Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
FASEB J ; 36(9): e22457, 2022 09.
Article in English | MEDLINE | ID: mdl-35997997

ABSTRACT

Tree and shrub barks have been used as folk medicine by numerous cultures across the globe for millennia, for a variety of indications, including as vasorelaxants and antispasmodics. Here, using electrophysiology and myography, we discovered that the KCNQ5 voltage-gated potassium channel mediates vascular smooth muscle relaxant effects of barks used in Native American folk medicine. Bark extracts (1%) from Birch, Cramp Bark, Slippery Elm, White Oak, Red Willow, White Willow, and Wild Cherry each strongly activated KCNQ5 expressed in Xenopus oocytes. Testing of a subset including both the most and the least efficacious extracts revealed that Red Willow, White Willow, and White Oak KCNQ-dependently relaxed rat mesenteric arteries; in contrast, Black Haw bark neither activated KCNQ5 nor induced vasorelaxation. Two compounds common to the active barks (gallic acid and tannic acid) had similarly potent and efficacious effects on both KCNQ5 activation and vascular relaxation, and this together with KCNQ5 modulation by other tannins provides a molecular basis for smooth muscle relaxation effects of Native American folk medicine bark extracts.


Subject(s)
KCNQ Potassium Channels , Vasodilator Agents , Animals , Humans , Mesenteric Arteries , Rats , Tannins/pharmacology , Vasodilator Agents/pharmacology , American Indian or Alaska Native
2.
Pain ; 160(1): 198-209, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30204648

ABSTRACT

Chronic neuropathic pain may be caused, in part, by loss of inhibition in spinal pain processing pathways due to attenuation of local GABAergic tone. Nociception and nocifensive behaviors are reduced after enhancement of tonically activated extrasynaptic GABAAR-mediated currents by agonist ligands for δ subunit-containing GABAARs. However, typical ligands that target δ subunit-containing GABAARs are limited due to sedative effects at higher doses. We used the spinal nerve ligation (SNL) and gp120 models of experimental neuropathic pain to evaluate compound 2-261, a nonbenzodiazepine site positive allosteric modulator of α4ß3δ GABAARs optimized to be nonsedative by selective activation of ß2/3-subunit-containing GABAARs over receptor subtypes incorporating ß1 subunits. Similar levels of 2-261 were detected in the brain and plasma after intraperitoneal administration. Although systemic 2-261 did not alter sensory thresholds in sham-operated animals, it significantly reversed SNL-induced thermal and tactile hypersensitivity in a GABAAR-dependent fashion. Intrathecal 2-261 produced conditioned place preference and elevated dopamine levels in the nucleus accumbens of nerve-injured, but not sham-operated, rats. In addition, systemic pretreatment with 2-261 blocked conditioned place preference from spinal clonidine in SNL rats. Moreover, 2-261 reversed thermal hyperalgesia and partially reversed tactile allodynia in the gp120 model of HIV-related neuropathic pain. The effects of 2-261 likely required interaction with the α4ß3δ GABAAR because 2-301, a close structural analog of 2-261 with limited extrasynaptic receptor efficacy, was not active. Thus, 2-261 may produce pain relief with diminished side effects through selective modulation of ß2/3-subunit-containing extrasynaptic GABAARs.


Subject(s)
Chronic Pain/drug therapy , Chronic Pain/metabolism , GABA Modulators/therapeutic use , Receptors, GABA/metabolism , Allosteric Regulation/drug effects , Animals , Chronic Pain/etiology , Conditioning, Operant , Disease Models, Animal , Dose-Response Relationship, Drug , GABA Modulators/chemistry , HIV Infections/complications , Hyperalgesia/physiopathology , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , Pain Measurement , Pain Threshold/drug effects , Patch-Clamp Techniques , Peripheral Nerve Injuries/complications , Physical Endurance/drug effects , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA/genetics
3.
Epilepsy Behav ; 82: 57-63, 2018 05.
Article in English | MEDLINE | ID: mdl-29587186

