Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Proc Natl Acad Sci U S A ; 105(1): 33-8, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18162540

ABSTRACT

Rapamycin is an immunosuppressive immunophilin ligand reported as having neurotrophic activity. We show that modification of rapamycin at the mammalian target of rapamycin (mTOR) binding region yields immunophilin ligands, WYE-592 and ILS-920, with potent neurotrophic activities in cortical neuronal cultures, efficacy in a rodent model for ischemic stroke, and significantly reduced immunosuppressive activity. Surprisingly, both compounds showed higher binding selectivity for FKBP52 versus FKBP12, in contrast to previously reported immunophilin ligands. Affinity purification revealed two key binding proteins, the immunophilin FKBP52 and the beta1-subunit of L-type voltage-dependent Ca(2+) channels (CACNB1). Electrophysiological analysis indicated that both compounds can inhibit L-type Ca(2+) channels in rat hippocampal neurons and F-11 dorsal root ganglia (DRG)/neuroblastoma cells. We propose that these immunophilin ligands can protect neurons from Ca(2+)-induced cell death by modulating Ca(2+) channels and promote neurite outgrowth via FKBP52 binding.


Subject(s)
Calcium Channels/chemistry , Sirolimus/chemistry , Tacrolimus Binding Proteins/chemistry , Animals , Calcium/metabolism , Electrophysiology/methods , Humans , Immunophilins/metabolism , Immunosuppressive Agents/pharmacology , Ligands , Models, Chemical , Neurites/metabolism , Neuroblastoma/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Protein Binding , Rats , Stroke/metabolism
2.
J Pharmacol Exp Ther ; 328(3): 766-76, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19050173

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (nAChR) has been implicated in Alzheimer's disease and schizophrenia, leading to efforts targeted toward discovering agonists and positive allosteric modulators (PAMs) of this receptor. In a Ca2+ flux fluorometric imaging plate reader assay, SB-206553 (3,5-dihydro-5-methyl -N-3-pyridinylbenzo [1, 2-b:4,5 -b']-di pyrrole-1(2H)-carboxamide), a compound known as a 5-hydroxytryptamine(2B/2C) receptor antagonist, produced an 8-fold potentiation of the evoked calcium signal in the presence of an EC(20) concentration of nicotine and a corresponding EC(50) of 1.5 muM for potentiation of EC(20) nicotine responses in GH4C1 cells expressing the alpha7 receptor. SB-206553 was devoid of direct alpha7 receptor agonist activity and selective against other nicotinic receptors. Confirmation of the PAM activity of SB-206553 on the alpha7 nAChR was obtained in patch-clamp electrophysiological experiments in GH4C1 cells, where it failed to evoke any detectable currents when applied alone, yet dramatically potentiated the currents evoked by an EC(20) (17 microM) and EC(100) (124 microM) of acetylcholine (ACh). Native nicotinic receptors in CA1 stratum radiatum interneurons of rat hippocampal slices could also be activated by ACh (200 microM), an effect that was entirely blocked by the alpha7-selective antagonist methyllycaconitine (MLA). These ACh currents were potentiated by SB-206553, which increased the area of the current response significantly, resulting in a 40-fold enhancement at 100 microM. In behavioral experiments in rats, SB-206553 reversed an MK-801 (dizocilpine maleate)-induced deficit in the prepulse inhibition of acoustic startle response, an effect attenuated in the presence of MLA. This latter observation provides further evidence in support of the potential therapeutic utility of alpha7 nAChR PAMs in schizophrenia.


Subject(s)
Indoles/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/physiology , Receptors, Serotonin, 5-HT2/physiology , Serotonin Antagonists/pharmacology , Animals , Cell Line , Electrophysiology , Hippocampus/drug effects , Hippocampus/physiology , Humans , Kidney/embryology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Nicotinic/drug effects , Receptors, Serotonin, 5-HT2/drug effects , Receptors, Serotonin, 5-HT2/genetics , Transfection , alpha7 Nicotinic Acetylcholine Receptor
3.
Psychopharmacology (Berl) ; 203(1): 41-52, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18949460

ABSTRACT

RATIONALE: Acid sensing ion channels (ASICs) are proton-gated ion channels located in the central and peripheral nervous systems. Of particular interest is ASIC1a, which is located in areas associated with fear and anxiety behaviors. Recent reports suggest a role for ASIC1a in preclinical models of fear conditioning and anxiety. OBJECTIVES: The present experiments evaluated various ASIC inhibitors in preclinical models of autonomic and behavioral parameters of anxiety. In addition, neurochemical studies evaluated the effects of an ASIC inhibitor (A-317567) on neurotransmitter levels in the amygdala. RESULTS: In electrophysiological studies using hippocampal primary neuronal cultures, three ASIC inhibitors (PcTX-1, A-317567, and amiloride) produced concentration-dependent inhibition of acid-evoked currents. In the stress-induced hyperthermia model, acute administration of psalmotoxin 1 (PcTX-1; 10-56 ng, i.c.v.), A-317567 (0.1-1.0 mg/kg, i.p.), and amiloride (10-100 mg/kg, i.p.) prevented stress-induced elevations in core body temperature. In the four-plate test, acute treatment with PcTX-1 (10-56 ng, i.c.v.) and A-317567 (0.01-0.1 mg/kg, i.p.), but not amiloride (3-100 mg/kg, i.p.), produced dose-dependent and significant increases in the number of punished crossings relative to vehicle-treated animals. Additionally, PcTX-1 (56-178 ng, i.c.v.), A-317567 (0.1-10 mg/kg, i.p.), and amiloride (10-100 mg/kg, i.p.) lacked significant anxiolytic-like activity in the elevated zero maze. In neurochemical studies, an infusion of A-317567 (100 microM) into the amygdala significantly elevated the extracellular levels of GABA, but not glutamate, in this brain region. CONCLUSIONS: These findings demonstrate that ASIC inhibition produces anxiolytic-like effects in some behavioral models and indicate a potential role for GABAergic mechanisms to underlie these anxiolytic-like effects.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Drug Evaluation, Preclinical , Nerve Tissue Proteins/antagonists & inhibitors , Sodium Channel Blockers/pharmacology , Acid Sensing Ion Channels , Amiloride/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/metabolism , Anxiety/psychology , Behavior, Animal/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Exploratory Behavior/drug effects , Fever/metabolism , Fever/prevention & control , Fever/psychology , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/metabolism , Isoquinolines/pharmacology , Male , Membrane Potentials , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microdialysis , Naphthalenes/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Peptides , Rats , Rats, Sprague-Dawley , Sodium Channels/metabolism , Spider Venoms/pharmacology , Stress, Psychological/complications , Stress, Psychological/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Neuron ; 41(4): 587-98, 2004 Feb 19.
Article in English | MEDLINE | ID: mdl-14980207

ABSTRACT

The family of calcium binding proteins called KChIPs associates with Kv4 family K(+) channels and modulates their biophysical properties. Here, using mutagenesis and X-ray crystallography, we explore the interaction between Kv4 subunits and KChIP1. Two regions in the Kv4.2 N terminus, residues 7-11 and 71-90, are necessary for KChIP1 modulation and interaction with Kv4.2. When inserted into the Kv1.2 N terminus, residues 71-90 of Kv4.2 are also sufficient to confer association with KChIP1. To provide a structural framework for these data, we solved the crystal structures of Kv4.3N and KChIP1 individually. Taken together with the mutagenesis data, the individual structures suggest that that the Kv4 N terminus is required for stable association with KChIP1, perhaps through a hydrophobic surface interaction, and that residues 71-90 in Kv4 subunits form a contact loop that mediates the specific association of KChIPs with Kv4 subunits.


Subject(s)
Calcium-Binding Proteins/chemistry , Cell Membrane/chemistry , Potassium Channels, Voltage-Gated , Potassium Channels/chemistry , Amino Acid Sequence/physiology , Animals , Binding Sites/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Kv Channel-Interacting Proteins , Membrane Potentials/genetics , Models, Molecular , Mutagenesis, Site-Directed/genetics , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Protein Subunits , Shal Potassium Channels
5.
Assay Drug Dev Technol ; 6(2): 181-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18471073

ABSTRACT

Heterologous functional expression of alpha7 nicotinic acetylcholine receptors (nAChRs) is difficult to achieve in mammalian cell lines, and the reasons have been associated with a lack of expression of the putative chaperone factor RIC-3. Here, we describe the generation and functional and pharmacological characterization of a Chinese hamster ovary (CHO)-K1 cell line co-expressing the human alpha7 nAChR and RIC-3. Stable recombinant cells expressing alpha7 nAChR on the plasma membrane were selected by binding of fluorochrome-conjugated alpha-bungarotoxin and fluorescence-activated cell sorting. The presence of functional alpha7 channels was demonstrated by whole cell patch clamp recordings. Nicotine and acetylcholine induced rapid desensitizing currents with 50% effective concentration values of 14 and 37 microM, respectively, with agonist-evoked currents detected in approximately 75% of the cell population. Surprisingly, when tested in a FLIPR (Molecular Devices, Sunnyvale, CA) Ca(2+) assay, activation of alpha7 nAChRs was measured only when nicotinic agonists were applied either in the presence of the positive allosteric modulator (PAM) PNU-120596 or after pretreatment of cells with the tyrosine kinase inhibitor genistein. No Ca(2+) influx was measured upon addition of agonists alone or together with allosteric potentiators such as 5-hydroxyindole that predominantly increase the apparent peak amplitude without robustly affecting the current desensitization rate, as exemplified by PNU-120596. These results show that functional alpha7 nAChRs can stably be expressed in the non-neuronal CHO-K1 cell line. This recombinant cell system is useful for characterization of alpha7 nAChRs and to study the mechanism of action of chemical modulators, in particular the detection of PAMs capable of slowing receptor desensitization kinetics.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Nicotinic/physiology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Electrophysiology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Genistein/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Isoxazoles/pharmacology , Kinetics , Microscopy, Fluorescence , Nicotinic Agonists/pharmacology , Phenylurea Compounds/pharmacology , RNA/biosynthesis , RNA/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection , alpha7 Nicotinic Acetylcholine Receptor
6.
Bioorg Med Chem ; 16(6): 3067-75, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18226531

ABSTRACT

Kv1.1 channels are expressed in many regions of the brain and spinal cord [Monaghan, M. M.; Trimmer, J. S.; Rhodes, K. J. J. Neurosci.2001, 21, 5973; Rasband, M. N.; Trimmer, J. S. J. Comp. Neurol.2001, 429, 166; Trimmer, J. S.; Rhodes, K. J. Ann. Rev. Physiol.2004, 66, 477]. When expressed alone, they produce a delayed rectifier slowly inactivating type current that contributes to hyperpolarizing the neuron following depolarization. In the hippocampus Kv1.1 is co-expressed with Kvbeta1 (and other beta subunits), which converts Kv1.1 into a transient, fast inactivating current, reducing its ability to hyperpolarize the cell and thus increasing neuronal excitability. To reduce neuronal excitability, screening for compounds that prevent inactivation of Kv1.1 channels by Kvbeta1 was performed using a yeast two-hybrid screen. A variety of compounds were discovered in this assay and subsequently determined to disrupt inactivation of the ionic currents, and hence were termed 'disinactivators'. Several of these disinactivators also inhibited pentylenetetrazole-induced seizures (PTZ) in mice. Compounds were found to act by several mechanisms to prevent Kvbeta1 inactivation of Kv1.1 channels, including enhancement of Ca(2+) release/influx and by direct mechanisms. Two structural classes were identified that act on a Kvbeta1N70-Kv1.1 chimera where the N-terminal 70 amino acids of Kvbeta1 were attached to the N-terminus of Kv1.1. It is likely that these disinactivators act directly on the Kvbeta1 N-terminus or its receptor site on Kv1.1, thus preventing it from blocking Kv1.1 channels. Compounds acting by this mechanism may be useful for reducing neuronal hyperexcitability in diseases such as epilepsy and neuropathic pain.


Subject(s)
Kv1.1 Potassium Channel/drug effects , Organic Chemicals/pharmacology , Small Molecule Libraries , Animals , Calcium/metabolism , Membrane Potentials/drug effects , Mice , Seizures/prevention & control , Structure-Activity Relationship , Two-Hybrid System Techniques
7.
Sci Rep ; 8(1): 13438, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30194389

ABSTRACT

Protein interacting with C kinase (PICK1) is a scaffolding protein that is present in dendritic spines and interacts with a wide array of proteins through its PDZ domain. The best understood function of PICK1 is regulation of trafficking of AMPA receptors at neuronal synapses via its specific interaction with the AMPA GluA2 subunit. Disrupting the PICK1-GluA2 interaction has been shown to alter synaptic plasticity, a molecular mechanism of learning and memory. Lack of potent, selective inhibitors of the PICK1 PDZ domain has hindered efforts at exploring the PICK1-GluA2 interaction as a therapeutic target for neurological diseases. Here, we report the discovery of PICK1 small molecule inhibitors using a structure-based drug design strategy. The inhibitors stabilized surface GluA2, reduced Aß-induced rise in intracellular calcium concentrations in cultured neurons, and blocked long term depression in brain slices. These findings demonstrate that it is possible to identify potent, selective PICK1-GluA2 inhibitors which may prove useful for treatment of neurodegenerative disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Carrier Proteins/antagonists & inhibitors , Dendritic Spines/metabolism , Neurodegenerative Diseases/metabolism , Nuclear Proteins/antagonists & inhibitors , Synapses/metabolism , Animals , Brain/pathology , Calcium/metabolism , Calcium Signaling , Carrier Proteins/metabolism , Cell Cycle Proteins , Dendritic Spines/pathology , Drug Design , Mice , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Nuclear Proteins/metabolism , PDZ Domains , Receptors, AMPA/metabolism , Synapses/pathology
8.
J Biomol Screen ; 12(8): 1059-67, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18087070

ABSTRACT

Enhancers of KCNQ channels are known to be effective in chronic pain models. To discover novel enhancers of KCNQ channels, the authors developed a medium-throughput electrophysiological assay by using the IonWorks platform. Screening of 20 CHO-K1 clones stably expressing KCNQ2/3 was performed on the IonWorks HT until the best clone (judged from seal rate, current level, and stability) was obtained. The KCNQ2/3 current amplitude in the cells was found to increase from 60 +/- 15 pA to 473 +/- 80 pA (at -10 mV), and the expression rate was increased by 56% when the cells were incubated at 27 degrees C overnight. The clone used for compound screening had a seal rate of greater than 90% and an overall success rate of greater than 70%. The voltage step protocol (hold cells at -80 mV and depolarize to -10 mV for 1 s) was designed to provide moderate current but still allow for pharmacological current enhancement. EC(50)s were generated from 8-point concentration-response curves with a control compound on each plate using compounds that were also tested with conventional patch clamp. The authors found that there was a very good correlation (R(2) > 0.9) between the 2 assays, thus demonstrating the highly predictive nature of the IonWorks assay.


Subject(s)
Electrophysiology/instrumentation , Electrophysiology/methods , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Rats , Reproducibility of Results , Rubidium/metabolism , Transfection
9.
Assay Drug Dev Technol ; 4(4): 443-50, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16945016

ABSTRACT

The slow delayed rectifier K+ current, Iks, encoded by KCNQ1 (KvLQT1)/KCNE1 (mink) genes, contributes to cardiac action potential repolarization and determines the heartbeat rate. Mutations in either KCNQ1 or KCNE1 that reduce Iks cause long-QT syndrome (LQTS), a disorder of ventricular repolarization that results in cardiac arrhythmia and sudden death. A well-recognized potential treatment for LQTS caused by reduction of Iks is to enhance functional activation of cardiac KCNQ1/KCNE1 channels. In the present study, we generated a stable Chinese hamster ovary cell line that expresses KCNQ1/KCNE1 channels confirmed by electrophysiology. Using a pharmacological tool compound R-L3 (L-364,373 [(3-R)-1,3-dihydro-5-(2-fluorophenyl)-3-(1H-indol- 3-ylmethyl)-1-methyl-2H-1,4-benzodiazepin-2-one]), which activates KCNQ1/mink channels, we then developed and validated a non-radioactive rubidium (Rb+) efflux assay that directly measures the functional activity of KCNQ1/KCNE1 channels by atomic absorption spectroscopy. Our results show that the validated Rb+ efflux assay can be used for screening of KCNQ1/KCNE1 openers that potentially treat LQTS in both inherited and acquired forms. In addition, the assay also can be used for evaluation of possible long-QT liability during cardiac selectivity of new chemical entities.


Subject(s)
Benzodiazepines/pharmacology , KCNQ1 Potassium Channel/physiology , Potassium Channels, Voltage-Gated/physiology , Rubidium/metabolism , Animals , Anthracenes/pharmacology , CHO Cells , Chromans/pharmacology , Cricetinae , Cyclic Nucleotide-Gated Cation Channels , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Gene Expression/drug effects , Gene Expression/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Ion Channels/physiology , KCNQ1 Potassium Channel/antagonists & inhibitors , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocardium/metabolism , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/genetics , Potassium Chloride/pharmacology , Rats , Reproducibility of Results , Risk Factors , Spectrophotometry, Atomic/methods , Tetraethylammonium/pharmacology
10.
Assay Drug Dev Technol ; 4(1): 49-56, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16506888

ABSTRACT

F11 cells are derived from a fusion between mouse embryonic neuroblastoma and rat dorsal root ganglion (DRG) neurons. These cells have been shown to retain many features of native DRG neurons, including synthesis of neurotransmitters, expression of neuropeptide receptors, and voltage-gated calcium channels. In this study, we describe the presence of KCNQ2/3 channels in F11 cells as determined by both reverse transcription-polymerase chain reaction and functional assessment. Electrophysiological recordings in whole-cell configuration performed in F11 cells revealed the functional expression of a KCNQ/M-current with characteristic slow deactivation kinetics, similar to the KCNQ/M-current recorded from dissociated DRG neurons. Deactivation tail currents elicited by conventional M-current protocols were enhanced by a specific KCNQ/M-channel opener, WAY-1, and inhibited by the specific blocker XE991 [10,10-bis(4-pyridinylmethyl)-9(10H)- anthracenone]. Using a non-radioactive atomic absorption Rb+ efflux assay, we further validated that Rb+ efflux can be induced in differentiated F11 cells by activation of KCNQ/M-channels. These findings have led us to conclude that F11 cells can be used as a DRG cell model to evaluate effects of KCNQ/M-channel modulators.


Subject(s)
Cyanates/pharmacology , Ganglia, Spinal/physiology , KCNQ2 Potassium Channel/physiology , Neurons, Afferent/physiology , Animals , DNA Primers , Elapid Venoms/pharmacology , KCNQ2 Potassium Channel/drug effects , KCNQ2 Potassium Channel/genetics , Kinetics , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Rats , Rubidium/metabolism , Tetraethylammonium/pharmacology
11.
Assay Drug Dev Technol ; 2(5): 525-34, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15671650

ABSTRACT

M-channels (M-current), encoded by KCNQ2/3 K(+) channel genes, have emerged as novel drug targets for a number of neurological disorders. The lack of direct high throughput assays combined with the low throughput of conventional electrophysiology (EP) has impeded rapid screening and evaluation of K(+)-channel modulators. Development of a sensitive and efficient assay for the direct measurement of M-current activity is critical for identifying novel M-channel modulators and subsequent investigation of their therapeutic potential. Using a stable CHO cell line expressing rat KCNQ2/3 K(+) channels confirmed by EP, we have developed and validated a nonradioactive rubidium (Rb(+)) efflux assay in a 96-well plate format. The Rb(+) efflux assay directly measures the activity of functional channels by atomic absorption spectroscopy using the automated Ion Channel Reader (ICR) 8000. The stimulated Rb(+) efflux from KCNQ2/3-expressing cells was blocked by the channel blockers XE991 and linopirdine with IC(50) values of 0.15 microM and 1.3 microM, respectively. Twelve compounds identified as KCNQ2/3 openers were further assessed in this assay, and their EC(50) values were compared with those obtained with EP. A higher positive correlation coefficient between these two assays (r = 0.60) was observed than that between FlexStation membrane potential and EP assays (r = 0.23). To simplify the assay and increase the throughput, we demonstrate that EC(50) values obtained by measuring Rb(+) levels in the supernatant are as robust and consistent as those obtained from the ratio of Rb(+) in supernatant/lysate. By measuring the supernatant only, the throughput of ICR8000 in an eight-point titration is estimated to be 40 compounds per day, which is suitable for a secondary confirmation assay.


Subject(s)
Potassium Channels, Voltage-Gated/metabolism , Rubidium/metabolism , Spectrophotometry, Atomic/methods , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Large-Conductance Calcium-Activated Potassium Channels , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Rats
12.
Neurochem Int ; 45(2-3): 273-83, 2004.
Article in English | MEDLINE | ID: mdl-15145543

ABSTRACT

Medium conditioned by cultured hippocampal glial contains an inhibitory factor that can hyperpolarize and suppress neuronal activity. Using biochemistry, electrophysiology, pharmacology, and mass spectrometry, we have identified the inhibitory factor as GABA (gamma-aminobutyric acid). Like GABA, the inhibitory factor increases chloride and potassium currents in neurons, which can be blocked by bicuculline. Mass spectrometry analysis of conditioned medium reveals peaks that are identical to that for GABA. Up to 500 micromolar GABA is found in conditioned medium from glial cultures. No GABA is found in conditioned medium from neuronal cultures. Hippocampal glia make much more GABA than cortical glia or glia from other brain regions. It is not clear how hippocampal glia synthesize GABA. Although they express GAD mRNA and adding glutamate to the culture medium increases the amount of GABA produced, other data suggest that glia do not use GAD to make GABA. Identifying the mechanism(s) by which GABA is produced by hippocampal glia would help clarify its role in modulating neuronal activity in the brain.


Subject(s)
Hippocampus/physiology , Neuroglia/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cells, Cultured , Chloride Channels/physiology , Culture Media, Conditioned , Mass Spectrometry , Neurons/physiology , Potassium Channels/physiology , Rats , Synapses/physiology
13.
J Med Chem ; 55(22): 10277-81, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23083093

ABSTRACT

α7 Nicotinic acetylcholine receptors (α7 nAChR) represent promising therapeutic candidates for the treatment of cognitive impairment associated with Alzheimer's disease (AD) and schizophrenia. A medicinal chemistry effort around previously reported compound 1 (SEN15924, WAY-361789) led to the identification of 12 (SEN78702, WYE-308775) a potent and selective full agonist of the α7 nAChR that demonstrated improved plasma stability, brain levels, and efficacy in behavioral cognition models.


Subject(s)
Brain/drug effects , Cognition/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Nicotinic Agonists/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Nicotinic/chemistry , Animals , CHO Cells , Calcium/metabolism , Chemistry, Pharmaceutical , Cricetinae , ERG1 Potassium Channel , Humans , Models, Molecular , Nicotinic Agonists/chemical synthesis , Piperidines/chemical synthesis , Pyrazoles/chemical synthesis , Rats , Receptors, Nicotinic/metabolism , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor
14.
J Med Chem ; 55(10): 4806-23, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22468936

ABSTRACT

Alpha-7 nicotinic acetylcholine receptors (α7 nAChR) are implicated in the modulation of many cognitive functions such as attention, working memory, and episodic memory. For this reason, α7 nAChR agonists represent promising therapeutic candidates for the treatment of cognitive impairment associated with Alzheimer's disease (AD) and schizophrenia. A medicinal chemistry effort, around our previously reported chemical series, permitted the discovery of a novel class of α7 nAChR agonists with improved selectivity, in particular against the α3 receptor subtype and better ADME profile. The exploration of this series led to the identification of 5-(4-acetyl[1,4]diazepan-1-yl)pentanoic acid [5-(4-methoxyphenyl)-1H-pyrazol-3-yl] amide (25, SEN15924, WAY-361789), a novel, full agonist of the α7 nAChR that was evaluated in vitro and in vivo. Compound 25 proved to be potent and selective, and it demonstrated a fair pharmacokinetic profile accompanied by efficacy in rodent behavioral cognition models (novel object recognition and auditory sensory gating).


Subject(s)
Azepines/chemical synthesis , Nicotinic Agonists/chemical synthesis , Pyrazoles/chemical synthesis , Receptors, Nicotinic/metabolism , Administration, Oral , Animals , Azepines/pharmacokinetics , Azepines/pharmacology , Brain/metabolism , Calcium/metabolism , Catalytic Domain , Cell Line , Cell Membrane Permeability , Cognition/drug effects , Dogs , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Humans , Male , Membrane Potentials/drug effects , Models, Molecular , Nicotinic Agonists/pharmacokinetics , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/chemical synthesis , Nicotinic Antagonists/pharmacokinetics , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Radioligand Assay , Rats , Rats, Long-Evans , Reflex, Startle/drug effects , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor
15.
Psychopharmacology (Berl) ; 218(4): 635-47, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21643676

ABSTRACT

RATIONALE: α7 nicotinic acetylcholine receptor (nAChR) agonists are proposed as candidate agents for the adjunctive treatment of cognitive deficits associated with schizophrenia. Despite the pursuit of such an approach clinically, it is surprising that the preclinical profile of pro-cognitive agents in conjunction with antipsychotic drugs is currently unexplored. OBJECTIVES: We determined if the memory-enhancing effects of the selective α7 nAChR agonist WYE-103914 were preserved in the presence of the atypical antipsychotic drug risperidone, and if the antipsychotic-like profile of risperidone was preserved in the presence of WYE-103914. METHODS: Using the rat novel object recognition (NOR) paradigm, the maintenance of memory-enhancing activity of the α7 nAChR agonist WYE-103914 in the presence of risperidone was examined. Similarly, in the standard tests of antipsychotic-like activity, apomorphine-induced climbing (AIC) in mice and conditioned avoidance responding (CAR) in rats, the preservation of antipsychotic-like activity of risperidone was evaluated in the presence of WYE-103914. RESULTS: WYE-103914 exhibited memory-enhancing activity in rat NOR, and this effect of WYE-103914 was retained in the presence of risperidone. In AIC, the atypical antipsychotic profile of risperidone was not significantly altered by WYE-103914. In contrast, WYE-103914 moderately potentiated the efficacy profile of risperidone in CAR, an effect that did not appear to be convincingly linked to a pharmacokinetic interaction. CONCLUSIONS: These data underscore the value of a preclinical evaluation of the adjunctive profile of a memory-enhancing agent in combination with antipsychotics and provide further support to augmentation with α7 nAChR agonists to address the cognitive deficits associated with schizophrenia.


Subject(s)
Cognition Disorders/drug therapy , Pyridines/pharmacology , Risperidone/pharmacology , Schizophrenia/drug therapy , Urea/analogs & derivatives , Animals , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Cognition Disorders/etiology , Drug Evaluation, Preclinical , Drug Interactions , Drug Therapy, Combination , Male , Memory/drug effects , Mice , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Schizophrenia/physiopathology , Urea/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
16.
J Med Chem ; 53(11): 4379-89, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20465311

ABSTRACT

Alpha-7 nicotinic acetylcholine receptor (alpha7 nAChR) agonists are promising therapeutic candidates for the treatment of cognitive impairment. We report a series of novel, potent small molecule agonists (4-18) of the alpha7 nAChR deriving from our continuing efforts in the areas of Alzheimer's disease and schizophrenia. One of the compounds of the series containing a urea moiety (16) was further shown to be a selective agonist of the alpha7 nAChR with excellent in vitro and in vivo profiles, brain penetration, and oral bioavailability and demonstrated in vivo efficacy in multiple behavioral cognition models. Structural modifications leading to the improved selectivity profile and the biological evaluation of this series of compounds are discussed.


Subject(s)
Nicotinic Agonists/chemical synthesis , Nicotinic Agonists/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/pharmacology , Administration, Oral , Animals , Humans , Inhibitory Concentration 50 , Male , Models, Molecular , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacokinetics , Protein Conformation , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Rats , Receptors, Nicotinic/chemistry , Structure-Activity Relationship , Substrate Specificity , Urea/administration & dosage , Urea/pharmacokinetics , alpha7 Nicotinic Acetylcholine Receptor
17.
Bioorg Med Chem ; 13(22): 6112-9, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16081294

ABSTRACT

Potassium channels and their associated subunits are important contributors to electrical excitability in many cell types. In this study, a yeast two-hybrid assay was used to identify inhibitors such as a diaryl-urea compound (CL-888) that binds to and modulates the formation of the Kv4/KChIP complex. CL-888 altered the apparent affinity of KChIP1 to Kv4.3-N in a Biacore assay, but did not dissociate the two proteins in size-exclusion chromatography experiments. Kv4.2/KChIP1 current amplitude and kinetics were altered with compound exposure, supporting the hypothesis of a compound-induced conformational change in the protein complex. Fluorescence spectroscopy of a unique tryptophan residue in KChIP1 was consistent with compound binding to the protein. Molecular modeling using the KChIP1 crystal structure indicates that compound binding may occur in a small tryptophan-containing binding pocket located on the hydrophilic side of the protein.


Subject(s)
Kv Channel-Interacting Proteins/metabolism , Shal Potassium Channels/metabolism , Urea/analogs & derivatives , Animals , Chromatography, Gel , Electrophysiology , Gene Transfer Techniques , Humans , Inhibitory Concentration 50 , Kv Channel-Interacting Proteins/genetics , Models, Chemical , Models, Molecular , Oocytes/physiology , Protein Binding , Protein Conformation/drug effects , Shal Potassium Channels/genetics , Spectrometry, Fluorescence , Two-Hybrid System Techniques , Urea/pharmacology , Xenopus
18.
Proc Natl Acad Sci U S A ; 101(43): 15535-40, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15486093

ABSTRACT

Voltage-gated Kv1.1/Kvbeta1.1 A-type channels, as a natural complex, can switch from fast to slow inactivation under oxidation/reduction conditions. The mode-switching of inactivation, which is mediated by a cysteine residue in the inactivation ball domain of the Kvbeta1.1 N terminus, can regulate membrane electrical excitability. In the present study, we identified a mechanism whereby inactivation in Kv1.1/Kvbeta1.1 channels is regulated by calcium influx. The rise in intracellular calcium, due to either influx from extracellular space or release from intracellular stores, eliminates fast inactivation induced by Kvbeta1.1, resulting in slower inactivation and increased steady-state current. This oxidation-independent calcium effect is mediated through the Kvbeta1.1 N terminus, not the C terminus. We propose that a coupling between calcium influx and inactivation of voltage-gated A-type K+ channels occurs as a result of membrane depolarization and may contribute to afterhyperpolarization as negative feedback to control neuronal excitability.


Subject(s)
Calcium/physiology , Ion Channel Gating , Potassium Channels, Voltage-Gated/physiology , Animals , Calcimycin/pharmacology , Humans , Kv1.1 Potassium Channel , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/drug effects , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL