Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Exp Biol Med (Maywood) ; 243(3): 228-236, 2018 02.
Article in English | MEDLINE | ID: mdl-29105512

ABSTRACT

Neurotoxicity has been linked with exposure to a number of common drugs and chemicals, yet efficient, accurate, and minimally invasive methods to detect it are lacking. Fluid-based biomarkers such as those found in serum, plasma, urine, and cerebrospinal fluid have great potential due to the relative ease of sampling but at present, data on their expression and translation are lacking or inconsistent. In this pilot study using a trimethyl tin rat model of central nervous system toxicity, we have applied state-of-the-art assessment techniques to identify potential individual biomarkers and patterns of biomarkers in serum, plasma, urine or cerebral spinal fluid that may be indicative of nerve cell damage and degeneration. Overall changes in metabolites and microRNAs were observed in biological fluids that were associated with neurotoxic damage induced by trimethyl tin. Behavioral changes and magnetic resonance imaging T2 relaxation and ventricle volume changes served to identify animals that responded to the adverse effects of trimethyl tin. Impact statement These data will help design follow-on studies with other known neurotoxicants to be used to assess the broad applicability of the present findings. Together this approach represents an effort to begin to develop and qualify a set of translational biochemical markers of neurotoxicity that will be readily accessible in humans. Such biomarkers could prove invaluable for drug development research ranging from preclinical studies to clinical trials and may prove to assist with monitoring of the severity and life cycle of brain lesions.


Subject(s)
Biomarkers , Body Fluids/chemistry , Central Nervous System/pathology , MicroRNAs/analysis , Neurons/pathology , Neurotoxicity Syndromes/diagnosis , Trimethyltin Compounds/toxicity , Amino Acids/analysis , Animals , Behavior, Animal/drug effects , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/urine , Humans , Magnetic Resonance Imaging , Male , Metabolome/physiology , MicroRNAs/genetics , Pilot Projects , Rats , Rats, Sprague-Dawley
2.
Toxicol Sci ; 148(2): 332-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26609132

ABSTRACT

Neurotoxicity has been linked to a number of common drugs and chemicals, yet efficient and accurate methods to detect it are lacking. There is a need for more sensitive and specific biomarkers of neurotoxicity that can help diagnose and predict neurotoxicity that are relevant across animal models and translational from nonclinical to clinical data. Fluid-based biomarkers such as those found in serum, plasma, urine, and cerebrospinal fluid (CSF) have great potential due to the relative ease of sampling compared with tissues. Increasing evidence supports the potential utility of fluid-based biomarkers of neurotoxicity such as microRNAs, F2-isoprostanes, translocator protein, glial fibrillary acidic protein, ubiquitin C-terminal hydrolase L1, myelin basic protein, microtubule-associated protein-2, and total tau. However, some of these biomarkers such as those in CSF require invasive sampling or are specific to one disease such as Alzheimer's, while others require further validation. Additionally, neuroimaging methodologies, including magnetic resonance imaging, magnetic resonance spectroscopy, and positron emission tomography, may also serve as potential biomarkers and have several advantages including being minimally invasive. The development of biomarkers of neurotoxicity is a goal shared by scientists across academia, government, and industry and is an ideal topic to be addressed via the Health and Environmental Sciences Institute (HESI) framework which provides a forum to collaborate on key challenging scientific topics. Here we utilize the HESI framework to propose a consensus on the relative potential of currently described biomarkers of neurotoxicity to assess utility of the selected biomarkers using a nonclinical model.


Subject(s)
Biomarkers/metabolism , Nervous System/drug effects , Neurotoxicity Syndromes/etiology , Toxicology/methods , Translational Research, Biomedical/methods , Animals , Disease Models, Animal , Genetic Markers , Humans , Nervous System/metabolism , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment
3.
J Neurosci ; 35(3): 1199-210, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25609634

