injeções Subcutâneas , Ketamina/administração & dosagem , Ketamina/antagonistas & inibidores , Masculino , Microdiálise , Microinjeções , Antagonistas Nicotínicos/administração & dosagem , Antagonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/metabolismo , Quinoxalinas/administração & dosagem , Quinoxalinas/farmacologia , Ratos , Receptores de AMPA/agonistas , Receptores de AMPA/antagonistas & inibidores , Espermina/administração & dosagem , Espermina/análogos & derivados , Espermina/farmacologia
7.
Br J Anaesth ; 110 Suppl 1: i3-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23533250

RESUMO

BACKGROUND: Ketamine-induced neuroapoptosis has been attributed to diverse stress-related mechanisms. Glycogen synthase kinase-3ß (GSK-3ß) is a multifunctional kinase that is active in neuronal development and linked to neurodegenerative disorders. We hypothesized that ketamine would enhance GSK-3ß-induced neuroapopotosis, and that lithium, an inhibitor of GSK-3ß, would attenuate this response in vivo. METHODS: Protein levels of cleaved caspase-3, protein kinase B (AKT), GSK-3ß, and cyclin D1 were measured in post-natal day 7 rat pups after 1.5, 3, 4.5, and 6 h exposure to ketamine. A cohort of rat pups was randomized to a 6 h exposure to ketamine with and without lithium. Neuroapoptosis was measured by cleaved caspase-3 and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labelling staining by immunohistochemistry. Protein levels of cleaved caspase-3 and -9 and the total and phosphorylated forms of AKT, GSK-3ß, and cyclin D1 (cell cycle protein) were also measured. RESULTS: Ketamine produced a duration-dependent increase in cleaved caspase-3 and cyclin D1, which corresponded to decreases in phosphorylated AKT and GSK-3ß. Co-administration of lithium with ketamine attenuated this response. CONCLUSIONS: Ketamine-induced neuroapoptosis is associated with a temporal decrease in GSK-3ß phosphorylation, and simultaneous administration of lithium mitigated this response. These findings suggest that GSK-3ß is activated during this ketamine-induced neuroapoptosis.


Assuntos
Anestésicos Dissociativos/toxicidade , Quinase 3 da Glicogênio Sintase/fisiologia , Ketamina/toxicidade , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/enzimologia , Anestésicos Dissociativos/antagonistas & inibidores , Anestésicos Dissociativos/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 3/biossíntese , Ativação Enzimática/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Ketamina/antagonistas & inibidores , Ketamina/farmacologia , Cloreto de Lítio/farmacologia , Cloreto de Lítio/uso terapêutico , Neurônios/enzimologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/prevenção & controle , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley
8.
J Biomed Sci ; 19: 11, 2012 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-22300389

RESUMO

BACKGROUND: Acute exposure of ethanol (alcohol) inhibits NMDA receptor function. Our previous study showed that acute ethanol inhibited the pressor responses induced by NMDA applied intrathecally; however, prolonged ethanol exposure may increase the levels of phosphorylated NMDA receptor subunits leading to changes in ethanol inhibitory potency on NMDA-induced responses. The present study was carried out to examine whether acute ethanol exposure influences the effects of ketamine, a noncompetitive NMDA receptor antagonist, on spinal NMDA-induced pressor responses. METHODS: The blood pressure responses induced by intrathecal injection of NMDA were recorded in urethane-anesthetized rats weighing 250-275 g. The levels of several phosphorylated residues on NMDA receptor GluN1 subunits were determined by western blot analysis. RESULTS: Intravenous injection of ethanol or ketamine inhibited spinal NMDA-induced pressor responses in a dose-dependent and reversible manner. Ketamine inhibition of NMDA-induced responses was synergistically potentiated by ethanol when ethanol was applied just before ketamine. However, ketamine inhibition was significantly reduced when applied at 10 min after ethanol administration. Western blot analysis showed that intravenous ethanol increased the levels of phosphoserine 897 on GluN1 subunits (pGluN1-serine 897), selectively phosphorylated by protein kinase A (PKA), in the lateral horn regions of spinal cord at 10 min after administration. Intrathecal administration of cAMPS-Sp, a PKA activator, at doses elevating the levels of pGluN1-serine 897, significantly blocked ketamine inhibition of spinal NMDA-induced responses. CONCLUSIONS: The results suggest that ethanol may differentially regulate ketamine inhibition of spinal NMDA receptor function depending on ethanol exposure time and the resulting changes in the levels of pGluN1-serine 897.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , AMP Cíclico/análogos & derivados , Etanol/toxicidade , Ketamina/farmacologia , N-Metilaspartato/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Medula Espinal/metabolismo , Tionucleotídeos/farmacologia , Animais , Western Blotting , AMP Cíclico/administração & dosagem , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Injeções Espinhais , Ketamina/antagonistas & inibidores , Masculino , N-Metilaspartato/administração & dosagem , Fosforilação , Fosfosserina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Medula Espinal/efeitos dos fármacos , Tionucleotídeos/administração & dosagem , Fatores de Tempo
9.
Acta Anaesthesiol Scand ; 56(8): 1058-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22694670

RESUMO

BACKGROUND: An increasing amount of both experimental and epidemiological data indicates that neonatal anaesthesia causes disruption of normal brain development in rodents and primates, as manifested by acute increased apoptosis and long-lasting altered behaviour and learning. It is necessary to seek strategies that avoid the possible adverse effects after anaesthesia. Our purpose is to show that increased apoptosis and behavioural alterations after ketamine exposure during this period may be prevented by clonidine, a compound already used by paediatric anaesthetists for sedation. METHODS: To investigate the protective properties of clonidine pre-treatment, five groups of 10-day-old mice were injected with either ketamine 50 mg/kg, clonidine 40 µg/kg, ketamine 50 mg/kg 30 min after 10 µg/kg clonidine, ketamine 50 mg/kg 30 min after 40 µg/kg clonidine or saline (control). Apoptosis was measured 24 h after treatment using Flouro-Jade staining. Spontaneous activity in a novel environment was tested at an age of 55 days. RESULTS: Pre-treatment with 40 µg/kg clonidine, but not 10 µg/kg clonidine, 30 min before ketamine exposure abolished ketamine-induced apoptosis and the behavioural changes observed in the young adult mice. The mice exposed to clonidine alone showed no differences from the saline-treated (control) mice. CONCLUSION: The administration of clonidine eliminated the adverse effects of ketamine in this mouse model, suggesting a possible strategy for protection. Alone, clonidine did not cause any adverse effects in these tests.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Anestésicos Dissociativos/antagonistas & inibidores , Anestésicos Dissociativos/toxicidade , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Clonidina/farmacologia , Ketamina/antagonistas & inibidores , Ketamina/toxicidade , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Meio Ambiente , Feminino , Fluoresceínas , Corantes Fluorescentes , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Compostos Orgânicos
10.
Anesthesiology ; 114(6): 1345-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21478733

RESUMO

BACKGROUND: There is poor experimental evidence concerning the effects of anesthetic doses of the noncompetitive N-methyl-D-aspartate receptor antagonist ketamine on rodents' memory abilities. The current study was designed (1) to investigate the consequences of posttraining administration of anesthetic ketamine (100 mg/kg intraperitoneally) on rats' recognition memory and (2) to evaluate the ability of the nitric oxide synthase inhibitor N-nitro-L-arginine methylester (L-NAME; 1, 3, and 10 mg/kg intraperitoneally) to counteract the expected behavioral deficits produced by anesthetic ketamine. Finally, in an attempt to clarify if the expected memory impairments produced by anesthetic ketamine were related to the anesthesia, we also tested the effects of a subanesthetic dose of it (3 mg/kg intraperitoneally) on rats' recognition memory. METHODS: The novel object recognition test, a procedure assessing recognition memory in rats, was selected. RESULTS: Posttraining administration of anesthetic (but not of subanesthetic) ketamine disrupted rats' performance in the novel object recognition paradigm. The discrimination index (D) was decreased by ketamine from 0.415 (using saline) to 0.128, thus indicating that the anesthetic dose of ketamine impaired recognition memory. L-NAME (1-3, but not 10, mg/kg) reversed this memory deficit produced by ketamine; the D index of 0.128 using ketamine treatment was increased by 1 and 3 mg/kg L-NAME to 0.427 and 0.478, respectively. CONCLUSIONS: The current results indicate that anesthetic ketamine impaired rats' posttraining memory components (storage and/or retrieval of information) and that a nitric oxide component modulates its behavioral effects.


