Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 205
1.
Psychopharmacology (Berl) ; 238(4): 1157-1169, 2021 Apr.
Article En | MEDLINE | ID: mdl-33483802

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.


Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy , Ketamine/therapeutic use , Limbic System/drug effects , Adolescent , Adult , Aged , Antidepressive Agents/administration & dosage , Clonidine/therapeutic use , Depressive Disorder, Treatment-Resistant/diagnostic imaging , Depressive Disorder, Treatment-Resistant/psychology , Female , Gyrus Cinguli/drug effects , Hallucinogens/adverse effects , Humans , Infusions, Intravenous , Ketamine/administration & dosage , Ketamine/antagonists & inhibitors , Limbic System/diagnostic imaging , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Psychiatric Status Rating Scales , Sympatholytics/therapeutic use , Treatment Outcome , Young Adult
2.
Sci Rep ; 10(1): 5265, 2020 03 24.
Article En | MEDLINE | ID: mdl-32210319

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.


Analgesia , Anesthesia , Dexmedetomidine/antagonists & inhibitors , Fentanyl/antagonists & inhibitors , Flumazenil/pharmacology , GABA-A Receptor Antagonists/pharmacology , Ketamine/antagonists & inhibitors , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Adjuvants, Anesthesia , Allosteric Regulation , Animals , Delayed Emergence from Anesthesia/drug therapy , Drug Combinations , Drug Evaluation, Preclinical , Female , Male , Maze Learning/drug effects , Naltrexone/pharmacology , Nociception/drug effects , Pain Measurement , Rats , Reflex, Righting/drug effects , Rotarod Performance Test
3.
Article En | MEDLINE | ID: mdl-31676464

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.


Central Nervous System Sensitization/drug effects , Ketamine/pharmacology , Locomotion/drug effects , Receptors, Dopamine D2/physiology , Receptors, Serotonin, 5-HT2/physiology , Animals , Dose-Response Relationship, Drug , Female , Ketamine/antagonists & inhibitors , Ketanserin/pharmacology , Male , Mice , Raclopride/pharmacology , Stereotyped Behavior/drug effects
4.
Gen Physiol Biophys ; 38(5): 427-434, 2019 Sep.
Article En | MEDLINE | ID: mdl-31411575

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.


Antioxidants/pharmacology , Ketamine/adverse effects , Ketamine/antagonists & inhibitors , Ovary/drug effects , Sulfonamides/pharmacology , Uterus/drug effects , Animals , Female , Glutathione/metabolism , Malondialdehyde/metabolism , Ovary/enzymology , Ovary/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Random Allocation , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Uterus/enzymology , Uterus/metabolism
5.
Int J Neuropsychopharmacol ; 22(10): 665-674, 2019 10 01.
Article En | MEDLINE | ID: mdl-31325908

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.


Ketamine/analogs & derivatives , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Serotonin/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Ketamine/administration & dosage , Ketamine/antagonists & inhibitors , Lipopolysaccharides , Male , Mice , Microdialysis , Microinjections , Norepinephrine/metabolism , Quinoxalines/pharmacology , Receptors, AMPA/metabolism , Stereoisomerism
6.
Neurosci Lett ; 699: 140-144, 2019 04 23.
Article En | MEDLINE | ID: mdl-30716423

The participation of endocannabinoids in central and peripheral antinociception induced by several compounds has been shown by our group. In this study, we investigated the effect of endocannabinoids on the central antinociception induced by ketamine. The nociceptive threshold for thermal stimulation was measured using the tail-flick test in Swiss mice. The drugs were administered intracerebroventricularly. Probabilities less than 5% (p < 0.05) were considered to be statistically significant (Two-way ANOVA/Bonferroni's test). The CB1-selective cannabinoid receptor antagonist AM251 (2 and 4 µg) completely reversed the central antinociception induced by ketamine (4 µg) in a dose-dependent manner. In contrast, the CB2-selective cannabinoid receptor antagonist AM630 (2 and 4 µg) did not antagonize this effect. Additionally, the administration of the anandamide amidase inhibitor MAFP (0.2 µg) and anandamide uptake inhibitor VDM11 (4 µg) significantly enhanced the antinociception induced by a low dose of ketamine (2 µg). It was concluded that central antinociception induced by ketamine involves the activation of CB1 cannabinoid receptors. Mobilization of cannabinoids might be required for the activation of those receptors, since inhibitors of the endogenous cannabinoids potentiate the effect of Ketamine.


