Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Mol Psychiatry ; 25(11): 2786-2799, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-30116027

RESUMEN

Recent clinical and preclinical studies suggest that selective activators of the M4 muscarinic acetylcholine receptor have potential as a novel treatment for schizophrenia. M4 activation inhibits striatal dopamine release by mobilizing endocannabinoids, providing a mechanism for local effects on dopamine signaling in the striatum but not in extrastriatal areas. G protein-coupled receptors (GPCRs) typically induce endocannabinoid release through activation of Gαq/11-type G proteins whereas M4 transduction occurs through Gαi/o-type G proteins. We now report that the ability of M4 to inhibit dopamine release and induce antipsychotic-like effects in animal models is dependent on co-activation of the Gαq/11-coupled mGlu1 subtype of metabotropic glutamate (mGlu) receptor. This is especially interesting in light of recent findings that multiple loss of function single nucleotide polymorphisms (SNPs) in the human gene encoding mGlu1 (GRM1) are associated with schizophrenia, and points to GRM1/mGlu1 as a gene within the "druggable genome" that could be targeted for the treatment of schizophrenia. Herein, we report that potentiation of mGlu1 signaling following thalamo-striatal stimulation is sufficient to inhibit striatal dopamine release, and that a novel mGlu1 positive allosteric modulator (PAM) exerts robust antipsychotic-like effects through an endocannabinoid-dependent mechanism. However, unlike M4, mGlu1 does not directly inhibit dopamine D1 receptor signaling and does not reduce motivational responding. Taken together, these findings highlight a novel mechanism of cross talk between mGlu1 and M4 and demonstrate that highly selective mGlu1 PAMs may provide a novel strategy for the treatment of positive symptoms associated with schizophrenia.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Antipsicóticos/metabolismo , Receptor Muscarínico M4/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Ácido Glutámico/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Proc Natl Acad Sci U S A ; 114(22): 5719-5724, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28507136

RESUMEN

Obsessive-compulsive disorder (OCD) is a chronic, disabling condition with inadequate treatment options that leave most patients with substantial residual symptoms. Structural, neurochemical, and behavioral findings point to a significant role for basal ganglia circuits and for the glutamate system in OCD. Genetic linkage and association studies in OCD point to SLC1A1, which encodes the neuronal glutamate/aspartate/cysteine transporter excitatory amino acid transporter 3 (EAAT3)/excitatory amino acid transporter 1 (EAAC1). However, no previous studies have investigated EAAT3 in basal ganglia circuits or in relation to OCD-related behavior. Here, we report a model of Slc1a1 loss based on an excisable STOP cassette that yields successful ablation of EAAT3 expression and function. Using amphetamine as a probe, we found that EAAT3 loss prevents expected increases in (i) locomotor activity, (ii) stereotypy, and (iii) immediate early gene induction in the dorsal striatum following amphetamine administration. Further, Slc1a1-STOP mice showed diminished grooming in an SKF-38393 challenge experiment, a pharmacologic model of OCD-like grooming behavior. This reduced grooming is accompanied by reduced dopamine D1 receptor binding in the dorsal striatum of Slc1a1-STOP mice. Slc1a1-STOP mice also exhibit reduced extracellular dopamine concentrations in the dorsal striatum both at baseline and following amphetamine challenge. Viral-mediated restoration of Slc1a1/EAAT3 expression in the midbrain but not in the striatum results in partial rescue of amphetamine-induced locomotion and stereotypy in Slc1a1-STOP mice, consistent with an impact of EAAT3 loss on presynaptic dopaminergic function. Collectively, these findings indicate that the most consistently associated OCD candidate gene impacts basal ganglia-dependent repetitive behaviors.