Assuntos
Ketamina/toxicidade , Transtornos da Memória/induzido quimicamente , Rememoração Mental/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Reconhecimento Psicológico/efeitos dos fármacos , Anestésicos Dissociativos/antagonistas & inibidores , Anestésicos Dissociativos/toxicidade , Animais , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Ketamina/antagonistas & inibidores , Masculino , Transtornos da Memória/enzimologia , Transtornos da Memória/prevenção & controle , Rememoração Mental/fisiologia , NG-Nitroarginina Metil Éster/uso terapêutico , Óxido Nítrico Sintase/metabolismo , Estudos Prospectivos , Ratos , Ratos Wistar , Reconhecimento Psicológico/fisiologia
11.
Psychopharmacology (Berl) ; 238(4): 1157-1169, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33483802

RESUMO

Ketamine produces a rapid antidepressant response in over 50% of adults with treatment-resistant depression. A long infusion of ketamine may provide durable remission of depressive symptoms, but the safety, efficacy, and neurobiological correlates are unknown. In this open-label, proof-of-principle study, adults with treatment-resistant depression (N = 23) underwent a 96-h infusion of intravenous ketamine (0.15 mg/kg/h titrated toward 0.6 mg/kg/h). Clonidine was co-administered to reduce psychotomimetic effects. We measured clinical response for 8 weeks post-infusion. Resting-state functional magnetic resonance imaging was used to assess functional connectivity in patients pre- and 2 weeks post-infusion and in matched non-depressed controls (N = 27). We hypothesized that responders to therapy would demonstrate response-dependent connectivity changes while all subjects would show treatment-dependent connectivity changes. Most participants completed infusion (21/23; mean final dose 0.54 mg/kg/h, SD 0.13). The infusion was well tolerated with minimal cognitive and psychotomimetic side effects. Depressive symptoms were markedly reduced (MADRS 29 ± 4 at baseline to 9 ± 8 one day post-infusion), which was sustained at 2 weeks (13 ± 8) and 8 weeks (15 ± 8). Imaging demonstrated a response-dependent decrease in hyperconnectivity of the subgenual anterior cingulate cortex to the default mode network, and a treatment-dependent decrease in hyperconnectivity within the limbic system (hippocampus, amygdala, medial thalamus, nucleus accumbens). In exploratory analyses, connectivity was increased between the limbic system and frontal areas, and smaller right hippocampus volume at baseline predicted larger MADRS change. A single prolonged infusion of ketamine provides a tolerated, rapid, and sustained response in treatment-resistant depression and normalizes depression-related hyperconnectivity in the limbic system and frontal lobe. ClinicalTrials.gov : Treatment Resistant Depression (Pilot), NCT01179009.


Assuntos
Antidepressivos/uso terapêutico , Transtorno Depressivo Resistente a Tratamento/tratamento farmacológico , Ketamina/uso terapêutico , Sistema Límbico/efeitos dos fármacos , Adolescente , Adulto , Idoso , Antidepressivos/administração & dosagem , Clonidina/uso terapêutico , Transtorno Depressivo Resistente a Tratamento/diagnóstico por imagem , Transtorno Depressivo Resistente a Tratamento/psicologia , Feminino , Giro do Cíngulo/efeitos dos fármacos , Alucinógenos/efeitos adversos , Humanos , Infusões Intravenosas , Ketamina/administração & dosagem , Ketamina/antagonistas & inibidores , Sistema Límbico/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/efeitos dos fármacos , Escalas de Graduação Psiquiátrica , Simpatolíticos/uso terapêutico , Resultado do Tratamento , Adulto Jovem
12.
Int J Neuropsychopharmacol ; 13(1): 71-82, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19288975