Analgesics/pharmacology , Cannabinoids/metabolism , Ketamine/pharmacology , Receptor, Cannabinoid, CB1/agonists , Animals , Arachidonic Acids/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Indoles/pharmacology , Infusions, Intraventricular , Ketamine/administration & dosage , Ketamine/antagonists & inhibitors , Male , Mice , Organophosphonates/pharmacology , Pain Measurement/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors
7.
Neuropharmacology ; 148: 87-95, 2019 04.
Article En | MEDLINE | ID: mdl-30597159

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.


Bicuculline/pharmacology , Flumazenil/pharmacology , Ketamine/adverse effects , Memory Disorders/prevention & control , Recognition, Psychology/drug effects , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Ketamine/antagonists & inhibitors , Male , Memory Disorders/chemically induced , Rats
8.
Neuropharmacology ; 148: 107-116, 2019 04.
Article En | MEDLINE | ID: mdl-30590060

The impairment of social behaviors induced by social defeat stress exposure as juveniles is resistant to some antidepressants and an antipsychotic, although the underlying mechanisms and/or therapeutic target are not yet clear. In this study, we investigated the involvement of the glutamatergic neuronal system in the impairment of social behaviors in this model, as this system is known to be involved in many central pathologies. Acute administration of ketamine, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist and subsequent stimulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, attenuated the expression of impairment of social behaviors. Lack of the NMDA receptor GluN2A subunit or acute administration of ifenprodil, an NMDA receptor GluN2B subunit antagonist, did not cause an effect. There were no significant changes in NMDA function, as determined by the ratios of phosphorylated NMDA receptor subunits in the prefrontal cortex and hippocampus. 2,3-Dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione, a selective AMPA receptor antagonist, prevented the effect of ketamine on the expression of impairment of social behaviors. On the contrary, the ratio of phosphorylated AMPA receptor GluA1 subunit in the hippocampus was significantly increased in the non-tested, defeated group. Ketamine increased the level of total protein, but not the ratio of phosphorylated GluA1 in the hippocampus of the non-tested, defeated group. In conclusion, exposure to social defeat stress as juveniles may induce the expression of impairment of social behaviors in adolescents via functional changes in GluA1. Activators of AMPA receptor signaling, such as ketamine, may constitute a novel treatment strategy for stress-related psychiatric disorders in adolescents with adverse juvenile experiences.


Ketamine/pharmacology , Receptors, AMPA/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior , Age Factors , Animals , Hippocampus/metabolism , Ketamine/antagonists & inhibitors , Male , Mice , Piperidines/pharmacology , Prefrontal Cortex/metabolism , Quinoxalines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Stress, Psychological/psychology
10.
Psychopharmacology (Berl) ; 235(7): 1875-1886, 2018 07.
Article En | MEDLINE | ID: mdl-29564482

BACKGROUND: Aberrant glutamate neurotransmission, and in particular dysfunction of the N-methyl-D-aspartate receptor (NMDAR), has been implicated in psychiatric disorders and represents a novel therapeutic target. Low-dose administration of the NMDA antagonist ketamine in healthy volunteers elicits a strong blood oxygenation level dependent (BOLD) imaging signal that can be attenuated by pretreatment with single, therapeutically effective doses of marketed medicines interacting with the glutamate system. OBJECTIVE: To test the attenuation of the ketamine-induced BOLD signal by pretreatment with either a metabotropic glutamate receptor (mGluR) 2/3 or a mGluR2 agonist in healthy volunteers METHODS: We used a ketamine challenge pharmacological magnetic resonance imaging (phMRI) paradigm to assess the modulatory effects of single acute doses of LY2140023 (pomaglumetad methionil), the methionine prodrug of the mGluR2/3 agonist LY404039 (10, 40, and 160 mg; N = 16 subjects) and of LY2979165, and the alanine prodrug of the selective orthosteric mGluR2 agonist 2812223 (20 and 60 mg; N = 16 subjects). RESULTS: A reduction in the ketamine-evoked BOLD phMRI signal relative to placebo was observed at the highest doses tested of both LY2140023 and LY2979165. A relationship was observed between reduction of the BOLD signal and increasing plasma levels of 2812223 in the LY2979165 cohort. CONCLUSIONS: These results identify pharmacologically active doses of the group II mGluR agonist prodrugs LY2140023 and LY2979165 in humans. They also extend the classes of compounds that have been experimentally shown to reverse the ketamine-evoked phMRI signal in humans, further supporting the use of this method as a neuroimaging biomarker for assessing functional effects.


