Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 347(1): 225-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23902940

ABSTRACT

Abnormal involuntary movements or dyskinesias are a serious complication of long-term l-DOPA treatment of Parkinson's disease, for which there are few treatment options. Accumulating preclinical data show that nicotine decreases l-DOPA-induced dyskinesias (LIDs), suggesting that it may be a useful antidyskinetic therapy for Parkinson's disease. Here, we investigated whether nicotinic acetylcholine receptor (nAChR) agonists reduced LIDs in nonhuman primates. We first tested the nonselective nAChR agonist 1, 6,7,8,9-tetrahydro-6,10-methano-6H-pyrazino[2,3-h][3]benzazepine (varenicline), which offers the advantage that it is approved by the U.S. Food and Drug Administration for use in humans. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys (n = 23) were first administered l-DOPA/carbidopa (10/2.5 mg/kg) twice daily 5 days/week until stably dyskinetic. Oral varenicline (0.03-0.10 mg/kg) decreased LIDs ∼50% compared with vehicle-treated monkeys, whereas nicotine treatment (300 µg/ml in drinking water) reduced LIDs by 70% in a parallel group of animals. We next tested the selective α4ß2*/α6ß2* nAChR agonist TC-8831 [3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane] on LIDs in the same set of monkeys after a 10-week washout. We also tested TC-8831 in another set of MPTP-lesioned monkeys (n = 16) that were nAChR drug-naïve. Oral TC-8831 (0.03-0.3 mg/kg) reduced LIDs in both sets by 30-50%. After a washout period, repeat TC-8831 dosing led to a greater decline in LIDs (60%) in both sets of monkeys that was similar to the effect of nicotine. Tolerance to any nAChR drug did not develop over the course of the study (3-4 months). NAChR drug treatment did not worsen parkinsonism or cognitive ability. These data suggest that nAChR agonists may be useful for the management of dyskinesias in l-DOPA-treated Parkinson's disease patients.


Subject(s)
Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Levodopa/toxicity , Nicotinic Agonists/therapeutic use , Parkinson Disease/drug therapy , Animals , Dyskinesia, Drug-Induced/physiopathology , Female , Male , Nicotine/therapeutic use , Parkinson Disease/physiopathology , Receptors, Nicotinic/physiology , Saimiri
2.
Pharmacol Biochem Behav ; 111: 1-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23872135

ABSTRACT

The degeneration of the nigrostriatal dopamine (DA) system underlies the motor deficits in Parkinson's disease (PD). In recent years, epidemiological reports that smokers have lower incidences of PD have brought attention to the nicotinic acetylcholine system as a potential target for novel therapeutics. Nicotine, an agonist of neuronal nicotinic receptors (NNRs), modulates functions relevant to PD via stimulation of dopaminergic transmission in the nigrostriatal pathway, particularly via activation of α6ß2* and α4ß2* NNRs. Recently, reduced support of DA neurons by neurotrophic growth factors has been described in PD. Fibroblast growth factor (FGF) is critical for the development and protection of adult DA neurons. In FGF-2 knockout mice and the related th-fgfr1(tk-) mouse model there is heightened sensitivity to DA neuronal oxidative neurotoxin 6-hydroxydopamine (6-OHDA). In the present study, FGF-deficient transgenic mice th-fgfr1(tk-) were used to analyze the effects of novel full (TC-8831) and partial (TC-8581) agonists of ß2-containing nicotinic receptors on impaired motor behavior following unilateral 6-OHDA lesions. The lesions generated spontaneous (drug-naïve) turning asymmetries that correlated exponentially with the depletion of DA biomarkers in the lesioned striata. These mice also exhibited a reduced capacity to remain on the accelerating rotarod. Oral administration of TC-8831, an NNR agonist with high specificity for ß2 subunits and a full agonist at producing DA release from striatal synaptosomes, attenuated unidirectional turning and improved motor coordination. In contrast, partial ß2 NNR agonist TC-8581 had no effect on behaviors in this model. This study demonstrates the potential of NNR targeting-compounds to facilitate motor function in PD.


Subject(s)
Azabicyclo Compounds/pharmacology , Cyclopropanes/pharmacology , Disease Models, Animal , Motor Activity/drug effects , Neurons/drug effects , Nicotinic Agonists/pharmacology , Parkinson Disease/physiopathology , Pyridines/pharmacology , Receptors, Nicotinic/physiology , Animals , Behavior, Animal , Cell Line , Dopamine/metabolism , Mice , Mice, Knockout , Neurons/metabolism
3.
Neuropharmacology ; 73: 337-47, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770260

ABSTRACT

Long-term L-DOPA treatment for Parkinson's disease (PD) is limited by motor complications, particularly L-DOPA-induced dyskinesia (LID). A therapy with the ability to ameliorate LID without reducing anti-parkinsonian benefit would be of great value. We assessed the ability of TC-8831, an agonist at nicotinic acetylcholine receptors (nAChR) containing α6ß2/α4ß2 subunit combinations, to provide such benefits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP) lesioned macaques with established LID. Animals were treated orally for consecutive 14-day periods with twice-daily vehicle (weeks 1-2) or TC-8831 (0.03, 0.1 or 0.3 mg/kg, weeks 3-8). L-DOPA was also administered, once-daily, (weeks 1-12, median-dose 30 mg/kg, p.o.). For the following two-weeks (weeks 9-10), TC-8831 was washed out, while once-daily L-DOPA treatment was maintained. The effects of once-daily amantadine (3 mg/kg, p.o.) were then assessed over weeks 11-12. LID, parkinsonism, duration and quality of ON-time were assessed weekly by a neurologist blinded to treatment. TC-8831 reduced the duration of 'bad' ON-time (ON-time with disabling dyskinesia) by up to 62% and decreased LID severity (median score 18 cf. 34 (vehicle), 0.1 mg/kg, 1-3 h period). TC-8831 also significantly reduced choreiform and dystonic dyskinesia (median scores 6 and 31 cf. 19 and 31 respectively (vehicle), both 0.03 mg/kg, 1-3 h). At no time did TC-8831 treatment result in a reduction in anti-parkinsonian benefit of L-DOPA. By comparison, amantadine also significantly reduced dyskinesia and decreased 'bad' ON-time (up to 61%) but at the expense of total ON-time (reduced by up to 23%). TC-8831 displayed robust anti-dyskinetic actions and improved the quality of ON-time evoked by L-DOPA without any reduction in anti-parkinsonian benefit.


Subject(s)
Azabicyclo Compounds/therapeutic use , Cyclopropanes/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , MPTP Poisoning/drug therapy , Nicotinic Agonists/therapeutic use , Amantadine/therapeutic use , Animals , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/complications , Female , Levodopa , MPTP Poisoning/complications , Macaca fascicularis
4.
Neuropharmacology ; 71: 191-203, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583932

ABSTRACT

L-Dopa-induced dyskinesias are a serious long-term side effect of dopamine replacement therapy for Parkinson's disease for which there are few treatment options. Our previous studies showed that nicotine decreased l-dopa-induced abnormal involuntary movements (AIMs). Subsequent work with knockout mice demonstrated that α6ß2* nicotinic receptors (nAChRs) play a key role. The present experiments were done to determine if α4ß2* nAChRs are also involved in l-dopa-induced dyskinesias. To approach this, we took advantage of the finding that α6ß2* nAChRs are predominantly present on striatal dopaminergic nerve terminals, while a significant population of α4ß2* nAChRs are located on other neurons. Thus, a severe dopaminergic lesion would cause a major loss in α6ß2*, but not α4ß2* nAChRs. Experiments were therefore done in which rats were unilaterally lesioned with 6-hydroxydopamine, at a dose that led to severe nigrostriatal damage. The dopamine transporter, a dopamine nerve terminal marker, was decreased by >99%. This lesion also decreased striatal α6ß2* nAChRs by 97%, while α4ß2* nAChRs were reduced by only 12% compared to control. A series of ß2* nAChR compounds, including TC-2696, TI-10165, TC-8831, TC-10600 and sazetidine reduced l-dopa-induced AIMs in these rats by 23-32%. TC-2696, TI-10165, TC-8831 were also tested for parkinsonism, with no effect on this behavior. Tolerance did not develop with up to 3 months of treatment. Since α4α5ß2 nAChRs are also predominantly on striatal dopamine terminals, these data suggest that drugs targeting α4ß2 nAChRs may reduce l-dopa-induced dyskinesias in late stage Parkinson's disease.


Subject(s)
Antiparkinson Agents/adverse effects , Azabicyclo Compounds/therapeutic use , Cyclopropanes/therapeutic use , Disease Models, Animal , Dyskinesia, Drug-Induced/prevention & control , Levodopa/adverse effects , Nicotinic Agonists/therapeutic use , Parkinson Disease/drug therapy , Receptors, Nicotinic/metabolism , Animals , Anti-Dyskinesia Agents/therapeutic use , Antiparkinson Agents/therapeutic use , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agonists/adverse effects , Dopamine Agonists/therapeutic use , Drug Tolerance , Female , Levodopa/therapeutic use , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/chemistry , Synaptosomes/drug effects , Synaptosomes/metabolism
5.
J Pain ; 13(12): 1162-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23182225

ABSTRACT

UNLABELLED: Alpha-7 nicotinic acetylcholine receptor (α7 nAChR) agonists attenuate pain and inflammation in preclinical models. This study tested whether systemic delivery of an α7 nAChR agonist attenuates neuropathic pain and associated immune-mediated pro-inflammation. Hind paw response thresholds to mechanical stimuli in male Sprague Dawley rats were assessed before and after sciatic chronic constriction injury (CCI) or sham surgery. Osmotic mini-pumps containing TC-7020, an α7 nAChR selective agonist, were implanted 10 to 14 days after surgery. TC-7020 (1, 3, and 10 mg/kg/d; s.c.) significantly attenuated CCI-induced allodynia, which lasted through 2 weeks of test compound administration. Spinal cords were collected after 2 weeks and processed for microglial and astrocyte activation markers within the ipsilateral L4-L6 dorsal horn. In addition, ipsilateral L4-5 dorsal root ganglia (DRGs) were processed for neuronal injury and satellite cell activation markers. CCI-induced central glial cell activation markers were not suppressed by TC-7020, even though TC-7020 is mildly blood-brain barrier permeable. However, TC-7020 downregulated the integrated density of activation transcription factor 3 (ATF3) but not the number of ATF positive cells. TC-7020 also downregulated phosphorylated extracellular signal kinase (p-ERK) and satellite cell activation in the CCI-affected DRGs. Therefore, systemic α7 nAChR agonist may be effective in treating neuropathic pain via reducing neuronal injury and immune cells activation occurring in the periphery. PERSPECTIVE: These studies demonstrated that TC-7020, an alpha7 nicotinic acetylcholine receptor agonist with partial blood-brain barrier permeability, reversed neuropathic pain in rats, likely via attenuation of inflammation in the DRG and/or the site of sciatic injury.


Subject(s)
Neuralgia/drug therapy , Nicotinic Agonists/administration & dosage , Quinuclidines/administration & dosage , Receptors, Nicotinic/physiology , Thiophenes/administration & dosage , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Male , Neuralgia/metabolism , Neuralgia/pathology , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
6.
Eur J Pharm Sci ; 47(5): 813-23, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23036283

ABSTRACT

Nicotinic α4ß2* agonists are known to be effective in a variety of preclinical pain models, but the underlying mechanisms of analgesic action are not well-understood. In the present study, we characterized activation and desensitization properties for a set of seventeen novel α4ß2*-selective agonists that display druggable physical and pharmacokinetic attributes, and correlated the in vitro pharmacology results to efficacies observed in a mouse formalin model of analgesia. ABT-894 and Sazetidine-A, two compounds known to be effective in the formalin assay, were included for comparison. The set of compounds displayed a range of activities at human (α4ß2)(2)ß2 (HS-α4ß2), (α4ß2)(2)α5 (α4ß2α5) and (α4ß2)(2)α4 (LS-α4ß2) receptors. We report the novel finding that desensitization of α4ß2* receptors may drive part of the antinociceptive outcome. Our molecular modeling approaches revealed that when receptor desensitization rather than activation activitiesat α4ß2* receptors are considered, there is a better correlation between analgesia scores and combined in vitro properties. Our results suggest that although all three α4ß2 subtypes assessed are involved, it is desensitization of α4ß2α5 receptors that plays a more prominent role in the antinociceptive action of nicotinic compounds. For modulation of Phase I responses, correlations are significantly improved from an r(2) value of 0.53 to 0.67 and 0.66 when HS- and LS-α4ß2 DC(50) values are considered, respectively. More profoundly, considering the DC(50) at α4ß2α5 takes the r(2) from 0.53 to 0.70. For Phase II analgesia scores, adding HS- or LS-α4ß2 desensitization potencies did not improve the correlations significantly. Considering the α4ß2α5 DC(50) value significantly increased the r(2) from 0.70 to 0.79 for Phase II, and strongly suggested a more prominent role for α4ß2α5 nAChRs in the modulation of pain in the formalin assay. The present studies demonstrate that compounds which are more potent at desensitization of α4ß2* receptors display better analgesia scores in the formalin test. Consideration of desensitization propertiesat α4ß2* receptors, especially at α4ß2α5, in multiple linear regression analyses significantly improves correlations with efficacies of analgesia. Thus, α4ß2* nicotinic acetylcholine receptor desensitization may contribute to efficacy in the mediation of pain, and represent a mechanism for analgesic effects mediated by nicotinic agonists.


Subject(s)
Analgesics/therapeutic use , Nicotinic Agonists/therapeutic use , Pain/drug therapy , Receptors, Nicotinic/physiology , Analgesics/pharmacology , Animals , Binding, Competitive , Cell Line , Cell Line, Tumor , Formaldehyde , HEK293 Cells , Humans , Male , Mice , Motor Activity/drug effects , Nicotinic Agonists/pharmacology , PC12 Cells , Pain/chemically induced , Pain/physiopathology , Rats
7.
Neuropharmacology ; 62(7): 2239-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22306792

ABSTRACT

The interaction of 13-desmethylspirolide C (SPX-desMe-C) and gymnodimine with several nicotinic and muscarinic acetylcholine receptors was investigated. Interaction at the muscarinic receptors was minimal. At nicotinic receptors, both SPX-desMe-C and gymnodimine displayed greatest affinity for the α7 receptor. The rank order for binding affinity (Ki) for SPX-desMe-C was α7 > α6ß3ß4α5 >> rat α3ß4, α1ßγδ > α4ß4, human α3ß4 > human α4ß2 > rat α4ß2 and for gymnodimine was α7, α6ß3ß4α5 > rat α3ß4 > human α3ß4, α4ß4 > rat α4ß2, human α4ß2 > α1ßγδ. Both molecules antagonized agonist-induced nicotinic responses. The antagonism rank order of potency (IC(50)) for SPX-desMe-C was α7 > low sensitivity (LS) α4ß2 > human α3ß4 > high sensitivity (HS) α4ß2, α1ßγδ > α4ß4 > rat α3ß4 and for gymnodimine was LS α4ß2 > human α3ß4 > α7 > HS α4ß2 > α4ß4 > rat α3ß4 > α1ßγδ. Neither gymnodimine nor SPX-desMe-C antagonism could be surmounted by increasing concentrations of nicotine. To elucidate the nature of this insurmountable blockade, we carried out homology modelling and molecular docking studies of both ligands with α7 nAChR. Their very high binding affinity results from very tight hydrophobic enclosures, in addition to previously reported hydrogen-bond and cation-π interactions. Also, the higher the hydrophilic surface area of the binding site of nAChRs, the weaker the binding affinity of both ligands. Together these results show the targets of action are nicotinic and define these marine toxins as additional tools to advance our understanding regarding interactions between antagonists and the nAChR ligand binding domain.


Subject(s)
Heterocyclic Compounds, 3-Ring/metabolism , Hydrocarbons, Cyclic/metabolism , Imines/metabolism , Marine Toxins/metabolism , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/metabolism , Spiro Compounds/metabolism , Animals , Binding Sites/physiology , Binding, Competitive/physiology , Female , Humans , Rats , Rats, Sprague-Dawley
8.
Biochem Pharmacol ; 82(8): 862-72, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21736871

ABSTRACT

Recent progress has been made in the understanding of the anatomical distribution, composition, and physiological role of nicotinic acetylcholine receptors containing the α6 subunit. Extensive study by many researchers has indicated that a collection of α6-containing receptors representing a nicotinic sub-family is relevant in preclinical models of nicotine self-administration and locomotor activity. Due to a number of technical difficulties, the state of the art of in vitro model systems expressing α6-containing receptors has lagged behind the state of knowledge of native α6 nAChR subunit composition. Several techniques, such as the expression of chimeric and concatameric α6 subunit constructs in oocytes and mammalian cell lines have been employed to overcome these obstacles. There remains a need for other critical tools, such as selective small molecules and radioligands, to advance the field of research and to allow the discovery and development of potential therapeutics targeting α6-containing receptors for smoking cessation, Parkinson's disease and other disorders.


Subject(s)
Biomedical Research , Drug Discovery , Receptors, Nicotinic , Animals , Biomedical Research/methods , Biomedical Research/trends , Drug Discovery/methods , Drug Discovery/trends , Humans , Ligands , Protein Binding , Protein Subunits , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/physiology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
9.
J Mol Neurosci ; 30(1-2): 19-20, 2006.
Article in English | MEDLINE | ID: mdl-17192610

ABSTRACT

To date, the primary treatments for Alzheimer's disease with proven efficacy have been acetylcholinesterase inhibitors that prevent the hydrolysis of acetylcholine (ACh) in the synaptic cleft, thereby prolonging its activity. Although these agents have some benefit in alleviating cognitive impairment, they have limited clinical utility because of insufficient efficacy and marginal tolerability. Within the last decade, there has been much experimental support for the use of therapeutics that directly target nicotinic ACh receptors (nAChRs) to improve cognitive function and slow neurodegenerative disease progression. These findings have spurred considerable research efforts to develop ligands selective for nAChRs, such as ABT-418 (Arneric et al., 1995), SIB-1553 (Bontempi et al., 2001), TC-2403 (Lippiello et al., 1996), and TC-2559 (Bencherif et al., 2000). There is abundant evidence that nAChR modulators have the potential to alleviate cognitive impairment in demented states. In addition to improving cognitive function, a large body of research implicates a role for nAChRs in neuroprotection, suggesting potential for disease modification. An impact of nAChR agonists on disease progression would provide an advantage over currently available treatments for memory loss. The profile of previous nAChR-targeted clinical candidates has not been adequate to warrant further development owing to poor oral bioavailability, side effects, and/or lack of efficacy. Thus, a challenge in nAChR drug design and development has been the reduction of undesirable effects that result from activity at specific nAChRs in the CNS and PNS, including cardiovascular toxicity, emesis, seizures, and hypothermia.


Subject(s)
Alzheimer Disease/drug therapy , Cognition/drug effects , Neuroprotective Agents/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Nicotinic/physiology , Administration, Oral , Alzheimer Disease/psychology , Animals , Humans , Memory/drug effects , Receptors, Nicotinic/drug effects
10.
Trends Pharmacol Sci ; 26(7): 352-60, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946748

ABSTRACT

Agonists of nicotinic acetylcholine receptors (nAChRs) produce long-lasting cognitive effects in animal models and humans. The duration of these cognitive effects can outlast the presence of the agonists in the system, and the persistence of cognitive enhancement is increased further by repeated exposure. The basis for this discrepancy appears be the cellular and systemic mechanisms of learning and memory. Agonists of nAChRs induce long-term potentiation (LTP), which is a strengthening of synaptic connections that is associated with learning and memory formation. Some of the cellular effects of nAChR agonists overlap with the known cellular mechanisms of LTP, including long-lasting increases in intracellular concentrations of Ca2+, activation of second-messenger systems and transcription factors, elevated levels of gene products and enhanced neurotransmitter release. A better understanding of this phenomenon might shed new light on the role of nAChR systems in memory formation and retrieval.


Subject(s)
Cognition Disorders/drug therapy , Nicotinic Agonists/therapeutic use , Cognition/drug effects , Cognition Disorders/psychology , Humans , Long-Term Potentiation , Nicotine/pharmacology , Nicotinic Agonists/pharmacokinetics
11.
J Pharmacol Exp Ther ; 312(2): 619-26, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15356217

ABSTRACT

Heterologous expression and lesioning studies were conducted to identify possible subunit assembly partners in nicotinic acetylcholine receptors (nAChR) containing alpha6 subunits (alpha6(*) nAChR). SH-EP1 human epithelial cells were transfected with the requisite subunits to achieve stable expression of human alpha6beta2, alpha6beta4, alpha6beta2beta3, alpha6beta4beta3, or alpha6beta4beta3alpha5 nAChR. Cells expressing subunits needed to form alpha6beta4beta3alpha5 nAChR exhibited saturable [(3)H]epibatidine binding (K(d) = 95.9 +/- 8.3 pM and B(max) = 84.5 +/- 1.6 fmol/mg of protein). The rank order of binding competition potency (K(i)) for prototypical nicotinic compounds was alpha-conotoxin MII (6 nM) > nicotine (156 nM) approximately methyllycaconitine (200 nM) > alpha-bungarotoxin (>10 microM), similar to that for nAChR in dopamine neurons displaying a distinctive pharmacology. 6-Hydroxydopamine lesioning studies indicated that beta3 and alpha5 subunits are likely partners of the alpha6 subunits in nAChR expressed in dopaminergic cell bodies. Similar to findings in rodents, quantitative real-time reverse transcription-polymerase chain reactions of human brain indicated that alpha6 subunit mRNA expression was 13-fold higher in the substantia nigra than in the cortex or the rest of the brain. Thus, heterologous expression studies suggest that the human alpha5 subunit makes a critical contribution to alpha6beta4beta3alpha5 nAChR assembly into a ligand-binding form with native alpha6(*)-nAChR-like pharmacology and of potential physiological and pathophysiological relevance.


Subject(s)
Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Animals , Binding, Competitive/drug effects , Cell Line , Cell Membrane/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Male , RNA/biosynthesis , RNA/isolation & purification , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Substantia Nigra/drug effects , Substantia Nigra/metabolism
12.
CNS Drug Rev ; 10(2): 147-66, 2004.
Article in English | MEDLINE | ID: mdl-15179444

ABSTRACT

The development of selective ligands targeting neuronal nicotinic acetylcholine receptors to alleviate symptoms associated with neurodegenerative diseases presents the advantage of affecting multiple deficits that are the hallmarks of these pathologies. TC-1734 is an orally active novel neuronal nicotinic agonist with high selectivity for neuronal nicotinic receptors. Microdialysis studies indicate that TC-1734 enhances the release of acetylcholine from the cortex. TC-1734, by either acute or repeated administration, exhibits memory enhancing properties in rats and mice and is neuroprotective following excitotoxic insult in fetal rat brain in cultures and against alterations of synaptic transmission induced by deprivation of glucose and oxygen in hippocampal slices. At submaximal doses, TC-1734 produced additive cognitive effects when used in combination with tacrine or donepezil. Unlike (-)-nicotine, behavioral sensitization does not develop following repeated administration of TC-1734. Its pharmacokinetic (PK) profile (half-life of 2 h) contrasts with the long lasting improvement in working memory (18 h) demonstrating that cognitive improvement extends beyond the lifetime of the compound. The very low acute toxicity of TC-1734 and its receptor activity profile provides additional mechanistic basis for its suggested potential as a clinical candidate. TC-1734 was very well tolerated in acute and chronic oral toxicity studies in mice, rats and dogs. Phase I clinical trials demonstrated TC-1734's favorable pharmacokinetic and safety profile by acute oral administration at doses ranging from 2 to 320 mg. The bioavailability, pharmacological, pharmacokinetic, and safety profile of TC-1734 provides an example of a safe, potent and efficacious neuronal nicotinic modulator that holds promise for the management of the hallmark symptomatologies observed in dementia.


Subject(s)
Antidepressive Agents/pharmacology , Cognition/drug effects , Neuroprotective Agents/pharmacology , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Sympathomimetics/pharmacology , Administration, Oral , Adolescent , Adrenocorticotropic Hormone/drug effects , Adrenocorticotropic Hormone/metabolism , Adult , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinesterase Inhibitors/pharmacology , Discrimination Learning/drug effects , Dogs , Humans , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Middle Aged , Motor Activity/drug effects , Neuroprotective Agents/metabolism , Nicotinic Agonists/metabolism , Pyridines/chemistry , Pyridines/metabolism , Rats , Receptors, Nicotinic/metabolism , Reference Values , Sympathomimetics/metabolism , Toxicity Tests, Chronic
13.
Bioorg Med Chem ; 11(24): 5333-43, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14642577

ABSTRACT

In recent years, there has been considerable effort to design and synthesize radiotracers suitable for use in Positron Emission Tomography (PET) imaging of the alpha4beta2 neuronal nicotinic acetylcholine receptor (nAChR) subtype. A new fluoropyridinyl derivative of (-)-cytisine (1), namely (-)-9-(2-fluoropyridinyl)cytisine (3, K(i) values of 24 and 3462 nM for the alpha4beta2 and alpha7 nAChRs subtypes, respectively) has been synthesized in four chemical steps from (-)-cytisine and labelled with fluorine-18 (T(1/2): 119.8 min) using an efficient two-step radiochemical process [(a). nucleophilic heteroaromatic ortho-radiofluorination using the corresponding N-Boc-protected nitro-derivative, (b). TFA removal of the Boc protective group]. Typically, 20-45 mCi (0.74-1.67 GBq) of (-)-9-(2-[18F]fluoropyridinyl)cytisine ([18F]-3, 2-3 Ci/micromol or 74-111 GBq/micromol) were easily obtained in 70-75 min starting from a 100 mCi (3.7 GBq) aliquot of a cyclotron-produced [18F]fluoride production batch (20-45% non decay-corrected yield based on the starting [18F]fluoride). The in vivo pharmacological profile of (-)-9-(2-[18F]fluoropyridinyl)cytisine ([18F]-3) was evaluated in rats with biodistribution studies and brain radioactivity monitoring using intracerebral radiosensitive beta-microprobes. The observed in vivo distribution of the radiotracer in brain was rather uniform, and did not match with the known regional densities of nAChRs. It was also significantly different from that of the parent compound (-)-[3H]cytisine. Moreover, competition studies with (-)-nicotine (5 mg/kg, 5 min before the radiotracer injection) did not reduce brain uptake of the radiotracer. These experiments clearly indicate that (-)-9-(2-[18F]fluoropyridinyl)cytisine ([18F]-3) does not have the required properties for imaging nAChRs using PET.


Subject(s)
Azocines/chemical synthesis , Brain Chemistry , Pyridines/chemical synthesis , Receptors, Nicotinic/analysis , Tomography, Emission-Computed , Animals , Azocines/chemistry , Female , Fluorine Radioisotopes , Isotope Labeling , Kinetics , Ligands , Male , Molecular Structure , Pyridines/chemistry , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...