Asunto(s)
Ganglios Basales/fisiología , Transportador 3 de Aminoácidos Excitadores/genética , Actividad Motora/genética , Trastorno Obsesivo Compulsivo/genética , Trastorno Obsesivo Compulsivo/fisiopatología , Anfetaminas/farmacología , Animales , Línea Celular , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Aseo Animal/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Receptores de Dopamina D1/metabolismo , Reflejo de Sobresalto/fisiología
3.
Bioorg Med Chem Lett ; 29(14): 1714-1718, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31113706

RESUMEN

This letter describes progress towards an M4 PAM preclinical candidate that resulted in the discovery of VU6005806/AZN-00016130. While the thieno[2,3-c]pyridazine core has been a consistent feature of key M4 PAMs, no work had previously been reported with respect to alternate functionality at the C3 position of the pyridazine ring. Here, we detail new chemistry and analogs that explored this region, and quickly led to VU6005806/AZN-00016130, which was profiled as a putative candidate. While, the ß-amino carboxamide moiety engendered solubility limited absorption in higher species precluding advancement (or requiring extensive pharmaceutical sciences formulation), VU6005806/AZN-00016130 represents a new, high quality preclinical in vivo probe.


Asunto(s)
Regulación Alostérica/inmunología , Receptor Muscarínico M4/inmunología , Estructura Molecular , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 29(16): 2224-2228, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31248774

RESUMEN

This letter describes progress towards an M4 PAM preclinical candidate inspired by an unexpected aldehyde oxidase (AO) metabolite of a novel, CNS penetrant thieno[2,3-c]pyridine core to an equipotent, non-CNS penetrant thieno[2,3-c]pyrdin-7(6H)-one core. Medicinal chemistry design efforts yielded two novel tricyclic cores that enhanced M4 PAM potency, regained CNS penetration, displayed favorable DMPK properties and afforded robust in vivo efficacy in reversing amphetamine-induced hyperlocomotion in rats.


Asunto(s)
Aldehído Oxidasa/metabolismo , Miotonía Congénita/metabolismo , Receptor Muscarínico M4/metabolismo , Animales , Descubrimiento de Drogas , Humanos , Ratas , Relación Estructura-Actividad
5.
Addict Biol ; 23(5): 1106-1116, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29044937

RESUMEN

Cocaine use disorder (CUD) remains a debilitating health problem in the United States for which there are no Food and Drug Administration-approved treatment options. Accumulating anatomical and electrophysiological evidence indicates that the muscarinic acetylcholine receptor (mAChR) subtype 5 (M5 ) plays a critical role in the regulation of the mesolimbic dopaminergic reward circuitry, a major site of action for cocaine and other psychostimulants. In addition, M5 knockout mice exhibit reduced cocaine self-administration behaviors with no differences in sugar pellet-maintained responding relative to wild-type mice. These findings suggest that selective inhibition of M5 mAChR may provide a novel pharmacological approach for targeting CUD. Recently, we reported the synthesis and characterization of ML375, a selective negative allosteric modulator (NAM) for the rat and human M5 mAChR with optimized pharmacokinetic properties for systemic dosing in rodents. In the present study, male Sprague-Dawley rats were trained to self-administer intravenous cocaine (0.1-0.75 mg/kg/infusion) under a 10-response fixed ratio or a progressive ratio schedule of reinforcement. Under both schedules of reinforcement, ML375 produced dose-related reductions in cocaine self-administration. ML375 also modestly reduced sugar pellet-maintained responding on the 10-response, fixed ratio schedule but had no effect under a progressive ratio schedule of reinforcement. Further, ML375 did not affect general motor output as assessed by a rotarod test. Collectively, these results provide the first demonstration that selective inhibition of M5 using the M5 NAM ML375 can attenuate both the reinforcing effects and the relative strength of cocaine and suggest that M5 NAMs may represent a promising, novel treatment approach for CUD.