RESUMO

The N-methyl-D-aspartate (NMDA) glutamate receptor antagonist ketamine may have rapid, albeit transient, antidepressant properties. This study in patients with treatment-resistant major depression (TRD) aimed to (1) replicate the acute efficacy of single-dose intravenous (i.v.) ketamine; (2) test the efficacy of the glutamate-modulating agent riluzole in preventing post-ketamine relapse; and (3) examine whether pretreatment with lamotrigine would attenuate ketamine's psychotomimetic effects and enhance its antidepressant activity. Twenty-six medication-free patients received open-label i.v. ketamine (0.5 mg/kg over 40 min). Two hours prior to infusion, patients were randomized to lamotrigine (300 mg) or placebo. Seventeen patients (65%) met response criterion (50% reduction from baseline on the Montgomery-Asberg Depression Rating Scale) 24 h following ketamine. Lamotrigine failed to attenuate the mild, transient side-effects associated with ketamine and did not enhance its antidepressant effects. Fourteen patients (54%) met response criterion 72 h following ketamine and proceeded to participate in a 32-d, randomized, double-blind, placebo-controlled, flexible-dose continuation trial of riluzole (100-200 mg/d). The main outcome measure was time-to-relapse. An interim analysis found no significant differences in time-to-relapse between riluzole and placebo groups [log-rank chi(2) = 0.17, d.f. = 1, p = 0.68], with 80% of patients relapsing on riluzole vs. 50% on placebo. The trial was thus stopped for futility. This pilot study showed that a sub-anaesthetic dose of i.v. ketamine is well-tolerated in TRD, and may have rapid and sustained antidepressant properties. Riluzole did not prevent relapse in the first month following ketamine. Further investigation of relapse prevention strategies post-ketamine is necessary.


Assuntos
Depressão/tratamento farmacológico , Depressão/prevenção & controle , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/antagonistas & inibidores , Riluzol/uso terapêutico , Triazinas/farmacologia , Adulto , Idoso , Depressão/diagnóstico , Interações Medicamentosas , Resistência a Medicamentos , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Feminino , Alucinógenos/antagonistas & inibidores , Humanos , Injeções Intravenosas , Ketamina/administração & dosagem , Lamotrigina , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Placebos , Prevenção Secundária , Fatores de Tempo , Resultado do Tratamento , Triazinas/uso terapêutico
14.
Artigo em Inglês | MEDLINE | ID: mdl-31676464

RESUMO

Ketamine has addictive potential, a troublesome fact due to its promising use as a therapeutic drug. An important phenomenon associated with drug addiction is behavioral sensitization, usually characterized as augmented locomotion. However, other behaviors may also be susceptible to sensitization, and/or interfere with locomotor activity. Thus, this study drew a comprehensive behavioral 'profiling' in an animal model of repeated administration of ketamine. Adult Swiss mice received single daily ketamine injections (30 or 50 mg/Kg, i.p.), which were followed by open field testing for 7 days (acquisition period, ACQ). A ketamine challenge (sensitization test, ST) was carried out after a 5-day withdrawal. Locomotion, rearing, grooming, rotation and falling were assessed during ACQ and ST. All behaviors were affected from the first ACQ day onwards, with no indication of competition between locomotion and the other behaviors. Only locomotion in response to 30 mg/Kg of ketamine both escalated during ACQ and expressed increased levels at ST, evidencing development and expression of locomotor sensitization. Considering the involvement of serotonin 5HT(2) and dopamine D(2) receptors on addiction mechanisms, we further tested the involvement of these receptors in ketamine-induced sensitization. Ketanserin (5HT2 antagonist, 3 mg/Kg, s.c.) prevented ketamine-evoked development of locomotor sensitization. However, ketanserin pretreatment during ACQ failed to inhibit its expression during ST. Raclopride (D2 antagonist, 0.5 mg/Kg, s.c.) evoked less robust reductions in locomotion but prevented the development of ketamine-evoked sensitization. Pretreatment during ACQ further inhibited the expression of sensitization during ST. These results indicate that a partial overlap in serotonergic and dopaminergic mechanisms underlies ketamine-induced locomotor sensitization.


Assuntos
Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Ketamina/farmacologia , Locomoção/efeitos dos fármacos , Receptores de Dopamina D2/fisiologia , Receptores 5-HT2 de Serotonina/fisiologia , Animais , Relação Dose-Resposta a Droga , Feminino , Ketamina/antagonistas & inibidores , Ketanserina/farmacologia , Masculino , Camundongos , Racloprida/farmacologia , Comportamento Estereotipado/efeitos dos fármacos
15.
Sci Rep ; 10(1): 5265, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32210319

RESUMO

Anesthetics are used to produce hypnosis and analgesic effects during surgery, but anesthesia for a long time after the operation is not conducive to the recovery of animals or patients. Therefore, finding appropriate treatments to counter the effects of anesthetics could enhance postoperative recovery. In the current study, we discovered the novel role of a GluN2A-selective positive allosteric modulator (PAM) in ketamine-induced anesthesia and investigated the effects of the PAM combined with nalmefene and flumazenil (PNF) in reversing the actions of an anesthetic combination (ketamine-fentanyl-dexmedetomidine, KFD). PAM treatment dose-dependently decreased the duration of the ketamine-induced loss of righting reflex (LORR). Compared with those in the KFD group, the duration of LORR and the analgesic effect of the KFD + PNF group were obviously decreased. Meanwhile, successive administration of PNF and KFD had no adverse effects on the cardiovascular and respiratory systems. Both the KFD group and the KFD + PNF group showed no changes in hepatic and renal function or cognitive function in rats. Moreover, the recovery of motor coordination of the KFD + PNF group was faster than that of the KFD group. In summary, our results suggest the potential application of the PNF combination as an antagonistic treatment strategy for anesthesia.


Assuntos
Analgesia , Anestesia , Dexmedetomidina/antagonistas & inibidores , Fentanila/antagonistas & inibidores , Flumazenil/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Ketamina/antagonistas & inibidores , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores de N-Metil-D-Aspartato/agonistas , Adjuvantes Anestésicos , Regulação Alostérica , Animais , Recuperação Demorada da Anestesia/tratamento farmacológico , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Feminino , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Naltrexona/farmacologia , Nociceptividade/efeitos dos fármacos , Medição da Dor , Ratos , Reflexo de Endireitamento/efeitos dos fármacos , Teste de Desempenho do Rota-Rod
16.
J Med Primatol ; 38(6): 404-10, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19735352

RESUMO

BACKGROUND: The potential of Atipamezole (ATI) to reverse Ketamine/Xylazine (KET/XYL) anesthesia in the Olive baboon (Papio anubis) was studied. METHODS: Anesthesia was induced with 10 mg/kg KET and 0.5 mg/kg XYL intramuscularly. Mean arousal time (MAT), heart rate (HR), systolic arterial blood pressure (SAP), rectal temperature, respiratory rate (RR), and hemoglobin oxygen saturation (SpO(2)) were monitored. Baboons were treated with: KET/XYL only, KET/XYL followed by 100 microg/kg ATI or by 200 microg/kg ATI administered 25 minutes after KET/XYL. RESULTS: Atipamezole rapidly reversed depressed HR and SAP (10 +/- 5.2 minutes), RR (5 +/- 2 minutes) and SpO(2) (3 +/- 6 minutes) and significantly decreased MAT (13 +/- 2.2 minutes) vs. KET/XYL alone (35 +/- 5 minutes). Emesis was absent and salivation was observed after administration of 200 microg/kg ATI only. CONCLUSIONS: Atipamezole at 100 microg/kg is sufficient for rapid and smooth reversal of KET/XYL anesthesia in the Olive baboon with minimal side effects.


Assuntos
Antagonistas Adrenérgicos alfa/farmacologia , Anestésicos Dissociativos/antagonistas & inibidores , Imidazóis/farmacologia , Ketamina/antagonistas & inibidores , Papio anubis , Xilazina/antagonistas & inibidores , Anestésicos Dissociativos/administração & dosagem , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Injeções Intramusculares , Ketamina/administração & dosagem , Masculino , Taxa Respiratória/efeitos dos fármacos , Xilazina/administração & dosagem
17.
Neurotoxicology ; 30(1): 151-4, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19038286