Amino Acids/administration & dosage , Excitatory Amino Acid Agonists/administration & dosage , Ketamine/administration & dosage , Magnetic Resonance Imaging/methods , Prodrugs/administration & dosage , Receptors, Metabotropic Glutamate/agonists , Administration, Oral , Adult , Cohort Studies , Cross-Over Studies , Double-Blind Method , Healthy Volunteers , Humans , Infusions, Intravenous , Ketamine/antagonists & inhibitors , Male , Middle Aged , Treatment Outcome , Young Adult
11.
Bosn J Basic Med Sci ; 18(4): 320-327, 2018 Nov 07.
Article En | MEDLINE | ID: mdl-29579407

Ketamine is a widely used anesthetic in pediatric clinical practice. Previous studies have demonstrated that ketamine induces neurotoxicity and has a modulatory effect on the cells of the immune system. Here, we evaluated the potential protective effect and underlying mechanisms of natural phenolic compound curcumin against ketamine-induced toxicity in rat thymocytes. Rat thymocytes were exposed to 100 µM ketamine alone or combined with increasing concentrations of curcumin (0.3, 1, and 3 µM) for 24 hours. Cell viability was analyzed with CCK-8 assay kit. Apoptosis was analyzed using flow cytometry and propidium iodide as well as Z-VAD-FMK and Z-LEHD-FMK inhibitors. Reactive oxygen species (ROS) production and mitochondrial membrane potential [MMP] were measured by flow cytometry. Colorimetric assay with DEVD-pNA substrate was used for assessing caspase-3 activity. Involvement of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway was tested with Wortmannin inhibitor. Ketamine induced toxicity in cells, increased the number of hypodiploid cells, caspase-3 activity and ROS production, and inhibited the MMP. Co-incubation of higher concentrations of curcumin (1 and 3 µM) with ketamine markedly decreased cytotoxicity, apoptosis rate, caspase-3 activity, and ROS production in rat thymocytes, and increased the MMP. Application of Z-VAD-FMK (a pan caspase inhibitor) or Z-LEHD-FMK (caspase-9 inhibitor) with ketamine effectively attenuated the ketamine-induced apoptosis in rat thymocytes. Administration of Wortmannin (a PI3K inhibitor) with curcumin and ketamine significantly decreased the protective effect of curcumin on rat thymocytes. Our results indicate that ketamine-induced toxicity in rat thymocytes mainly occurs through the mitochondria-mediated apoptotic pathway and that the PI3K/Akt signaling pathway is involved in the anti-apoptotic effect of curcumin.


Curcumin/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Ketamine/antagonists & inhibitors , Ketamine/toxicity , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Reactive Oxygen Species/metabolism , Thymocytes/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Membrane Potential, Mitochondrial/drug effects , Oligopeptides/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects
12.
Drug Res (Stuttg) ; 68(3): 159-167, 2018 03.
Article En | MEDLINE | ID: mdl-28962045

OBJECTIVES: Morin is a naturally occurring flavonoid with strong anti-oxidant and anti-inflammatory properties. Studies have shown that flavones modulate neurotransmission through enhancement of gamma amino butyric acid activity in the central nervous system; which led to the hypothesis that they could exert tranquilizing effects in rodents. Hence, this study was designed to evaluate the antipsychotic effect of morin on experimental animal models. METHODS: The antipsychotic effect of morin (25, 50 and 100 mg/kg) administered intraperitoneally (i.p.) was assessed on novelty-induced locomotion, apomorphine-induced stereotypy, ketamine-induced stereotypy, ketamine-induced hyperlocomotion and ketamine-enhanced immobility in forced swim test (FST). Catalepsy and rota rod tests were also carried out to evaluate the extrapyramidal side effects of morin. RESULTS: Morin (25, 50 and 100 mg/kg, i.p.) pretreatments significantly (p<0.05) demonstrated anti-schizophrenia-like behavior by inhibiting ketamine-induced hyperlocomotion in mice. Moreover, morin (50 and 100 mg/kg, i.p.) significantly (p<0.05) reduced spontaneous locomotor activity. Also, morin suppressed apomorphine-induced stereotypy and ketamine-induced stereotypy. The increase in immobility in FST due to ketamine administration was reduced by morin in a significant dose-dependent manner. Furthermore, the antipsychotic activity of morin was not associated with extrapyramidal side effects, as evidenced by decreased decent latency and increased motoric coordination and performance in mice. CONCLUSION: The results of the study revealed that morin demonstrated antipsychotic-like property devoid of extrapyramidal side effects in experimental animal models and may be beneficial in the treatment of schizophrenia-like behaviors; particularly in patients with behavioral hyperactivity and negative symptoms.