Asunto(s)
Trastornos Relacionados con Cocaína/prevención & control , Cocaína/administración & dosificación , Receptor Muscarínico M5/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante , Modelos Animales de Enfermedad , Inhibidores de Captación de Dopamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Esquema de Refuerzo , Recompensa , Autoadministración
6.
Bioorg Med Chem Lett ; 27(2): 171-175, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27939174

RESUMEN

This letter describes the chemical optimization of a novel series of M4 positive allosteric modulators (PAMs) based on a 5-amino-thieno[2,3-c]pyridazine core, developed via iterative parallel synthesis, and culminating in the highly utilized rodent in vivo tool compound, VU0467154 (5). This is the first report of the optimization campaign (SAR and DMPK profiling) that led to the discovery of VU0467154, and details all of the challenges faced in allosteric modulator programs (steep SAR, species differences in PAM pharmacology and subtle structural changes affecting CNS penetration).


Asunto(s)
Piridazinas/farmacología , Receptor Muscarínico M4/agonistas , Tiofenos/farmacología , Animales , Humanos , Ligandos , Proteínas de Transporte de Nucleósidos/metabolismo , Piridazinas/administración & dosificación , Piridazinas/síntesis química , Piridazinas/farmacocinética , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tiofenos/administración & dosificación , Tiofenos/síntesis química , Tiofenos/farmacocinética
7.
Bioorg Med Chem Lett ; 27(11): 2296-2301, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28442253

RESUMEN

This letter describes the further chemical optimization of the 5-amino-thieno[2,3-c]pyridazine series (VU0467154/VU0467485) of M4 positive allosteric modulators (PAMs), developed via iterative parallel synthesis, culminating in the discovery of the non-human primate (NHP) in vivo tool compound, VU0476406 (8p). VU0476406 is an important in vivo tool compound to enable translation of pharmacodynamics from rodent to NHP, and while data related to a Parkinson's disease model has been reported with 8p, this is the first disclosure of the optimization and discovery of VU0476406, as well as detailed pharmacology and DMPK properties.


Asunto(s)
Descubrimiento de Drogas , Piridazinas/farmacología , Tiofenos/farmacología , Investigación Biomédica Traslacional , Regulación Alostérica , Animales , Cristalografía por Rayos X , Enlace de Hidrógeno , Piridazinas/química , Ratas , Relación Estructura-Actividad , Tiofenos/química
8.
J Pharmacol Exp Ther ; 356(1): 123-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26503377

RESUMEN

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor subtype 5 (mGlu5) have potential applications in the treatment of fragile X syndrome, levodopa-induced dyskinesia in Parkinson disease, Alzheimer disease, addiction, and anxiety; however, clinical and preclinical studies raise concerns that complete blockade of mGlu5 and inverse agonist activity of current mGlu5 NAMs contribute to adverse effects that limit the therapeutic use of these compounds. We report the discovery and characterization of a novel mGlu5 NAM, N,N-diethyl-5-((3-fluorophenyl)ethynyl)picolinamide (VU0477573) that binds to the same allosteric site as the prototypical mGlu5 NAM MPEP but displays weak negative cooperativity. Because of this weak cooperativity, VU0477573 acts as a "partial NAM" so that full occupancy of the MPEP site does not completely inhibit maximal effects of mGlu5 agonists on intracellular calcium mobilization, inositol phosphate (IP) accumulation, or inhibition of synaptic transmission at the hippocampal Schaffer collateral-CA1 synapse. Unlike previous mGlu5 NAMs, VU0477573 displays no inverse agonist activity assessed using measures of effects on basal [(3)H]inositol phosphate (IP) accumulation. VU0477573 acts as a full NAM when measuring effects on mGlu5-mediated extracellular signal-related kinases 1/2 phosphorylation, which may indicate functional bias. VU0477573 exhibits an excellent pharmacokinetic profile and good brain penetration in rodents and provides dose-dependent full mGlu5 occupancy in the central nervous system (CNS) with systemic administration. Interestingly, VU0477573 shows robust efficacy, comparable to the mGlu5 NAM MTEP, in models of anxiolytic activity at doses that provide full CNS occupancy of mGlu5 and demonstrate an excellent CNS occupancy-efficacy relationship. VU0477573 provides an exciting new tool to investigate the efficacy of partial NAMs in animal models.


