Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
1.
Biomolecules ; 9(11)2019 11 18.
Article in English | MEDLINE | ID: mdl-31752219

ABSTRACT

N-methyl-d-aspartate/glutamate receptor (NMDAR) is one of the major voltage-sensitive ligand-gated cation channel. Several noncompetitive NMDAR antagonists contribute to pathophysiology of schizophrenia and mood disorders; however, the effects of inhibition of NMDAR on several transmitter system have not been well clarified. Thus, this study determined the selective NMDAR antagonist, MK801 (dizocilpine), on thalamocortical, mesothalamic, and mesocortical transmissions associated with l-glutamate, GABA, serotonin, norepinephrine, and dopamine using multiprobe microdialysis. Perfusion with MK801 into the medial prefrontal cortex (mPFC) increased and decreased respective regional releases of monoamine and GABA without affecting l-glutamate. The mPFC MK801-induced monoamine release is generated by the regional GABAergic disinhibition. Perfusion with MK801 into the reticular thalamic nucleus (RTN) decreased GABA release in the mediodorsal thalamic nucleus (MDTN) but increased releases of l-glutamate and catecholamine without affecting serotonin in the mPFC. The RTN MK801-induced l-glutamate release in the mPFC was generated by GABAergic disinhibition in the MDTN, but RTN MK801-induced catecholamine release in the mPFC was generated by activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate/glutamate receptor (AMPAR) which received l-glutamate release from thalamocortical glutamatergic terminals in the mPFC. Perfusion with MK801 into the dorsal raphe nucleus (DRN) decreased GABA release in the DRN but selectively increased serotonin release in the MDTN and mPFC. These DRN MK801-induced serotonin releases in the both mPFC and MDTN were also generated by GABAergic disinhibition in the DRN. These results indicate that the GABAergic disinhibition induced by NMDAR inhibition plays important roles in the MK801-induced releases of l-glutamate and monoamine in thalamic nuclei and cortex.


Subject(s)
Dizocilpine Maleate , Mediodorsal Thalamic Nucleus/metabolism , Prefrontal Cortex/metabolism , Synaptic Transmission/drug effects , Animals , Dizocilpine Maleate/pharmacokinetics , Dizocilpine Maleate/pharmacology , Male , Rats , Rats, Sprague-Dawley
2.
J Affect Disord ; 245: 325-334, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30419533

ABSTRACT

BACKGROUND: The discovery of the zinc-sensing receptor, has provided new possibilities for explaining the neurobiology of zinc. Recent studies indicate that the GPR39 zinc receptor may play an important role in the pathogenesis of depression as well as in the antidepressant mechanism of action. METHODS: In this study we evaluated the time-course of the antidepressant response of the GPR39 agonist (TC-G 1008), imipramine, ZnCl2 and MK-801 in the forced swim test in mice 30 min, 3 h, 6 h and 24 h after acute drug administration as well as after 14-day treatment. Zinc level was measured in serum of mice. BDNF protein level was evaluated in hippocampus following both acute and chronic TC-G 1008 treatment. RESULTS: A single administration of the GPR39 agonist caused an antidepressant-like effect lasting up to 24 h following the injection, which is longer than the effect of imipramine, ZnCl2 and MK-801. Chronic treatment with these compounds caused a decrease in immobility time in the FST. Serum zinc concentrations showed an increased level following chronic ZnCl2 administration, but not following administration of TC-G 1008, imipramine or MK-801. We also observed some tendencies for increased BDNF following acute TC-G 1008 treatment. LIMITATIONS: TC-G 1008 is new drug designed to study GPR39 therefore additional pharmacodynamic and pharmacokinetic properties in preclinical studies are required. CONCLUSION: This study shows for the first time the long-lasting antidepressant effect of the GPR39 agonist in comparison with imipramine, ZnCl2 and MK-801. Our findings suggest that GPR39 should be considered as a target in efforts to develop new antidepressant drugs.


Subject(s)
Antidepressive Agents/pharmacokinetics , Depression/drug therapy , Pyrimidines/pharmacokinetics , Receptors, G-Protein-Coupled/agonists , Sulfonamides/pharmacokinetics , Animals , Antidepressive Agents/administration & dosage , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Chlorides/pharmacokinetics , Dizocilpine Maleate/pharmacokinetics , Hippocampus/metabolism , Imipramine/pharmacology , Male , Mice , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Swimming , Time Factors , Zinc/blood , Zinc Compounds/pharmacokinetics
3.
Neuropharmacology ; 143: 122-129, 2018 12.
Article in English | MEDLINE | ID: mdl-30227149

ABSTRACT

NMDA receptors (NMDARs) are essential components in glutamatergic synaptic signaling. The NMDAR antagonist MK-801 has been a valuable pharmacological tool in evaluating NMDAR function because it binds with high affinity to the NMDAR ion channel pore and is non-competitive with ligand binding. MK-801 has also been used to selectively inhibit NMDAR current in only the cell being recorded by including the drug in the intracellular recording solution. Here, we report that intracellular MK-801 (iMK-801) only partially inhibits synaptic NMDAR currents at +40 mV at both cortical layer 4 to layer 2/3 and hippocampal Schaffer collateral to CA1 synapses. Furthermore, iMK-801 incompletely inhibits heterologously expressed NMDAR currents at -60 mV, consistent with a model of iMK-801 having a very slow binding rate and consequently ∼30,000 times lower affinity than MK-801 applied to the extracellular side of the receptor. While iMK-801 can be used as a qualitative tool to study reduced postsynaptic NMDAR function, it cannot be assumed to completely block NMDARs at concentrations typically used in experiments.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Transmission/drug effects , Animals , Binding Sites , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Extracellular Space/drug effects , Extracellular Space/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/metabolism , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/physiology , Tissue Culture Techniques
4.
Neuropharmacology ; 112(Pt A): 144-149, 2017 01.
Article in English | MEDLINE | ID: mdl-27520396

ABSTRACT

To avoid legislation based on chemical structure, research chemicals, frequently used for recreational purposes, are continually being synthesized. N-Ethyl-1,2-diphenylethanamine (ephenidine) is a diarylethylamine that has recently become popular with recreational users searching for dissociative hallucinogenic effects. In the present study, the pharmacological basis of its neural actions has been investigated, initially by assessing its profile in central nervous system receptor binding assays and subsequently in targeted electrophysiological studies. Ephenidine was a potent inhibitor of 3H-MK-801 binding (Ki: 66 nM), implying that it acts at the PCP site of the N-methyl-d-aspartate (NMDA) receptor. It also showed modest activity at dopamine (379 nM) and noradrenaline (841 nM) transporters and at sigma 1 (629 nM) and sigma 2 (722 nM) binding sites. In experiments of extracellular recording of field excitatory postsynaptic potentials (fEPSPs) from area CA1 of rat hippocampal slices, ephenidine, 1 and 10 µM, respectively, produced a 25% and a near maximal inhibition of the NMDA receptor mediated fEPSP after 4 h superfusion. By contrast, ephenidine (50 µM) did not affect the AMPA receptor mediated fEPSPs. In whole cell patch clamp recordings, from hippocampal pyramidal cells, ephenidine (10 µM) blocked NMDA receptor-mediated EPSCs in a highly voltage-dependent manner. Additionally, ephenidine, 10 µM, blocked the induction of long term potentiation (LTP) in CA1 induced by theta burst stimulation. The present data show that the new psychoactive substance, ephenidine, is a selective NMDA receptor antagonist with a voltage-dependent profile similar to ketamine. Such properties help explain the dissociative, cognitive and hallucinogenic effects in man. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Subject(s)
Ethanolamines/pharmacology , Hippocampus/drug effects , Ketamine/pharmacology , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain/drug effects , Brain/metabolism , Dizocilpine Maleate/pharmacokinetics , Ethanolamines/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/physiology , Ketamine/pharmacokinetics , Long-Term Potentiation/drug effects , Male , Psychotropic Drugs/pharmacokinetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
5.
Neuropsychopharmacology ; 41(9): 2232-40, 2016 08.
Article in English | MEDLINE | ID: mdl-26837462

ABSTRACT

Schizophrenia patients exhibit dysfunctional gamma oscillations in response to simple auditory stimuli or more complex cognitive tasks, a phenomenon explained by reduced NMDA transmission within inhibitory/excitatory cortical networks. Indeed, a simple steady-state auditory click stimulation paradigm at gamma frequency (~40 Hz) has been reproducibly shown to reduce entrainment as measured by electroencephalography (EEG) in patients. However, some investigators have reported increased phase locking factor (PLF) and power in response to 40 Hz auditory stimulus in patients. Interestingly, preclinical literature also reflects this contradiction. We investigated whether a graded deficiency in NMDA transmission can account for such disparate findings by administering subanesthetic ketamine (1-30 mg/kg, i.v.) or vehicle to conscious rats (n=12) and testing their EEG entrainment to 40 Hz click stimuli at various time points (~7-62 min after treatment). In separate cohorts, we examined in vivo NMDA channel occupancy and tissue exposure to contextualize ketamine effects. We report a robust inverse relationship between PLF and NMDA occupancy 7 min after dosing. Moreover, ketamine could produce inhibition or disinhibition of the 40 Hz response in a temporally dynamic manner. These results provide for the first time empirical data to understand how cortical NMDA transmission deficit may lead to opposite modulation of the auditory steady-state response (ASSR). Importantly, our findings posit that 40 Hz ASSR is a pharmacodynamic biomarker for cortical NMDA function that is also robustly translatable. Besides schizophrenia, such a functional biomarker may be of value to neuropsychiatric disorders like bipolar and autism spectrum where 40 Hz ASSR deficits have been documented.


Subject(s)
Cerebral Cortex/physiology , Evoked Potentials, Auditory , Gamma Rhythm , Receptors, N-Methyl-D-Aspartate/physiology , Acoustic Stimulation , Animals , Biomarkers , Cerebral Cortex/drug effects , Dizocilpine Maleate/pharmacokinetics , Electroencephalography , Excitatory Amino Acid Antagonists/administration & dosage , Ketamine/administration & dosage , Male , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Processing, Computer-Assisted
6.
Behav Brain Res ; 304: 51-9, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26876136

ABSTRACT

Repeated social defeat of adolescent male rats results in adult mesocortical dopamine hypofunction, impaired working memory, and increased contextual anxiety-like behavior. Given the role of glutamate in dopamine regulation, cognition, and fear and anxiety, we investigated potential changes to N-methyl-D-aspartic acid (NMDA) receptors following adolescent social defeat. As both NMDA receptors and mesocortical dopamine are implicated in the expression and extinction of conditioned fear, a separate cohort of rats was challenged with a classical fear conditioning paradigm to investigate whether fear learning is altered by adolescent defeat. Quantitative autoradiography was used to measure 3H-MK-801 binding to NMDA receptors in regions of the medial prefrontal cortex, caudate putamen, nucleus accumbens, amygdala and hippocampus. Assessment of fear learning was achieved using an auditory fear conditioning paradigm, with freezing toward the auditory tone used as a measure of conditioned fear. Compared to controls, adolescent social defeat decreased adult NMDA receptor expression in the infralimbic region of the prefrontal cortex and central amygdala, while increasing expression in the CA3 region of the hippocampus. Previously defeated rats also displayed decreased conditioned freezing during the recall and first extinction periods, which may be related to the observed decreases and increases in NMDA receptors within the central amygdala and CA3, respectively. The alteration in NMDA receptors seen following adolescent social defeat suggests that dysfunction of glutamatergic systems, combined with mesocortical dopamine deficits, likely plays a role in the some of the long-term behavioral consequences of social stressors in adolescence seen in both preclinical and clinical studies.


Subject(s)
CA3 Region, Hippocampal/metabolism , Fear/psychology , Learning Disabilities/etiology , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior , Stress, Psychological , Analysis of Variance , Animals , CA3 Region, Hippocampal/drug effects , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , Protein Binding/drug effects , Protein Binding/physiology , Rats , Stress, Psychological/complications , Stress, Psychological/etiology , Stress, Psychological/pathology , Tritium/pharmacokinetics
7.
Eur J Pharmacol ; 766: 1-8, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26325093

ABSTRACT

N-methyl-D-aspartate (NMDA) receptor antagonists, including open channel blockers and GluN2B receptor subtype selective antagonists, have been developed for the treatment of depression. The current study investigated effects of systemically administered NMDA channel blockers and GluN2B receptor antagonists on NMDA receptor activity in rodents using in vivo [(3)H]MK-801 binding. The receptor occupancy of GluN2B antagonists was measured using ex vivo [(3)H]Ro 25-6981 binding. Ketamine, a NMDA receptor channel blocker, produced a dose/exposure- and time-dependent inhibition of in vivo [(3)H]MK-801 binding that was maximal at ~100%. The complete inhibition of in vivo [(3)H]MK-801 binding was also observed with NMDA receptor channel blockers, AZD6765 (Lanicemine) and MK-801 (Dizocilpine). CP-101,606 (Traxoprodil), a GluN2B antagonist, produced a dose/exposure- and time-dependent inhibition of in vivo [(3)H]MK-801 binding that was maximal at ~60%. Partial inhibition was also observed with other GluN2B antagonists including MK-0657 (CERC-301), EVT-101, Ro 25-6981 and radiprodil. For all GluN2B antagonists tested, partial [(3)H]MK-801 binding inhibition was achieved at doses saturating GluN2B receptor occupancy. Combined treatment with ketamine (10mg/kg, i.p.) and Ro 25-6981(10mg/kg, i.p.) produced a level of inhibition of in vivo [(3)H]MK-801 binding that was similar to treatment with either agent alone. In conclusion, this in vivo [(3)H]MK-801 binding study shows that NMDA receptor activity in the rodent forebrain can be inhibited completely by channel blockers, but only partially (~60%) by GluN2B receptor antagonists. At doses effective in preclinical models of depression, ketamine may preferentially inhibit the same population of NMDA receptors as Ro 25-6981, namely those containing the GluN2B subunit.


Subject(s)
Dizocilpine Maleate/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Brain/metabolism , Excitatory Amino Acid Antagonists/blood , Excitatory Amino Acid Antagonists/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Male , Mice , Radioligand Assay , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Article in English | MEDLINE | ID: mdl-25584778

ABSTRACT

High fat (HF) diets are known to induce changes in synaptic plasticity in the forebrain leading to learning and memory impairments. Previous studies of oleanolic acid derivatives have found that these compounds can cross the blood-brain barrier to prevent neuronal cell death. We examined the hypothesis that the oleanolic acid derivative, bardoxolone methyl (BM) would prevent diet-induced cognitive deficits in mice fed a HF diet. C57BL/6J male mice were fed a lab chow (LC) (5% of energy as fat), a HF (40% of energy as fat), or a HF diet supplemented with 10mg/kg/day BM orally for 21weeks. Recognition memory was assessed by performing a novel object recognition test on the treated mice. Downstream brain-derived neurotrophic factor (BDNF) signalling molecules were examined in the prefrontal cortex (PFC) and hippocampus of mice via Western blotting and N-methyl-d-aspartate (NMDA) receptor binding. BM treatment prevented HF diet-induced impairment in recognition memory (p<0.001). In HF diet fed mice, BM administration attenuated alterations in the NMDA receptor binding density in the PFC (p<0.05), however, no changes were seen in the hippocampus (p>0.05). In the PFC and hippocampus of the HF diet fed mice, BM administration improved downstream BDNF signalling as indicated by increased protein levels of BDNF, phosphorylated tropomyosin related kinase B (pTrkB) and phosphorylated protein kinase B (pAkt), and increased phosphorylated AMP-activated protein kinase (pAMPK) (p<0.05). BM administration also prevented the HF diet-induced increase in the protein levels of inflammatory molecules, phosphorylated c-Jun N-terminal kinase (pJNK) in the PFC, and protein tyrosine phosphatase 1B (PTP1B) in both the PFC and hippocampus. In summary, these findings suggest that BM prevents HF diet-induced impairments in recognition memory by improving downstream BDNF signal transduction, increasing pAMPK, and reducing inflammation in the PFC and hippocampus.