RESUMO

Glutamate activation of the NMDA receptor is essential for neuronal differentiation, migration, and survival. Treatment with NMDA receptor antagonists, such as ketamine (KET) or phencyclidine (PCP), can trigger apoptosis in neonatal rats. However, L-carnitine (LC) treatment appears to prevent glutamate-induced toxicity in the developing CNS. Previously, we described altered preweaning behaviors (i.e., abnormal home cage, slant board and forelimb hang behaviors) resulting from neonatal PCP and KET treatment. Those adverse effects of KET were somewhat ameliorated by LC [Boctor SY, Wang C, Ferguson SA. Neonatal PCP is more potent than ketamine at modifying preweaning behaviors of Sprague-Dawley rats. Toxicol Sci 2008;106:172-9]. Here, a portion of those subjects were evaluated for prepulse inhibition (PPI) of the acoustic startle response at postnatal day (PND) 25 since previous reports described PCP-induced effects on this response. Rats were subcutaneously treated with: saline; 10 mg/kg PCP (1x/day) on PNDs 7, 9 and 11; 20 mg/kg KET (6 injections every 2h on PND 7); or a similar regimen of ketamine and 250 mg/kg LC on PND 7, with a single injection of 250 mg/kg LC on PNDs 8-11 (KLC). Male and female rats were assessed using a standard PPI paradigm with prepulses of 68, 78 and 82 dB. Body weight was decreased 17-21% and whole brain weight was decreased 10% in PCP-treated rats. Specifically, cerebellar weight was significantly less in PCP-treated rats relative to control. Despite the magnitude of those PCP-induced changes, startle response in normal pulse only trials and percent of PPI in PCP-, KET-, and KLC-treated groups were comparable to controls. Average latency to maximum startle was 2.6 ms less in females than males (p<0.007); there were no other significant sex effects. The lack of neonatal PCP treatment on later PPI is similar to that reported by Rasmussen et al. [Rasmussen BA, O'Neil J, Manaye KF, Perry DC, Tizabi Y. Long-term effects of developmental PCP administration on sensorimotor gating in male and female rats. Psychopharmacology (Berl) 2007; 190: 43-9.], and indicates that neonatal PCP-induced effects on PPI [Wang C, McInnis J, Ross-Sanchez M, Shinnick-Gallagher P, Wiley JL, Johnson KM. Long-term behavioral and neurodegenerative effects of perinatal phencyclidine administration: implications for schizophrenia. Neuroscience 2001; 107: 535-50.] appear difficult to replicate.


Assuntos
Inibição Psicológica , Ketamina/administração & dosagem , Fenciclidina/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Carnitina/farmacologia , Cerebelo/efeitos dos fármacos , Feminino , Ketamina/antagonistas & inibidores , Ketamina/farmacologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Fenciclidina/administração & dosagem , Fenciclidina/antagonistas & inibidores , Período Pós-Parto , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/efeitos dos fármacos , Caracteres Sexuais
18.
Anesth Analg ; 108(2): 491-5, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19151277

RESUMO

BACKGROUND: Orexins (OXs) regulate wakefulness, and a lack of OX Type-I receptors cause narcolepsy. OX selectively increases norepinephrine (NE) release from rat cerebral cortical slices, and brain noradrenergic neurons are involved in the sleep-wakefulness cycle. Ketamine increases NE release from the rat cerebral cortex. We hypothesized that OX would affect ketamine anesthesia's interactions with brain noradrenergic neuronal activity. METHODS: We used Sprague Dawley rats. We studied 1) in vivo effects of orexin A (OXA) and SB-334867-A (Orexin-1 receptor antagonist) on ketamine-induced anesthesia time, 2) in vivo effects of OXA on ketamine-induced increase in NE release from the frontal cortex assessed using microdialysis, and 3) in vitro effects of ketamine on OXA-evoked NE release from rat cerebrocortical slices. RESULTS: 1) Intracerebroventricular OXA 1 nmol significantly decreased ketamine anesthesia time by 20%-30% at 50, 100, and 125 mg/kg intraperitoneal (IP) ketamine. SB-334867-A fully reversed the decrease produced by OXA. 2) OXA also decreased the release of NE induced by ketamine even though OXA increased the release of NE in rat prefrontal cortex. Maximum NE release in Group OX + K (intracerebroventricular OXA 1 nmol + IP ketamine 100 mg/kg) was 271% and was significantly smaller than that in Group K (ketamine 100 mg/kg IP, 390% of baseline, P = 0.029). 3) Ketamine inhibited OX-evoked NE release with clinically relevant IC(50) values. CONCLUSION: Orexinergic neurons may be an important target for ketamine. OXA antagonized ketamine anesthesia via Orexin-1 receptor with noradrenergic neurons.