Antipsychotic Agents/pharmacology , Catalepsy/prevention & control , Flavonoids/pharmacology , Immobility Response, Tonic/drug effects , Stereotyped Behavior/drug effects , Animals , Apomorphine/antagonists & inhibitors , Apomorphine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Locomotion/drug effects , Male , Mice , Rotarod Performance Test
13.
Neurotoxicol Teratol ; 69: 63-72, 2018.
Article En | MEDLINE | ID: mdl-29225006

Ketamine, a phencyclidine derivative, is an antagonist of the Ca2+-permeable N-methyl-d-aspartate (NMDA)-type glutamate receptors. It is a pediatric anesthetic and has been implicated in developmental neurotoxicity. Ketamine has also been shown to deplete ATP in mammalian cells. Our previous studies showed that acetyl l-carnitine (ALCAR) prevented ketamine-induced cardiotoxicity and neurotoxicity in zebrafish embryos. Based on our finding that ALCAR's protective effect was blunted by oligomycin A, an inhibitor of ATP synthase, we further investigated the effects of ketamine and ALCAR on ATP levels, mitochondria and ATP synthase in zebrafish embryos. The results demonstrated that ketamine reduced ATP levels in the embryos but not in the presence of ALCAR. Ketamine reduced total mitochondrial protein levels and mitochondrial potential, which were prevented with ALCAR co-treatment. To determine the cause of ketamine-induced ATP deficiency, we explored the status of ATP synthase. The results showed that a subunit of ATP synthase, atp5α1, was transcriptionally down-regulated by ketamine, but not in the presence of ALCAR, although ketamine caused a significant upregulation in another ATP synthase subunit, atp5ß and total ATP synthase protein levels. Most of the ATP generated by heart mitochondria are utilized for its contraction and relaxation. Ketamine-treated embryos showed abnormal heart structure, which was abolished with ALCAR co-treatment. This study offers evidence for a potential mechanism by which ketamine could cause ATP deficiency mediated by mitochondrial dysfunction.


Ketamine/adverse effects , Mitochondria/metabolism , Zebrafish , Acetylcarnitine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Embryo, Nonmammalian/drug effects , Ketamine/antagonists & inhibitors , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/metabolism , Proteins/metabolism
14.
J Basic Clin Physiol Pharmacol ; 28(6): 601-608, 2017 Nov 27.
Article En | MEDLINE | ID: mdl-28902622

BACKGROUND: Neuropathic pain is a chronic pain condition, which is resistant to therapy. Ascorbate was released because of the activation of glutaminergic neurons. Due to the important role of N-methyl-D-aspartate (NMDA) receptors in the pathophysiology of neuropathic pain, this study investigated the analgesic efficacy of ascorbic acid (AA) in neuropathic pain condition and the role of NMDA receptors in this effect. METHODS: For this purpose, adult male rats were randomly allocated to experimental groups (n=8 in each group). Neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve. During the second week after CCI, animals received a single injection of 1, 3, 5, or 10 mg/kg of AA intraperitoneally and pain threshold was determined 15 and 60 min later. The antinociceptive effect of chronic administration was also evaluated by intraperitoneal injection (IP) of 3 mg/kg AA for 3 weeks. To determine the role of NMDA receptors, separate groups of animals 30 min after single injection of AA (1 mg/kg) animals received i.p. injection of ketamine (5 mg/kg), MK-801 (0.01 mg/kg), or glutamate (1000 nmol) and were tested 20 min afterwards. Data analyzed by ANOVA and Newman-Keuls tests and p<0.05 were considered as significant. RESULTS: IP of 3, 5 and 10 mg/kg increased the pain threshold during the second week after CCI (p<0.05, F=3 in tactile allodynia and p<0.01, F=3.2 in thermal and mechanical hyperalgeisa). Chronic administration of AA also produced antinociceptive effect. Ascorbic acid (1 mg/kg, i.p.) inhibited MK-801 and ketamine-induced antinociception response significantly (p<0.001, F=2). It also prevented the analgesic effect of glutamate administration (p<0.001, F=2). CONCLUSIONS: The results indicated that AA produced a dose-dependent antinociceptive effect that seems to mediate through its interaction with NMDA receptors.