Asunto(s)
Agonistas del GABA/farmacología , Ácidos Picolínicos/farmacología , Receptor del Glutamato Metabotropico 5/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Animales , Ansiolíticos/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Agonistas del GABA/farmacocinética , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ácidos Picolínicos/farmacocinética , Piridinas/metabolismo , Ensayo de Unión Radioligante , Ratas , Receptor del Glutamato Metabotropico 5/metabolismo , Transmisión Sináptica/efectos de los fármacos
9.
Bioorg Med Chem Lett ; 26(13): 3029-3033, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27185330

RESUMEN

This Letter describes the chemical optimization of a novel series of M4 positive allosteric modulators (PAMs) based on a 5,6-dimethyl-4-(piperidin-1-yl)thieno[2,3-d]pyrimidine core, identified from an MLPCN functional high-throughput screen. The HTS hit was potent and selective, but not CNS penetrant. Potency was maintained, while CNS penetration was improved (rat brain:plasma Kp=0.74), within the original core after several rounds of optimization; however, the thieno[2,3-d]pyrimidine core was subject to extensive oxidative metabolism. Ultimately, we identified a 6-fluoroquinazoline core replacement that afforded good M4 PAM potency, muscarinic receptor subtype selectivity and CNS penetration (rat brain:plasma Kp>10). Moreover, this campaign provided fundamentally distinct M4 PAM chemotypes, greatly expanding the available structural diversity for this exciting CNS target.


Asunto(s)
Piperidinas/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , Receptor Muscarínico M4/metabolismo , Tiofenos/farmacología , Regulación Alostérica , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Microsomas Hepáticos/metabolismo , Piperidinas/síntesis química , Piperidinas/metabolismo , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Quinazolinas/síntesis química , Quinazolinas/metabolismo , Ratas , Receptor Muscarínico M4/agonistas , Receptor Muscarínico M4/antagonistas & inhibidores , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/metabolismo
10.
J Neurosci ; 32(25): 8532-44, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22723693

RESUMEN

M(1) muscarinic acetylcholine receptors (mAChRs) represent a viable target for treatment of multiple disorders of the central nervous system (CNS) including Alzheimer's disease and schizophrenia. The recent discovery of highly selective allosteric agonists of M(1) receptors has provided a major breakthrough in developing a viable approach for the discovery of novel therapeutic agents that target these receptors. Here we describe the characterization of two novel M(1) allosteric agonists, VU0357017 and VU0364572, that display profound differences in their efficacy in activating M(1) coupling to different signaling pathways including Ca(2+) and ß-arrestin responses. Interestingly, the ability of these agents to differentially activate coupling of M(1) to specific signaling pathways leads to selective actions on some but not all M(1)-mediated responses in brain circuits. These novel M(1) allosteric agonists induced robust electrophysiological effects in rat hippocampal slices, but showed lower efficacy in striatum and no measureable effects on M(1)-mediated responses in medial prefrontal cortical pyramidal cells in mice. Consistent with these actions, both M(1) agonists enhanced acquisition of hippocampal-dependent cognitive function but did not reverse amphetamine-induced hyperlocomotion in rats. Together, these data reveal that M(1) allosteric agonists can differentially regulate coupling of M(1) to different signaling pathways, and this can dramatically alter the actions of these compounds on specific brain circuits important for learning and memory and psychosis.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Encéfalo/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M1/agonistas , Animales , Arrestinas/metabolismo , Células CHO , Calcio/metabolismo , Línea Celular , Cuerpo Estriado/fisiología , Cricetinae , Cricetulus , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Espacio Extracelular/fisiología , Miedo/psicología , Perfilación de la Expresión Génica , Hipocampo/fisiología , Humanos , Masculino , Aprendizaje por Laberinto , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Corteza Prefrontal/fisiología , Ratas , Ratas Sprague-Dawley
11.
Adv Neurobiol ; 30: 37-99, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36928846

RESUMEN

Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.