Subject(s)
Diet, High-Fat/adverse effects , Memory Disorders , Oleanolic Acid/analogs & derivatives , Prefrontal Cortex/drug effects , Recognition, Psychology/drug effects , Signal Transduction/drug effects , Animals , Autoradiography , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Drug Administration Schedule , Exploratory Behavior/drug effects , Male , Memory Disorders/etiology , Memory Disorders/pathology , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Oleanolic Acid/therapeutic use , Prefrontal Cortex/metabolism , Protein Binding/drug effects , Statistics, Nonparametric , Tritium/pharmacokinetics
9.
Neurobiol Dis ; 73: 60-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281318

ABSTRACT

Current concepts suggest that exposure to THC during adolescence may act as a risk factor for the development of psychiatric disorders later in life. However, the molecular underpinnings of this vulnerability are still poorly understood. To analyze this, we investigated whether and how THC exposure in female rats interferes with different maturational events occurring in the prefrontal cortex during adolescence through biochemical, pharmacological and electrophysiological means. We found that the endocannabinoid system undergoes maturational processes during adolescence and that THC exposure disrupts them, leading to impairment of both endocannabinoid signaling and endocannabinoid-mediated LTD in the adult prefrontal cortex. THC also altered the maturational fluctuations of NMDA subunits, leading to larger amounts of gluN2B at adulthood. Adult animals exposed to THC during adolescence also showed increased AMPA gluA1 with no changes in gluA2 subunits. Finally, adolescent THC exposure altered cognition at adulthood. All these effects seem to be triggered by the disruption of the physiological role played by the endocannabinoid system during adolescence. Indeed, blockade of CB1 receptors from early to late adolescence seems to prevent the occurrence of pruning at glutamatergic synapses. These results suggest that vulnerability of adolescent female rats to long-lasting THC adverse effects might partly reside in disruption of the pivotal role played by the endocannabinoid system in the prefrontal cortex maturation.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Developmental Disabilities/chemically induced , Dronabinol/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Age Factors , Animals , Cyclohexanols/pharmacokinetics , Dizocilpine Maleate/pharmacokinetics , Estradiol/blood , Estrous Cycle/drug effects , Excitatory Amino Acid Antagonists/pharmacokinetics , Female , In Vitro Techniques , Neurites/drug effects , Piperidines/pharmacology , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/ultrastructure , Pyrazoles/pharmacology , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism , Synaptic Potentials/drug effects , Tritium/pharmacokinetics
10.
Brain Dev ; 37(4): 376-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25034178

ABSTRACT

BACKGROUND AND PURPOSE: Most therapeutic agents are administered intravenously (IV) in clinical settings and intraperitoneally (IP) in preclinical studies with neonatal rodents; however, it remains unclear whether intraperitoneal (IP) injection is truly an acceptable alternative for intravenous (IV) injection in preclinical studies. The objective of our study is to clarify the differences in the therapeutic effects of drugs and in the distribution of infused cells after an IP or IV injection in animals with brain injury. METHODS: Dexamethasone or MK-801, an N-methyl-d-aspartate receptor antagonist was administered either IP or IV in a mouse model of neonatal hypoxic-ischemic encephalopathy. Green fluorescent protein-expressing mesenchymal stem cells (MSCs) or mononuclear cells (MNCs) were injected IP or IV in the mouse model. Two hours and 24h after the administration of the cells, we investigated the cell distributions by immunohistochemical staining. We also investigated distribution of IV administered MNCs labeled with 2-[18F]fluoro-2-deoxy-d-glucose in a juvenile primate, a macaque with stroke 1h after the administration. RESULTS: IP and IV administration of dexamethasone attenuated the brain injury to a similar degree. IP administration of MK-801 attenuated brain injury, whereas IV administration of MK-801 did not. The IV group showed a significantly greater number of infused cells in the lungs and brains in the MSC cohort and in the spleen, liver, and lung in the MNC cohort compared to the IP group. In the macaque, MNCs were detected in the spleen and liver in large amounts, but not in the brain and lungs. CONCLUSIONS: This study demonstrated that the administration route influences the effects of drugs and cell distribution. Therefore, a preclinical study may need to be performed using the optimal administration route used in a clinical setting.


Subject(s)
Dexamethasone/administration & dosage , Dizocilpine Maleate/administration & dosage , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/administration & dosage , Animals , Animals, Newborn , Bone Marrow Transplantation , Brain/drug effects , Brain/metabolism , Carotid Artery Diseases , Dexamethasone/pharmacokinetics , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacokinetics , Femoral Vein , Fluorodeoxyglucose F18 , Hypoxia-Ischemia, Brain/metabolism , Injections, Intraperitoneal , Injections, Intravenous , Leukocytes, Mononuclear/metabolism , Macaca , Male , Mice , Neuroprotective Agents/pharmacokinetics , Random Allocation , Rats, Inbred Lew , Rats, Transgenic , Treatment Outcome
11.
J Neurosci ; 34(34): 11426-38, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143622

ABSTRACT

Overstimulation of NMDA-type glutamate receptors is believed to be responsible for neuronal death of the CNS in various disorders, including cerebral and spinal cord ischemia. However, the intrinsic and physiological mechanisms of modulation of these receptors are essentially unknown. Here we report that cholestane-3ß,5α,6ß-triol (triol), a major metabolite of cholesterol, is an endogenous neuroprotectant and protects against neuronal injury both in vitro and in vivo via negative modulation of NMDA receptors. Treatment of cultured neurons with triol protects against glutamate-induced neurotoxicity, and administration of triol significantly decreases neuronal injury after spinal cord ischemia in rabbits and transient focal cerebral ischemia in rats. An inducible elevation of triol is associated with ischemic preconditioning and subsequent neuroprotection in the spinal cord of rabbits. This neuroprotection is effectively abolished by preadministration of a specific inhibitor of triol synthesis. Physiological concentrations of triol attenuate [Ca(2+)]i induced by glutamate and decrease inward NMDA-mediated currents in cultured cortical neurons and HEK-293 cells transiently transfected with NR1/NR2B NMDA receptors. Saturable binding of [(3)H]triol to cerebellar granule neurons and displacement of [(3)H]MK-801 binding to NMDA receptors by triol suggest that direct blockade of NMDA receptors may underlie the neuroprotective properties. Our findings suggest that the naturally occurring oxysterol, the major cholesterol metabolite triol, functions as an endogenous neuroprotectant in vivo, which may provide novel insights into understanding and developing potential therapeutics for disorders in the CNS.


Subject(s)
Brain Injuries/prevention & control , Cholestanols/therapeutic use , Neuroprotective Agents/therapeutic use , Spinal Cord Ischemia/prevention & control , Adult , Animals , Brain Injuries/etiology , Cells, Cultured , Central Nervous System/cytology , Cholestanols/blood , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Female , Glutamic Acid/pharmacology , Humans , Infarction, Middle Cerebral Artery/complications , Male , Neurons/drug effects , Neurons/physiology , Protein Binding/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution/drug effects , Tissue Distribution/physiology , Young Adult
12.
Neuropharmacology ; 86: 174-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25063581

ABSTRACT

Ketamine is used preclinically and clinically to study schizophrenia and depression. Accordingly, it is imperative to understand the temporal relationship between the central concentrations and N-methyl-d-aspartate receptor (NMDAR) interactions of both ketamine and norketamine, its primary active metabolite, across species to assess the translatability of animal models to humans and the back-translation of clinical observations to the preclinical realm. However, such an interspecies normalization of ketamine and norketamine exposures at different clinical and preclinical doses (and their different routes and regimens) is lacking. This work defines the NMDAR occupancy (RO) time course following single doses of ketamine in rats, nonhuman primates (nhp) and humans to allow direct interspecies comparisons of specific ketamine-mediated pharmacodynamics via RO normalization. Total plasma concentration (Cp)-time profiles of ketamine and norketamine were generated from rats and nhp following a single, memory-impairing dose of ketamine; neuropharmacokinetics were determined in rats. [(3)H]MK-801-displacement studies in rats determined estimated mean (95% confidence interval) unbound plasma concentrations (Cp,u) for ketamine and norketamine producing 50% RO (IC50) of 1420 (990, 2140) nM and 9110 (5870, 13700) nM, respectively. Together, these datasets transformed Cp,u-time data to predicted RO (ROpred)-time profiles for rats, nhp and humans at behaviorally relevant ketamine doses. Subsequently, this approach helped determine an infusion paradigm in rats producing a ROpred-time profile mirroring that for a clinically antidepressant infusion. The described indication-independent methodology allows normalization to RO at any time following any ketamine dose (regardless of route or regimen) in any species by simply quantifying the Cp of ketamine and norketamine. Matching temporal RO relationships in animals and humans should allow direct comparisons of specific ketamine-dependent NMDAR-based pharmacodynamics.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Animals , Depression/drug therapy , Depression/physiopathology , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Ketamine/analogs & derivatives , Ketamine/pharmacokinetics , Macaca fascicularis , Male , Memory/drug effects , Memory/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Species Specificity , Tritium
13.
J Neurosci Res ; 92(11): 1599-607, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24953757

ABSTRACT

Although both the onset of schizophrenia and human phencyclidine (PCP) abuse typically present within the interval from adolescence to early adulthood, the majority of preclinical research employing the PCP model of schizophrenia has been conducted on neonatal or adult animals. The present study was designed to evaluate the behavioral and neurochemical sequelae of subchronic exposure to PCP in adolescence. Male 35-42-day-old Sprague Dawley rats were subcutaneously administered either saline (10 ml · kg(-1) ) or PCP hydrochloride (10 mg · kg(-1) ) once daily for a period of 14 days (n = 6/group). The animals were allowed to withdraw from treatment for 2 weeks, and their social and exploratory behaviors were subsequently assessed in adulthood by using the social interaction test. To examine the effects of adolescent PCP administration on the regulation of N-methyl-D-aspartate receptors (NMDARs), quantitative autoradiography was performed on brain sections of adult, control and PCP-withdrawn rats by using 20 nM (3) H-MK-801. Prior subchronic exposure to PCP in adolescence had no enduring effects on the reciprocal contact and noncontact social behavior of adult rats. Spontaneous rearing in response to the novel testing arena and time spent investigating its walls and floor were reduced in PCP-withdrawn animals compared with control. The long-term behavioral effects of PCP occurred in the absence of persistent deficits in spontaneous locomotion or self-grooming activity and were not mediated by altered NMDAR density. Our results document differential effects of adolescent PCP administration on the social and exploratory behaviors of adult rats, suggesting that distinct neurobiological mechanisms are involved in mediating these behaviors.


Subject(s)
Behavioral Symptoms/chemically induced , Exploratory Behavior/drug effects , Hallucinogens/toxicity , Interpersonal Relations , Phencyclidine/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Age Factors , Animals , Autoradiography , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Male , Motor Activity/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Tritium/pharmacokinetics
14.
Neuropharmacology ; 81: 55-63, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24486380

ABSTRACT

Physical exercise has become a potentially beneficial therapy for reducing neurodegeneration symptoms in Alzheimer's disease. Previous studies have shown that cognitive deterioration, anxiety and the startle response observed in 7-month-old 3xTg-AD mice were ameliorated after 6 months of free access to a running wheel. Also, alterations in synaptic response to paired-pulse stimulation were improved. The present study further investigated some molecular mechanisms underlying the beneficial effects of 6 months of voluntary exercise on synaptic plasticity in 7-month-old 3xTg-AD mice. Changes in binding parameters of [(3)H]-flunitrazepam to GABAA receptor and of [(3)H]-MK-801 to NMDA receptor in cerebral cortex of 3xTgAD mice were restored by voluntary exercise. In addition, reduced expression levels of NMDA receptor NR2B subunit were reestablished. The synaptic proteins synaptophysin and PSD-95 and the neuroprotective proteins GDNF and SIRT1 were downregulated in 3xTgAD mice and were recovered by exercise treatment. Overall, in this paper we highlight the fact that different interrelated mechanisms are involved in the beneficial effects of exercise on synaptic plasticity alterations in the 3xTg-AD mouse model.


Subject(s)
Alzheimer Disease/rehabilitation , Apoptosis Regulatory Proteins/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Physical Conditioning, Animal/methods , Synapses/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cerebral Cortex/drug effects , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacokinetics , Flunitrazepam/pharmacokinetics , GABA Modulators/pharmacokinetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Presenilin-1/genetics , Protein Binding/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Tritium/pharmacokinetics
15.
Neuroscience ; 259: 84-93, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24295633

ABSTRACT

In this study, we investigated the effect of neonatal olfactory bulbectomy (nOBX) on behavioral paradigms related to olfaction such as exploratory behavior, locomotor activity in a novel environment and social interaction. We also studied the effect of nOBX on the activity of the N-methyl-d-aspartate (NMDA) subtype of glutamate receptors during development. The behavioral effects of nOBX (postnatal day 7, PD7) were investigated in pre- (PD30) and post-pubertal (PD60) Wistar rats. NMDA receptor activity was measured with [(125)I]MK-801 in the brain regions associated with the olfactory circuitry. A significant increase in the novelty-induced locomotion was seen in the pre-pubertal nOBX rats. Although the locomotor effect was less marked than in pre-pubertal rats, the nOBX rats tested post-pubertally failed to habituate to the novel situation as quickly as the sham- and normal- controls. Pre-pubertally, the head-dipping behavior was enhanced in nOBX rats compared with sham-operated and normal controls, while normal exploratory behavior was observed between groups in adulthood. In contrast, social interaction was increased in post-pubertal animals that underwent nOBX. Both pre- and post-pubertal nOBX rats recovered olfaction. Interestingly, pre-pubertal rats showed a significant increase in the [(125)I]MK-801 binding in the piriform cortex, dorsal hippocampus, inner and outer layers of the frontal cortex and outer layer of the cingulate cortex. At post-pubertal age, no significant differences in [(125)I]MK-801 binding were observed between groups at any of the brain regions analyzed. These results suggest that nOBX produces pre-pubertal behavioral disturbances and NMDA receptor changes that are transitory with recovery of olfaction early in adulthood.


Subject(s)
Exploratory Behavior/physiology , Motor Activity/physiology , Olfactory Bulb/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Smell/physiology , Age Factors , Animals , Animals, Newborn , Dizocilpine Maleate/pharmacokinetics , Female , Interpersonal Relations , Iodine Radioisotopes/pharmacokinetics , Male , Olfactory Bulb/growth & development , Olfactory Bulb/surgery , Olfactory Pathways , Pregnancy , Protein Binding/physiology , Radioligand Assay , Rats , Rats, Wistar , Reaction Time/physiology , Statistics, Nonparametric , Time Factors
16.
Auton Neurosci ; 171(1-2): 41-8, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23146621

ABSTRACT

The paraventricular nucleus (PVN) of the hypothalamus is an important region of the brain involved in the regulation of sympathetic vasomotor tone. Accumulating evidence supports the idea that a change in hypothalamic γ-aminobutyric acid (GABA)-ergic inhibitory and glutamatergic excitatory inputs contribute to the exacerbated sympathetic drive in chronic heart failure (HF). The purpose of this study was to determine whether a possible imbalance between glutamatergic and GABAergic inputs to the PVN contributes to increased sympathetic outflow in HF in two different sympathetic territories. Renal (RSNA) and splanchnic sympathetic nerve activity (SSNA), mean arterial blood pressure (MAP) and heart rate were recorded from urethane-anesthetized HF or sham rats. The NMDA-glutamate and GABA-A receptor densities within the PVN were quantified in HF and sham rats by autoradiography. Bilateral microinjection of kynurenic acid (4nmol) into the PVN decreased MAP and RSNA and SSNA in HF but not in sham rats. Furthermore, in response to GABA-A blockade in the PVN by bicuculline (400 pmol), hypertension and SSNA were reduced in HF compared to sham. The quantification of ionotropic NMDA receptors and GABA-A receptors in the PVN showed a significant reduction of GABA-A in HF rats; however, the NMDA density in the PVN did not differ between groups. Thus, this study provides evidence that the sympathoexcitation is maintained by an imbalance between GABAergic and glutamatergic inputs in the PVN in HF. The reduced GABAergic input results in relatively augmented glutamatergic actions in the PVN of HF rats.


Subject(s)
Blood Pressure/physiology , GABAergic Neurons/physiology , Heart Failure/physiopathology , Heart Rate/physiology , Paraventricular Hypothalamic Nucleus/physiopathology , Splanchnic Nerves/physiopathology , Animals , Autoradiography , Blood Pressure/drug effects , Disease Models, Animal , Dizocilpine Maleate/pharmacokinetics , Echocardiography , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Agonists/pharmacokinetics , GABAergic Neurons/drug effects , Heart Failure/pathology , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Kidney/innervation , Kynurenic Acid/pharmacology , Ligation/adverse effects , Male , Microinjections , Muscimol/pharmacokinetics , Paraventricular Hypothalamic Nucleus/drug effects , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacokinetics , Rats , Rats, Wistar , Splanchnic Nerves/drug effects , Terpenes/pharmacokinetics , Tritium/pharmacokinetics
17.
Neuroscience ; 215: 149-59, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22554779

ABSTRACT

The striatum is particularly vulnerable to mitochondrial dysfunction and this problem is linked to pathology created by environmental neurotoxins, stimulants like amphetamine, and metabolic disease and ischemia. We studied the course of recovery following a single systemic injection of the mitochondrial complex II inhibitor 3-nitropropionic acid (3-NP) and found 3-NP caused lasting changes in motor behavior that were associated with altered activity-dependent plasticity at corticostriatal synapses in Fischer 344 rats. The changes in synapse behavior varied with the time after exposure to the 3-NP injection. The earliest time point studied, 24h after 3-NP, revealed 3-NP-induced an exaggeration of D1 Dopamine (DA) receptor dependent long-term potentiation (LTP) that reversed to normal by 48 h post-3-NP exposure. Thereafter, the likelihood and degree of inducing D2 DA receptor dependent long-term depression (LTD) gradually increased, relative to saline controls, peaking at 1 month after the 3-NP exposure. NMDA receptor binding did not change over the same post 3-NP time points. These data indicate even brief exposure to 3-NP can have lasting behavioral effects mediated by changes in the way DA and glutamate synapses interact.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/cytology , Enzyme Inhibitors/pharmacology , Movement/drug effects , Nitro Compounds/pharmacology , Propionates/pharmacology , Synapses/drug effects , Adrenergic Agents/toxicity , Analysis of Variance , Animals , Benzazepines/pharmacology , Biophysics , Dizocilpine Maleate/pharmacokinetics , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacokinetics , Excitatory Postsynaptic Potentials/drug effects , Functional Laterality , In Vitro Techniques , Oxidopamine/toxicity , Protein Binding/drug effects , Rats , Rats, Inbred F344 , Substantia Nigra/injuries , Substantia Nigra/physiology , Sulpiride/pharmacology , Time Factors , Tritium/pharmacokinetics , Tyrosine 3-Monooxygenase/metabolism
18.
Neurobiol Learn Mem ; 97(4): 370-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22426465

ABSTRACT

A promising target for memory improvement is phosphodiesterase type 5 (PDE5), which selectively hydrolyzes cyclic guanosine monophosphate (cGMP). In rodents, PDE5 inhibitors (PDE5-Is) have been shown to improve memory performance in many behavioral paradigms. However, it is questioned whether the positive effects in animal studies result from PDE5 inhibition in the central nervous system or the periphery. Therefore, we studied the effects of PDE5 inhibition on memory and determined whether compound penetration of the blood-brain barrier (BBB) is required for this activity. Two selective PDE5-Is, vardenafil and UK-343,664, were tested in the object recognition task (ORT) in both a MK-801- and scopolamine-induced memory deficit model, and a time-delay model without pharmacological intervention. Compounds were dosed 30 min before the learning trial of the task. To determine if the PDE5-Is crossed the BBB, their concentrations were determined in plasma and brain tissue collected 30 min after oral administration. Vardenafil improved object recognition memory in all three variants of the ORT. UK-343,664 was ineffective at either preventing MK-801-induced memory disruption or time-dependent memory decay. However, UK-343,664 attenuated the memory impairment of scopolamine. Vardenafil crossed the BBB whereas UK-343,664 did not. Further, co-administration of UK-343,664 and scopolamine did not alter the brain partitioning of either molecule. This suggests that the positive effect of UK-343,664 on scopolamine-induced memory decay might arise from peripheral PDE5 inhibition. The results herein suggest that there may be multiple mechanisms that mediate the efficacy of PDE5 inhibition to improve memory performance in tasks such as the ORT and that these involve PDE5 located both within and outside of the brain. To further elucidate the underlying mechanisms, the cellular and subcellular localization of PDE5 needs to be determined.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Imidazoles/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Pyrimidinones/pharmacology , Recognition, Psychology/physiology , Animals , Blood-Brain Barrier , Cyclic Nucleotide Phosphodiesterases, Type 5/drug effects , Dizocilpine Maleate/pharmacokinetics , Dizocilpine Maleate/pharmacology , Imidazoles/pharmacokinetics , Male , Phosphodiesterase 5 Inhibitors/pharmacokinetics , Piperazines/pharmacokinetics , Pyrimidinones/pharmacokinetics , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Scopolamine/pharmacokinetics , Scopolamine/pharmacology , Sulfones/pharmacokinetics , Sulfones/pharmacology , Triazines/pharmacokinetics , Triazines/pharmacology , Vardenafil Dihydrochloride
19.
Neurosci Lett ; 503(1): 68-72, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21871531

ABSTRACT

The non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist (+)MK-801 is widely used in animal research (over 3000 publications), however its extracellular brain concentration has never been reported. Here, we show using in vivo microdialysis that systemic injection of (+)MK-801 at doses of 0.05, 0.1 or 0.2mg/kg resulted in peak brain ECF concentration of 6, 14 or 34 nM, respectively. Moreover, (+)MK-801 resulted in a dose-dependent learning impairment in the Morris water maze as well as hyperactivity in the open field. These data demonstrate for the first time that (+)MK-801 at doses producing behavioural alterations expected from NMDA receptor blockade reaches extracellular brain concentrations corresponding to the affinity at NMDA receptors.


Subject(s)
Behavior, Animal/drug effects , Dizocilpine Maleate/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacokinetics , Putamen/drug effects , Analysis of Variance , Animals , Area Under Curve , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Microdialysis/methods , Putamen/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization/methods , Statistics, Nonparametric
20.
PLoS One ; 6(6): e20210, 2011.
Article in English | MEDLINE | ID: mdl-21701689

ABSTRACT

The role of 5-HT7 receptor has been demonstrated in various animal models of mood disorders; however its function in cognition remains largely speculative. This study evaluates the effects of SB-269970, a selective 5-HT7 antagonist, in a translational model of working memory deficit and investigates whether it modulates cortical glutamate and/or dopamine neurotransmission in rats. The effect of SB-269970 was evaluated in the delayed non-matching to position task alone or in combination with MK-801, a non-competitive NMDA receptor antagonist, and, in separate experiments, with scopolamine, a non-selective muscarinic antagonist. SB-269970 (10 mg/kg) significantly reversed the deficits induced by MK-801 (0.1 mg/kg) but augmented the deficit induced by scopolamine (0.06 mg/kg). The ability of SB-269970 to modulate MK-801-induced glutamate and dopamine extracellular levels was separately evaluated using biosensor technology and microdialysis in the prefrontal cortex of freely moving rats. SB-269970 normalized MK-801 -induced glutamate but not dopamine extracellular levels in the prefrontal cortex. Rat plasma and brain concentrations of MK-801 were not affected by co-administration of SB-269970, arguing for a pharmacodynamic rather than a pharmacokinetic mechanism. These results indicate that 5-HT7 receptor antagonists might reverse cognitive deficits associated with NMDA receptor hypofunction by selectively normalizing glutamatergic neurotransmission.


Subject(s)
Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Memory Disorders/drug therapy , Memory, Short-Term/drug effects , Receptors, Serotonin/metabolism , Synaptic Transmission/drug effects , Animals , Cerebral Cortex/drug effects , Dizocilpine Maleate/pharmacokinetics , Dizocilpine Maleate/therapeutic use , Male , Memory Disorders/metabolism , Phenols/pharmacokinetics , Phenols/therapeutic use , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/therapeutic use , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...