Assuntos
Anestesia , Anestésicos Dissociativos/antagonistas & inibidores , Encéfalo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Ketamina/antagonistas & inibidores , Neurônios/fisiologia , Neuropeptídeos/farmacologia , Norepinefrina/fisiologia , Anestésicos Dissociativos/farmacologia , Animais , Benzoxazóis/farmacologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Técnicas In Vitro , Injeções Intraventriculares , Ketamina/farmacologia , Masculino , Microdiálise , Naftiridinas , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Norepinefrina/metabolismo , Orexinas , Ratos , Receptores de Neuropeptídeos/antagonistas & inibidores , Ureia/análogos & derivados , Ureia/farmacologia
19.
J Wildl Dis ; 45(3): 877-80, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19617505

RESUMO

Twenty adult, free-ranging, female Svalbard reindeer (Rangifer tarandus platyrhynchus) were immobilized with medetomidine-ketamine from 30 September through 9 October 1999 at Svalbard, Norway (78 degrees 55'N, 11 degrees 56'E). The animals were approached on foot, and the drugs were administered into the heavy muscles of the shoulder or the thigh by dart syringe injection from 15-25 m. The mean (SD) induction time in 10 animals immobilized with 0.113 (0.009) mg/kg of medetomidine and 2.26 (0.19) mg/kg of ketamine (group 2) was significantly shorter (P < 0.05) than in 10 animals immobilized with 0.215 (0.043) mg/kg of medetomidine and 1.08 (0.21) mg/kg of ketamine (group 1): 6.5 (3.2) versus 14.3 (10.6) min, respectively. Inductions were calm, major clinical side effects were not detected, and there were no significant differences between groups regarding rectal temperature, pulse rate, respiratory rate, or relative arterial oxygen saturation. The 5 mg of atipamezole/1 mg of medetomidine were given half intramuscularly and half subcutaneously for reversal, and the animals were standing within 9.5 (4.5, group 1) and 13.0 (6.4, group 2) min, respectively, after administration of the antagonist.


Assuntos
Anestésicos Combinados/administração & dosagem , Hipnóticos e Sedativos/administração & dosagem , Imobilização/veterinária , Rena/fisiologia , Antagonistas Adrenérgicos alfa/farmacologia , Anestésicos Combinados/antagonistas & inibidores , Anestésicos Dissociativos , Animais , Animais Selvagens/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Relação Dose-Resposta a Droga , Feminino , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Hipnóticos e Sedativos/antagonistas & inibidores , Imidazóis/farmacologia , Imobilização/métodos , Injeções Intramusculares/veterinária , Injeções Subcutâneas/veterinária , Ketamina/administração & dosagem , Ketamina/antagonistas & inibidores , Medetomidina/administração & dosagem , Medetomidina/antagonistas & inibidores
20.
Neuropharmacology ; 148: 87-95, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30597159

RESUMO

Experimental evidence indicates that anesthetic doses of the non-competitive NMDA receptor antagonist ketamine impair memory abilities in rodents. The mechanism by which anesthetic ketamine produces its adverse behavioural effects is not yet clarified. In this context, it has been proposed that the effects of anesthetic ketamine on memory might be attributed to its agonistic properties on the GABA type A receptor. The present study was designed to address this issue. Thus, we investigated the ability of the benzodiazepine receptor antagonist flumazenil (1, 3, 6 mg/kg, i.p.) and the GABAA receptor antagonist bicuculline (0.5, 1.5, 3 mg/kg, i.p.) to counteract recognition memory deficits produced by anesthetic ketamine (100 mg/kg, i.p.) in rats. For this purpose, the novel object recognition task, a behavioural paradigm assessing recognition memory abilities in rodents was used. Compounds were coadministered 24 h before testing or retention. Pre (24 h before testing) or post-training (24 h before retention) administration of flumazenil (6 mg/kg, i.p.) counteracted anesthetic ketamine-induced performance deficits in the novel object recognition memory task. Conversely, bicuculline failed to attenuate the recognition memory deficits caused by anesthetic ketamine. Our findings propose a functional interaction between anesthetic ketamine and the GABAA receptor allosteric modulator flumazenil on recognition memory.


Assuntos
Bicuculina/farmacologia , Flumazenil/farmacologia , Ketamina/efeitos adversos , Transtornos da Memória/prevenção & controle , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas GABAérgicos/farmacologia , Ketamina/antagonistas & inibidores , Masculino , Transtornos da Memória/induzido quimicamente , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA