Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Am J Psychiatry ; 176(5): 388-400, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30606046

ABSTRACT

OBJECTIVE: The N-methyl-d-aspartate receptor antagonist ketamine produces rapid and sustained antidepressant actions even in patients with treatment-resistant depression. Vascular endothelial growth factor (VEGF) has been implicated in the effects of conventional monoamine-based antidepressants, but the role of VEGF in the rapid antidepressant actions of ketamine remains unclear. The authors examined whether neuronal VEGF signaling in the medial prefrontal cortex (mPFC) mediates the rapid antidepressant actions of ketamine. METHODS: The authors used a combination of approaches, including conditional, neuron-specific knockout of VEGF or its receptor, Flk-1; antibody neutralization; viral-mediated knockdown of Flk-1; and pharmacological inhibitors. Further in vitro and in vivo experiments were performed to examine whether neuronal VEGF signaling was required for the neurotrophic and synaptogenic actions of ketamine that underlie its behavioral actions. RESULTS: The behavioral actions of systemic ketamine are blocked by forebrain excitatory neuron-specific deletion of either VEGF or Flk-1 or by intra-mPFC infusion of a VEGF neutralizing antibody. Moreover, intra-mPFC infusions of VEGF are sufficient to produce rapid ketamine-like behavioral actions, and these effects are blocked by neuron-specific Flk-1 deletion. The results also show that local knockdown of Flk-1 in mPFC excitatory neurons in adulthood blocks the behavioral effects of systemic ketamine. Moreover, inhibition of neuronal VEGF signaling blocks the neurotrophic and synaptogenic effects of ketamine. CONCLUSIONS: Together, these findings indicate that neuronal VEGF-Flk-1 signaling in the mPFC plays an essential role in the antidepressant actions of ketamine.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Neurons/drug effects , Prefrontal Cortex/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor Receptor-2/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Behavior, Animal/drug effects , Gene Knockdown Techniques , Gene Knockout Techniques , In Vitro Techniques , Mice , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/metabolism , Quinazolines/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Neuropharmacology ; 111: 242-252, 2016 12.
Article in English | MEDLINE | ID: mdl-27634096

ABSTRACT

Recent preclinical and clinical studies demonstrate that three functionally different compounds, the NMDA receptor channel blocker ketamine, mGlu2/3 receptor antagonist LY341495, and NMDA receptor glycine site agent GLYX-13 produce rapid and long lasting antidepressant effects. Furthermore, these agents are reported to stimulate ERK and mTORC1 signaling in brain. Here we used rat primary cortical culture neurons to further examine the cellular actions of these agents. The results demonstrate that low concentrations of all three compounds rapidly increase levels of the phosphorylated and activated forms of ERK and a downstream target of mTORC1, p70S6 kinase, in a concentration and time dependent manner. In addition, each compound rapidly increases BDNF release into the culture media. Further studies demonstrate that induction of BDNF release, as well as stimulation of phospho-ERK is blocked by incubation with an AMPA receptor antagonist. The requirement for AMPA receptor stimulation suggests that the effects of these rapid agents are activity dependent. This possibility is supported by studies demonstrating that neuronal silencing, via incubation with the GABAA receptor agonist muscimol, completely blocks phospho-ERK and BDNF release by each agent. Finally, incubation with each drug for 24 h increases the number and length of neuronal branches. Together, the results demonstrate that these three different rapid acting antidepressant agents increase ERK signaling and BDNF release in an activity dependent manner that leads to increased neuronal complexity. Further studies will be required to determine the exact mechanisms underlying these effects in cultured neurons and in rodent models.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , MAP Kinase Signaling System/drug effects , Amino Acids/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Dendrites/drug effects , GABAergic Neurons/metabolism , Ketamine/pharmacology , Oligopeptides/pharmacology , Primary Cell Culture , Rats , Receptor, trkB/metabolism , Receptors, AMPA/metabolism , Xanthenes/pharmacology
3.
Neurobiol Dis ; 82: 254-261, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26102021

ABSTRACT

Clinical studies demonstrate that scopolamine, a non-selective muscarinic acetylcholine receptor (mAchR) antagonist, produces rapid therapeutic effects in depressed patients, and preclinical studies report that the actions of scopolamine require glutamate receptor activation and the mechanistic target of rapamycin complex 1 (mTORC1). The present study extends these findings to determine the role of the medial prefrontal cortex (mPFC) and specific muscarinic acetylcholine receptor (M-AchR) subtypes in the actions of scopolamine. The administration of scopolamine increases the activity marker Fos in the mPFC, including the infralimbic (IL) and prelimbic (PrL) subregions. Microinfusions of scopolamine into either the IL or the PrL produced significant antidepressant responses in the forced swim test, and neuronal silencing of IL or PrL blocked the antidepressant effects of systemic scopolamine. The results also demonstrate that the systemic administration of a selective M1-AChR antagonist, VU0255035, produced an antidepressant response and stimulated mTORC1 signaling in the PFC, similar to the actions of scopolamine. Finally, we used a chronic unpredictable stress model as a more rigorous test of rapid antidepressant actions and found that a single dose of scopolamine or VU0255035 blocked the anhedonic response caused by CUS, an effect that requires the chronic administration of typical antidepressants. Taken together, these findings indicate that mPFC is a critical mediator of the behavioral actions of scopolamine and identify the M1-AChR as a therapeutic target for the development of novel and selective rapid-acting antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptor, Muscarinic M1/metabolism , Scopolamine/pharmacology , Anhedonia/drug effects , Anhedonia/physiology , Animals , Chronic Disease , Dietary Sucrose , Disease Models, Animal , Male , Mechanistic Target of Rapamycin Complex 1 , Microinjections , Multiprotein Complexes/metabolism , Muscarinic Antagonists/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptor, Muscarinic M1/antagonists & inhibitors , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/metabolism , Thiadiazoles/pharmacology , Time Factors , Tissue Culture Techniques
4.
Proc Natl Acad Sci U S A ; 112(19): 6188-93, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918363

ABSTRACT

Current treatments for major depressive disorder (MDD) have a time lag and are ineffective for a large number of patients. Development of novel pharmacological therapies requires a comprehensive understanding of the molecular events that contribute to MDD pathophysiology. Recent evidence points toward aberrant activity of synaptic proteins as a critical contributing factor. In the present studies, we used viral-mediated gene transfer to target a key mediator of activity-dependent synaptic protein synthesis downstream of mechanistic target of rapamycin complex 1 (mTORC1) known as p70 S6 kinase 1 (S6K1). Targeted delivery of two mutants of S6K1, constitutively active or dominant-negative, to the medial prefrontal cortex (mPFC) of rats allowed control of the mTORC1/S6K1 translational pathway. Our results demonstrate that increased expression of S6K1 in the mPFC produces antidepressant effects in the forced swim test without altering locomotor activity. Moreover, expression of active S6K1 in the mPFC blocked the anhedonia caused by chronic stress, resulting in a state of stress resilience. This antidepressant response was associated with increased neuronal complexity caused by enhanced S6K1 activity. Conversely, expression of dominant-negative S6K1 in the mPFC resulted in prodepressive behavior in the forced swim test and was sufficient to cause anhedonia in the absence of chronic stress exposure. Together, these data demonstrate a critical role for S6K1 activity in depressive behaviors, and suggest that pathways downstream of mTORC1 may underlie the pathophysiology and treatment of MDD.


Subject(s)
Depressive Disorder, Major/metabolism , Gene Expression Regulation, Enzymologic , Prefrontal Cortex/metabolism , Ribosomal Protein S6 Kinases/physiology , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal , Disease Models, Animal , Ketamine/chemistry , Male , Maze Learning , Neurons/metabolism , Phenotype , Phosphorylation , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/genetics , Signal Transduction , Sirolimus/chemistry , Swimming
5.
Int J Neuropsychopharmacol ; 18(1)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25539510

ABSTRACT

BACKGROUND: Recent studies demonstrate that the rapid antidepressant ketamine increases spine number and function in the medial prefrontal cortex (mPFC), and that these effects are dependent on activation of glutamate α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors and brain-derived neurotrophic factor (BDNF). In vitro studies also show that activation of AMPA receptors stimulates BNDF release via activation of L-type voltage-dependent calcium channels (VDCC). METHODS: Based on this evidence, we examined the role of BDNF release and the impact of L-type VDCCs on the behavioral actions of ketamine. RESULTS: The results demonstrate that infusion of a neutralizing BDNF antibody into the mPFC blocks the behavioral effects of ketamine in the forced swim test (FST). In addition, we show that pretreatment with nifedipine or verapamil, two structurally-different L-type calcium channel antagonists, blocks the behavioral effects of ketamine in the FST. Finally, we show that ketamine treatment stimulates BDNF release in primary cortical neurons and that this effect is blocked by inhibition of AMPA receptors or L-type VDCCs. CONCLUSIONS: Taken together, these results indicate that the antidepressant effects of ketamine are mediated by activation of L-type VDCCs and the release of BDNF. They further elucidate the cellular mechanisms underlying this novel rapid-acting antidepressant.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Ketamine/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Cells, Cultured , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Male , Neurons/drug effects , Neurons/physiology , Nifedipine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Verapamil/pharmacology
6.
Neuropsychopharmacology ; 38(11): 2268-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23680942

ABSTRACT

A single dose of the short-acting NMDA antagonist ketamine produces rapid and prolonged antidepressant effects in treatment-resistant patients with major depressive disorder (MDD), which are thought to occur via restoration of synaptic connectivity. However, acute dissociative side effects and eventual fading of antidepressant effects limit widespread clinical use of ketamine. Recent studies in medial prefrontal cortex (mPFC) show that the synaptogenic and antidepressant-like effects of a single standard dose of ketamine in rodents are dependent upon activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) signaling pathway together with inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3), which relieves its inhibitory in influence on mTOR. Here, we found that the synaptogenic and antidepressant-like effects of a single otherwise subthreshold dose of ketamine were potentiated when given together with a single dose of lithium chloride (a nonselective GSK-3 inhibitor) or a preferential GSK-3ß inhibitor; these effects included rapid activation of the mTORC1 signaling pathway, increased inhibitory phosphorylation of GSK-3ß, increased synaptic spine density/diameter, increased excitatory postsynaptic currents in mPFC layer V pyramidal neurons, and antidepressant responses that persist for up to 1 week in the forced-swim test model of depression. The results demonstrate that low, subthreshold doses of ketamine combined with lithium or a selective GSK-3 inhibitor are equivalent to higher doses of ketamine, indicating the pivotal role of the GSK-3 pathway in modulating the synaptogenic and antidepressant responses to ketamine. The possible mitigation by GSK-3 inhibitors of the eventual fading of ketamine's antidepressant effects remains to be explored.


Subject(s)
Antidepressive Agents/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Ketamine/pharmacology , Lithium Chloride/pharmacology , Synapses/drug effects , Animals , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glycogen Synthase Kinase 3/metabolism , Immobility Response, Tonic/drug effects , Indoles/pharmacology , Male , Maleimides/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Signal Transduction/drug effects , Synapses/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Biol Psychiatry ; 73(12): 1189-98, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23295207

ABSTRACT

Antidepressants that produce rapid and robust effects, particularly for severely ill patients, represent one of the largest unmet medical needs for the treatment of depression. Currently available drugs that modulate monoamine neurotransmission provide relief for only a subset of patients, and this minimal efficacy requires several weeks of chronic treatment. The recent discovery that the glutamatergic agent ketamine produces rapid antidepressant responses within hours has opened a new area of research to explore the molecular mechanisms through which ketamine produces these surprising responses. Clinical and preclinical findings have exposed some of the unique actions of ketamine and identified a cell-signaling pathway known as the mammalian target of rapamycin. Activation of mammalian target of rapamycin and increased synaptogenesis in the prefrontal cortex are crucial in mediating the antidepressant effects of ketamine. Importantly, the synaptic actions of ketamine allow rapid recovery from the insults produced by exposure to repeated stress that cause neuronal atrophy and loss of synaptic connections. In the following review, we explore some of the clinical and preclinical findings that have thrust ketamine to the forefront of rapid antidepressant research and unveiled some of its unique molecular and cellular actions.


Subject(s)
Antidepressive Agents/pharmacology , Neurogenesis/drug effects , Sirolimus/metabolism , Synapses/drug effects , Animals , Antidepressive Agents/therapeutic use , Humans , Mammals
8.
J Mol Psychiatry ; 1(1): 15, 2013.
Article in English | MEDLINE | ID: mdl-25408908

ABSTRACT

BACKGROUND: Depression is a prevalent neuropsychiatric disorder that affects an estimated 350 million people worldwide. Currently available treatments for depression are lacking in both speed of onset and efficacy. Recent pharmacological efforts have targeted the glutamatergic neurotransmitter system using the N-methyl-D-aspartate (NMDA) receptor antagonist ketamine to produce rapid and robust antidepressant effects, however the widespread clinical use of ketamine is limited due to side effects and abuse liability. More recently, work evaluating metabotropic mGluR2/3 receptor antagonists has demonstrated many similarities with ketamine. METHODS: Male, Sprague-Dawley rats were exposed to a chronic unpredictable stress paradigm, which produces decreased sucrose preference, a measure of anhedonia. Rats were then treated with vehicle or a single injection of the mGluR2/3 antagonist LY341495 (3 mg/kg, i.p.) and tested at 24 hrs, 48 hrs or 10 days after a single treatment. RESULTS: We demonstrate that a single treatment with LY341495 produces a rapid (within 1-2 days) and long-lasting (10 days) reversal of anhedonia caused by chronic unpredictable stress in rats. This model provides a rigorous test of rapid-acting agents as typical antidepressants require several weeks of treatment to produce a response. CONCLUSIONS: These data suggest that LY341495 has the ability to produce rapid and robust antidepressant effects similar to ketamine. Together, the results highlight the potential for similar compounds to produce rapid and lasting efficacy for the treatment of depression.

9.
Int J Neuropsychopharmacol ; 15(4): 429-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22114864

ABSTRACT

Recent studies demonstrate that ketamine, a fast-acting antidepressant, rapidly activates the mammalian target of rapamycin (mTOR) and increases synaptogenesis in the prefrontal cortex. Because of the side-effect and abuse potential of ketamine we are investigating alternative agents that produce similar effects. Here, we demonstrate that a single dose of LY 341495, an mGluR2/3 antagonist, produces ketamine-like biochemical and behavioural actions. LY 341495 administration rapidly (1 h) activates the mTOR pathway (mTOR, p70S6K, 4E-BP1) and subsequently (24 h later) increases levels of synaptic proteins (PSD-95, GluR1 and Synapsin I), similar to the effects of ketamine. Finally, the antidepressant effects of LY 341495 in the rat forced swim test are completely blocked by the mTOR inhibitor, rapamycin. The results indicate that the antidepressant actions of LY 341495 are mediated by activation of mTOR and suggest that this and other mGluR2/3 antagonists could produce rapid antidepressant effects in depressed patients.


Subject(s)
Gene Expression Regulation/physiology , Receptors, Metabotropic Glutamate/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids/pharmacology , Animals , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Ketamine/pharmacology , Male , Membrane Proteins/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/ultrastructure , Rats , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Signal Transduction/drug effects , Swimming/psychology , Synapsins/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Xanthenes/pharmacology
10.
Curr Opin Cell Biol ; 23(6): 730-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21996102

ABSTRACT

Depression is associated with structural alterations in limbic brain regions that control emotion and mood. Studies of chronic stress in animal models and postmortem tissue from depressed subjects demonstrate that these structural alterations result from atrophy and loss of neurons and glial cells. These findings indicate that depression and stress-related mood disorders can be considered mild neurodegenerative disorders. Importantly, there is evidence that these structural alterations can be blocked or even reversed by elimination of stress and by antidepressant treatments. A major focus of current investigations is to characterize the molecular signaling pathways and factors that underlie these effects of stress, depression, and antidepressant treatment. Recent advances in this research area are discussed and potential novel targets for antidepressant development are highlighted.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/pathology , Animals , Atrophy/drug therapy , Brain/pathology , Depressive Disorder/drug therapy , Depressive Disorder/pathology , Humans , Mood Disorders/drug therapy , Mood Disorders/pathology , Neuroglia/pathology , Neurons/pathology
11.
J Pharmacol Exp Ther ; 338(1): 345-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508084

ABSTRACT

Metabotropic glutamate receptor 7 (mGluR7) remains the most elusive of the eight known mGluRs primarily because of the limited availability of tool compounds to interrogate its potential therapeutic utility. The discovery of N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) as the first orally active, brain-penetrable, mGluR7-selective allosteric agonist by Mitsukawa and colleagues (Proc Natl Acad Sci USA 102:18712-18717, 2005) provides a means to investigate this receptor system directly. AMN082 demonstrates mGluR7 agonist activity in vitro and interestingly has a behavioral profile that supports utility across a broad spectrum of psychiatric disorders including anxiety and depression. The present studies were conducted to extend the in vitro and in vivo characterization of AMN082 by evaluating its pharmacokinetic and metabolite profile. Profiling of AMN082 in rat liver microsomes revealed rapid metabolism (t(1/2) < 1 min) to a major metabolite, N-benzhydrylethane-1,2-diamine (Met-1). In vitro selectivity profiling of Met-1 demonstrated physiologically relevant transporter binding affinity at serotonin transporter (SERT), dopamine transporter (DAT), and norepinephrine transporter (NET) (323, 3020, and 3410 nM, respectively); whereas the parent compound AMN082 had appreciable affinity at NET (1385 nM). AMN082 produced antidepressant-like activity and receptor occupancy at SERT up to 4 h postdose, a time point at which AMN082 is significantly reduced in brain and plasma while the concentration of Met-1 continues to increase in brain. Acute Met-1 administration produced antidepressant-like activity as would be expected from its in vitro profile as a mixed SERT, NET, DAT inhibitor. Taken together, these data suggest that the reported in vivo actions of AMN082 should be interpreted with caution, because they may involve other mechanisms in addition to mGluR7.


Subject(s)
Benzhydryl Compounds/pharmacology , Biogenic Monoamines/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Benzhydryl Compounds/metabolism , Biogenic Monoamines/physiology , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Male , Mice , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
12.
Biol Psychiatry ; 69(8): 754-61, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21292242

ABSTRACT

BACKGROUND: Despite widely reported clinical and preclinical studies of rapid antidepressant actions of glutamate N-methyl-D-aspartate (NMDA) receptor antagonists, there has been very little work examining the effects of these drugs in stress models of depression that require chronic administration of antidepressants or the molecular mechanisms that could account for the rapid responses. METHODS: We used a rat 21-day chronic unpredictable stress (CUS) model to test the rapid actions of NMDA receptor antagonists on depressant-like behavior, neurochemistry, and spine density and synaptic function of prefrontal cortex neurons. RESULTS: The results demonstrate that acute treatment with the noncompetitive NMDA channel blocker ketamine or the selective NMDA receptor 2B antagonist Ro 25-6981 rapidly ameliorates CUS-induced anhedonic and anxiogenic behaviors. We also found that CUS exposure decreases the expression levels of synaptic proteins and spine number and the frequency/amplitude of synaptic currents (excitatory postsynaptic currents) in layer V pyramidal neurons in the prefrontal cortex and that these deficits are rapidly reversed by ketamine. Blockade of the mammalian target of rapamycin protein synthesis cascade abolishes both the behavioral and biochemical effects of ketamine. CONCLUSIONS: The results indicate that the structural and functional deficits resulting from long-term stress exposure, which could contribute to the pathophysiology of depression, are rapidly reversed by NMDA receptor antagonists in a mammalian target of rapamycin dependent manner.


Subject(s)
Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Prefrontal Cortex/drug effects , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Synapses/drug effects , Animals , Behavior, Animal/physiology , Blotting, Western , Choice Behavior/drug effects , Choice Behavior/physiology , Dendritic Spines/drug effects , Dendritic Spines/physiology , Electrophysiology , Ketamine/pharmacology , Neurons/physiology , Phenols/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Synapses/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
13.
Bioorg Med Chem Lett ; 21(1): 195-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21126874

ABSTRACT

A series of 3-(pyridin-2-yl-ethynyl)benzamide negative allosteric modulators of the metabotropic glutamate receptor 5 (mGluR5 NAMs) have been prepared. Starting from HTS hit 1 (IC(50): 926 nM), potent mGluR5 NAMs showing excellent potencies (IC(50)s<50 nM) and good physicochemical profiles were prepared by monitoring LipE values. One compound 26 showed excellent mGluR5 binding (K(i): 21 nM) and antagonism (IC(50): 8 nM), an excellent rat PK profile (CL: 12 mL/min/kg, %F: 85) and showed oral activity in a mouse 4-Plate Behavioral model of anxiety (MED: 30 mpk) and a mouse Stress Induced Hyperthermia model of anxiety (MED 17.8 mpk).


Subject(s)
Benzamides/chemistry , Pyridines/chemistry , Receptors, Metabotropic Glutamate/chemistry , Allosteric Regulation , Animals , Anxiety Disorders/drug therapy , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Disease Models, Animal , High-Throughput Screening Assays , Mice , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism
14.
Science ; 329(5994): 959-64, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20724638

ABSTRACT

The rapid antidepressant response after ketamine administration in treatment-resistant depressed patients suggests a possible new approach for treating mood disorders compared to the weeks or months required for standard medications. However, the mechanisms underlying this action of ketamine [a glutamate N-methyl-D-aspartic acid (NMDA) receptor antagonist] have not been identified. We observed that ketamine rapidly activated the mammalian target of rapamycin (mTOR) pathway, leading to increased synaptic signaling proteins and increased number and function of new spine synapses in the prefrontal cortex of rats. Moreover, blockade of mTOR signaling completely blocked ketamine induction of synaptogenesis and behavioral responses in models of depression. Our results demonstrate that these effects of ketamine are opposite to the synaptic deficits that result from exposure to stress and could contribute to the fast antidepressant actions of ketamine.


Subject(s)
Antidepressive Agents/pharmacology , Ketamine/pharmacology , Neuropeptides/biosynthesis , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/drug effects , Animals , Antidepressive Agents/pharmacokinetics , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Depression/drug therapy , Depression/metabolism , Intracellular Signaling Peptides and Proteins/agonists , Ketamine/pharmacokinetics , Male , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Phenols/pharmacology , Piperidines/pharmacology , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sirolimus/pharmacology , Synapses/metabolism , TOR Serine-Threonine Kinases , Time Factors
15.
Neurobiol Dis ; 39(2): 148-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20381618

ABSTRACT

Rimonabant was the first clinically marketed cannabinoid (CB)(1) receptor antagonist developed to treat obesity. Unfortunately, CB(1) receptor antagonism produced adverse psychiatric events in patients. To determine whether this occurs pre-clinically, we investigated the effects of rimonabant in rodent models of mood disorders. Chronic treatment with rimonabant increased immobility time in the rat forced swim test and reduced the consumption of sucrose-sweetened water in an assay postulated to model anhedonia. These responses were similar to the effects elicited by chronic mild stress in these behavioral models, which, taken together, are indicative of a depression-like phenotype. Additionally, chronic treatment with rimonabant produced decreases in frontal cortex serotonin levels, marked reductions in hippocampal cell proliferation, survival, and BDNF levels, and elevations in the concentrations of pro-inflammatory cytokines including interferon gamma and TNF alpha. These preclinical findings mimic clinical reports and implicate possible mechanisms responsible for the unfavorable psychiatric events reported following chronic rimonabant use.


Subject(s)
Phenotype , Piperidines/adverse effects , Pyrazoles/adverse effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Stress, Psychological/chemically induced , Stress, Psychological/physiopathology , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry/methods , Food Preferences/drug effects , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Immobility Response, Tonic/drug effects , Male , Microdialysis/methods , Rats , Rats, Sprague-Dawley , Rimonabant , Stress, Psychological/pathology , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
16.
Proc Natl Acad Sci U S A ; 107(18): 8457-62, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20404172

ABSTRACT

Phosphodiesterase 11A (PDE11A) is the most recently identified family of phosphodiesterases (PDEs), the only known enzymes to break down cyclic nucleotides. The tissue expression profile of this dual specificity PDE is controversial, and little is understood of its biological function, particularly in the brain. We seek here to determine if PDE11A is expressed in the brain and to understand its function, using PDE11A(-/-) knockout (KO) mice. We show that PDE11A mRNA and protein are largely restricted to hippocampus CA1, subiculum, and the amygdalohippocampal area, with a two- to threefold enrichment in the ventral vs. dorsal hippocampus, equal distribution between cytosolic and membrane fractions, and increasing levels of protein expression from postnatal day 7 through adulthood. Interestingly, PDE11A KO mice show subtle psychiatric-disease-related deficits, including hyperactivity in an open field, increased sensitivity to the glutamate N-methyl-D-aspartate receptor antagonist MK-801, as well as deficits in social behaviors (social odor recognition memory and social avoidance). In addition, PDE11A KO mice show enlarged lateral ventricles and increased activity in CA1 (as per increased Arc mRNA), phenotypes associated with psychiatric disease. The increased sensitivity to MK-801 exhibited by PDE11A KO mice may be explained by the biochemical dysregulation observed around the glutamate alpha-amino-3-hydroxy-5-methyl-4-isozazolepropionic (AMPA) receptor, including decreased levels of phosphorylated-GluR1 at Ser845 and the prototypical transmembrane AMPA-receptor-associated proteins stargazin (gamma2) and gamma8. Together, our data provide convincing evidence that PDE11A expression is restricted in the brain but plays a significant role in regulating brain function.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Hippocampus/enzymology , Mental Disorders/enzymology , 3',5'-Cyclic-GMP Phosphodiesterases/deficiency , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Animals , Behavior, Animal , Female , Gene Expression Regulation, Enzymologic , Glutamine/metabolism , Hippocampus/pathology , Male , Mental Disorders/genetics , Mental Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Social Behavior
17.
Psychopharmacology (Berl) ; 209(4): 303-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20224888

ABSTRACT

INTRODUCTION: The effects of angiotensin (Ang) IV result from binding to a constitutively active metallopeptidase known as the AT(4) receptor (or oxytocinase/insulin-regulated membrane aminopeptidase). While in vitro evidence indicates that Ang IV inhibits the peptidase activity of AT(4) receptors, leading to increases in the concentrations of several peptides, including oxytocin, the consequence of inhibiting AT(4) peptidase activity in vivo remains unresolved. DISCUSSION: Microdialysis coupled to immunoassay techniques revealed that systemic and intra-amygdala injection of Nle-Ang IV, a metabolically stable derivative of Ang IV, significantly elevated extracellular levels of oxytocin in the rat amygdala. Based on earlier reports describing the anxiolytic-like effects of oxytocin, we investigated whether disrupting AT(4) peptidase activity would yield similar responses. In the mouse four-plate test, acute treatment with either Nle-Ang IV or LVV-hemorphin-7, a related AT(4) receptor ligand, elicited significant increases in the number of punished crossings. These behavioral responses were comparable to the anxiolytic-like effects of oxytocin and to the standard anxiolytic agent, chlordiazepoxide. Cotreatment with either the AT(4) receptor antagonist, divalinal, or the selective oxytocin receptor antagonist, WAY-162720, reversed the anxiolytic-like effects of Nle-Ang IV, while combining ineffective doses of Nle-Ang IV and oxytocin increased the number of punished crossings in this assay. Conversely, Nle-Ang IV and LVV-hemorphin-7 were inactive in the mouse tail suspension test of antidepressant activity. These findings represent the first in vivo demonstration of the peptidase activity of AT(4) receptors, confirm the anxiolytic-like properties of Ang IV, and reveal a unique and previously uncharacterized relationship between AT(4) and oxytocin receptor systems.


Subject(s)
Amygdala/drug effects , Angiotensin II/analogs & derivatives , Anti-Anxiety Agents/administration & dosage , Anxiety/prevention & control , Oxytocin/metabolism , Receptors, Angiotensin/agonists , Receptors, Oxytocin/agonists , Amygdala/metabolism , Angiotensin II/administration & dosage , Angiotensin Receptor Antagonists , Animals , Anxiety/metabolism , Anxiety/psychology , Behavior, Animal/drug effects , Depression/drug therapy , Depression/metabolism , Depression/psychology , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hemoglobins/administration & dosage , Immunoassay , Injections, Intraperitoneal , Injections, Intraventricular , Injections, Subcutaneous , Ligands , Male , Mice , Microdialysis , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Time Factors , Up-Regulation
18.
Int J Neuropsychopharmacol ; 13(9): 1193-205, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20047711

ABSTRACT

Biogenic amines such as norepinephrine, dopamine, and serotonin play a well-described role in the treatment of mood disorders and some types of pain. As alpha2A-adrenoceptors regulate the release of these neurotransmitters, we examined the therapeutic potential of BRL 44408, a potent (Ki=8.5 nM) and selective (>50-fold) alpha2A-adrenoceptor antagonist (K(B)=7.9 nM). In rats, BRL 44408 penetrated the central nervous system resulting in peak brain and plasma concentrations of 586 ng/g and 1124 ng/ml, respectively. In a pharmacodynamic assay, pretreatment with BRL 44408 to rats responding under a fixed-ratio 30 operant response paradigm resulted in a rightward shift of the clonidine dose-response curve, an effect indicative of alpha2-adrenoceptor antagonism in vivo. Consistent with presynaptic autoreceptor antagonism and tonic regulation of neurotransmitter release, acute administration of BRL 44408 elevated extracellular concentrations of norepinephrine and dopamine, but not serotonin, in the medial prefrontal cortex. Additionally, BRL 44408, probably by inhibiting alpha2A heteroceptors, produced a significant increase in cortical levels of acetylcholine. In the forced swim test and schedule-induced polydipsia assay, BRL 44408 produced an antidepressant-like response by dose-dependently decreasing immobility time and adjunctive water intake, respectively, while in a model of visceral pain, BRL 44408 exhibited analgesic activity by decreasing para-phenylquinone (PPQ)-induced abdominal stretching. Finally, BRL 44408 did not produce deficits in overall motor coordination nor alter general locomotor activity. This preclinical characterization of the neurochemical and behavioural profile of BRL 44408 suggests that selective antagonism of alpha2A-adrenoceptors may represent an effective treatment strategy for mood disorders and visceral pain.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Analgesics/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Imidazoles/pharmacology , Isoindoles/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacokinetics , Analgesics/pharmacokinetics , Animals , Antidepressive Agents/pharmacokinetics , Biogenic Monoamines/metabolism , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Drug Evaluation, Preclinical , Imidazoles/pharmacokinetics , Isoindoles/pharmacokinetics , Male , Mice , Microdialysis , Radioligand Assay , Rats , Rats, Sprague-Dawley , Swimming , Thirst/drug effects
19.
Neuropharmacology ; 58(1): 69-77, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19615387

ABSTRACT

The widely reported effects of oxytocin (OT) on CNS function has generated considerable interest in the therapeutic potential for targeting this system for a variety of human psychiatric diseases, including anxiety disorders, autism, schizophrenia, and depression. The utility of synthetic OT, as both a research tool and neurotherapeutic, is limited by the physiochemical properties inherent in most neuropeptides, notably its short half-life and poor blood brain barrier penetration. Subsequently, the discovery and development of non-peptide molecules that act as selective agonists of the oxytocin receptor (OTR) has been an important goal of the field. In this study, we report the receptor and behavioral pharmacology of WAY-267464, a first generation small-molecule OTR agonist. WAY-267464 is a high-affinity, potent, and selective (vs. V1a, V2, V1b) agonist of the OTR. In assays measuring both behavioral (four-plate test, elevated zero maze) and autonomic (stress-induced hyperthermia) parameters of the anxiety response, WAY-267464 exhibits an anxiolytic-like profile similar to OT. We have demonstrated that the anxiolytic-like profile of WAY-267464 is mediated through central sites of action. WAY-267464 also significantly reverses disruption in prepulse inhibition of the acoustic startle reflex induced by either MK-801 or amphetamine, similar to the antipsychotic-like effects previously reported for OT. Interestingly, in the mouse tail suspension test, WAY-267464 failed to produce changes in immobility that are seen with OT, raising the question of whether the antidepressant-like activity of OT may be working independently of the OTR. A selective OTR antagonist also failed to block the effects of OT on immobility in the TST. The significance of these findings for shaping the clinical development of OTR agonists is discussed.


Subject(s)
Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Oxytocin/pharmacology , Receptors, Oxytocin/agonists , Acoustic Stimulation/adverse effects , Animals , Avoidance Learning/drug effects , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , CHO Cells , Cricetinae , Cricetulus , Fever/drug therapy , Fever/etiology , Hindlimb Suspension/methods , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neural Inhibition/drug effects , Oxytocin/agonists , Protein Binding/drug effects , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Reflex, Startle/physiology , Stress, Psychological/complications
20.
Pharmacol Biochem Behav ; 92(4): 649-54, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19303035

ABSTRACT

One of the few preclinical models used to identify mood stabilizers is an assay in which amphetamine-induced hyperactivity (AMPH) is potentiated by the benzodiazepine chlordiazepoxide (CDP), an effect purportedly blocked by mood stabilizers. Our data here challenge this standard interpretation of the AMPH-CDP model. We show that the potentiating effects of AMPH-CDP are not explained by a pharmacokinetic interaction as both drugs have similar brain and plasma exposures whether administered alone or in combination. Of concern, however, we find that combining CDP (1-12 mg/kg) with AMPH (3 mg/kg) results in an inverted-U dose response in outbred CD-1 as well as inbred C57Bl/6N and 129S6 mice (peak hyperactivity at 3 mg/kg CDP+3 mg/kg AMPH). Such an inverted-U dose response complicates interpreting whether a reduction in hyperactivity produced by a mood stabilizer reflects a "blockade" or a "potentiation" of the mixture. In fact, we show that the prototypical mood stabilizer valproic acid augments the effects of CDP on hypolocomotion and anxiolytic-like behavior (increases punished crossings by Swiss-Webster mice in the four-plate test). We argue that these data, in addition to other practical and theoretical concerns surrounding the model, limit the utility of the AMPH-CDP mixture model in drug discovery.


Subject(s)
Affect/drug effects , Amphetamine/administration & dosage , Chlordiazepoxide/administration & dosage , Animals , Antimanic Agents/administration & dosage , Anxiety/drug therapy , Bipolar Disorder/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Synergism , Male , Mice , Mice, Inbred C57BL , Models, Neurological , Motor Activity/drug effects , Valproic Acid/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...