ABSTRACT

OBJECTIVE: Subunit-specific positive allosteric modulators (PAMs) of gamma-aminobutyric acid-A (GABA-A) receptors are commonly used to uncover the role of GABA-A receptor isoforms in brain function. Recently, we have designed novel PAMs selective for ß2/3-subunit containing GABA-A receptors (ß2/3-selective PAMs) that are nonbenzodiazepine site-mediated and do not show an α-subunit isoform selectivity, yet exhibit anxiolytic efficacy with reduced potential for sedation, cognitive impairment, and tolerance. In this study, we used three novel ß2/3-selective PAMs (2-261, 2-262, and 10029) with differential ß2/3-subunit potency to identify the role of ß2/3-selective receptor isoforms in limbic epileptogenesis. METHODS: Experimental epileptogenesis was induced in mice by daily hippocampus stimulations until each mouse showed generalized (stage 5) seizures. Patch-clamp electrophysiology was used to record GABA-gated currents. Brain levels of ß2/3-selective PAMs were determined for mechanistic correlations. RESULTS: Treatment with the ß2/3-selective PAMs 2-261 (30mg/kg), 2-262 (10mg/kg), and 10029 (30mg/kg), 30min prior to stimulations, significantly suppressed the rate of development of kindled seizure activity without affecting the afterdischarge (AD) signal, indicating their disease-modifying activity. The ß2/3-selective agents suppressed chemical epileptogenesis in the pentylenetetrazol model. Test doses of these agents were devoid of acute antiseizure activity in the kindling model. CONCLUSION: These findings demonstrate that ß2/3-selective PAMs can moderately retard experimental epileptogenesis, indicating the protective role of ß2/3-subunit GABA-A receptor isoforms in the development of epilepsy.


Subject(s)
Epilepsy/drug therapy , Epilepsy/physiopathology , GABA Modulators/therapeutic use , Hippocampus/physiopathology , Kindling, Neurologic/physiology , Receptors, GABA-A/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , GABA Modulators/pharmacology , Hippocampus/drug effects , Kindling, Neurologic/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Protein Isoforms , Seizures/physiopathology , Temporal Lobe/drug effects , Temporal Lobe/physiopathology
4.
Neuropharmacology ; 126: 38-47, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28842344

ABSTRACT

Autism spectrum disorder (ASD) is associated with two core symptoms (social communication deficits and stereotyped repetitive behaviors) in addition to a number of comorbidities. There are no FDA-approved drugs for the core symptoms and the changes that underlie these behaviors are not fully understood. One hypothesis is an imbalance of the excitation (E)/inhibition (I) ratio with excessive E and diminished I occurring in specific neuronal circuits. Data suggests that both gamma-aminobutyric acidA (GABAA) and α7 nicotinic acetylcholine receptors (nAChRs) significantly impact E/I. BTBR T+tf/J (BTBR) mice are a model that display an autism-like phenotype with impaired social interaction and stereotyped behavior. A ß2/3-subunit containing GABAA receptor (GABAAR) subtype selective positive allosteric modulator (PAM), 2-261, and an α7 nAChR subtype selective PAM, AVL-3288, were tested in social approach and repetitive self-grooming paradigms. 2-261 was active in the social approach but not the self-grooming paradigm, whereas AVL-3288 was active in both. Neither compound impaired locomotor activity. Modulating α7 nAChRs alone may be sufficient to correct these behavioral and cognitive deficits. GABAergic and nicotinic compounds are already in various stages of clinical testing for treatment of the core symptoms and comorbidities associated with ASD. Our findings and those of others suggest that compounds that have selective activities at GABAAR subtypes and the α7 nAChR may address not only the core symptoms, but many of the associated comorbidities as well and warrant further investigation in other models of ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Behavior, Animal/drug effects , Receptors, GABA-A/physiology , alpha7 Nicotinic Acetylcholine Receptor/physiology , Allosteric Regulation , Anilides/administration & dosage , Animals , Autism Spectrum Disorder/prevention & control , Disease Models, Animal , Grooming/drug effects , Humans , Isoxazoles/administration & dosage , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Social Behavior
5.
Psychopharmacology (Berl) ; 231(17): 3517-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24682501

