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










Publication year range
1.
Bioorg Med Chem Lett ; 26(12): 2915-2919, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27131990

ABSTRACT

This letter describes the further chemical optimization of the picolinamide-derived family of mGlu4 PAMs wherein we identified a 3-amino substituent to the picolinamide warhead that engendered potency, CNS penetration and in vivo efficacy. From this optimization campaign, VU0477886 emerged as a potent (EC50=95nM, 89% Glu Max) mGlu4 PAM with an attractive DMPK profile (brain:plasma Kp=1.3), rat CLp=4.0mL/min/kg, t1/2=3.7h) and robust efficacy in our standard preclinical Parkinson's disease model, haloperidol-induced catalepsy (HIC).


Subject(s)
Amides/pharmacology , Central Nervous System/drug effects , Drug Discovery , Picolines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Allosteric Regulation/drug effects , Amides/chemistry , Amides/metabolism , Animals , Central Nervous System/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Molecular Structure , Picolines/chemistry , Picolines/metabolism , Rats , Structure-Activity Relationship
2.
Neuropsychopharmacology ; 41(4): 1166-78, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26315507

ABSTRACT

Cocaine abuse remains a public health concern for which pharmacotherapies are largely ineffective. Comorbidities between cocaine abuse, depression, and anxiety support the development of novel treatments targeting multiple symptom clusters. Selective negative allosteric modulators (NAMs) targeting the metabotropic glutamate receptor 5 (mGlu5) subtype are currently in clinical trials for the treatment of multiple neuropsychiatric disorders and have shown promise in preclinical models of substance abuse. However, complete blockade or inverse agonist activity by some full mGlu5 NAM chemotypes demonstrated adverse effects, including psychosis in humans and psychotomimetic-like effects in animals, suggesting a narrow therapeutic window. Development of partial mGlu5 NAMs, characterized by their submaximal but saturable levels of blockade, may represent a novel approach to broaden the therapeutic window. To understand potential therapeutic vs adverse effects in preclinical behavioral assays, we examined the partial mGlu5 NAMs, M-5MPEP and Br-5MPEPy, in comparison with the full mGlu5 NAM MTEP across models of addiction and psychotomimetic-like activity. M-5MPEP, Br-5MPEPy, and MTEP dose-dependently decreased cocaine self-administration and attenuated the discriminative stimulus effects of cocaine. M-5MPEP and Br-5MPEPy also demonstrated antidepressant- and anxiolytic-like activity. Dose-dependent effects of partial and full mGlu5 NAMs in these assays corresponded with increasing in vivo mGlu5 occupancy, demonstrating an orderly occupancy-to-efficacy relationship. PCP-induced hyperlocomotion was potentiated by MTEP, but not by M-5MPEP and Br-5MPEPy. Further, MTEP, but not M-5MPEP, potentiated the discriminative-stimulus effects of PCP. The present data suggest that partial mGlu5 NAM activity is sufficient to produce therapeutic effects similar to full mGlu5 NAMs, but with a broader therapeutic index.


Subject(s)
Alkynes/administration & dosage , Alkynes/pharmacology , Alkynes/pharmacokinetics , Brain/drug effects , Cocaine/administration & dosage , Drug-Seeking Behavior/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Pyridines/pharmacokinetics , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Anti-Anxiety Agents/administration & dosage , Antidepressive Agents/administration & dosage , Conditioning, Classical/drug effects , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Male , Mice , Motor Activity/drug effects , Phencyclidine/administration & dosage , Rats, Sprague-Dawley , Self Administration , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
3.
J Med Chem ; 58(18): 7485-500, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26335039

ABSTRACT

Previous preclinical work has demonstrated the therapeutic potential of antagonists of the group II metabotropic glutamate receptors (mGlus). Still, compounds that are selective for the individual group II mGlus (mGlu2 and mGlu3) have been scarce. There remains a need for such compounds with the balance of properties suitable for convenient use in a wide array of rodent behavioral studies. We describe here the discovery of a selective mGlu3 NAM 106 (VU0650786) suitable for in vivo work. Compound 106 is a member of a series of 5-aryl-6,7-dihydropyrazolo[1,5-a]pyrazine-4(5H)-one compounds originally identified as a mGlu5 positive allosteric modulator (PAM) chemotype. Its suitability for use in rodent behavioral models has been established by extensive in vivo PK studies, and the behavioral experiments presented here with compound 106 represent the first examples in which an mGlu3 NAM has demonstrated efficacy in models where prior efficacy had previously been noted with nonselective group II antagonists.


Subject(s)
Anti-Anxiety Agents/chemistry , Antidepressive Agents/chemistry , Brain/metabolism , Heterocyclic Compounds, 2-Ring/chemistry , Pyridines/chemistry , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Animals , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Calcium/metabolism , Dogs , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Madin Darby Canine Kidney Cells , Mice , Microsomes, Liver/metabolism , Permeability , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
4.
Neuron ; 86(4): 1029-1040, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25937172

ABSTRACT

Schizophrenia is associated with disruptions in N-methyl-D-aspartate glutamate receptor subtype (NMDAR)-mediated excitatory synaptic signaling. The metabotropic glutamate receptor subtype 5 (mGlu5) is a closely associated signaling partner with NMDARs and regulates NMDAR function in forebrain regions implicated in the pathology of schizophrenia. Efficacy of mGlu5 positive allosteric modulators (PAMs) in animal models of psychosis and cognition was previously attributed to potentiation of NMDAR function. To directly test this hypothesis, we identified VU0409551 as a novel mGlu5 PAM that exhibits distinct stimulus bias and selectively potentiates mGlu5 coupling to Gαq-mediated signaling but not mGlu5 modulation of NMDAR currents or NMDAR-dependent synaptic plasticity in the rat hippocampus. Interestingly, VU0409551 produced robust antipsychotic-like and cognition-enhancing activity in animal models. These data provide surprising new mechanistic insights into the actions of mGlu5 PAMs and suggest that modulation of NMDAR currents is not critical for in vivo efficacy. VIDEO ABSTRACT.


Subject(s)
Antipsychotic Agents/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Allosteric Regulation/drug effects , Animals , Cognition/drug effects , Cognition/physiology , Glutamic Acid/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Signal Transduction/drug effects
5.
Neuropharmacology ; 95: 121-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25749357

ABSTRACT

Metabotropic glutamate receptor 4 (mGlu4) negatively modulates GABA and glutamate release in the 'indirect pathway' of the basal ganglia, and has thus been proposed as a potential target to treat motor symptoms in Parkinson's disease. Here, we present an extensive comparison of the behavioural effects produced by the mGlu4 positive allosteric modulator (PAM), VU0364770, and the mGlu4 orthosteric agonist, LSP1-2111, in rats with unilateral 6-OHDA lesions. The compounds' activity was initially assessed in a test of haloperidol-induced catalepsy in intact rats, and effective doses were then evaluated in the hemiparkinsonian animal model. Neither of the two compounds modified the development of dyskinetic behaviours elicited by chronic treatment with full doses of l-DOPA. When given together with l-DOPA to rats with already established dyskinesias, neither VU0364770 nor LSP1-2111 modified the abnormal involuntary movement scores. VU0364770 potentiated, however, the motor stimulant effect of a subthreshold l-DOPA dose in certain behavioural tests, whereas LSP1-2111 lacked this ability. Taken together, these results indicate that a pharmacological stimulation of mGlu4 lacks intrinsic antidyskinetic activity, but may have DOPA-sparing activity in Parkinson's disease. For the latter indication, mGlu4 PAMs appear to provide a better option than orthosteric agonists.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/drug therapy , Excitatory Amino Acid Agents/pharmacology , Levodopa/toxicity , Parkinsonian Disorders/drug therapy , Receptors, Metabotropic Glutamate/agonists , Aminobutyrates/pharmacology , Animals , Antiparkinson Agents/pharmacology , Catalepsy/drug therapy , Catalepsy/metabolism , Dyskinesia, Drug-Induced/metabolism , Haloperidol , Levodopa/pharmacology , Male , Oxidopamine , Parkinsonian Disorders/metabolism , Phosphinic Acids/pharmacology , Picolinic Acids/pharmacology , Random Allocation , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism
6.
J Med Chem ; 57(23): 10192-7, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25423411

ABSTRACT

A duplexed, functional multiaddition high throughput screen and subsequent iterative parallel synthesis effort identified the first highly selective and CNS penetrant glucagon-like peptide-1R (GLP-1R) positive allosteric modulator (PAM). PAM (S)-9b potentiated low-dose exenatide to augment insulin secretion in primary mouse pancreatic islets, and (S)-9b alone was effective in potentiating endogenous GLP-1R to reverse haloperidol-induced catalepsy.


Subject(s)
Indoles/chemical synthesis , Pyrrolidines/chemical synthesis , Receptors, Glucagon/drug effects , Allosteric Regulation/drug effects , Animals , Catalepsy/chemically induced , Catalepsy/drug therapy , Central Nervous System Agents/therapeutic use , Drug Synergism , Exenatide , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Haloperidol , High-Throughput Screening Assays , Indoles/metabolism , Indoles/pharmacokinetics , Indoles/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Male , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Peptides/pharmacology , Pyrrolidines/metabolism , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Structure-Activity Relationship , Venoms/pharmacology
7.
J Pharmacol Exp Ther ; 340(2): 404-21, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22088953

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disorder associated with severe motor impairments caused by the loss of dopaminergic innervation of the striatum. Previous studies have demonstrated that positive allosteric modulators (PAMs) of metabotropic glutamate receptor 4 (mGlu4), including N-phenyl-7-(hydroxyimino) cyclopropa[b]chromen-1a-carboxamide, can produce antiparkinsonian-like effects in preclinical models of PD. However, these early mGlu4 PAMsexhibited unsuitable physiochemical properties for systemic dosing, requiring intracerebroventricular administration and limiting their broader utility as in vivo tools to further understand the role of mGlu4 in the modulation of basal ganglia function relevant to PD. In the present study, we describe the pharmacologic characterization of a systemically active mGlu4 PAM, N-(3-chlorophenyl)picolinamide (VU0364770), in several rodent PD models. VU0364770 showed efficacy alone or when administered in combination with L-DOPA or an adenosine 2A (A2A) receptor antagonist currently in clinical development (preladenant). When administered alone, VU0364770 exhibited efficacy in reversing haloperidol-induced catalepsy, forelimb asymmetry-induced by unilateral 6-hydroxydopamine (6-OHDA) lesions of the median forebrain bundle, and attentional deficits induced by bilateral 6-OHDA nigrostriatal lesions in rats. In addition, VU0364770 enhanced the efficacy of preladenant to reverse haloperidol-induced catalepsy when given in combination. The effects of VU0364770 to reverse forelimb asymmetry were also potentiated when the compound was coadministered with an inactive dose of L-DOPA, suggesting that mGlu4 PAMs may provide L-DOPA-sparing activity. The present findings provide exciting support for the potential role of selective mGlu4 PAMs as a novel approach for the symptomatic treatment of PD and a possible augmentation strategy with either L-DOPA or A2A antagonists.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Picolinic Acids/therapeutic use , Receptors, Metabotropic Glutamate/agonists , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adenosine A2 Receptor Antagonists/blood , Adenosine A2 Receptor Antagonists/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Calcium Signaling/drug effects , Catalepsy/chemically induced , Catalepsy/drug therapy , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Glutamic Acid/pharmacology , HEK293 Cells , Haloperidol/pharmacology , Humans , Levodopa/metabolism , Male , Monoamine Oxidase/metabolism , Motor Neuron Disease/chemically induced , Motor Neuron Disease/drug therapy , Motor Neuron Disease/metabolism , Motor Neuron Disease/pathology , Motor Neuron Disease/physiopathology , Oxidopamine/pharmacology , Picolinic Acids/blood , Picolinic Acids/metabolism , Picolinic Acids/pharmacokinetics , Picolinic Acids/pharmacology , Protein Binding , Psychomotor Performance/drug effects , Pyrimidines/blood , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reaction Time/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/genetics
8.
J Pharmacol Exp Ther ; 340(3): 595-603, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22135383

ABSTRACT

Antagonists of the muscarinic acetylcholine receptors (mAChRs) were among the first treatments for Parkinson's disease. However, the clinical utility of mAChR antagonists is limited by adverse effects associated with the blockade of multiple mAChR subtypes. Understanding the roles of specific mAChR subtypes in regulating basal ganglia and motor function could lead to the development of novel agents that have antiparkinsonian activity with fewer adverse effects. Using the novel, highly selective M1 antagonist N-[3-oxo-3-[4-(4-pyridinyl)-1-piperazinyl]propyl]-2,1,3-benzothiadiazole-4-sulfonamide (VU0255035) and the M1 positive allosteric modulator benzylquinolone carboxylic acid, we investigated the roles of M1 receptors in cholinergic excitation and regulation of synaptic transmission in striatal medium spiny neurons (MSNs) and neurons in the subthalamic nucleus (STN) and substantia nigra pars reticulata (SNr). Electrophysiological studies demonstrate that M1 activation has excitatory effects on MSNs but plays little or no role in mAChR-mediated increases in firing frequency or the regulation of synaptic transmission in STN and SNr neurons. On the basis of this profile, M1-selective antagonists may have weak antiparkinsonian activity but would not have the full efficacy observed in nonselective mAChR antagonists. Consistent with this, the M1-selective antagonist VU0255035 partially reversed reserpine-induced akinesia and decreased haloperidol-induced catalepsy in rats but did not have the full efficacy observed with the nonselective mAChR antagonist scopolamine. These results suggest that the M1 receptor participates in the overall regulation of basal ganglia function and antiparkinsonian effects of mAChR antagonists but that other mAChR subtype(s) also play important roles at multiple levels of the basal ganglia motor circuit.


Subject(s)
Basal Ganglia/physiology , Parkinson Disease/drug therapy , Receptor, Muscarinic M1/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/antagonists & inhibitors , Substantia Nigra/physiology , Subthalamic Nucleus/physiology , Synaptic Transmission
9.
J Med Chem ; 54(21): 7639-47, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21966889

ABSTRACT

There is an increasing amount of literature data showing the positive effects on preclinical antiparkinsonian rodent models with selective positive allosteric modulators of metabotropic glutamate receptor 4 (mGlu(4)). However, most of the data generated utilize compounds that have not been optimized for druglike properties, and as a consequence, they exhibit poor pharmacokinetic properties and thus do not cross the blood-brain barrier. Herein, we report on a series of N-4-(2,5-dioxopyrrolidin-1-yl)phenylpicolinamides with improved PK properties with excellent potency and selectivity as well as improved brain exposure in rodents. Finally, ML182 was shown to be orally active in the haloperidol induced catalepsy model, a well-established antiparkinsonian model.


Subject(s)
Antiparkinson Agents/chemical synthesis , Isoindoles/chemical synthesis , Picolinic Acids/chemical synthesis , Receptors, Metabotropic Glutamate/physiology , Administration, Oral , Allosteric Regulation , Animals , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/pharmacology , Biological Availability , CHO Cells , Catalepsy/chemically induced , Catalepsy/drug therapy , Cricetinae , Cricetulus , Haloperidol , Humans , In Vitro Techniques , Isoindoles/pharmacokinetics , Isoindoles/pharmacology , Microsomes, Liver/metabolism , Picolinic Acids/pharmacokinetics , Picolinic Acids/pharmacology , Rats , Structure-Activity Relationship
10.
ACS Chem Neurosci ; 2(12): 730-742, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22368764

ABSTRACT

T-type Ca(2+) channel inhibitors hold tremendous therapeutic potential for the treatment of pain, epilepsy, sleep disorders, essential tremor and other neurological disorders; however, a lack of truly selective tools has hindered basic research, and selective tools from the pharmaceutical industry are potentially burdened with intellectual property (IP) constraints. Thus, an MLPCN high-throughput screen (HTS) was conducted to identify novel T-type Ca(2+) channel inhibitors free from IP constraints, and freely available through the MLPCN, for use by the biomedical community to study T-type Ca(2+) channels. While the HTS provided numerous hits, these compounds could not be optimized to the required level of potency to be appropriate tool compounds. Therefore, a scaffold hopping approach, guided by SurflexSim, ultimately afforded ML218 (CID 45115620) a selective T-Type Ca(2+) (Ca(v)3.1, Ca(v)3.2, Ca(v)3.3) inhibitor (Ca(v)3.2, IC(50) = 150 nM in Ca(2+) flux; Ca(v)3.2 IC(50) = 310 nM and Ca(v)3.3 IC(50) = 270 nM, respectively in patch clamp electrophysiology) with good DMPK properties, acceptable in vivo rat PK and excellent brain levels. Electrophysiology studies in subthalamic nucleus (STN) neurons demonstrated robust effects of ML218 on the inhibition of T-Type calcium current, inhibition of low threshold spike and rebound burst activity. Based on the basal ganglia circuitry in Parkinson's disease (PD), the effects of ML218 in STN neurons suggest a therapeutic role for T-type Ca(2+) channel inhibitors, and ML218 was found to be orally efficacious in haloperidol-induced catalepsy, a preclinical PD model, with comparable efficacy to an A(2A) antagonist, a clinically validated PD target. ML218 proves to be a powerful new probe to study T-Type Ca(2+) function in vitro and in vivo, and freely available.

11.
Pain ; 151(3): 806-815, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926192

ABSTRACT

This investigation determined whether the activation of the kappa opioid receptor (KOR) in the spinal cord produces estrogen-dependent, sex-specific modulation of acute and inflammation-induced persistent nociception. We demonstrate for the first time that KOR antinociception and gene expression are enhanced by exogenous or endogenous estrogen in the female. The lack of KOR antinociception and KOR gene expression are not altered by the hormonal status (testosterone or estrogen) in males. Cannulae were implanted intrathecally in male, gonadectomized male (GDX), intact and ovariectomized female (OVX) Sprague-Dawley rats. Estradiol was injected subcutaneously, 48h before testing (GDX+E and OVX+E). Intrathecal injection of U50,488H, a selective KOR agonist, dose dependently increased heat-evoked tail flick latencies (TFLs) in proestrous and OVX+E groups, but not in male, GDX, GDX+E, OVX, and diestrous groups. Further, estrogen dose-dependently enhanced the effect of U50,488H in OVX rats. KOR selective antagonist, nor-binaltorphimine (Nor-BNI), blocked the antinociceptive effect of U50,488H. U50,488H reversed the carrageenan-induced thermal hyperalgesia in OVX+E rats, but not in male or OVX rats. However, U50,488H treatment did not alter mechanical thresholds in any group, with or without inflammation. KOR gene expression was enhanced in proestrous and OVX+E groups as compared to any other group. We conclude that selective activation of KOR in the spinal cord produces sex-specific, stimulus- and estrogen-dependent attenuation of acute and inflammatory pain in the rat via estrogen-induced upregulation of the KOR gene expression in the spinal cord. These findings may further implicate estrogen dependence of KOR effects in learning, epilepsy, stress response, addiction etc.


Subject(s)
Analgesia/methods , Estradiol/metabolism , Pain/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Analysis of Variance , Animals , Area Under Curve , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrous Cycle/metabolism , Female , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Injections, Spinal , Lumbosacral Region , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Orchiectomy , Ovariectomy , Pain/drug therapy , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Reverse Transcriptase Polymerase Chain Reaction , Sex Characteristics , Spinal Cord/drug effects , Spinal Cord/metabolism , Testosterone/metabolism , Testosterone/pharmacology
12.
J Pharmacol Exp Ther ; 327(3): 941-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18772318

ABSTRACT

Previous clinical and animal studies suggest that selective activators of M(1) and/or M(4) muscarinic acetylcholine receptors (mAChRs) have potential as novel therapeutic agents for treatment of schizophrenia and Alzheimer's disease. However, highly selective centrally penetrant activators of either M(1) or M(4) have not been available, making it impossible to determine the in vivo effects of selective activation of these receptors. We previously identified VU10010 [3-amino-N-(4-chlorobenzyl)-4, 6-dimethylthieno[2,3-b]pyridine-2-carboxamide] as a potent and selective allosteric potentiator of M(4) mAChRs. However, unfavorable physiochemical properties prevented use of this compound for in vivo studies. We now report that chemical optimization of VU10010 has afforded two centrally penetrant analogs, VU0152099 [3-amino-N-(benzo[d][1,3]dioxol-5-ylmethyl)-4,6-dimethylthieno[2,3-b]pyridine carboxamide] and VU0152100 [3-amino-N-(4-methoxybenzyl)-4,6-dimethylthieno[2,3-b]pyridine carboxamide], that are potent and selective positive allosteric modulators of M(4). VU0152099 and VU0152100 had no agonist activity but potentiated responses of M(4) to acetylcholine. Both compounds were devoid of activity at other mAChR subtypes or at a panel of other GPCRs. The improved physiochemical properties of VU0152099 and VU0152100 allowed in vivo dosing and evaluation of behavioral effects in rats. Interestingly, these selective allosteric potentiators of M(4) reverse amphetamine-induced hyperlocomotion in rats, a model that is sensitive to known antipsychotic agents and to nonselective mAChR agonists. This is consistent with the hypothesis that M(4) plays an important role in regulating midbrain dopaminergic activity and raises the possibility that positive allosteric modulation of M(4) may mimic some of the antipsychotic-like effects of less selective mAChR agonists.


Subject(s)
Allosteric Regulation , Motor Activity/drug effects , Pyridines/pharmacology , Receptor, Muscarinic M4/agonists , Thiophenes/pharmacology , Acetylcholine/pharmacology , Animals , Dopamine , Mesencephalon , Rats
13.
Mol Pharmacol ; 74(5): 1345-58, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18664603

ABSTRACT

Parkinson's disease (PD) is caused by the death of dopamine neurons in the basal ganglia and results in motor symptoms such as tremor and bradykinesia. Activation of metabotropic glutamate receptor 4 (mGluR4) has been shown to modulate neurotransmission in the basal ganglia and results in antiparkinsonian effects in rodent PD models. N-Phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) is a positive allosteric modulator (PAM) of mGluR4 that has been used to further validate the role of mGluR4 in PD, but the compound suffers from a lack of selectivity, relatively low potency, and poor solubility. Via high-throughput screening, we discovered more than 400 novel PAMs of mGluR4. Compounds derived from a novel chemical scaffold were characterized in vitro at both rat and human mGluR4 using two distinct assays of mGluR4 function. The lead compound was approximately 8-fold more potent than PHCCC, enhanced the potency of glutamate at mGluR4 by 8-fold, and did not show any significant potentiator or antagonist activity at other mGluR subtypes. Resolution of the regioisomers of the lead revealed that the cis regioisomer, (+/-)-cis-2-(3,5-dichlorphenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041), contained the majority of the mGluR4 PAM activity and also exhibited partial agonist activity at mGluR4 at a site that was distinct from the glutamate binding site, suggesting that this compound is a mixed allosteric agonist/PAM of mGluR4. VU0155041 was soluble in an aqueous vehicle, and intracerebroventricular administration of 31 to 316 nmol of VU0155041 dose-dependently decreased haloperidol-induced catalepsy and reserpine-induced akinesia in rats. These exciting results provide continued support for mGluR4 as a therapeutic target in PD.


Subject(s)
Antiparkinson Agents/therapeutic use , Parkinson Disease/drug therapy , Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation , Animals , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/chemistry , Antiparkinson Agents/pharmacology , CHO Cells , Corpus Striatum/drug effects , Cricetinae , Cricetulus , Humans , In Vitro Techniques , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...