Asunto(s)
Trastornos Mentales , Animales , Trastornos Mentales/tratamiento farmacológico , Modelos Animales
12.
Neuropharmacology ; 227: 109424, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36720403

RESUMEN

Recent evidence suggests that inhibition of the M5 muscarinic acetylcholine receptor (mAChR) may provide a novel non-opioid mechanism for the treatment of opioid use disorder (OUD). Previous studies from our group and others have demonstrated that acute administration of the long-acting M5 negative allosteric modulator (NAM) ML375 attenuates established self-administration of cocaine, ethanol, oxycodone, and remifentanil in rats. In the present study, we characterized the effects of acute and repeated administration of the novel, short-acting M5 NAM VU6008667 on the reinforcing effects of oxycodone and reinstatement of oxycodone-seeking behaviors in male Sprague-Dawley rats, as well as on physiological withdrawal from oxycodone. Acute VU6008667 decreased oxycodone self-administration under both fixed ratio 3 (FR3) and progressive ratio (PR) schedules of reinforcement and attenuated cue-induced reinstatement of lever pressing following extinction from oxycodone self-administration, a commonly used relapse model. When administered daily to opioid-naïve rats, VU6008667 prevented acquisition of oxycodone self-administration behavior. VU6008667 had minimal effects on naloxone-precipitated withdrawal. After acute administration, VU6008667 did not inhibit sucrose self-administration and, when given chronically, delayed but did not prevent acquisition of sucrose maintained self-administration. VU6008667 also did not impact oxycodone induced anti-nociception or motor coordination, but mildly decreased novelty exploration. Finally, acute or daily VU6008667 administration did not impair cued fear conditioning. Overall, these results suggest that inhibition of the M5 mAChR may provide a novel, non-opioid based treatment for distinct aspects of OUD by inhibiting opioid intake in established OUD, reducing relapse during abstinence, and by reducing the risk of developing OUD.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Animales , Masculino , Ratas , Oxicodona , Ratas Sprague-Dawley , Receptores Muscarínicos , Autoadministración , Sacarosa/farmacología
13.
J Pharmacol Exp Ther ; 340(2): 404-21, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22088953

RESUMEN

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.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Levodopa/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Ácidos Picolínicos/uso terapéutico , Receptores de Glutamato Metabotrópico/agonistas , Ácido 3,4-Dihidroxifenilacético/metabolismo , Antagonistas del Receptor de Adenosina A2/sangre , Antagonistas del Receptor de Adenosina A2/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Señalización del Calcio/efectos de los fármacos , Catalepsia/inducido químicamente , Catalepsia/tratamiento farmacológico , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ácido Glutámico/farmacología , Células HEK293 , Haloperidol/farmacología , Humanos , Levodopa/metabolismo , Masculino , Monoaminooxidasa/metabolismo , Enfermedad de la Neurona Motora/inducido químicamente , Enfermedad de la Neurona Motora/tratamiento farmacológico , Enfermedad de la Neurona Motora/metabolismo , Enfermedad de la Neurona Motora/patología , Enfermedad de la Neurona Motora/fisiopatología , Oxidopamina/farmacología , Ácidos Picolínicos/sangre , Ácidos Picolínicos/metabolismo , Ácidos Picolínicos/farmacocinética , Ácidos Picolínicos/farmacología , Unión Proteica , Desempeño Psicomotor/efectos de los fármacos , Pirimidinas/sangre , Pirimidinas/metabolismo , Pirimidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato Metabotrópico/genética
14.
Handb Exp Pharmacol ; (208): 121-66, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22222698

RESUMEN

The muscarinic cholinergic system constitutes an important part of the neuronal circuitry that modulates normal cognition. Muscarinic receptor antagonists are well known to produce or exacerbate impairments in attention, learning, and memory. Conversely, both direct-acting muscarinic receptor agonists and indirect-acting muscarinic cholinergic agonists, such as acetylcholinesterase inhibitors, have shown cognition-enhancing properties, including improvements in normal cognitive function, reversal of cognitive deficits induced by muscarinic receptor antagonists, and attenuation of cognitive deficits in psychiatric and neurological disorders, such as Alzheimer's disease and schizophrenia. However, until recently, the lack of small molecule ligands that antagonize or activate specific muscarinic acetylcholine receptor (mAChR) subtypes with high selectivity has been a major obstacle in defining the relative contributions of individual mAChRs to different aspects of cognitive function and for the development of novel therapeutic agents. These limitations may be potentially overcome by the recent discovery of novel mAChR subtype-selective compounds, notably allosteric agonists and positive allosteric modulators, which exhibit greater selectivity for individual mAChR subtypes than previous mAChR orthosteric agonists. In preclinical studies, these novel ligands have shown promising efficacy in several models for the enhancement of cognition. In this chapter, we will review the muscarinic cholinergic circuitry and pharmacology of mAChR agonists and antagonists relevant to the modulation of different aspects of cognition in animals and clinical populations.


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Nootrópicos/farmacología , Receptores Muscarínicos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Acetilcolina/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/psicología , Humanos , Antagonistas Muscarínicos/farmacología , Receptores Muscarínicos/metabolismo
15.
J Med Chem ; 65(8): 6273-6286, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35417155

RESUMEN

The muscarinic acetylcholine receptor (mAChR) subtype 5 (M5) represents a novel potential target for the treatment of multiple addictive disorders, including opioid use disorder. Through chemical optimization of several functional high-throughput screening hits, VU6019650 (27b) was identified as a novel M5 orthosteric antagonist with high potency (human M5 IC50 = 36 nM), M5 subtype selectivity (>100-fold selectivity against human M1-4) and favorable physicochemical properties for systemic dosing in preclinical addiction models. In acute brain slice electrophysiology studies, 27b blocked the nonselective muscarinic agonist oxotremorine-M-induced increases in neuronal firing rates of midbrain dopamine neurons in the ventral tegmental area, a part of the mesolimbic dopaminergic reward circuitry. Moreover, 27b also inhibited oxycodone self-administration in male Sprague-Dawley rats within a dose range that did not impair general motor output.


Asunto(s)
Trastornos Relacionados con Opioides , Receptor Muscarínico M5 , Animales , Neuronas Dopaminérgicas , Masculino , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1 , Receptores Muscarínicos
16.
Neurobiol Dis ; 39(3): 283-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20423730

RESUMEN

Angelman syndrome (AS) is a neurogenetic disorder caused by loss of maternal UBE3A expression or mutation-induced dysfunction of its protein product, the E3 ubiquitin-protein ligase, UBE3A. In humans and rodents, UBE3A/Ube3a transcript is maternally imprinted in several brain regions, but the distribution of native UBE3A/Ube3a(1) protein expression has not been comprehensively examined. To address this, we systematically evaluated Ube3a expression in the brain and peripheral tissues of wild-type (WT) and Ube3a maternal knockout mice (AS mice). Immunoblot and immunohistochemical analyses revealed a marked loss of Ube3a protein in hippocampus, hypothalamus, olfactory bulb, cerebral cortex, striatum, thalamus, midbrain, and cerebellum in AS mice relative to WT littermates. Also, Ube3a expression in heart and liver of AS mice showed greater than the predicted 50% reduction relative to WT mice. Co-localization studies showed Ube3a expression to be primarily neuronal in all brain regions and present in GABAergic interneurons as well as principal neurons. These findings suggest that neuronal function throughout the brain is compromised in AS.


Asunto(s)
Síndrome de Angelman/metabolismo , Encéfalo/metabolismo , Hígado/metabolismo , Miocardio/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Análisis de Varianza , Síndrome de Angelman/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/metabolismo , Distribución Tisular , Ubiquitina-Proteína Ligasas/genética , Ácido gamma-Aminobutírico/metabolismo
17.
Neuropsychopharmacology ; 45(13): 2219-2228, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32868847

RESUMEN

Degeneration of basal forebrain cholinergic circuitry represents an early event in the development of Alzheimer's disease (AD). These alterations in central cholinergic function are associated with disruptions in arousal, sleep/wake architecture, and cognition. Changes in sleep/wake architecture are also present in normal aging and may represent a significant risk factor for AD. M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs) have been reported to enhance cognition across preclinical species and may also provide beneficial effects for age- and/or neurodegenerative disease-related changes in arousal and sleep. In the present study, electroencephalography was conducted in young animals (mice, rats and nonhuman primates [NHPs]) and in aged mice to examine the effects of the selective M1 PAM VU0453595 in comparison with the acetylcholinesterase inhibitor donepezil, M1/M4 agonist xanomeline (in NHPs), and M1 PAM BQCA (in rats) on sleep/wake architecture and arousal. In young wildtype mice, rats, and NHPs, but not in M1 mAChR KO mice, VU0453595 produced dose-related increases in high frequency gamma power, a correlate of arousal and cognition enhancement, without altering duration of time across all sleep/wake stages. Effects of VU0453595 in NHPs were observed within a dose range that did not induce cholinergic-mediated adverse effects. In contrast, donepezil and xanomeline increased time awake in rodents and engendered dose-limiting adverse effects in NHPs. Finally, VU0453595 attenuated age-related decreases in REM sleep duration in aged wildtype mice. Development of M1 PAMs represents a viable strategy for attenuating age-related and dementia-related pathological disturbances of sleep and arousal.


Asunto(s)
Enfermedades Neurodegenerativas , Roedores , Regulación Alostérica , Animales , Nivel de Alerta , Ratones , Primates , Piridinas , Pirroles , Ratas , Receptor Muscarínico M1 , Sueño
18.
J Neurosci ; 28(41): 10422-33, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18842902

RESUMEN

Recent studies suggest that subtype-selective activators of M(1)/M(4) muscarinic acetylcholine receptors (mAChRs) may offer a novel approach for the treatment of psychotic symptoms associated with schizophrenia and Alzheimer's disease. Previously developed muscarinic agonists have provided clinical data in support of this hypothesis, but failed in clinical development because of a lack of true subtype specificity and adverse effects associated with activation of other mAChR subtypes. We now report characterization of a novel highly selective agonist for the M(1) receptor with no agonist activity at any of the other mAChR subtypes, termed TBPB [1-(1'-2-methylbenzyl)-1,4'-bipiperidin-4-yl)-1H-benzo[d]imidazol-2(3H)-one]. Mutagenesis and molecular pharmacology studies revealed that TBPB activates M(1) through an allosteric site rather than the orthosteric acetylcholine binding site, which is likely critical for its unprecedented selectivity. Whole-cell patch-clamp recordings demonstrated that activation of M(1) by TBPB potentiates NMDA receptor currents in hippocampal pyramidal cells but does not alter excitatory or inhibitory synaptic transmission, responses thought to be mediated by M(2) and M(4). TBPB was efficacious in models predictive of antipsychotic-like activity in rats at doses that did not produce catalepsy or peripheral adverse effects of other mAChR agonists. Finally, TBPB had effects on the processing of the amyloid precursor protein toward the non-amyloidogenic pathway and decreased Abeta production in vitro. Together, these data suggest that selective activation of M(1) may provide a novel approach for the treatment of symptoms associated with schizophrenia and Alzheimer's disease.