Analgesics/pharmacology , Ascorbic Acid/pharmacology , Neuralgia/drug therapy , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sciatic Nerve/injuries , Analgesics/therapeutic use , Animals , Ascorbic Acid/therapeutic use , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/pharmacology , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Ligation/adverse effects , Male , Neuralgia/complications , Pain Threshold/drug effects , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/drug therapy , Rats , Sciatic Nerve/surgery
15.
Biochem Biophys Res Commun ; 489(4): 420-425, 2017 08 05.
Article En | MEDLINE | ID: mdl-28577999

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.


Brain-Derived Neurotrophic Factor/genetics , Hippocampus/cytology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Ketamine/pharmacology , Neurons/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Ketamine/antagonists & inhibitors , Neurons/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Rats , Structure-Activity Relationship
16.
Metab Brain Dis ; 32(5): 1475-1489, 2017 10.
Article En | MEDLINE | ID: mdl-28508340

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.


Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Haloperidol/pharmacology , Ketamine/antagonists & inhibitors , Sertraline/pharmacology , Animals , Anxiety/psychology , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Grooming/drug effects , Interpersonal Relations , Ketamine/toxicity , Male , Memory Disorders/chemically induced , Memory Disorders/psychology , Memory, Short-Term/drug effects , Mice , Motor Activity/drug effects , Olanzapine
17.
PLoS One ; 12(5): e0177036, 2017.
Article En | MEDLINE | ID: mdl-28472198

Single sub-anesthetic doses of ketamine can exacerbate the symptoms of patients diagnosed with schizophrenia, yet similar ketamine treatments rapidly reduce depressive symptoms in major depression. Acute doses of the atypical antipsychotic drug clozapine have also been shown to counteract ketamine-induced psychotic effects. In the interest of understanding whether these drug effects could be modeled with alterations in neuroplasticity, we examined the impact of acutely-administered ketamine and clozapine on in vivo long-term potentiation (LTP) in the rat's hippocampus-to-prefrontal cortex (H-PFC) pathway. We found that a low dose of ketamine depressed H-PFC LTP, whereas animals that were co-administrated the two drugs displayed LTP that was similar to a saline-treated control. To address which signaling molecules might mediate such effects, we also examined phosphorylation and total protein levels of GSK3ß, GluA1, TrkB, ERK, and mTOR in prefrontal and hippocampal sub-regions. Among the statistically significant effects that were detected (a) both ketamine and clozapine increased the phosphorylation of Ser9-GSK3ß throughout the prefrontal cortex and of Ser2481-mTOR in the dorsal hippocampus (DH), (b) clozapine increased the phosphorylation of Ser831-GluA1 throughout the prefrontal cortex and of Ser845-GluA1 in the ventral hippocampus, (c) ketamine treatment increased the phosphorylation of Thr202/Tyr204-ERK in the medial PFC (mPFC), and (d) clozapine treatment was associated with decreases in the phosphorylation of Tyr705-TrkB in the DH and of Try816-TrkB in the mPFC. Further analyses involving phosphorylation effect sizes also suggested Ser831-GluA1 in the PFC displayed the highest degree of clozapine-responsivity relative to ketamine. These results provide evidence for how ketamine and clozapine treatments affect neuroplasticity and signaling pathways in the stress-sensitive H-PFC network. They also demonstrate the potential relevance of H-PFC pathway neuroplasticity for modeling ketamine-clozapine interactions in regards to psychosis.