ABSTRACT

BACE1 is a key protease controlling the formation of amyloid ß, a peptide hypothesized to play a significant role in the pathogenesis of Alzheimer's disease (AD). Therefore, the development of potent and selective inhibitors of BACE1 has been a focus of many drug discovery efforts in academia and industry. Herein, we report the nonclinical and early clinical development of LY2886721, a BACE1 active site inhibitor that reached phase 2 clinical trials in AD. LY2886721 has high selectivity against key off-target proteases, which efficiently translates in vitro activity into robust in vivo amyloid ß lowering in nonclinical animal models. Similar potent and persistent amyloid ß lowering was observed in plasma and lumbar CSF when single and multiple doses of LY2886721 were administered to healthy human subjects. Collectively, these data add support for BACE1 inhibition as an effective means of amyloid lowering and as an attractive target for potential disease modification therapy in AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Picolinic Acids/pharmacology , Protease Inhibitors/pharmacology , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Disease Models, Animal , Dogs , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Mice , Picolinic Acids/pharmacokinetics , Picolinic Acids/therapeutic use , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/therapeutic use
4.
Psychopharmacology (Berl) ; 226(2): 189-200, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23397050

ABSTRACT

RATIONALE: Treatment of attention-deficit/hyperactivity disorder (ADHD) has for many years relied on psychostimulants, particularly various formulations of amphetamines and methylphenidate. These are central nervous system stimulants and are scheduled because of their abuse potential. Atomoxetine (atomoxetine hydrochloride; Strattera®) was approved in 2002 for treatment of ADHD, and was the first nonstimulant medication approved for this disorder. It was classified as an unscheduled medication indicating a low potential for abuse. However, the abuse potential of atomoxetine has not been reviewed. OBJECTIVES: In this article, we review the evidence regarding abuse potential of atomoxetine, a selective inhibitor of the presynaptic norepinephrine transporter, which is unscheduled/unrestricted in all countries where it is approved. METHODS: Results from receptor binding, in vitro electrophysiology, in vivo microdialysis, preclinical behavioral, and human laboratory studies have been reviewed. RESULTS: Atomoxetine has no appreciable affinity for, or action at, central receptors through which drugs of abuse typically act, i.e., dopamine transporters, GABA(A) receptors, and opioid µ receptors. In behavioral experiments in rodents, atomoxetine does not increase locomotor activity, and in drug discrimination studies, its profile is similar to that of drugs without abuse potential. Atomoxetine does not serve as a reinforcer in monkey self-administration studies, and human laboratory studies suggest that atomoxetine does not induce subjective effects indicative of abuse. CONCLUSION: Neurochemical, preclinical, and early clinical studies predicted and supported a lack of abuse potential of atomoxetine, which is consistent with the clinical trial and postmarketing spontaneous event data in the past 10 years.


Subject(s)
Adrenergic Uptake Inhibitors/adverse effects , Adrenergic Uptake Inhibitors/pharmacology , Attention Deficit Disorder with Hyperactivity/drug therapy , Propylamines/adverse effects , Propylamines/pharmacology , Substance-Related Disorders/physiopathology , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Atomoxetine Hydrochloride , Behavior, Addictive/metabolism , Drug-Seeking Behavior/drug effects , Humans , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Propylamines/therapeutic use , Protein Binding/physiology , Receptors, Neurotransmitter/metabolism
5.
Neuropharmacology ; 66: 89-98, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22313530

ABSTRACT

Group II metabotropic glutamate (mGlu) receptor agonists were efficacious in randomized clinical research trials for schizophrenia and generalized anxiety disorder. The regional quantification of mGlu(2) and mGlu(3) receptors remains unknown. A selective and structurally novel mGlu(2/3) receptor agonist, 2-amino-4-fluorobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY459477) was tritiated and the distribution of mGlu(2) and mGlu(3) receptors was studied in transgenic mice lacking either mGlu(2), mGlu(3) or both receptors. LY459477 is an agonist with 1-2 nM potency for rodent and human mGlu(2) and mGlu(3) receptors. The functional selectivity of LY459477 was demonstrated by over 640-fold selectivity and the displacement binding selectivity was greater than 320-fold for all glutamate receptors except mGlu(6) (∼230-fold). More than 1000-fold selectivity was demonstrated for all non-glutamate receptors known to be targeted by antipsychotic drugs. Like atypical antipsychotic drugs, LY459477 reversed in vitro electrophysiological effects of a serotonergic hallucinogen and behavioral effects of phencyclidine or amphetamine. There was virtually no binding of [(3)H]LY459477 to any brain region in mice with a deletion of both mGlu(2) and mGlu(3) receptors. Regions enriched in mGlu(2) receptors included the medial prefrontal cortex, select hippocampal regions, the medial mammillary nucleus, the medial habenula, and the cerebellar granular cell layer. Regions enriched in mGlu(3) receptors were the dorsolateral entorhinal cortex, the hippocampal CA1 field, the piriform cortex, the substantia nigra, the thalamic reticular nucleus, and primary sensory thalamic nuclei. These findings suggest [(3)H]LY459477 should be a useful tool to further define the role of mGlu(2) and mGlu(3) receptors throughout the brain with respect to major neuropsychiatric syndromes. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Subject(s)
Amino Acids, Dicarboxylic , Brain/metabolism , Bridged Bicyclo Compounds , Excitatory Amino Acid Agonists , Receptors, Metabotropic Glutamate/metabolism , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Radioligand Assay/methods , Rats , Receptors, Metabotropic Glutamate/genetics , Tritium
6.
J Neurosci ; 31(46): 16507-16, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090477

ABSTRACT

According to the amyloid cascade hypothesis, cerebral deposition of amyloid-ß peptide (Aß) is critical for Alzheimer's disease (AD) pathogenesis. Aß generation is initiated when ß-secretase (BACE1) cleaves the amyloid precursor protein. For more than a decade, BACE1 has been a prime target for designing drugs to prevent or treat AD. However, development of such agents has turned out to be extremely challenging, with major hurdles in cell penetration, oral bioavailability/metabolic clearance, and brain access. Using a fragment-based chemistry strategy, we have generated LY2811376 [(S)-4-(2,4-difluoro-5-pyrimidin-5-yl-phenyl)-4-methyl-5,6-dihydro-4H-[1,3]thiazin-2-ylamine], the first orally available non-peptidic BACE1 inhibitor that produces profound Aß-lowering effects in animals. The biomarker changes obtained in preclinical animal models translate into man at doses of LY2811376 that were safe and well tolerated in healthy volunteers. Prominent and long-lasting Aß reductions in lumbar CSF were measured after oral dosing of 30 or 90 mg of LY2811376. This represents the first translation of BACE1-driven biomarker changes in CNS from preclinical animal models to man. Because of toxicology findings identified in longer-term preclinical studies, this compound is no longer progressing in clinical development. However, BACE1 remains a viable target because the adverse effects reported here were recapitulated in LY2811376-treated BACE1 KO mice and thus are unrelated to BACE1 inhibition. The magnitude and duration of central Aß reduction obtainable with BACE1 inhibition positions this protease as a tractable small-molecule target through which to test the amyloid hypothesis in man.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Adult , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Aspartic Acid Endopeptidases/analysis , Cells, Cultured , Cerebral Cortex/cytology , Crystallography/methods , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/blood , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Models, Chemical , Mutation/genetics , Peptide Fragments/cerebrospinal fluid , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Thiazines/pharmacology , Thiazines/therapeutic use , Time Factors , Young Adult
7.
Cephalalgia ; 30(10): 1159-69, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20855361

ABSTRACT

INTRODUCTION: Lasmiditan (also known as COL-144 and LY573144; 2,4,6-trifluoro-N-[6-[(1-methylpiperidin-4-yl)carbonyl]pyridin-2yl]benzamide) is a high-affinity, highly selective serotonin (5-HT) 5-HT(1F) receptor agonist. RESULTS: In vitro binding studies show a K(i) value of 2.21 nM at the 5-HT(1F) receptor, compared with K(i) values of 1043 nM and 1357 nM at the 5-HT(1B) and 5-HT(1D) receptors, respectively, a selectivity ratio greater than 470-fold. Lasmiditan showed higher selectivity for the 5-HT(1F) receptor relative to other 5-HT(1) receptor subtypes than the first generation 5-HT(1F) receptor agonist LY334370. Unlike the 5-HT(1B/1D) receptor agonist sumatriptan, lasmiditan did not contract rabbit saphenous vein rings, a surrogate assay for human coronary artery constriction, at concentrations up to 100 µM. In two rodent models of migraine, oral administration of lasmiditan potently inhibited markers associated with electrical stimulation of the trigeminal ganglion (dural plasma protein extravasation, and induction of the immediate early gene c-Fos in the trigeminal nucleus caudalis). CONCLUSIONS: Lasmiditan presents a unique pyridinoyl-piperidine scaffold not found in any other antimigraine class. Its chemical structure and pharmacological profile clearly distinguish it from the triptans. The potency and selectivity of lasmiditan make it ideally suited to definitively test the involvement of 5-HT(1F) receptors in migraine headache therapy.


Subject(s)
Benzamides/pharmacology , Indoles/pharmacology , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Animals , Humans , Male , Muscle, Smooth, Vascular/drug effects , Rabbits , Rats , Rats, Wistar , Receptors, Serotonin/metabolism , Trigeminal Ganglion/drug effects , Receptor, Serotonin, 5-HT1F
8.
Bioorg Med Chem Lett ; 17(20): 5600-4, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17804228

ABSTRACT

Analogues of pindolol, 1-(1H-indol-4-yloxy)-3-isopropylamino-propan-2-ol, were synthesized and evaluated as 5-HT(1A) receptor antagonists. The structural features required for optimal binding to the 5-HT1A receptor are as follows: S-2-propanol linker, 4-indoloxy substituent, and a large lipophilic cyclic amine substituent.


Subject(s)
Indoles/chemistry , Propanolamines/chemistry , Propanolamines/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Antagonists , Animals , Corticosterone/blood , Hydroxylation , Isomerism , Male , Molecular Structure , Oxidation-Reduction , Propanolamines/chemical synthesis , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
J Pharmacol Exp Ther ; 321(1): 308-17, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17204749

ABSTRACT

Group II metabotropic glutamate (mGlu) receptor agonists, including (1S,2S,5R,6S)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylate monohydrate (LY354740) and (-)-2-oxa-4-aminobicyclo[3.1.0]hexane-4,6-dicarboxylate (LY379268), have demonstrated efficacy in animal models of anxiety and schizophrenia, and LY354740 decreased anxiety in human subjects. Herein, we report the in vitro pharmacological profile and pharmacokinetic properties of another potent, selective, and structurally novel mGlu2/3 receptor agonist, (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039) and provide comparisons with LY354740. Similar to LY354740, LY404039 is a nanomolar potent agonist at recombinant human mGlu2 and mGlu3 receptors (K(i) = 149 and 92, respectively) and in rat neurons expressing native mGlu2/3 receptors (Ki = 88). LY404039 is highly selective for mGlu2/3 receptors, showing more than 100-fold selectivity for these receptors, versus ionotropic glutamate receptors, glutamate transporters, and other receptors targeted by known anxiolytic and antipsychotic medications. Functionally, LY404039 potently inhibited forskolin-stimulated cAMP formation in cells expressing human mGlu2 and mGlu3 receptors. Electrophysiological studies indicated that LY404039 suppressed electrically evoked excitatory activity in the striatum, and serotonin-induced l-glutamate release in the prefrontal cortex; effects reversed by LY341495. These characteristics suggest LY404039 modulates glutamatergic activity in limbic and forebrain areas relevant to psychiatric disorders; and that, similar to LY354740, it works through a mechanism that may be devoid of negative side effects associated with current antipsychotics and anxiolytics. Interestingly, despite the slightly lower potency (approximately 2-5-fold) of LY404039 versus LY354740 in binding, functional, and electrophysiological assays, LY404039 demonstrated higher plasma exposure and better oral bioavailability in pharmacokinetic experiments. Collectively, the current data indicate that LY404039 may be valuable in the treatment of neuropsychiatric disorders, including anxiety and psychosis.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cyclic S-Oxides/pharmacology , Cyclic S-Oxides/pharmacokinetics , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/pharmacokinetics , Receptors, Metabotropic Glutamate/drug effects , Animals , Binding Sites , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacokinetics , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line , Cerebral Cortex/drug effects , Colforsin/pharmacology , Cyclic AMP/biosynthesis , Cyclic S-Oxides/chemistry , Electric Stimulation , Excitatory Amino Acid Agonists/chemistry , Excitatory Postsynaptic Potentials/drug effects , Humans , Hydrolysis , In Vitro Techniques , Male , Phosphatidylinositols/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Serotonin/pharmacology , Serotonin Antagonists/pharmacology
10.
J Pharmacol Exp Ther ; 318(2): 772-81, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16690725

ABSTRACT

The excitatory neurotransmitter glutamate has been implicated in both migraine and persistent pain. The identification of the kainate receptor GLU(K5) in dorsal root ganglia, the dorsal horn, and trigeminal ganglia makes it a target of interest for these indications. We examined the in vitro and in vivo pharmacology of the competitive GLU(K5)-selective kainate receptor antagonist LY466195 [(3S,4aR,6S,8aR)-6-[[(2S)-2-carboxy-4,4-difluoro-1-pyrrolidinyl]-methyl]decahydro-3-isoquinolinecarboxylic acid)], the most potent GLU(K5) antagonist described to date. Comparisons were made to the competitive GLU(K5)/alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor antagonist LY293558 [(3S,4aR,6R,8aR)-6-[2-(1(2)H-tetrazole-5-yl)ethyl]-decahydroisoquinoline-3-carboxylic acid], other decahydroisoquinoline GLU(K5) receptor antagonists, and the noncompetitive AMPA receptor antagonist LY300168 [1-(4-aminophenyl)-4-methyl-7,8-methylenedioxy-5H-2,3-benzodi-azepine]. When characterized electrophysiologically in rat dorsal root ganglion neurons, LY466195 antagonized kainate (30 microM)-induced currents with an IC50 value of 0.045 +/- 0.011 microM. In HEK293 cells transfected with GLU(K5), GLU(K2)/GLU(K5), or GLU(K5)/GLU(K6) receptors, LY466195 produced IC50 values of 0.08 +/- 0.02, 0.34 +/- 0.17, and 0.07 +/- 0.02 microM, respectively. LY466195 was efficacious in a dural plasma protein extravasation (PPE) model of migraine with an ID100 value of 100 microg/kg i.v. LY466195 was also efficacious in the c-fos migraine model, with a dose of 1 microg/kg i.v. significantly reducing the number of Fos-positive cells in the rat nucleus caudalis after electrical stimulation of the trigeminal ganglion. Furthermore, LY466195 showed no contractile activity in the rabbit saphenous vein in vitro. The diethyl ester prodrug of LY466195 was also efficacious in the same PPE and c-fos models after oral administration at doses of 10 and 100 microg/kg, respectively while having no N-methyl-D-aspartate antagonist-like behavioral effects at oral doses up to 100 mg/kg.


Subject(s)
Isoquinolines/pharmacology , Receptors, Kainic Acid/antagonists & inhibitors , Animals , Benzodiazepines/pharmacology , Binding, Competitive/drug effects , Blood Proteins/metabolism , Calcium/metabolism , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Humans , In Vitro Techniques , Ligands , Male , Migraine Disorders/metabolism , Motor Activity/drug effects , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Neurons/drug effects , Neurons/metabolism , Phencyclidine/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Rabbits , Rats , Receptors, AMPA/antagonists & inhibitors , Saphenous Vein/cytology , Saphenous Vein/drug effects , Transfection
11.
Psychopharmacology (Berl) ; 185(2): 240-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16470401

ABSTRACT

RATIONALE: Although convergent evidence exists for a role of glutamate in the regulation of anxiety, the involvement of specific glutamate receptor subtypes has yet to be defined. OBJECTIVE: To evaluate the potential for blockade of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)/kainate receptors to produce anxioltyic-like effects with the AMPA/GLU(K5) (kainate) antagonist (3S,4aR,6R,8aR)-6-[2-(1(2)H-tetrazole-5yl)ethyl]decahydroisoquinoline-3carboxylic acid (LY293558) MATERIALS AND METHODS: Punished responding of rats was used to determine the efficacy of LY293558. Other in vivo and in vitro studies further characterized the specificity of LY293558 for AMPA/kainate receptors. RESULTS: LY293558 had a rank order of potency of GLU(K5) > or = GLU(K5/6) approximately GLU(A2i) approximately GLU(K2/5) approximately GLU(A1i) approximately GLU(A2o) approximately GLU(A3i) approximately GLU(A1o) > or = GLU(A3o) > or = GLU(A4i) approximately GLU(A4o) and >100 microM affinity for rat cortical GABA(A) receptors. Comparison of the blockade of AMPA- vs N-methyl-D-aspartate (NMDA)-induced inward currents demonstrated that LY293558 was five-fold more potent as an antagonist at AMPA vs NMDA receptors in vitro. In keeping with the low affinity of LY293558 for NMDA receptors, LY293558 was not effective in preventing NMDA-induced seizures in mice. LY293558 increased punished responding, a preclinical predictor of anxiolytic efficacy, at a dose that decreased unpunished responding (10 mg/kg, i.p.). Chlordiazepoxide produced comparable increases in both punished and unpunished responding. The NMDA antagonist dizocilpine [(+)-MK-801] also increased both punished and unpunished responding. CONCLUSIONS: These data along with those in the literature suggest that AMPA and/or kainate receptor blockade may be an important component to producing anxiolytic-like effects and may therefore be a target for compounds with efficacy in the therapeutic treatment of anxiety disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , Carboxylic Acids/pharmacology , Isoquinolines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Tetrazoles/pharmacology , Animals , Cells, Cultured , Chlordiazepoxide/pharmacology , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Eating/drug effects , Humans , In Vitro Techniques , Male , N-Methylaspartate , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Protein Subunits/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, Kainic Acid/physiology , Seizures/chemically induced , Seizures/drug therapy
12.
J Pharmacol Exp Ther ; 315(3): 1265-77, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16141369

ABSTRACT

FMPD [6-fluoro-10-[3-(2-methoxyethyl)-4-methyl-piperazin-1-yl]-2-methyl-4H-3-thia-4,9-diaza-benzo[f]azulene] is a potential novel antipsychotic with high affinity for dopamine D2 (Ki= 6.3 nM), 5-HT(2A) (Ki= 7.3 nM), and 5-HT6 (Ki= 8.0 nM) human recombinant receptors and lower affinity for histamine H1 (Ki= 30 nM) and 5-HT2C (Ki= 102 nM) human recombinant receptors than olanzapine. Oral administration of FMPD increased rat nucleus accumbens 3,4-dihyroxyphenylacetic acid concentrations (ED200 = 6 mg/kg), blocked 5-HT2A agonist-induced increases in rat serum corticosterone levels (ED50= 1.8 mg/kg), and inhibited the ex vivo binding of [125I]SB-258585 [4-iodo-N-[4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyl]-benzenesulfonamide] to striatal 5-HT6 receptors (ED50= 10 mg/kg) but failed to inhibit ex vivo binding of [3H]pyrilamine to hypothalamic histamine H1 receptors at doses of up to 30 mg/kg. In electrophysiology studies, acute administration of FMPD selectively elevated the number of spontaneously active A10 (versus A9) dopamine neurons and chronic administration selectively decreased the number of spontaneously active A10 (versus A9) dopamine neurons. FMPD did not produce catalepsy at doses lower than 25 mg/kg p.o. In Fos-induction studies, FMPD had an atypical antipsychotic profile in the striatum and nucleus accumbens and increased Fos expression in orexin-containing neurons of the hypothalamus. FMPD produced only a transient elevation of prolactin levels. These data indicate that FMPD is an orally available potent antagonist of dopamine D2, 5-HT2A, and 5-HT6 receptors and a weak antagonist of H1 and 5-HT2C receptors. FMPD has the potential to have efficacy in treating schizophrenia and bipolar mania with a low risk of treatment-emergent extrapyramidal symptoms, prolactin elevation, and weight gain. Clinical trials are needed to test these hypotheses.


Subject(s)
Antipsychotic Agents/pharmacology , Piperazines/pharmacology , Receptors, Histamine H1/metabolism , 3,4-Dihydroxyphenylacetic Acid/analysis , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Antipsychotic Agents/metabolism , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Body Weight/drug effects , Catalepsy/chemically induced , Cocaine/pharmacology , Corticosterone/blood , Drug Evaluation, Preclinical , Electrochemistry , Electrophysiology , Fasting , Female , Immunohistochemistry , Male , Molecular Structure , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Olanzapine , Piperazines/chemistry , Piperazines/metabolism , Prolactin/blood , Quipazine/pharmacology , Radioligand Assay , Rats , Rats, Inbred F344 , Rats, Inbred Strains , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology , Thiophenes/therapeutic use , Time Factors
13.
Article in English | MEDLINE | ID: mdl-14642972

ABSTRACT

The interactions of the atypical antipsychotic drugs (APD) clozapine, olanzapine, risperidone, quetiapine and ziprasidone with muscarinic receptors were reviewed. Only clozapine and olanzapine have marked affinity for muscarinic receptors in radioligand binding studies; however, the affinity of these compounds is considerably lower than classical muscarinic antagonists. Although functional assays in cell lines transfected with muscarinic receptors suggest that olanzapine and clozapine have weak partial agonist activity at muscarinic receptors, particularly M4 receptors, studies in vitro and in vivo indicate that the compounds function as antagonists. In animal studies and in humans, clozapine has pronounced antimuscarinic effects whereas olanzapine has weak antimuscarinic effects. However, olanzapine significantly occupies central muscarinic receptors in humans. Overall, the role of muscarinic receptors in the antipsychotic effects of clozapine and olanzapine is controversial and complex.


Subject(s)
Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Cholinergic Agents/pharmacology , Psychotic Disorders/drug therapy , Receptors, Muscarinic/metabolism , Acetylcholine/metabolism , Animals , Antipsychotic Agents/chemistry , Behavior/drug effects , Binding, Competitive , Dose-Response Relationship, Drug , Drug Interactions , Humans , Kinetics , Models, Animal , Neural Inhibition/drug effects , Receptors, Muscarinic/classification , Receptors, Muscarinic/drug effects , Second Messenger Systems/drug effects
14.
J Biol Chem ; 278(46): 46107-16, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-12949072

ABSTRACT

Functional gamma-secretase inhibitors (FGSIs) can block the cleavage of several transmembrane proteins including amyloid precursor protein (APP), and the cell fate regulator Notch-1. FGSIs, by inhibiting APP processing, block the generation of amyloid beta (Abeta) peptides and may slow the development of Alzheimer's disease. FGSIs used to inhibit APP processing may disrupt Notch processing, thus interfering with cell fate determination. Described herein is a FGSI-mediated gastrointestinal toxicity characterized by cell population changes in the ileum of rats, which are indicative of Notch signaling disruption. Microarray analysis of ileum from FGSI-treated rats revealed differential expression responses in a number of genes indicative of Notch signaling perturbation, including the serine protease adipsin. We were able to show that FGSI-treated rats had elevated levels of adipsin protein in gastrointestinal contents and feces, and by immunohistochemistry demonstrated that adipsin containing ileum crypt cells were increased in FGSI-treated rats. The mouse Adipsin proximal promoter contains a putative binding site for the Notch-induced transcriptional regulator Hes-1, which we demonstrate is able to bind Hes-1. Additional studies in 3T3-L1 preadipocytes demonstrate that this FGSI inhibits Hes-1 expression while up-regulating adipsin expression. Overexpression of Hes-1 was able to down-regulate adipsin expression and block pre-adipocyte differentiation. We propose that adipsin is a Hes-1-regulated gene that is de-repressed during FGSI-mediated disruption of Notch/Hes-1 signaling. Additionally, the aberrant expression of adipsin, and its presence in feces may serve as a noninvasive biomarker of gastrointestinal toxicity associated with perturbed Notch signaling.


Subject(s)
Amyloid beta-Peptides/chemistry , Digestive System/metabolism , Endopeptidases/metabolism , Serine Endopeptidases/chemistry , 3T3-L1 Cells , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases , Base Sequence , Binding Sites , Cell Differentiation , Complement Factor D , Down-Regulation , Endopeptidases/chemistry , Enzyme Inhibitors/pharmacology , Ileum/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Models, Chemical , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats , Receptors, Notch , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...