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










Publication year range
1.
Mov Disord ; 30(14): 1901-1911, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26573698

ABSTRACT

BACKGROUND: ABT-126 is a novel, safe, and well-tolerated α7 nicotinic receptor agonist in a Phase 2 Alzheimer's disease study. We tested the antidyskinetic effect of ABT-126 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated squirrel monkeys with moderate and more severe nigrostriatal damage. METHODS: Monkeys (n = 21, set 1) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1-2×. When parkinsonian, they were gavaged with levodopa (10 mg/kg)/carbidopa (2.5 mg/kg) twice daily and dyskinesias rated. They were then given nicotine in drinking water (n = 5), or treated with vehicle (n = 6) or ABT-126 (n = 10) twice daily orally 30 min before levodopa. Set 1 was then re-lesioned 1 to 2 times for a total of 3 to 4 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine injections. The antidyskinetic effect of ABT-126, nicotine, and the ß2* nicotinic receptor agonist ABT-894 was re-assessed. Another group of monkeys (n = 23, set 2) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine only 1× to 2×. They were treated with levodopa/carbidopa, administered the α7 agonist ABT-107 (n = 6), ABT-894 (n = 6), nicotine (n = 5), or vehicle (n = 6) and dyskinesias evaluated. All monkeys were euthanized and the dopamine transporter measured. RESULTS: With moderate nigrostriatal damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1×-2×), ABT-126 dose-dependently decreased dyskinesias (∼60%), with similar results seen with ABT-894 (∼60%) or nicotine (∼60%). With more severe damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 3-4×), ABT-126 and nicotine reduced dyskinesias, but ABT-894 did not. The dopamine transporter was 41% and 8.9% of control, with moderate and severe nigrostriatal damage, respectively. No drug modified parkinsonism. CONCLUSION: The novel α7 nicotinic receptor drug ABT-126 reduced dyskinesias in monkeys with both moderate and severe nigrostriatal damage. ABT-126 may be useful to reduce dyskinesias in both early- and later-stage Parkinson's disease.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Nicotinic Agonists/therapeutic use , Parkinsonian Disorders/drug therapy , Substantia Nigra/pathology , alpha7 Nicotinic Acetylcholine Receptor/agonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/pathology , Female , Levodopa/therapeutic use , Male , Nicotinic Agonists/pharmacology , Parkinsonian Disorders/pathology , Saimiri
2.
Exp Neurol ; 263: 277-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25261754

ABSTRACT

The finding that smoking is inversely correlated with Parkinson's disease and that nicotine attenuates nigrostriatal damage in Parkinsonian animals supports the idea that nicotine may be neuroprotective. Nicotine is thought to exert this effect by acting at nicotinic receptors (nAChRs), including the α7 subtype. The objective of this study was twofold: first, to test the protective potential of ABT-107, an agonist with high selectivity for α7 nAChRs; and second, to investigate its cellular mechanism of action. Rats were implanted with minipumps containing ABT-107 (0.25mg/kg/d). In addition, we tested the effect of nicotine (1mg/kg/d) as a positive control, and also DMXB (2mg/kg/d) which acts primarily with α7 but also α4ß2* nAChRs. Two weeks after minipump placement, the rats were lesioned by unilateral administration of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle. Lesioning alone decreased contralateral forelimb use and adjusted stepping, two measures of Parkinsonism. ABT-107 and nicotine treatment significantly improved these behaviors at all weeks tested, with variable improvement with DMXB. We next investigated the cellular mechanism involved. The striatal dopamine transporter (DAT), a marker of dopaminergic integrity, was reduced ~70% with lesioning. ABT-107 or nicotine treatment significantly increased DAT levels in lesioned striatum; these drugs did not alter DAT levels in intact striatum. ABT-107 and nicotine also significantly improved basal dopamine release from lesioned striatum, as well as nicotine-stimulated dopamine release mediated via α4ß2* and α6ß2* nAChRs. These data suggest that α7 nAChR agonists may improve motor behaviors associated with nigrostriatal damage by enhancing striatal dopaminergic function.


Subject(s)
Brain/drug effects , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/metabolism , Quinuclidines/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Adrenergic Agents/toxicity , Animals , Autoradiography , Brain/metabolism , Male , Nicotinic Agonists/pharmacology , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley
3.
J Pharmacol Exp Ther ; 351(1): 25-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25034405

ABSTRACT

Previous studies in Parkinsonian rats and monkeys have shown that ß2-selective nicotinic acetylcholine receptor (nAChR) agonists reduce l-Dopa-induced dyskinesias (LIDs), a serious complication of l-Dopa therapy for Parkinson's disease. Since rodent studies also suggested an involvement of α7 nAChRs in LIDs, we tested the effect of the potent, selective α7 agonist ABT-107 [5-(6-[(3R)-1-azabicyclo[2.2.2]oct-3-yloxy] pyridazin-3-yl)-1H-indole]. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-lesioned monkeys were gavaged with l-Dopa/carbidopa (10 and 2.5 mg/kg, respectively) twice daily, which resulted in stable LIDs. A dose-response study (0.03-1.0 mg/kg) showed that oral ABT-107 decreased LIDs by 40-60%. LIDs returned to control levels only after a 6-week ABT-107 washout, suggesting that long-term molecular changes were involved. Subsequent readministration of ABT-107 decreased LIDs by 50-60%, indicating that tolerance did not develop. ABT-107 had no effect on Parkinsonism or cognitive performance. We next tested ABT-107 together with the ß2 agonist ABT-894 [(3-(5,6-dichloro-pyridin-3-yl)-1(S),5 (S)-3,6-diazabicyclo[3.2.0]heptane], previously shown to reduce LIDs in Parkinsonian monkeys. In one study, the monkeys were first given oral ABT-894 (0.01 mg/kg), which maximally decreased LIDs by 50-60%; they were then also treated with 0.1 mg/kg ABT-107, a dose that maximally reduced LIDs. The effect of combined treatment on LIDs was similar to that with either drug alone. Comparable results were observed in a group of monkeys first treated with ABT-107 and then also given ABT-894. Thus, α7 and ß2 nAChR-selective drugs may function via a final common mechanism to reduce LIDs. The present results suggest that drugs targeting either α7 or ß2 nAChRs may be useful as antidyskinetic agents in Parkinson's disease.


Subject(s)
Dyskinesia, Drug-Induced/drug therapy , Indoles/therapeutic use , MPTP Poisoning/drug therapy , Nicotinic Agonists/therapeutic use , Quinuclidines/therapeutic use , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Antiparkinson Agents/adverse effects , Female , Indoles/pharmacology , Levodopa/adverse effects , Male , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Saimiri
4.
Mov Disord ; 29(4): 508-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24515328

ABSTRACT

Levodopa-induced dyskinesias (LIDs) are a serious complication of levodopa therapy for Parkinson's disease for which there is little treatment. Accumulating evidence shows that nicotinic acetylcholine receptor (nAChR) drugs decrease LIDs in parkinsonian animals. Here, we examined the effect of two ß2 nAChR agonists, ABT-089 and ABT-894, that previously were approved for phase 2 clinical trials for other indications. Two sets of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys were administered levodopa/carbidopa (10 mg/kg and 2.5 mg/kg, respectively) twice daily 5 days a week until they were stably dyskinetic. Each set had a vehicle-treated group, an nAChR agonist-treated group, and a nicotine-treated group as a positive control. Set A monkeys had previously received other nAChR drugs (nAChR drug-primed), whereas Set B monkeys were initially nAChR drug-naive. Both sets were administered the partial agonist ABT-089 (range, 0.01-1.0 mg/kg) orally 5 days a week twice daily 30 minutes before levodopa with each dose given for 1 to 5 weeks. ABT-089 decreased LIDs by 30% to 50% compared with vehicle-treated monkeys. Nicotine reduced LIDs by 70% in a parallel group. After 4 weeks of washout, the effect of the full agonist ABT-894 (range, 0.0001-0.10 mg/kg) was assessed on LIDs in Set A and Set B. ABT-894 reduced LIDs by 70%, similar to nicotine. Both drugs acted equally well at α4ß2* and α6ß2* nAChRs; however, ABT-089 was 30 to 60 times less potent than ABT-894. Tolerance did not develop for the time periods tested (range, 3-4 months). The nAChR drugs did not worsen parkinsonism or cognitive ability. Emesis, a common problem with nAChR drugs, was not observed. ABT-894 and ABT-089 appear to be good candidate nAChR drugs for the management of LIDs in Parkinson's disease.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , MPTP Poisoning/drug therapy , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Animals , Antiparkinson Agents/therapeutic use , Female , Levodopa/therapeutic use , Male , Saimiri , Treatment Outcome
5.
Am J Pathol ; 184(2): 520-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269557

ABSTRACT

Alzheimer disease (AD) is a progressive neurodegenerative disorder with associated memory loss, spatial disorientation, and other psychiatric problems. Cholinergic system dysfunction is an early and salient feature of AD, and enhancing cholinergic signaling with acetylcholinesterase inhibitors is currently the primary strategy for improving cognition. The beneficial effects of acetylcholinesterase inhibitors, however, are typically short-lived and accompanied by adverse effects. Recent evidence suggests that activating α7 nicotinic acetylcholine receptors (α7 nAChR) may facilitate the specific modulation of brain cholinergic signaling, leading to cognitive enhancement and possibly to amelioration of AD pathologic findings. In the present study, we determined the effect of long-term treatment with the selective α7 nAChR agonist A-582941 in aged 3xTg-AD mice with robust AD-like pathology, which is particularly significant not only because this is the only mouse model that co-develops amyloid plaques and neurofibrillary tangles but also because it enabled us to explore whether A-582941 is able to restore brain function after the severe damage associated with AD. Analysis of ß-amyloid deposits, tau phosphorylation, and inflammatory cells revealed that, overall, pathologic findings were unchanged. Rather, α7 nAChR activation induced expression of c-Fos and brain-derived neurotrophic factor and phosphorylation of cyclic adenosine monophosphate response element binding and neurotrophic tyrosine receptor kinase type 2. More important, A-582941 completely restored cognition in aged 3xTg-AD mice to the level of that in age-matched nontransgenic mice. These novel findings indicate that activating α7 nAChR is a promising treatment for cognitive impairment in AD.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Cognition/drug effects , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Memory/drug effects , Mice , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Nootropic Agents/pharmacology , Phosphorylation/drug effects , Plaque, Amyloid/metabolism , Plaque, Amyloid/physiopathology , Pyridazines/pharmacology , Pyrroles/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , tau Proteins/metabolism
6.
Biochem Pharmacol ; 82(8): 883-90, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21684265

ABSTRACT

This review highlights some of the many contributions of the late Dr. Jerry J. Buccafusco to the neurobiology of nicotinic acetylcholine receptors (nAChRs) and cognition over a 25 year period. The article is written by two of Dr. Buccafusco's professional colleagues, one from academia and one from the pharmaceutical industry. While Dr. Buccafusco's expertise in the cholinergic field was extensive, his insights into the practical relevance of his work (with a long-term goal of formulating new drug development strategies) were unique, and a great asset to both the basic science community and pharmaceutical companies. In 1988, Dr. Buccafusco's laboratory was the first to report the cognitive enhancing action of low doses of nicotine in non-human primates. Since that time he studied a large number of novel pro-cognitive agents from several pharmacological classes in rodents as well as monkeys. Based on years of observing paradoxical effects of nicotinic ligands in vitro and in vivo, Dr. Buccafusco made the provocative argument that it might be possible to develop new chemical entities (with pro-cognitive actions) that have the ability to desensitize nAChRs without producing an antecedent agonist action. Some of his more recent work focused on development of single molecular entities that act on multiple CNS targets (including nAChRs) to enhance cognition, provide neuroprotection, and/or provide additional therapeutic actions (e.g., antipsychotic effects). Dr. Buccafusco's influence will live on in the work of the numerous graduate students, postdoctoral fellows, and junior faculty that he mentored over the years who now serve in prestigious positions throughout the world.


Subject(s)
Cognition/physiology , Receptors, Nicotinic/physiology , Animals , Drug Discovery/history , History, 20th Century , History, 21st Century , Humans , Receptors, Nicotinic/history , Receptors, Nicotinic/metabolism , United States
7.
Biochem Pharmacol ; 82(8): 967-76, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21620806

ABSTRACT

Positive modulation of the neuronal nicotinic acetylcholine receptor (nAChR) α4ß2 subtype by selective positive allosteric modulator NS-9283 has shown to potentiate the nAChR agonist ABT-594-induced anti-allodynic activity in preclinical neuropathic pain. To determine whether this benefit can be extended beyond neuropathic pain, the present study examined the analgesic activity and adverse effect profile of co-administered NS-9283 and ABT-594 in a variety of preclinical models in rats. The effect of the combined therapy on drug-induced brain activities was also determined using pharmacological magnetic resonance imaging. In carrageenan-induced thermal hyperalgesia, co-administration of NS-9283 (3.5 µmol/kg, i.p.) induced a 6-fold leftward shift of the dose-response of ABT-594 (ED(50)=26 vs. 160 nmol/kg, i.p.). In the paw skin incision model of post-operative pain, co-administration of NS-9283 similarly induced a 6-fold leftward shift of ABT-594 (ED(50)=26 vs. 153 nmol/kg). In monoiodo-acetate induced knee joint pain, co-administration of NS-9283 enhanced the potency of ABT-594 by 5-fold (ED(50)=1.0 vs. 4.6 nmol/kg). In pharmacological MRI, co-administration of NS-9283 was shown to lead to a leftward shift of ABT-594 dose-response for cortical activation. ABT-594 induced CNS-related adverse effects were not exacerbated in presence of an efficacious dose of NS-9283 (3.5 µmol/kg). Acute challenge of NS-9283 produced no cross sensitization in nicotine-conditioned animals. These results demonstrate that selective positive allosteric modulation at the α4ß2 nAChR potentiates nAChR agonist-induced analgesic activity across neuropathic and nociceptive preclinical pain models without potentiating ABT-594-mediated adverse effects, suggesting that selective positive modulation of α4ß2 nAChR by PAM may represent a novel analgesic approach.


Subject(s)
Analgesics/therapeutic use , Azetidines/therapeutic use , Nicotinic Agonists/therapeutic use , Oxadiazoles/therapeutic use , Pain/drug therapy , Pyridines/therapeutic use , Receptors, Nicotinic/metabolism , Allosteric Regulation , Analgesics/administration & dosage , Analgesics/adverse effects , Animals , Azetidines/administration & dosage , Azetidines/adverse effects , Behavior, Animal/drug effects , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Drug Therapy, Combination , Magnetic Resonance Imaging , Male , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/adverse effects , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Oxadiazoles/administration & dosage , Oxadiazoles/adverse effects , Pain/metabolism , Pyridines/administration & dosage , Pyridines/adverse effects , Rats , Rats, Sprague-Dawley
8.
J Pharmacol Exp Ther ; 336(3): 716-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21172907

ABSTRACT

Neuronal acetylcholine nicotinic receptors (nAChRs) are targets for the development of novel treatments of brain diseases. However, adverse effects (for example, emesis or nausea) associated with high drug maximal exposures or C(max) at nAChRs often hinder the advancement of experimental compounds in clinical trials. Therefore, it is essential to explore the feasibility of maintaining exposures below a predetermined C(max) while sustaining targeted CNS effects. By use of a [¹²³I]5-IA [5-[¹²³I]iodo-3-[2(S)-azetidinylmethoxy]pyridine] displacement SPECT imaging paradigm in nonhuman primates, we compared brain nAChR binding activity elicited by either a bolus injection or by slow infusion of an identical dose of a novel neuronal nicotinic agonist, ABT-089 [2-methyl-3-(2-(S)-pyrrolidinylmethoxy)pyridine dihydrochloride], where the slow infusion scheme was derived from a two-compartment pharmacokinetic modeling designed to limit the C(max). We determined [¹²³I]5-IA displacement using doses of ABT-089 (0.04, 0.4, and 1.0 mg/kg i.v.) that encompassed efficacious drug exposures in nonhuman primates and examined the relationship between ABT-089 displacement ratios and plasma exposures. Our results indicated that calculated displacement ratios were quite similar between the two different dosing regimens despite substantial differences in C(max). In addition, displacement ratios correlated well with drug exposures calculated as the area-under-curve (AUC) of plasma concentration and varied in a dose-dependent manner, suggesting that displacement ratios are driven by the AUC of drug plasma exposure but not C(max). Our data demonstrate the feasibility of predicting plasma exposures using a two-compartment pharmacokinetic model and its potential for optimizing dosing regimens.


Subject(s)
Azetidines/pharmacokinetics , Brain/diagnostic imaging , Models, Biological , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Pyrrolidines/administration & dosage , Pyrrolidines/pharmacokinetics , Tomography, Emission-Computed, Single-Photon/methods , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Female , Papio , Papio anubis
9.
Brain Res ; 1354: 74-84, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20682302

ABSTRACT

The histamine H(3) receptor is predominantly expressed in the central nervous system and plays a role in diverse physiological mechanisms. In the present study, the effects of GSK189254, a potent and selective H(3) antagonist, were characterized in preclinical pain models in rats. Systemic GSK189254 produced dose-dependent efficacy (ED(50)=0.77 mg/kg i.p.) in a rat model of monoiodoacetate (MIA) induced osteoarthritic (OA) pain as evaluated by hindlimb grip force. The role of H(3) receptors in regulating pain perception was further demonstrated using other structurally distinct H(3) antagonists. GSK189254 also displayed efficacy in a rat surrogate model indicative of central sensitization, namely phase 2 response of formalin-induced flinching, and attenuated tactile allodynia in the spinal nerve ligation model of neuropathic pain (ED(50)=1.5mg/kg i.p.). In addition, GSK189254 reversed persistent (CFA) (ED(50)=2.1mg/kg i.p,), whereas was ineffective in acute (carrageenan) inflammatory pain. When administered intrathecally (i.t.) to the lumbar spinal cord, GSK189254 produced robust effects in relieving the OA pain (ED(50)=0.0027 mg/kg i.t.). The systemic GSK189254 effect was completely reversed by the alpha-adrenergic receptor antagonist phentolamine (i.p. and i.t.) but not by the opioid receptor antagonist naloxone (i.p.). Furthermore, the i.t. GSK189254 effect was abolished when co-administered with phentolamine (i.t.). These results suggest that the spinal cord is an important site of action for H(3) antagonism and the effect can be associated with activation of the noradrenergic system. Our data also provide support that selective H(3) antagonists may represent a class of agents for the treatment of pain disorders.


Subject(s)
Histamine H3 Antagonists/pharmacology , Neurons/drug effects , Norepinephrine/metabolism , Pain Measurement/drug effects , Pain/drug therapy , Receptors, Histamine H3/metabolism , Adrenergic alpha-Antagonists/pharmacology , Analysis of Variance , Animals , Benzazepines/pharmacology , Dose-Response Relationship, Drug , Formaldehyde , Hand Strength , Injections, Spinal , Male , Motor Activity/drug effects , Neurons/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pain/chemically induced , Pain/metabolism , Pain Perception/drug effects , Phentolamine/pharmacology , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 334(3): 875-86, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20504913

ABSTRACT

We previously reported that alpha7 nicotinic acetylcholine receptor (nAChR) agonism produces efficacy in preclinical cognition models correlating with activation of cognitive and neuroprotective signaling pathways associated with Alzheimer's disease (AD) pathology. In the present studies, the selective and potent alpha7 nAChR agonist 5-(6-[(3R)-1-azabicyclo[2.2.2]oct-3-yloxy] pyridazin-3-yl)-1H-indole (ABT-107) was evaluated in behavioral assays representing distinct cognitive domains. Studies were also conducted to address potential issues that may be associated with the clinical development of an alpha7 nAChR agonist. Specifically, ABT-107 improved cognition in monkey delayed matching to sample, rat social recognition, and mouse two-trial inhibitory avoidance, and continued to improve cognitive performance at injection times when exposure levels continued to decline. Rats concurrently infused with ABT-107 and donepezil at steady-state levels consistent with clinical exposure showed improved short-term recognition memory. Compared with nicotine, ABT-107 did not produce behavioral sensitization in rats or exhibit psychomotor stimulant activity in mice. Repeated (3 days) daily dosing of ABT-107 increased extracellular cortical acetylcholine in rats, whereas acute administration increased cortical extracellular signal-regulated kinase and cAMP response element-binding protein phosphorylation in mice, neurochemical and biochemical events germane to cognitive function. ABT-107 increased cortical phosphorylation of the inhibitory residue (Ser9) of glycogen synthase kinase-3, a primary tau kinase associated with AD pathology. In addition, continuous infusion of ABT-107 in tau/amyloid precursor protein transgenic AD mice reduced spinal tau hyperphosphorylation. These findings show that targeting alpha7 nAChRs may have potential utility for symptomatic alleviation and slowing of disease progression in the treatment AD, and expand the understanding of the potential therapeutic viability associated with the alpha7 nAChR approach in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/drug effects , Acetylcholine/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/toxicity , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Donepezil , Electroencephalography/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indans/pharmacology , Indoles/pharmacokinetics , Indoles/pharmacology , Macaca mulatta , Male , Mice , Mice, Knockout , Nicotinic Agonists/pharmacokinetics , Nootropic Agents/pharmacology , Phosphorylation , Piperidines/pharmacology , Psychomotor Performance/drug effects , Quinuclidines/pharmacokinetics , Quinuclidines/pharmacology , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Social Perception , alpha7 Nicotinic Acetylcholine Receptor , tau Proteins/genetics , tau Proteins/toxicity
11.
J Neurosci ; 30(9): 3518-30, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203212

ABSTRACT

One-second-long increases in prefrontal cholinergic activity ("transients") were demonstrated previously to be necessary for the incorporation of cues into ongoing cognitive processes ("cue detection"). Nicotine and, more robustly, selective agonists at alpha4beta2* nicotinic acetylcholine receptors (nAChRs) enhance cue detection and attentional performance by augmenting prefrontal cholinergic activity. The present experiments determined the role of beta2-containing and alpha7 nAChRs in the generation of prefrontal cholinergic and glutamatergic transients in vivo. Transients were evoked by nicotine, the alpha4beta2* nAChR agonist ABT-089 [2-methyl-3-(2-(S)-pyrrolindinylmethoxy) pyridine dihydrochloride], or the alpha7 nAChR agonist A-582941 [2-methyl-5-(6-phenyl-pyridazin-3-yl)-octahydro-pyrrolo[3,4-c]pyrrole]. Transients were recorded in mice lacking beta2 or alpha7 nAChRs and in rats after removal of thalamic glutamatergic or midbrain dopaminergic inputs to prefrontal cortex. The main results indicate that stimulation of alpha4beta2* nAChRs evokes glutamate release and that the presence of thalamic afferents is necessary for the generation of cholinergic transients. ABT-089-evoked transients were completely abolished in mice lacking beta2* nAChRs. The amplitude, but not the decay rate, of nicotine-evoked transients was reduced by beta2* knock-out. Conversely, in mice lacking the alpha7 nAChR, the decay rate, but not the amplitude, of nicotine-evoked cholinergic and glutamatergic transients was attenuated. Substantiating the role of alpha7 nAChR in controlling the duration of release events, stimulation of alpha7 nAChR produced cholinergic transients that lasted 10- to 15-fold longer than those evoked by nicotine. alpha7 nAChR-evoked cholinergic transients are mediated in part by dopaminergic activity. Prefrontal alpha4beta2* nAChRs play a key role in evoking and facilitating the transient glutamatergic-cholinergic interactions that are necessary for cue detection and attentional performance.


Subject(s)
Acetylcholine/metabolism , Glutamic Acid/metabolism , Prefrontal Cortex/metabolism , Receptors, Nicotinic/metabolism , Synaptic Transmission/physiology , Afferent Pathways/anatomy & histology , Afferent Pathways/metabolism , Afferent Pathways/surgery , Animals , Denervation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Agonists/pharmacology , Presynaptic Terminals/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Signal Transduction/physiology , Synapses/genetics , Synapses/metabolism , Thalamus/anatomy & histology , Thalamus/metabolism , Thalamus/surgery , Time Factors , Ventral Tegmental Area/anatomy & histology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/surgery , alpha7 Nicotinic Acetylcholine Receptor
12.
Pharmacol Biochem Behav ; 95(2): 146-57, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20064548

ABSTRACT

Several studies have tested nicotinic receptor ligands in the 5-Choice Serial Reaction Time Task (5-CSRTT) with varying results. Some investigators have increased attentional demands by modifying task parameters or using aged or poor performing rats to observe treatment effects. This study examined the alpha4beta2 nicotinic agonist ABT-594 in the 5-CSRTT using a variety of manipulations to determine optimal conditions for observing enhancement. ABT-594 had no effect in drug-naïve adult rats that self-initiated trials. Constant trial presentation decreased accuracy and omissions, with the latter significantly attenuated by acute administration of ABT-594 (0.019-0.062 micromol/kg). Sub-chronic treatment (0.019 micromol/kg) initially impaired drug-naïve subjects, but significant improvements in accuracy and decreased omissions were observed after 5 days of dosing. In 18-22 month-old rats, attentional demands were altered by interspersing blocks of trials with different stimulus durations. Acute ABT-594 (0.062 micromol/kg) enhanced accuracy performance in poor performing rats (<70% accuracy) but not in those that performed well (>80% accuracy), while omissions were decreased in both groups. Sub-chronic treatment with (0.019 micromol/kg) decreased omissions in all rats, but enhanced accuracy primarily in poor performing rats. These experiments demonstrate that an alpha4beta2 nicotinic agonist can enhance attention, but accuracy effects may only be observed under specific conditions. Moreover, a reduction in omissions was more reliably observed than improvements in accuracy, resulting in a net increase in signals successfully detected.


Subject(s)
Azetidines/pharmacology , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Reaction Time , Receptors, Nicotinic/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats
13.
Brain Res ; 1311: 136-47, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19944081

ABSTRACT

Mutant Tg2576 mice which possess the human "Swedish" APP mutation have been shown to demonstrate both Abeta plaque pathology and memory deficits in behavioral tasks. These mice are routinely maintained on a mixed C57BL/6xSJL genetic background which exhibits a high frequency of retinal degeneration allele and high variability in many behavioral assays. The same APP mutation is also available maintained on a 129 genetic background, providing more genetic homogeneity, but little data are published regarding the effects of the mutation on this background. We investigated whether transgenic mice expressing the Swedish mutation on the 129 background show similar behavioral deficits and Abeta pathology as those on the mixed background. Mice on the 129 background were tested at 6-7, 11-12, or 18-19 months of age in locomotor activity, Y-maze spontaneous alternation, and contextual fear conditioning. Differences were detected between WT and Tg mice in locomotor activity at 6-7 and 18-19 months, Y-maze at 6-7 and 11-12 months, and fear conditioning at 6-7, 11-12, and 18-19 months. In contrast, Tg mice on the mixed B6/SJL background tested at 6-7 months only demonstrated significant impairment in the contextual fear conditioning assay and in the Y-maze in one of 2 cohorts tested. Despite the behavioral differences observed, similar Abeta pathology was observed between Tg mice on the two genetic backgrounds. These results indicate that mice on the 129 genetic background may generate more consistent and robust behavioral differences, providing a useful model for testing therapeutic agents for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/genetics , Behavior, Animal , Disease Models, Animal , Receptors, Cell Surface/genetics , Aging , Alzheimer Disease/blood , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/blood , Animals , Brain/pathology , Conditioning, Classical , Fear , Humans , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Motor Activity/genetics , Mutation , Plaque, Amyloid/pathology , Protease Nexins , Species Specificity
14.
Neuropharmacology ; 58(2): 537-43, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19703478

ABSTRACT

Most animal models of pain cannot separate the sensory and affective components of pain. One model that has been used to assess affective pain is the place escape avoidance paradigm (PEAP). The aim of the current study is two-fold. First, validate PEAP with Complete Freund's Adjuvant (CFA)-induced inflammation for the assessment of the affective component of pain using the reference analgesics celecoxib, diclofenac and duloxetine; fluoxetine and scopolamine were tested as negative controls. Secondly, determine if there is a difference in efficacy in PEAP in comparison to the effects of the same compounds on von Frey-evoked mechanical allodynia in CFA animals. All compounds were tested in mechanical allodynia, place escape/avoidance, and for potentially confounding side effects in locomotor activity. Results show that celecoxib, diclofenac, and duloxetine significantly increased the time spent on the side associated with stimulation of the injured paw, whereas fluoxetine and scopolamine had no effect. Higher doses of celecoxib, diclofenac, duloxetine, and fluoxetine were required to attenuate von Frey-evoked mechanical allodynia. In the side effect assays, only fluoxetine decreased locomotor activity at doses used in PEAP. These results show that in inflammatory pain induced by CFA injection, PEAP is more sensitive to the effects of pain relieving compounds than mechanical allodynia. Fluoxetine showed efficacy in the mechanical allodynia test, but not PEAP, whereas duloxetine showed efficacy in mechanical allodynia and PEAP. These studies show that methods other than reflex based measures of pain such as affective pain models could be more predictive of efficacy/potency in the clinic.


Subject(s)
Disease Models, Animal , Inflammation/psychology , Pain/psychology , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/therapeutic use , Animals , Behavior, Animal/drug effects , Celecoxib , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/therapeutic use , Diclofenac/administration & dosage , Diclofenac/therapeutic use , Dose-Response Relationship, Drug , Duloxetine Hydrochloride , Escape Reaction/drug effects , Fluoxetine/administration & dosage , Fluoxetine/therapeutic use , Inflammation/physiopathology , Male , Motor Activity/drug effects , Neuropsychological Tests , Pain/chemically induced , Pain/drug therapy , Pain Measurement , Physical Stimulation , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Scopolamine/administration & dosage , Scopolamine/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Thiophenes/administration & dosage , Thiophenes/therapeutic use , Treatment Outcome
15.
Eur J Pharmacol ; 613(1-3): 39-45, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19376109

ABSTRACT

This study was conducted to characterize movement-induced pain in a rat model of knee joint osteoarthritis and validate this behavioral assessment by evaluating the effects of clinically used analgesic compounds. Unilateral intra-articular administration of a chondrocyte glycolytic inhibitor monoiodoacetate, was used to induce knee joint osteoarthritis in Sprague-Dawley rats. In this osteoarthritis model, histologically erosive disintegration of the articular surfaces of the ipsilateral joint are observed which closely mimic the clinical picture of osteoarthritis. Movement-induced pain behavior was measured using hind limb compressive grip force evaluation. The animals exhibited pain behaviors epitomized by a long-lasting decrement in bilateral compressive hind limb grip force following unilateral knee injury. The effects of clinically used reference analgesics were evaluated 20 days following i.a. injection of monoiodoacetate. Full analgesic activity was observed for tramadol, celecoxib and diclofenac; moderate effects for indomethacin, duloxetine and gabapentin but weak or no effects for acetaminophen, ibuprofen and lamotrigine. As morphine reduced grip force in naïve rats, its analgesic effects could not be accurately evaluated in this model. Finally, the effects of celecoxib were maintained following chronic dosing. The results indicate that this in vivo model utilizing a movement-induced pain behavior spawned by knee joint osteoarthritis may provide a valuable tool in examining the role of potential analgesic targets in osteoarthritic pain. As the model is clinically relevant, it will further enhance the mechanistic understanding of chronic arthritic joint pain and help in developing newer and better therapeutic strategies to manage osteoarthritis pain.


Subject(s)
Analgesics/pharmacology , Movement , Osteoarthritis/drug therapy , Pain/drug therapy , Pain/etiology , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Analgesics/therapeutic use , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Male , Osteoarthritis/complications , Pain/complications , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Time Factors
16.
Behav Brain Res ; 198(1): 83-90, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-18996151

ABSTRACT

The purinergic P2X(7) receptor is a ligand-gated ion channel found on peripheral macrophages and microglia in the nervous system. Activation of P2X(7) receptors results in the rapid release of interleukin-1 beta (IL-1 beta). Cytokines like IL-1 beta are suggested to be involved in the pathophysiology of depression. The aim of this study was to behaviorally profile P2X(7) receptor knockout (KO) mice in behavioral models of depression- and anxiety-like behaviors. P2X(7) receptor KO and wild type (WT) mice were tested in multiple models including; forced swim test, tail suspension test, elevated plus maze, novelty suppressed feeding, spontaneous locomotor activity, and food intake. P2X(7) receptor KO mice exhibited an antidepressant-like profile in tail suspension test and forced swim test; an effect that was not associated with changes in spontaneous locomotor activity. In addition, P2X(7) receptor KO mice showed higher responsivity to a subefficacious dose of the antidepressant drug imipramine (15 mg/kg) in forced swim test. No significant differences between genotypes were observed in models of anxiety. These data support the relevance of pro-inflammatory cytokines in depressive-like states, and suggest that P2X(7) receptor antagonists could be of potential interest for the treatment of affective disorders.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Depression/physiopathology , Mice, Knockout/physiology , Receptors, Purinergic P2/genetics , Analysis of Variance , Animals , Antidepressive Agents, Tricyclic/administration & dosage , Antidepressive Agents, Tricyclic/pharmacology , Anxiety/immunology , Behavior, Animal/drug effects , Depression/immunology , Disease Models, Animal , Eating , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/physiology , Hindlimb Suspension/physiology , Imipramine/administration & dosage , Imipramine/pharmacology , Interleukin-1beta/analysis , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Polymerase Chain Reaction , Receptors, Purinergic P2/deficiency , Swimming
17.
J Neurosci ; 28(14): 3769-80, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18385335

ABSTRACT

Because modulation of cortical cholinergic neurotransmission has been hypothesized to represent a necessary mechanism mediating the beneficial cognitive effects of nicotine and nicotinic acetylcholine receptor (nAChR) subtype-selective agonists, we used choline-sensitive microelectrodes for the real-time measurement of ACh release in vivo, to characterize cholinergic transients evoked by nicotine and the alpha4beta2*-selective nAChR partial agonist 2-methyl-3-(2-(S)-pyrrolindinylmethoxy)pyridine dihydrochloride (ABT-089), a clinically effective cognition enhancer. In terms of cholinergic signal amplitudes, ABT-089 was significantly more potent than nicotine in evoking ACh cholinergic transients. Moreover, cholinergic signals evoked by ABT-089 were characterized by faster signal rise time and decay rate. The nAChR antagonist mecamylamine attenuated the cholinergic signals evoked by either compound. Cholinergic signals evoked by ABT-089 were more efficaciously attenuated by the relatively beta2*-selective nAChR antagonist dihydro-beta-erythroidine. The alpha7 antagonist methyllycaconitine did not affect choline signal amplitudes but partly attenuated the relatively slow decay rate of nicotine-evoked cholinergic signals. Furthermore, the AMPA receptor antagonist DNQX as well as the NMDA receptor antagonist APV more potently attenuated cholinergic signals evoked by ABT-089. Using glutamate-sensitive microelectrodes to measure glutamatergic transients, ABT-089 was more potent than nicotine in evoking glutamate release. Glutamatergic signals were highly sensitive to tetrodotoxin-induced blockade of voltage-regulated sodium channels. Together, the present evidence indicates that compared with nicotine, ABT-089 evokes more potent and sharper cholinergic transients in prefrontal cortex. Glutamatergic mechanisms necessarily mediate the cholinergic effects of nAChR agonists in the prefrontal cortex.


Subject(s)
Acetylcholine/metabolism , Glutamic Acid/metabolism , Nicotinic Agonists/pharmacology , Prefrontal Cortex/drug effects , Pyridines/pharmacology , Pyrrolidines/pharmacology , Analysis of Variance , Animals , Choline/metabolism , Dihydro-beta-Erythroidine/pharmacology , Dose-Response Relationship, Drug , Electrochemistry/methods , Evoked Potentials/drug effects , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Male , Mecamylamine/pharmacology , Neostigmine/pharmacology , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Quinoxalines/pharmacology , Rats , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
18.
Expert Opin Drug Discov ; 3(9): 1027-40, 2008 Sep.
Article in English | MEDLINE | ID: mdl-23506178

ABSTRACT

BACKGROUND: Nicotine has a variety of effects in humans and in experimental animals that are suggestive of therapeutic potential. However, nicotine has a variety of adverse effects that preclude its development as a therapeutic agent, with the exception of its short-term use as a smoking cessation aide. OBJECTIVE: In this paper, we describe the clinical evidence supporting the potential of new nicotinic acetylcholine receptor (nAChR) agonists for the treatment of CNS conditions and review discovery strategies for targeting neuronal nAChRs as well as some of the opportunities and challenges still ahead. METHOD: The scientific literature and company websites are used for this literature review. CONCLUSION: Advances in the understanding of nAChRs during the past two decades have revealed evidence of considerable subtype diversity and hence the possibility of targeted nAChR ligands as improved therapeutics. Several new nAChR agonists have advanced to Phase II clinical trials and have produced signals of efficacy in Alzheimer's disease, attention deficit hyperactivity disorder, cognitive deficits of schizophrenia and pain. In addition, the neuronal nAChR partial agonist varenicline recently received regulatory approval for use in smoking cessation.

19.
J Neurosci ; 27(39): 10578-87, 2007 Sep 26.
Article in English | MEDLINE | ID: mdl-17898229

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (nAChR) plays an important role in cognitive processes and may represent a drug target for treating cognitive deficits in neurodegenerative and psychiatric disorders. In the present study, we used a novel alpha7 nAChR-selective agonist, 2-methyl-5-(6-phenyl-pyridazin-3-yl)-octahydro-pyrrolo[3,4-c]pyrrole (A-582941) to interrogate cognitive efficacy, as well as examine potential cellular mechanisms of cognition. Exhibiting high affinity to native rat (Ki = 10.8 nM) and human (Ki = 16.7 nM) alpha7 nAChRs, A-582941 enhanced cognitive performance in behavioral assays including the monkey delayed matching-to-sample, rat social recognition, and mouse inhibitory avoidance models that capture domains of working memory, short-term recognition memory, and long-term memory consolidation, respectively. In addition, A-582941 normalized sensory gating deficits induced by the alpha7 nAChR antagonist methyllycaconitine in rats, and in DBA/2 mice that exhibit a natural sensory gating deficit. Examination of signaling pathways known to be involved in cognitive function revealed that alpha7 nAChR agonism increased extracellular-signal regulated kinase 1/2 (ERK1/2) phosphorylation in PC12 cells. Furthermore, increases in ERK1/2 and cAMP response element-binding protein (CREB) phosphorylation were observed in mouse cingulate cortex and/or hippocampus after acute A-582941 administration producing plasma concentrations in the range of alpha7 binding affinities and behavioral efficacious doses. The MEK inhibitor SL327 completely blocked alpha7 agonist-evoked ERK1/2 phosphorylation. Our results demonstrate that alpha7 nAChR agonism can lead to broad-spectrum efficacy in animal models at doses that enhance ERK1/2 and CREB phosphorylation/activation and may represent a mechanism that offers potential to improve cognitive deficits associated with neurodegenerative and psychiatric diseases, such as Alzheimer's disease and schizophrenia.


Subject(s)
Central Nervous System Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mental Processes/drug effects , Receptors, Nicotinic , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Cognition/drug effects , Cognition/physiology , Humans , Learning/drug effects , Learning/physiology , Macaca mulatta , Male , Mental Processes/physiology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Pyridazines/pharmacology , Pyrroles/pharmacology , Rats , Signal Transduction , Treatment Outcome , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
20.
Biochem Pharmacol ; 74(8): 1212-23, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17689498

ABSTRACT

Attention deficit/hyperactivity disorder (ADHD) is the most commonly diagnosed neurobehavioral disorder in children and adolescents, and in about half of these patients, significant symptomology continues into adulthood. Although impulsivity and hyperactivity are the most salient features of ADHD, cognitive deficits, particularly impairments in attention and executive function, are an important component, particularly in adolescents and adults, with over 90% of adults seeking treatment for ADHD manifesting cognitive dysfunction. Currently available medications treat the core ADHD symptoms but typically do not adequately address cognitive aspects of ADHD, underscoring the need for new therapeutics. Dopamine and norepinephrine are hypothesized to be particularly important in ADHD, but there is emerging evidence that cholinergic neurotransmission, particularly involving neuronal nicotinic acetylcholine receptors (nAChRs), may play a role in the pathophysiology of ADHD. Nicotine has demonstrated procognitive effects in both humans and experimental animals and has produced signals of efficacy in small proof-of-concept adult ADHD trials. Although adverse effects associated with nicotine preclude its development as a therapeutic, a number of novel nAChR agonists with improved safety/tolerability profiles have been discovered. Of these, ABT-418 and ABT-089 have both demonstrated signals of efficacy in adults with ADHD. Notably, tolerability issues that might be expected of a nAChR agonist, such as nausea and emesis, were not observed at efficacious doses of ABT-089. Further understanding of the effects of novel neuronal nAChR agonists on specific aspects of cognitive functioning in ADHD is required to assess the full potential of this approach.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Cognition/drug effects , Isoxazoles/therapeutic use , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Animals , Humans , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/physiology , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...