Clozapine/pharmacology , Hippocampus/drug effects , Ketamine/antagonists & inhibitors , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Signal Transduction/drug effects , Animals , Blotting, Western , Hippocampus/metabolism , Hippocampus/physiology , Ketamine/pharmacology , Male , Nerve Tissue Proteins/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley
18.
Neurotoxicol Teratol ; 60: 82-86, 2017.
Article En | MEDLINE | ID: mdl-27826117

Prolonged ketamine exposure in neonates at anesthetic doses is known to cause long-term impairments of learning and memory. A current theoretical mechanism explains this phenomenon as being neuro-excitotoxicity mediated by compensatory upregulation of N-methyl-d-aspartate receptors (NMDARs), which then initiates widespread neuroapoptosis. Additionally, the excitatory behavior of GABAergic synaptic transmission mediated by GABAA receptors (GABAARs), occurring during the early neuronal development period, is proposed as contributing to the susceptibility of neonatal neurons to ketamine-induced injury. This is due to differential developmental expression patterns of Na+-K+-2Cl- co-transporter (NKCC1) and K+-Cl- co-transporter. Studies have shown that bumetanide, an NKCC1 inhibitor, allows neurons to become inhibitory rather than excitatory early in development. We thus hypothesized that bumetanide co-administration during ketamine treatment would reduce over excitation and protect the neurons from excitotoxicity. In this initial study, the Morris Water Maze test was used to assess the effects of co-administration of ketamine and bumetanide to neonatal Sprague-Dawley rats on long-term learning and memory changes seen later in life. It was revealed that bumetanide, when co-treated with ketamine neonatally, significantly impeded behavioral deficits typically seen in animals exposed to ketamine alone. Therefore, these findings suggest a new mechanism by which neonatal ketamine induced learning impairments can be prevented.


Bumetanide/pharmacology , Ketamine/adverse effects , Ketamine/antagonists & inhibitors , Memory Disorders/prevention & control , Solute Carrier Family 12, Member 2/drug effects , Spatial Learning/drug effects , Animals , Animals, Newborn , Drug Combinations , Female , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Rats
19.
Anesthesiology ; 125(2): 333-45, 2016 08.
Article En | MEDLINE | ID: mdl-27341276

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.


Anesthesia, General/methods , Heterocyclic Compounds, 4 or More Rings/pharmacology , Sulfonic Acids/pharmacology , Anesthetics, Dissociative/toxicity , Anesthetics, Intravenous/toxicity , Animals , Blood Pressure/drug effects , Cell Survival/drug effects , Electroencephalography/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Etomidate/antagonists & inhibitors , Etomidate/toxicity , Ketamine/antagonists & inhibitors , Ketamine/toxicity , Male , Mutagens/toxicity , Necrosis/prevention & control , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects
20.
Psychopharmacology (Berl) ; 233(11): 2077-2097, 2016 06.
Article En | MEDLINE | ID: mdl-27004790

OBJECTIVE: The aim of our study was to test whether ketamine produces an antidepressant effect in animal model of olfactory bulbectomy and assess the role of mammalian target of rapamycin (mTOR) pathway in ketamine's antidepressant effect. METHODS: Bulbectomized (OBX) rats and sham controls were assigned to four subgroups according to the treatment they received (ketamine, saline, ketamine + rapamycin, and saline + rapamycin). The animals were subjected to open field (OF), elevated plus maze (EPM), passive avoidance (PA), Morris water maze (MWM), and Carousel maze (CM) tests. Blood samples were collected before and after drug administration for analysis of phosphorylated mTOR level. After behavioral testing, brains were removed for evaluation of brain-derived neurotrophic factor (BDNF) in prefrontal cortex (PFC) and hippocampus. RESULTS: Ketamine normalized hyperactivity of OBX animals in EPM and increased the time spent in open arms. Rapamycin pretreatment resulted in elimination of ketamine effect in EPM test. In CM test, ketamine + rapamycin administration led to cognitive impairment not observed in saline-, ketamine-, or saline + rapamycin-treated OBX rats. Prefrontal BDNF content was significantly decreased, and level of mTOR was significantly elevated in OBX groups. CONCLUSIONS: OBX animals significantly differed from sham controls in most of the tests used. Treatment had more profound effect on OBX phenotype than controls. Pretreatment with rapamycin eliminated the anxiolytic and antidepressant effects of ketamine in task-dependent manner. The results indicate that ketamine + rapamycin application resulted in impaired stress responses manifested by cognitive deficits in active place avoidance (CM) test. Intensity of stressor (mild vs. severe) used in the behavioral tests had opposite effect on controls and on OBX animals.


Antidepressive Agents/pharmacology , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Sirolimus/pharmacology , Animals , Anxiety/psychology , Avoidance Learning/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Olfactory Bulb/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
...