ABSTRACT

RATIONALE: In order to improve upon the pharmacological properties of the neuroactive steroid ganaxolone, it was used as the starting point in the design of novel neurosteroids that replace the 17ß-acetyl side chain with an isoxazole bioisostere. OBJECTIVES: UCI-50027 (3-[3α-hydroxy-3ß-methyl-5α-androstan-17ß-yl]-5-(hydroxymethyl)isoxazole) was designed as an orally active neuroactive steroid specifically targeted at the gamma-aminobutyric acid(A) receptor (GABAAR). METHODS: UCI-50027 was tested in vitro in Xenopus oocytes expressing human GABAARs and in vivo as an anticonvulsant, for ataxic effects and for anxiolytic activity. RESULTS: In vitro, UCI-50027 dose-dependently enhanced the activity of GABA at human α1ß2γ2L, α2ß1γ2L, and α4ß3δ GABAARs. Consistent with its action as a positive allosteric modulator (PAM), it had no direct activity in the absence of GABA. UCI-50027 protected against acute pentylenetetrazol (PTZ)-induced convulsions with an ED50 of 6 mg/kg p.o. In the rotarod (RR) paradigm in mice, the AD50 (the ataxic dose where half of the animals fail the RR test) was found to be 38 mg/kg p.o., giving a therapeutic index (TI = RR AD50/PTZ ED50)∼6 versus 2.8 for ganaxolone. In the mouse-elevated plus maze (EPM) model for anxiety, UCI-50027 showed a minimum effective dose (MED) ≤0.3 mg/kg p.o. Thus, the TI (TI = RR AD50/EPM MED) for the compound as an anxiolytic is ≥127 versus 3.3 for ganaxolone. CONCLUSIONS: UCI-50027 is an orally active neuroactive steroid with pharmacological activity consistent with a GABAAR PAM that has an improved separation between anticonvulsant/anxiolytic and rotarod effects, potent activity as an anticonvulsant and anxiolytic when compared to ganaxolone.


Subject(s)
Androstanes/pharmacology , Neurotransmitter Agents/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Male , Mice , Motor Activity/drug effects , Neurotransmitter Agents/pharmacokinetics , Patch-Clamp Techniques , Pentylenetetrazole/antagonists & inhibitors , Postural Balance/drug effects , Pregnanolone/analogs & derivatives , Pregnanolone/pharmacokinetics , Pregnanolone/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Xenopus
6.
J Psychopharmacol ; 28(5): 472-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24108410

ABSTRACT

GABAergic anxiolytics have well-documented centrally mediated side effects including sedation, potentiation of ethanol, tolerance, abuse liability and memory impairment. Most research directed towards identifying an anxioselective GABAergic therapeutic has been based upon the theory that these side effects could be mitigated by avoiding α1/5-subunit GABAA receptors while specifically targeting those with the α2/3-subunit. Unfortunately, there are prominent exceptions to this theory and it has yet to be translated into clinical success. We previously demonstrated that ß2/3-subunit-selective GABAA receptor-positive allosteric modulators act as anxiolytics with reduced sedation and ethanol potentiation regardless of their activity at α1-subunit GABAA receptors. The prototypical ß2/3-subunit-selective positive allosteric modulator, 2-261, is further characterized here for additional side effects commonly associated with central GABAA receptor activation. In mice, 10 times the anxiolytic dose (10 mg/kg) of 2-261 does not induce behavioral tolerance in the elevated plus maze following a 2 week subchronic treatment. In rats, an anxiolytic dose (10 mg/kg) of 2-261 is inactive in conditioned place preference, suggesting a reduced abuse liability. In rats, 10 times the anxiolytic dose (100 mg/kg) of 2-261 does not have a significant amnestic effect in the radial arm maze, suggesting a greater therapeutic index for memory impairment. These results suggest that ß2/3-subunit subtype-selective GABAA receptor-positive allosteric modulators not only have reduced sedative liability, but also a reduction in other central side effects commonly associated with broader GABAA receptor activation. ß2/3-subunit-selective compounds may represent a novel design template for anxiolytics with benzodiazepine-like efficacy and mitigated side effects.


Subject(s)
Allosteric Regulation/drug effects , Anti-Anxiety Agents/administration & dosage , GABA-A Receptor Agonists/administration & dosage , Receptors, GABA-A/metabolism , Animals , Ethanol/metabolism , Hypnotics and Sedatives/administration & dosage , Mice , Rats , Rats, Sprague-Dawley
7.
J Med Chem ; 56(21): 8352-65, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24098954

ABSTRACT

A series of novel arylpyrid-3-ylmethanones (7a-aa) were designed as modulators of α7 nicotinic acetylcholine receptors (nAChRs). The methanones were found to be type I positive allosteric modulators (PAMs) of human α7 nAChRs expressed in Xenopus ooctyes. Structure-activity relationship (SAR) studies resulted in the identification of compound 7v as a potent and efficacious type I PAM with maximum modulation of a nicotine EC5 response of 1200% and EC50 = 0.18 µM. Compound 7z was active in reversing the effect of scopolamine in the novel object recognition (NOR) paradigm with a minimum effective ip dose of 1.0 mg/kg (2.7 µmol/kg). This effect was blocked by the selective α7 nAChR antagonist methyllycaconitine (MLA). These compounds are potent type I positive allosteric modulators of α7 nAChRs that may have therapeutic value in restoring impaired sensory gating and cognitive deficits in schizophrenia and Alzheimer's disease.


Subject(s)
Drug Design , Pyridines/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Binding Sites/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Molecular Structure , Oocytes/chemistry , Oocytes/drug effects , Oocytes/metabolism , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship , Xenopus , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis
8.
Neuropharmacology ; 61(1-2): 95-104, 2011.
Article in English | MEDLINE | ID: mdl-21419142

ABSTRACT

Our current study aims to evaluate the mechanisms of tranylcypromine (TCP)-mediated enhancement of nicotine self-administration. We replicated our previous findings which demonstrate that 1 h pretreatment with TCP (3 mg/kg, i.p.) enhances nicotine self-administration (7.5 µg/kg/inj, i.v.) when compared with vehicle-treated rodents. We tested whether TCP-mediated enhancement of nicotine self-administration was due to MAO inhibition or off-target effects by (i) extending the TCP pretreatment time from 1 to 20 h, and (ii) evaluating the role of the individual TCP stereoisomers in nicotine self-administration studies. While 20 h and (-)TCP pretreatment induced significant inhibition of MAO (60-90%), animals found nicotine only weakly reinforcing. Furthermore, while both (+) and (±)TCP treatment induced nearly 100% MAO inhibition, (+)TCP pretreated animals took longer to acquire nicotine self-administration compared to (±)TCP pretreated animals. Stable nicotine self-administration in (+)TCP pretreated animals was influenced by nicotinic receptor activation but not nicotine-paired cues. The opposite was found in (±)TCP pretreated animals. Treatment with (-) or (±)TCP increased dopamine and serotonin overflow, while the (+) and (±)TCP treatment enhanced monoamine overflow subsequent to nicotine. Together, our data suggests TCP enhancement of nicotine self-administration are mediated through mechanisms independent of MAO inhibition, including nicotine-paired cues and monoamine uptake inhibition.


Subject(s)
Behavior, Addictive/chemically induced , Behavior, Addictive/metabolism , Monoamine Oxidase Inhibitors/toxicity , Nicotine/administration & dosage , Tranylcypromine/toxicity , Animals , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Male , Monoamine Oxidase/metabolism , Rats , Rats, Sprague-Dawley , Self Administration , Serotonin/metabolism
9.
J Pharmacol Exp Ther ; 336(3): 908-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21159751

ABSTRACT

α5 Subunit-containing GABA(A) receptors (GABA(A)Rs) and α7 neuronal nicotinic-acetylcholine receptors (nAChRs) are members of the Cys-loop family of ligand-gated ion channels (LGICs) that mediate cognitive and attentional processes in the hippocampus. α5 GABA(A)Rs alter network activity by tonic inhibition of CA1/CA3 pyramidal cells of the hippocampus. Postsynaptic α7 nAChRs in the hippocampus regulate inhibitory GABAergic interneuron activity required for synchronization of pyramidal neurons in the CA1, whereas presynaptic α7 nAChRs regulate glutamate release. Can simultaneous allosteric modulation of these LGICs produce synergistic effects on cognition? We show that combined transient application of two allosteric modulators that individually 1) inhibit α5 GABA(A)Rs and 2) enhance α7 nAChRs causes long-term potentiation (LTP) of mossy fiber stimulation-induced excitatory postsynaptic currents (EPSC) from CA1 pyramidal neurons of rat hippocampal slices. The LTP effect evoked by two compounds is replicated by 3-(2,5-difluorophenyl)-6-(N-ethylindol-5-yl)-1,2,4-triazolo[4,3-b]pyridazine (522-054), a compound we designed to simultaneously inhibit α5 GABA(A)Rs and enhance α7 nAChRs. Selective antagonists for either receptor block sustained EPSC potentiation produced by 522-054. In vivo, 522-054 enhances performance in the radial arm maze and facilitates attentional states in the five-choice serial reaction time trial with similar receptor antagonist sensitivity. These observations may translate into therapeutic utility of dual action compounds in diseases of hippocampal-based cognitive impairment.


Subject(s)
Cognition/physiology , Hippocampus/physiology , Ligand-Gated Ion Channels/physiology , Long-Term Potentiation/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Cognition/drug effects , Female , Hippocampus/drug effects , Indoles/chemistry , Indoles/pharmacology , Long-Term Potentiation/drug effects , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, GABA-A/physiology , Receptors, Nicotinic/physiology , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
10.
J Pharmacol Exp Ther ; 332(3): 1040-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19940102

ABSTRACT

GABA(A) receptor (R) positive allosteric modulators that selectively modulate GABA(A)Rs containing beta(2)- and/or beta(3)- over beta(1)-subunits have been reported across diverse chemotypes. Examples include loreclezole, mefenamic acid, tracazolate, and etifoxine. In general,"beta(2/3)-selective" GABA(A)R positive allosteric modulators are nonbenzodiazepines (nonBZs), do not show alpha-subunit isoform selectivity, yet have anxiolytic efficacy with reduced ataxic/sedative effects in animal models and humans. Here, we report on an enantiomeric pair of nonBZ GABA(A)R positive allosteric modulators that demonstrate differential beta-subunit isoform selectivity. We have tested this enantiomeric pair along with a series of other beta(2/3)-subunit selective, alpha-subunit isoform-selective, BZ and nonBZ GABA(A) positive allosteric modulators using electrophysiological, pharmacokinetic, and behavioral assays to test the hypothesis that ataxia may be correlated with the extent of modulation at beta(1)-subunit-containing GABA(A)Rs. Our findings provide an alternative strategy for designing anxioselective allosteric modulators of the GABA(A)R with BZ-like anxiolytic efficacy by reducing or eliminating activity at beta(1)-subunit-containing GABA(A)Rs.


Subject(s)
Anti-Anxiety Agents/pharmacology , Ataxia/prevention & control , GABA Modulators/pharmacology , Receptors, GABA-A/physiology , Allosteric Regulation , Amides/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacokinetics , Ataxia/physiopathology , Ataxia/psychology , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , GABA Modulators/chemistry , GABA Modulators/pharmacokinetics , Humans , Male , Mice , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Protein Isoforms/physiology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Xenopus laevis
11.
J Pharmacol Exp Ther ; 323(3): 907-15, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17873105

ABSTRACT

Drugs that antagonize nicotinic acetylcholine receptors (nAChRs) can be used to inhibit nicotine-induced behavior in both humans and animals. The aim of our experiments is to establish a proof-of-principle that antagonism of nAChRs by negative allosteric modulation can alter behavior in a relevant animal model of addiction, nicotine self-administration. We have identified a novel, negative allosteric modulator of nAChRs, UCI-30002 [N-(1,2,3,4-tetrahydro-1-naphthyl)-4-nitroaniline], with selectivity for the major neuronal nAChR subtypes over muscle-type nAChRs. After systemic administration, UCI-30002 significantly reduces nicotine self-administration in rats on both fixed ratio and progressive ratio schedules of reinforcement. The minimum effective dose that significantly alters nicotine self-administration corresponds to brain concentrations of UCI-30002 that produce at least 30% inhibition of the major neuronal nAChR subtypes measured in vitro. UCI-30002 has no effect on responding for food reinforcement in rats on either type of schedule, indicating that there is no effect on general responding or natural reward. UCI-30002 represents validation of the concept that negative allosteric modulators may have significant benefits as a strategy for treating nicotine addiction and encourages the development of subtype-selective modulators.


Subject(s)
Aniline Compounds/therapeutic use , Naphthalenes/therapeutic use , Nicotine/administration & dosage , Nicotinic Antagonists/therapeutic use , Receptors, Nicotinic/metabolism , Tobacco Use Disorder , Allosteric Site , Aniline Compounds/administration & dosage , Aniline Compounds/adverse effects , Animals , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Electrophysiology , Feeding Behavior/drug effects , Feeding Behavior/ethnology , Ligands , Male , Mice , Motor Activity/drug effects , Naphthalenes/administration & dosage , Naphthalenes/adverse effects , Nicotine/adverse effects , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/adverse effects , Nicotinic Antagonists/pharmacokinetics , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Protein Binding , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/prevention & control , Self Administration , Tobacco Use Disorder/drug therapy , Tobacco Use Disorder/metabolism , Xenopus laevis
12.
J Med Chem ; 50(14): 3369-79, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17571865

ABSTRACT

A series of enaminone esters and amides have been developed as potent allosteric modulators of gamma-aminobutyric acidA (GABAA) receptors. The compounds bind to a novel modulatory site that is independent of the benzodiazepine (BZ), isosteric GABA, and neuroactive steroid binding sites. Structure-activity relationship (SAR) studies resulted in the synthesis of the c-Bu amide 16h with an in vitro potency of 7 nM based on inhibition of [35S]TBPS binding. The activity of the enaminones as positive allosteric modulators was confirmed with electrophysiological measurements in oocytes expressing alpha1beta2gamma2L GABAA receptors. The i-Pr, s-Bu, c-Pr, and c-Bu amides (16e-h) were orally active in mice with profound central nervous system depressant effects. The i-Pr amide 16e was an orally active anxiolytic in the mouse light-dark paradigm.


Subject(s)
Amides/pharmacology , Receptors, GABA-A/drug effects , Administration, Oral , Animals , Magnetic Resonance Spectroscopy , Male , Mice , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
Proc Natl Acad Sci U S A ; 104(19): 8059-64, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470817

ABSTRACT

Activation of brain alpha7 nicotinic acetylcholine receptors (alpha7 nAChRs) has broad therapeutic potential in CNS diseases related to cognitive dysfunction, including Alzheimer's disease and schizophrenia. In contrast to direct agonist activation, positive allosteric modulation of alpha7 nAChRs would deliver the clinically validated benefits of allosterism to these indications. We have generated a selective alpha7 nAChR-positive allosteric modulator (PAM) from a library of GABAA receptor PAMs. Compound 6 (N-(4-chlorophenyl)-alpha-[[(4-chloro-phenyl)amino]methylene]-3-methyl-5-isoxazoleacet-amide) evokes robust positive modulation of agonist-induced currents at alpha7 nAChRs, while preserving the rapid native characteristics of desensitization, and has little to no efficacy at other ligand-gated ion channels. In rodent models, it corrects sensory-gating deficits and improves working memory, effects consistent with cognitive enhancement. Compound 6 represents a chemotype for allosteric activation of alpha7 nAChRs, with therapeutic potential in CNS diseases with cognitive dysfunction.


Subject(s)
GABA Modulators/pharmacology , Nootropic Agents/pharmacology , Receptors, GABA-A/drug effects , Receptors, Nicotinic/drug effects , Animals , Brain/metabolism , Cell Survival/drug effects , Cognition/drug effects , Dizocilpine Maleate/pharmacology , Humans , Male , Mice , Mice, Inbred DBA , Rats , Rats, Sprague-Dawley , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
14.
J Drug Target ; 14(3): 127-36, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16753826

ABSTRACT

The endogenous opioid peptide leu-enkephalin (ENK) was chemically modified by a method known as reversible aqueous lipidization (REAL) with a novel amine-reacting lipophilic dimethylmaleic anhydride analog, 3,4-bis(decylthiomethyl)-2,5-furandione. The binding affinity of the product, REAL-ENK, to opioid receptors was greatly reduced. This prodrug was stable in neutral and basic phosphate buffers but underwent rapid hydrolysis under acidic conditions in the presence of 50% acetonitrile. It also showed increased stability toward enzymatic degradations in various tissue preparations. The half-lives of REAL-ENK in mouse small intestinal mucosal homogenate and liver homogenate were 12 and 80 min, representing a 12- and 32-fold increase over those of ENK itself. In contrast to ENK (t(1/2) 6.7 min), REAL-ENK was stable in mouse plasma. More importantly, REAL-ENK produced significant and sustained antinociception mediated by peripheral opioid receptors in a rodent inflammatory pain model. Pharmacokinetic studies employing a radioimmunoassay (RIA) demonstrated that significantly higher and sustained plasma peptide levels were detected up to 24 h following the oral administration of REAL-ENK in normal mice. The peak concentration and area under the curve of oral REAL-ENK were 4.4 and 21 times higher than that of oral ENK. Our results indicate that like its disulfide-based counterpart, amine-based REAL may be an enabling technology which can be applied to enhance metabolic stability, increase oral absorption, and preserve and possibly prolong the pharmacological activity of peptide drugs.


Subject(s)
Enkephalin, Leucine/administration & dosage , Lipids/chemistry , Administration, Oral , Animals , Caco-2 Cells , Enkephalin, Leucine/chemistry , Enkephalin, Leucine/pharmacokinetics , Half-Life , Humans , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Liver/metabolism , Mice , Radioimmunoassay , Tissue Distribution
15.
J Pharmacol Exp Ther ; 318(3): 1083-93, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16728593

ABSTRACT

Voltage-gated Na(+) channels may play important roles in establishing pathological neuronal hyperexcitability associated with chronic pain in humans. Na(+) channel blockers, such as carbamazepine (CBZ) and lamotrigine (LTG), are efficacious in treating neuropathic pain; however, their therapeutic utility is compromised by central nervous system side effects. We reasoned that it may be possible to gain superior control over pain states and, in particular, a better therapeutic index, by designing broad-spectrum Na(+) channel blockers with higher potency, faster onset kinetics, and greater levels of state dependence than existing drugs. 2-[4-(4-Chloro-2-fluorophenoxy)phenyl]-pyrimidine-4-carboxamide (PPPA) is a novel structural analog of the state-dependent Na(+) channel blocker V102862 [4-(4-fluorophenoxy)benzaldehyde semicarbazone]. Tested on recombinant rat Na(v)1.2 channels and native Na(+) currents in cultured rat dorsal root ganglion neurons, PPPA was approximately 1000 times more potent, had 2000-fold faster binding kinetics, and > or =10-fold higher levels of state dependence than CBZ and LTG. Tested in rat pain models against mechanical endpoints, PPPA had minimal effective doses of 1 to 3 mg/kg p.o. in partial sciatic nerve ligation, Freund's complete adjuvant, and postincisional pain. In all cases, efficacy was similar to clinically relevant comparators. Importantly, PPPA did not produce motor deficits in the accelerating Rotarod assay of ataxia at doses up to 30 mg/kg p.o., indicating a therapeutic index >10, which was superior to CBZ and LTG. Our experiments suggest that high-potency, broad-spectrum, state-dependent Na(+) channel blockers will have clinical utility for treating neuropathic, inflammatory, and postsurgical pain. Optimizing the biophysical parameters of broad-spectrum voltage-gated Na(+) channel blockers may lead to improved pain therapeutics.


Subject(s)
Pain/drug therapy , Pyrimidines/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Carbamazepine/pharmacology , Humans , Hyperalgesia/drug therapy , Lamotrigine , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Semicarbazones/pharmacology , Tetrodotoxin/pharmacology , Triazines/pharmacology
16.
J Med Chem ; 47(6): 1547-52, 2004 Mar 11.
Article in English | MEDLINE | ID: mdl-14998340

ABSTRACT

A series of 3-(4-phenoxyphenyl)-1H-pyrazoles were synthesized and characterized as potent state-dependent sodium channel blockers. A limited SAR study was carried out to delineate the chemical requirements for potency. The results indicate that the distal phenyl group is critical for activity but will tolerate lipophilic (+pi) electronegative (+sigma) substituents at the ortho and/or para position. Substitution at the pyrazole nitrogen with a H-bond donor improves potency. Compound 18 showed robust activity in the rat Chung neuropathy paradigm.


Subject(s)
Analgesics/chemical synthesis , Pyrazoles/chemical synthesis , Sodium Channel Blockers/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Cell Line , Humans , Male , Pain/drug therapy , Pain/etiology , Patch-Clamp Techniques , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Structure-Activity Relationship
17.
Nat Med ; 10(1): 31-2, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14647497

ABSTRACT

Patients taking fluoroquinolone antibiotics such as norfloxacin exhibit a low incidence of convulsions and anxiety. These side effects probably result from antagonism of the neurotransmitter gamma-aminobutyric acid (GABA) at the brain GABA(A) receptor complex (GRC). Modification of norfloxacin yields molecules such as compound 4 that potentiate GABA action with alpha(2) subunit selectivity. Compound 4 is anxiolytic but does not cause sedation, and may represent a new class of ligands that have anxiolytic activity without sedative liability.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anti-Infective Agents/pharmacology , Fluoroquinolones/pharmacology , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Fluoroquinolones/chemistry , Fluoroquinolones/metabolism , Humans , Protein Binding , Receptors, GABA-A/metabolism , Recombinant Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...