Asunto(s)
Sitio Alostérico/fisiología , Amiloide/metabolismo , Antipsicóticos/farmacología , Bencimidazoles/farmacología , Piperidinas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptor Muscarínico M1/química , Receptor Muscarínico M1/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/metabolismo , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Conductividad Eléctrica , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Piperidinas/administración & dosificación , Piperidinas/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Transmisión Sináptica/efectos de los fármacos , Transfección
19.
Eur J Neurosci ; 30(2): 207-16, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19659923

RESUMEN

Parkinson's Disease (PD) is marked by prominent motor symptoms that reflect striatal dopamine insufficiency. However, non-motor symptoms, including depression, are common in PD. It has been suggested that these changes reflect pathological involvement of non-dopaminergic systems. We examined regional changes in serotonin (5-HT) and norepinephrine (NE) systems in mice treated with two different 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment paradigms, at survival times of 3 or 16 weeks after the last MPTP injection. MPTP caused a decrease in striatal dopamine concentration, the magnitude of which depended on the treatment regimen and survival interval after MPTP treatment. There was significant involvement of other subcortical areas receiving a dopamine innervation, but no consistent changes in 5-HT or NE levels in subcortical sites. In contrast, we observed an enduring decrease in 5-HT and NE concentrations in both the somatosensory cortex and medial prefrontal cortex (PFC). Immunohistochemical studies also revealed a decrease in the density of PFC NE and 5-HT axons. The decrease in the cortical serotonergic innervation preferentially involved the thick beaded but not smooth fine 5-HT axons. Similar changes in the 5-HT innervation of post-mortem samples of the PFC from idiopathic PD cases were seen. Our findings point to a major loss of the 5-HT and NE innervations of the cortex in MPTP-induced parkinsonism, and suggest that loss of the beaded cortical 5-HT innervation is associated with a predisposition to the development of depression in PD.


Asunto(s)
Corteza Cerebral/metabolismo , Norepinefrina/metabolismo , Trastornos Parkinsonianos/metabolismo , Serotonina/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/análogos & derivados , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Anciano , Anciano de 80 o más Años , Animales , Recuento de Células/métodos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Desnervación/métodos , Dopamina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Trastornos Parkinsonianos/patología
20.
ACS Chem Neurosci ; 10(8): 3740-3750, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31268669

RESUMEN

Opioid use disorder (OUD) is a debilitating neuropsychiatric condition characterized by compulsive opioid use, dependence, and repeated relapse after periods of abstinence. Given the high risk of developing OUD following prescription opioid use, the continued need for opioid-induced analgesia, and the limitations of current OUD treatments, it is necessary to develop novel, non-opioid-based treatments for OUD and decrease abuse potential of prescription opioids. Recent evidence suggests that negative allosteric modulation (NAM) of the M5 muscarinic acetylcholine receptor (M5 mAChR) may provide an alternative therapeutic approach for the treatment of OUD. Previous studies demonstrated localization of M5 mAChR expression within the mesocorticolimbic reward circuitry and that the selective M5 NAM ML375 attenuates both cocaine and alcohol self-administration in rats. In the present study, the effects of ML375 were evaluated in rats self-administering the µ-opioid agonists oxycodone or remifentanil on a progressive ratio (PR) schedule or on cue reactivity (a rodent model of relapse) in the absence of oxycodone following 72 h of abstinence. ML375 reduced the PR break point for oxycodone and remifentanil self-administration and attenuated cue-elicited responding. Importantly, ML375 did not affect sucrose pellet-maintained responding on a PR schedule or opioid-induced antinociception using the hot-plate and tail-flick assays. We also confirm expression of M5 mAChR mRNA in the ventral tegmental area and show that this is primarily on dopamine (tyrosine hydroxylase mRNA-positive) neurons. Taken together, these findings suggest that selective functional antagonism of the M5 mAChR may represent a novel, non-opioid-based treatment for OUD.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Narcóticos/administración & dosificación , Nocicepción/efectos de los fármacos , Oxicodona/administración & dosificación , Receptor Muscarínico M5/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Señales (Psicología) , Masculino , Ratas , Ratas Sprague-Dawley , Remifentanilo/administración & dosificación , Recompensa , Autoadministración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA