Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Pharmacother ; 105: 540-544, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29886374

ABSTRACT

Neurotrophin dysregulation has been implicated in a large number of neurodegenerative and neuropsychiatric diseases. Unfortunately, neurotrophins cannot cross the blood brain barrier thus, novel means of up regulating their expression are greatly needed. It has been demonstrated previously that neurotrophins are up regulated in response to increases in brain activity. Therefore, molecules that act as cognitive enhancers may provide a clinical means of up regulating neurotrophin expression. Ampakines are a class of molecules that act as positive allosteric modulators of AMPA-type glutamate receptors. Currently, they are being developed to prevent opioid-induced respiratory depression without sacrificing the analgesic properties of the opioids. In addition, these molecules increase neuronal activity and have been shown to restore age-related deficits in LTP in aged rats. In the current study, we examined whether two different ampakines could increase levels of BDNF and NGF at doses that are active in behavioral measures of cognition. Results demonstrate that ampakines CX516 and CX691 induce differential increases in neurotrophins across several brain regions. Notable increases in NGF were observed in the dentate gyrus and piriform cortex while notable BDNF increases were observed in basolateral and lateral nuclei of the amygdala. Taken together, our data demonstrates that chronic administration of clinically relevant doses of ampakines have the ability to elevate neurotrophin expression in different brain regions, and may have therapeutic benefit in multiple neurodegenerative and/or neuropsychiatric disorders.


Subject(s)
Brain/metabolism , Nerve Growth Factors/genetics , Receptors, AMPA/agonists , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Dioxoles/pharmacology , Male , Nerve Growth Factors/metabolism , Piperidines/pharmacology , Piriform Cortex/drug effects , Piriform Cortex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
2.
Anticancer Res ; 38(1): 265-269, 2018 01.
Article in English | MEDLINE | ID: mdl-29277782

ABSTRACT

BACKGROUND/AIM: The 5-year survival rate of glioblastoma (GBM) is ~10%, demonstrating that a new therapeutic modality for this cancer is desperately needed. Complicating the search for such a modality is that most large molecules cannot pass through the blood brain barrier, so molecules demonstrating efficacy in vitro may not be useful in vivo because they never reach the brain. Recently, the selective serotonin reuptake inhibitor (SSRI) fluoxetine (FLX) was identified as an effective agent in targeting GBM in vitro and in vivo by agonizing AMPA-glutamate receptors (AMPARs), eliciting massive calcium influx and mitochondrial calcium overload and apoptosis. MATERIALS AND METHODS: In the current study, we used a colorimetric cell viability assay to determine if we could enhance the oncolytic effect of FLX in vitro by pre-treating cells with an AMPAR-positive allosteric modulator (Ampakine). RESULTS: Our results demonstrated for the first time that concentrations of the Class I ampakine CX614, which increase AMPAR agonist binding affinity, possess oncolytic activity as a sole agent and synergistically reduce GBM viability when paired with FLX. FLX also demonstrates a dose-dependent induction of apoptosis in cancer cells originating outside the CNS that overexpress calcium-permeable AMPARs. Likewise, CX614 inhibits cancer cell viability in a dose-dependent fashion and its combination with FLX synergistically reduces cell viability. These effects of CX614 were not seen with the Class II ampakines, CX717 and CX1739. CONCLUSION: CX614 inhibits the growth of multiple cancers in vitro and bolsters the oncolytic activity of FLX in several cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Fluoxetine/pharmacology , Oxazines/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/drug therapy , Humans , Receptors, AMPA
3.
J Pharmacol Exp Ther ; 321(3): 1208-25, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17325229

ABSTRACT

Bicifadine (1-p-tolyl-3-azabicyclo[3.1.0]hexane) inhibits monoamine neurotransmitter uptake by recombinant human transporters in vitro with a relative potency of norepinephrine > serotonin > dopamine (approximately 1:2:17). This in vitro profile is supported by microdialysis studies in freely moving rats, where bicifadine (20 mg/kg i.p.) increased extrasynaptic norepinephrine and serotonin levels in the prefrontal cortex, norepinephrine levels in the locus coeruleus, and dopamine levels in the striatum. Orally administered bicifadine is an effective antinociceptive in several models of acute, persistent, and chronic pain. Bicifadine potently suppressed pain responses in both the Randall-Selitto and kaolin models of acute inflammatory pain and in the phenyl-p-quinone-induced and colonic distension models of persistent visceral pain. Unlike many transport inhibitors, bicifadine was potent and completely efficacious in both phases of the formalin test in both rats and mice. Bicifadine also normalized the nociceptive threshold in the complete Freund's adjuvant model of persistent inflammatory pain and suppressed mechanical and thermal hyperalgesia and mechanical allodynia in the spinal nerve ligation model of chronic neuropathic pain. Mechanical hyperalgesia was also reduced by bicifadine in the streptozotocin model of neuropathic pain. Administration of the D(2) receptor antagonist (-)-sulpiride reduced the effects of bicifadine in the mechanical hyperalgesia assessment in rats with spinal nerve ligations. These results indicate that bicifadine is a functional triple reuptake inhibitor with antinociceptive and antiallodynic activity in acute, persistent, and chronic pain models, with activation of dopaminergic pathways contributing to its antihyperalgesic actions.


Subject(s)
Analgesics/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Pain/drug therapy , Acute Disease , Analgesics/metabolism , Animals , Brain/drug effects , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Chronic Disease , Desipramine/pharmacology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice , Microdialysis , Motor Activity/drug effects , Neurotransmitter Transport Proteins/antagonists & inhibitors , Neurotransmitter Transport Proteins/metabolism , Norepinephrine/metabolism , Pain/metabolism , Pain/physiopathology , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reaction Time/drug effects , Receptors, Neurotransmitter/antagonists & inhibitors , Receptors, Neurotransmitter/metabolism , Serotonin/metabolism
4.
J Pharmacol Exp Ther ; 319(3): 1244-52, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16971504

ABSTRACT

Studies using mice with point mutations of GABA(A) receptor alpha subunits suggest that the sedative and anxiolytic properties of 1,4-benzodiazepines are mediated, respectively, by GABA(A) receptors bearing the alpha(1) and alpha(2) subunits. This hypothesis predicts that a compound with high efficacy at GABA(A) receptors containing the alpha(1) subunit would produce sedation, whereas an agonist acting at alpha(2) subunit-containing receptors (with low or null efficacy at alpha(1)-containing receptors) would be anxioselective. Electrophysiological studies using recombinant GABA(A) receptors expressed in Xenopus oocytes indicate that maximal potentiation of GABA-stimulated currents by the pyrazolo-[1,5-a]-pyrimidine, DOV 51892, at alpha(1)beta(2)gamma(2S) constructs of the GABA(A) receptor was significantly higher (148%) than diazepam. In contrast, DOV 51892 was considerably less efficacious and/or potent than diazepam in enhancing GABA-stimulated currents mediated by constructs containing alpha(2), alpha(3), or alpha(5) subunits. In vivo, DOV 51892 increased punished responding in the Vogel conflict test, an effect blocked by flumazenil, and increased the percentage of time spent in the open arms of the elevated plus-maze. However, DOV 51892 had no consistent effects on motor function or muscle relaxation at doses more than 1 order of magnitude greater than the minimal effective anxiolytic dose. Although the mutant mouse data predict that the high-efficacy potentiation of GABA(A1a) receptor-mediated currents by DOV 51892 would be sedating, behavioral studies demonstrate that DOV 51892 is anxioselective, indicating that GABA potentiation mediated by alpha(1) subunit-containing GABA(A) receptors may be neither the sole mechanism nor highly predictive of the sedative properties of benzodiazepine recognition site modulators.


Subject(s)
Anti-Anxiety Agents/pharmacology , Chloride Channels/metabolism , Diazepam/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/physiology , Animals , Anti-Anxiety Agents/chemical synthesis , Ataxia/chemically induced , Ataxia/pathology , Chloride Channels/drug effects , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hand Strength/physiology , Hypnotics and Sedatives/pharmacology , Male , Motor Activity/drug effects , Muscle Relaxation/drug effects , Oocytes/metabolism , Postural Balance/drug effects , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Radioligand Assay , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, GABA-A/drug effects , Xenopus laevis
5.
Curr Protoc Pharmacol ; Chapter 5: Unit 5.38, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-22294128

ABSTRACT

Animal tests of anxiety are used to screen novel compounds for anxiolytic or anxiogenic activity, to investigate the neurobiology of anxiety, and to assess the impact of other occurrences such as exposure to predator odors or early rearing experiences. This unit presents protocols for the most commonly used animal tests of anxiety. The Geller-Seifter conflict test, the social interaction test, light/dark exploration, the elevated plus-maze, defensive burying, and the thirsty rat conflict. The protocols are described in terms of drug screening tests, but can be modified easily for other purposes.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/chemically induced , Anxiety/drug therapy , Psychological Tests , Animals , Behavior, Animal , Conditioning, Operant/drug effects , Conflict, Psychological , Darkness , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Exploratory Behavior/drug effects , Gerbillinae , Humans , Lighting , Male , Maze Learning/drug effects , Rats , Social Behavior , Thirst/drug effects
6.
Eur J Pharmacol ; 500(1-3): 441-51, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15464051

ABSTRACT

Benzodiazepines remain widely used for the treatment of anxiety disorders despite a side-effect profile that includes sedation, myorelaxation, amnesia, and ataxia, and the potential for abuse. gamma-Aminobutyric acid(A) (GABA(A)) receptor partial agonists, subtype-selective agents, and compounds combining both of these features are being developed in an attempt to achieve benzodiazepine-like efficacy without these potentially limiting side effects. This article reviews the nonclinical and clinical studies of "anxioselective" anxiolytics that target GABA(A) receptors and discusses potential mechanisms subserving an anxioselective profile.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety Disorders/drug therapy , GABA-A Receptor Agonists , Animals , Anti-Anxiety Agents/therapeutic use , Benzodiazepinones/pharmacology , Benzodiazepinones/therapeutic use , Carbolines/pharmacology , Carbolines/therapeutic use , Clinical Trials as Topic , Fluorobenzenes/pharmacology , Fluorobenzenes/therapeutic use , Humans , Indoles/pharmacology , Indoles/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Quinolones/pharmacology , Quinolones/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use
7.
Curr Protoc Neurosci ; Chapter 8: Unit 8.3, 2004 May.
Article in English | MEDLINE | ID: mdl-18428606

ABSTRACT

Animal tests of anxiety are used to screen novel compounds for anxiolytic or anxiogenic activity, to investigate the neurobiology of anxiety, and to assess the impact of other occurrences such as exposure to predator odors or early rearing experiences. This unit presents protocols for the most commonly used animal tests of anxiety. The Geller-Seifter conflict test, the social interaction test, light/dark exploration, the elevated plus-maze, defensive burying, and the thirsty rat conflict. The protocols are described in terms of drug screening tests, but can be modified easily for other purposes.


Subject(s)
Anxiety/diagnosis , Behavioral Sciences/methods , Biomedical Research/methods , Neurosciences/methods , Psychological Tests , Animals , Avoidance Learning , Conflict, Psychological , Darkness , Defense Mechanisms , Electroshock , Exploratory Behavior , Female , Interpersonal Relations , Light , Male , Maze Learning , Rats , Thirst
8.
Life Sci ; 73(25): 3175-9, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-14561522

ABSTRACT

The majority of antidepressants in current use selectively inhibit the reuptake of serotonin and/or norepinephrine. "Broad spectrum" antidepressants are compounds that inhibit the reuptake of norepinephrine, serotonin and dopamine, the three biogenic amines most closely linked to depression. The pharmacological profile of one such compound has recently been described (European Journal of Pharmacology, 461 (2003) 99). DOV 21,947, an azabicyclo[3.1.0]hexane, potently inhibits norepinephrine, serotonin and dopamine reuptake by the corresponding human transporter proteins. DOV 21,947 is orally active in the forced swim and tail suspension tests, preclinical procedures that are highly predictive of antidepressant action in patients. A closely related compound, DOV 216,303 is safe and well-tolerated in Phase I studies. The plasma concentrations of DOV 216,303 following both single and multiple doses appear sufficient to inhibit norepinephrine, serotonin, and dopamine reuptake. Based on the pivotal role proposed for dopamine in depression, it has been hypothesized that a broad spectrum antidepressant will produce a more rapid onset and/or higher efficacy than agents inhibiting the reuptake of serotonin and/or norepinephrine.


Subject(s)
Antidepressive Agents/therapeutic use , Aza Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Depression/drug therapy , Neurotransmitter Uptake Inhibitors/therapeutic use , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Immobilization , Male , Mice , Swimming , Treatment Outcome
9.
Eur J Pharmacol ; 461(2-3): 99-104, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12586204

ABSTRACT

DOV 21,947 [(+)-1-(3,4-dichlorophenyl)-3-azabicyclo-[3.1.0]hexane hydrochloride] inhibits the reuptake of [3H]serotonin, [3H]norepinephrine, and [3H]dopamine in human embryonic kidney (HEK) 293 cells expressing the corresponding human recombinant transporters (IC(50) values of 12, 23, and 96 nM, respectively). This compound also inhibits [125I]RTI 55 (3beta-(4-iodophenyl)tropane-2beta-carboxylic acid methyl ester) binding to the corresponding transporter proteins in membranes prepared from these cells (K(i) values of 99, 262, and 213 nM, respectively). DOV 21,947 reduces the duration of immobility in the forced swim test (using rats) with an oral minimum effective dose of 5 mg/kg. This antidepressant-like effect manifests in the absence of significant increases in motor activity at doses of up to 20 mg/kg. DOV 21,947 also produces a dose-dependent reduction in immobility in the tail suspension test, with a minimum effective oral dose of 5 mg/kg. The ability of DOV 21,947 to inhibit the reuptake of three biogenic amines closely linked to the etiology of depression may result in a therapeutic profile different from antidepressants that inhibit the reuptake of serotonin and/or norepinephrine.


Subject(s)
Antidepressive Agents/pharmacology , Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cocaine/analogs & derivatives , Nerve Tissue Proteins , Neurotransmitter Uptake Inhibitors/pharmacology , Animals , Behavior, Animal/drug effects , Binding, Competitive/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cocaine/metabolism , Dopamine/physiology , Dopamine Plasma Membrane Transport Proteins , Dose-Response Relationship, Drug , Gene Expression , Hindlimb Suspension , Humans , Iodine Radioisotopes , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Norepinephrine/physiology , Norepinephrine Plasma Membrane Transport Proteins , Rats , Rats, Wistar , Serotonin/physiology , Serotonin Plasma Membrane Transport Proteins , Swimming , Symporters/genetics , Symporters/metabolism , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...