Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Addict Biol ; 27(5): e13220, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36001441

RESUMEN

Glutamate signalling through the N-methyl-d-aspartate receptor (NMDAR) activates the enzyme neuronal nitric oxide synthase (nNOS) to produce the signalling molecule nitric oxide (NO). We hypothesized that disruption of the protein-protein interaction between nNOS and the scaffolding protein postsynaptic density 95 kDa (PSD95) would block NMDAR-dependent NO signalling and represent a viable therapeutic route to decrease opioid reward and relapse-like behaviour without the unwanted side effects of NMDAR antagonists. We used a conditioned place preference (CPP) paradigm to evaluate the impact of two small-molecule PSD95-nNOS inhibitors, IC87201 and ZL006, on the rewarding effects of morphine. Both IC87201 and ZL006 blocked morphine-induced CPP at doses that lacked intrinsic rewarding or aversive properties. Furthermore, in vivo fast-scan cyclic voltammetry (FSCV) was used to ascertain the impact of ZL006 on morphine-induced increases in dopamine (DA) efflux in the nucleus accumbens shell (NAc shell) evoked by electrical stimulation of the medial forebrain bundle (MFB). ZL006 attenuated morphine-induced increases in DA efflux at a dose that did not have intrinsic effects on DA transmission. We also employed multiple intravenous drug self-administration approaches to examine the impact of ZL006 on the reinforcing effects of morphine. Interestingly, ZL006 did not alter acquisition or maintenance of morphine self-administration, but reduced lever pressing in a morphine relapse test after forced abstinence. Our results provide behavioural and neurochemical support for the hypothesis that inhibition of PSD95-nNOS protein-protein interactions decreases morphine reward and relapse-like behaviour, highlighting a previously unreported application for these novel therapeutics in the treatment of opioid addiction.


Asunto(s)
Morfina , Recompensa , Animales , Homólogo 4 de la Proteína Discs Large , Morfina/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Núcleo Accumbens/metabolismo , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Recurrencia
2.
Bioorg Med Chem ; 28(21): 115727, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065437

RESUMEN

Specific tuning of cannabinoid 1 receptor (CB1R) activity by small-molecule allosteric modulators is a therapeutic modality with multiple properties inherently advantageous to therapeutic applications. We previously generated a library of unique CB1R positive allosteric modulators (PAMs) derived from GAT211, which has three pharmacophoric sites critical to its ago-PAM activity. To elaborate our CB1R PAM library, we report the rational design and molecular-pharmacology profiling of several 2-phenylindole analogs modified at the "site-III" aromatic ring. The comprehensive structure-activity relationship (SAR) investigation demonstrates that attaching small lipophilic functional groups on the ortho-position of the GAT211 site-III phenyl ring could markedly enhance CB1R ago-PAM activity. Select site-III modifications also improved GAT211's water solubility. The SAR reported both extends the structural diversity of this compound class and demonstrates the utility of GAT211's site-III for improving the parent compound's drug-like properties of potency and/or aqueous solubility.


Asunto(s)
Agonistas de Receptores de Cannabinoides/química , Indoles/química , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico , Agonistas de Receptores de Cannabinoides/metabolismo , Agonistas de Receptores de Cannabinoides/farmacología , Humanos , Indoles/metabolismo , Indoles/farmacología , Cinética , Simulación del Acoplamiento Molecular , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Solubilidad , Relación Estructura-Actividad , beta-Arrestinas/metabolismo
3.
Molecules ; 25(2)2020 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-31968549

RESUMEN

Cannabinoid receptor 1 (CB1) activation has been reported to reduce transient receptor potential cation channel subfamily V member 1 (TRPV1)-induced inflammatory responses and is anti-nociceptive and anti-inflammatory in corneal injury. We examined whether allosteric ligands, can modulate CB1 signaling to reduce pain and inflammation in corneal hyperalgesia. Corneal hyperalgesia was generated by chemical cauterization of cornea in wildtype and CB2 knockout (CB2-/-) mice. The novel racemic CB1 allosteric ligand GAT211 and its enantiomers GAT228 and GAT229 were examined alone or in combination with the orthosteric CB1 agonist Δ8-tetrahydrocannabinol (Δ8-THC). Pain responses were assessed following capsaicin (1 µM) stimulation of injured corneas at 6 h post-cauterization. Corneal neutrophil infiltration was also analyzed. GAT228, but not GAT229 or GAT211, reduced pain scores in response to capsaicin stimulation. Combination treatments of 0.5% GAT229 or 1% GAT211 with subthreshold Δ8-THC (0.4%) significantly reduced pain scores following capsaicin stimulation. The anti-nociceptive effects of both GAT229 and GAT228 were blocked with CB1 antagonist AM251, but remained unaffected in CB2-/- mice. Two percent GAT228, or the combination of 0.2% Δ8-THC with 0.5% GAT229 also significantly reduced corneal inflammation. CB1 allosteric ligands could offer a novel approach for treating corneal pain and inflammation.


Asunto(s)
Lesiones de la Cornea/tratamiento farmacológico , Dronabinol/análogos & derivados , Hiperalgesia/tratamiento farmacológico , Indoles/administración & dosificación , Inflamación/tratamiento farmacológico , Receptor Cannabinoide CB1/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Cauterización , Lesiones de la Cornea/complicaciones , Lesiones de la Cornea/etiología , Modelos Animales de Enfermedad , Dronabinol/administración & dosificación , Dronabinol/farmacología , Sinergismo Farmacológico , Técnicas de Inactivación de Genes , Hiperalgesia/metabolismo , Indoles/farmacología , Inflamación/etiología , Inflamación/metabolismo , Ligandos , Ratones , Receptor Cannabinoide CB1/química , Receptor Cannabinoide CB2/genética , Transducción de Señal
4.
J Med Chem ; 63(2): 542-568, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31756109

RESUMEN

Cannabinoid 1 receptor (CB1R) allosteric ligands hold a far-reaching therapeutic promise. We report the application of fluoro- and nitrogen-walk approaches to enhance the drug-like properties of GAT211, a prototype CB1R allosteric agonist-positive allosteric modulator (ago-PAM). Several analogs exhibited improved functional potency (cAMP, ß-arrestin 2), metabolic stability, and aqueous solubility. Two key analogs, GAT591 (6r) and GAT593 (6s), exhibited augmented allosteric-agonist and PAM activities in neuronal cultures, improved metabolic stability, and enhanced orthosteric agonist binding (CP55,940). Both analogs also exhibited good analgesic potency in the CFA inflammatory-pain model with longer duration of action over GAT211 while being devoid of adverse cannabimimetic effects. Another analog, GAT592 (9j), exhibited moderate ago-PAM potency and improved aqueous solubility with therapeutic reduction of intraocular pressure in murine glaucoma models. The SAR findings and the enhanced allosteric activity in this class of allosteric modulators were accounted for in our recently developed computational model for CB1R allosteric activation and positive allosteric modulation.


Asunto(s)
Antiinflamatorios no Esteroideos/síntesis química , Flúor/química , Indoles/química , Nitrógeno/química , Receptor Cannabinoide CB1/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Antiinflamatorios no Esteroideos/farmacología , Biotransformación , Adyuvante de Freund , Células HEK293 , Humanos , Indoles/farmacocinética , Indoles/farmacología , Inflamación/inducido químicamente , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Estereoisomerismo , Relación Estructura-Actividad
5.
ACS Med Chem Lett ; 10(8): 1216-1221, 2019 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-31413808

RESUMEN

Positive allosteric modulation of the cannabinoid 1 receptor (CB1R) has demonstrated distinct therapeutic advantages that address several limitations associated with orthosteric agonism and has opened a promising therapeutic avenue for further drug development. To advance the development of CB1R positive allosteric modulators, it is important to understand the molecular architecture of CB1R allosteric site(s). The goal of this work was to use Force-Biased MMC Simulated Annealing to identify binding sites for GAT228 (R), a partial allosteric agonist, and GAT229 (S), a positive allosteric modulator (PAM) at the CB1R. Our studies suggest that GAT228 binds in an intracellular (IC) TMH1-2-4 exosite that would allow this compound to act as a CB1 allosteric agonist as well as a CB1 PAM. In contrast, GAT229 binds at the extracellular (EC) ends of TMH2/3, just beneath the EC1 loop. At this site, this compound can act as CB1 PAM only. Finally, these results were successfully validated through the synthesis and biochemical evaluation of a focused library of compounds.

6.
Neuropharmacology ; 151: 1-12, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30940536

RESUMEN

Huntington's disease (HD) is an inherited progressive neurodegenerative disease characterized by motor, cognitive, and behavioural changes. One of the earliest changes to occur in HD is a reduction in cannabinoid 1 receptor (CB1) levels in the striatum, which is strongly correlated with HD pathogenesis. CB1 positive allosteric modulators (PAM) enhance receptor affinity for, and efficacy of activation by, orthosteric ligands, including the endocannabinoids anandamide and 2-arachidonoylglycerol. The goal of this study was to determine whether the recently characterized CB1 allosteric modulators GAT211 (racemic), GAT228 (R-enantiomer), and GAT229 (S-enantiomer), affected the signs and symptoms of HD. GAT211, GAT228, and GAT229 were evaluated in normal and HD cell models, and in a transgenic mouse model of HD (7-week-old male R6/2 mice, 10 mg/kg/d, 21 d, i.p.). GAT229 was a CB1 PAM that improved cell viability in HD cells and improved motor coordination, delayed symptom onset, and normalized gene expression in R6/2 HD mice. GAT228 was an allosteric agonist that did not enhance endocannabinoid signaling or change symptom progression in R6/2 mice. GAT211 displayed intermediate effects between its enantiomers. The compounds used here are not drugs, but probe compounds used to determine the potential utility of CB1 PAMs in HD. Changes in gene expression, and not protein, were quantified in R6/2 HD mice because HD pathogenesis is associated with dysregulation of mRNA levels. The data presented here provide the first proof of principle for the use of CB1 PAMs to treat the signs and symptoms of HD.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/uso terapéutico , Cuerpo Estriado/efectos de los fármacos , Enfermedad de Huntington/tratamiento farmacológico , Indoles/uso terapéutico , Receptor Cannabinoide CB1/metabolismo , Animales , Agonistas de Receptores de Cannabinoides/farmacología , Supervivencia Celular/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica/efectos de los fármacos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Indoles/farmacología , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología
7.
Biol Psychiatry ; 84(10): 722-733, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28823711

RESUMEN

BACKGROUND: Activation of cannabinoid CB1 receptors suppresses pathological pain but also produces unwanted central side effects. We hypothesized that a positive allosteric modulator of CB1 signaling would suppress inflammatory and neuropathic pain without producing cannabimimetic effects or physical dependence. We also asked whether a CB1 positive allosteric modulator would synergize with inhibitors of endocannabinoid deactivation and/or an orthosteric cannabinoid agonist. METHODS: GAT211, a novel CB1 positive allosteric modulator, was evaluated for antinociceptive efficacy and tolerance in models of neuropathic and/or inflammatory pain. Cardinal signs of direct CB1-receptor activation were evaluated together with the propensity to induce reward or aversion and physical dependence. Comparisons were made with inhibitors of endocannabinoid deactivation (JZL184, URB597) or an orthosteric cannabinoid agonist (WIN55,212-2). All studies used 4 to 11 subjects per group. RESULTS: GAT211 suppressed allodynia induced by complete Freund's adjuvant and the chemotherapeutic agent paclitaxel in wild-type but not CB1 knockout mice. GAT211 did not impede paclitaxel-induced tumor cell line toxicity. GAT211 did not produce cardinal signs of direct CB1-receptor activation in the presence or absence of pathological pain. GAT211 produced synergistic antiallodynic effects with fatty acid amide hydrolase and monoacylglycerol lipase inhibitors in paclitaxel-treated mice. Therapeutic efficacy was preserved over 19 days of chronic dosing with GAT211, but it was not preserved with the monoacylglycerol lipase inhibitor JZL184. The CB1 antagonist rimonabant precipitated withdrawal in mice treated chronically with WIN55,212-2 but not in mice treated with GAT211. GAT211 did not induce conditioned place preference or aversion. CONCLUSIONS: Positive allosteric modulation of CB1-receptor signaling shows promise as a safe and effective analgesic strategy that lacks tolerance, dependence, and abuse liability.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Hiperalgesia/tratamiento farmacológico , Indoles/farmacología , Receptor Cannabinoide CB1/metabolismo , Animales , Benzamidas/farmacología , Benzodioxoles/farmacología , Benzoxazinas/farmacología , Carbamatos/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Hiperalgesia/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfolinas/farmacología , Naftalenos/farmacología , Neuralgia/etiología , Paclitaxel , Piperidinas/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Recompensa
8.
Pharmacol Res ; 129: 475-481, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29158048

RESUMEN

The cannabinoid signaling system is found throughout the CNS and its involvement in several pathological processes makes it an attractive therapeutic target. Because orthosteric CB1 cannabinoid receptor ligands have undesirable adverse effects there has been great interest in the development of allosteric modulators - both negative (NAMs) and positive (PAMs) - of these receptors. NAMs of CB1 appeared first on the scene, followed more recently by PAMs. Because allosteric modulation can vary depending on the orthosteric ligand it is important to study their function in a system that employs endogenous cannabinoids. We have recently surveyed first generation NAMs using cultured autaptic hippocampal neurons. These neurons express depolarization induced suppression of excitation (DSE), a form of synaptic plasticity that is mediated by CB1 and 2-arachidonoyl glycerol (2-AG); they are therefore an excellent neuronal model of endogenous cannabinoid signaling in which to test CB1 modulators. In this study we find that while two related compounds, GAT211 and ZCZ011, each show PAM-like responses in autaptic hippocampal neurons, they also exhibit complex pharmacology. Notably we were able to separate the PAM- and agonist-like responses of GAT211 by examining the enantiomers of this racemic compound: GAT228 and GAT229. We find that GAT229 exhibits PAM-like behavior while GAT228 appears to directly activate the CB1 receptor. Both GAT229 and ZCZ011 represent the first PAMs that we have found to be effective in using this 2-AG utilizing neuronal model system. Because these compounds may exhibit both probe selectivity and biased signaling it will be important to test them with anandamide as well as other signaling pathways.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Indoles/farmacología , Neuronas/efectos de los fármacos , Receptor Cannabinoide CB1/fisiología , Tiofenos/farmacología , Regulación Alostérica , Animales , Hipocampo/citología , Hipocampo/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Transducción de Señal , Estereoisomerismo
9.
J Ocul Pharmacol Ther ; 33(8): 582-590, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28719234

RESUMEN

PURPOSE: Orthosteric cannabinoid receptor 1 (CB1) activation leads to decreases in intraocular pressure (IOP). However, use of orthosteric CB1 agonists chronically has several disadvantages, limiting their usefulness as clinically relevant drugs. Allosteric modulators interact with topographically distinct sites to orthosteric ligands and may be useful to circumvent some of these disadvantages. The purpose of this study was to investigate the effects of the novel CB1-positive allosteric modulator (PAM) GAT229 on IOP. METHODS: IOP was measured using rebound tonometry in anesthetized normotensive C57Bl/6 mice and in a genetic model of ocular hypertension [nose, eyes, ears (nee) mice] before drug administration, and at 1, 6, and 12 h thereafter. RESULTS: In normotensive mice, topical administration of 5 µL GAT229 alone at either 0.2% or 2% did not reduce IOP. However, a subthreshold dose (0.25%) of the nonselective orthosteric CB1 agonist WIN 55,212-2, when combined with 0.2% GAT229, significantly reduced IOP compared with vehicle at 6 and 12 h. Similarly, combination of subthreshold Δ9-tetrahydrocannabinol (a nonselective orthosteric CB1 agonist; 1 mg/kg) with topical 0.2% GAT229 produced IOP lowering at 6 h. In nee mice, administration of topical 0.2% GAT229 or 10 mg/kg GAT229 alone was sufficient to lower IOP at 6 and 12 h, and 12 h, respectively. CONCLUSIONS: The CB1 PAM GAT229 reduces IOP in ocular hypertensive mice and enhanced CB1-mediated IOP reduction when combined with subthreshold CB1 orthosteric ligands in normotensive mice. Administration of CB1 PAMs may provide a novel approach to reduce IOP with fewer of the disadvantages associated with orthosteric CB1 activation.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Indoles/farmacología , Presión Intraocular/efectos de los fármacos , Hipertensión Ocular/tratamiento farmacológico , Administración Tópica , Animales , Benzoxazinas/farmacología , Agonistas de Receptores de Cannabinoides/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Indoles/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Naftalenos/farmacología , Receptor Cannabinoide CB1/efectos de los fármacos , Factores de Tiempo , Tonometría Ocular
10.
Neuroscience ; 349: 303-317, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28285942

RESUMEN

Excessive activation of NMDA receptor (NMDAR) signaling within the spinal dorsal horn contributes to central sensitization and the induction and maintenance of pathological pain states. However, direct antagonism of NMDARs produces undesirable side effects which limit their clinical use. NMDAR activation produces central sensitization, in part, by initiating a signaling cascade that activates the enzyme neuronal nitric oxide synthase (nNOS) and generates the signaling molecule nitric oxide. NMDAR-mediated activation of nNOS requires a scaffolding protein, postsynaptic density protein 95kDa (PSD95), which tethers nNOS to NMDARs. Thus, disrupting the protein-protein interaction between PSD95 and nNOS may inhibit pro-nociceptive signaling mechanisms downstream of NMDARs and suppress central sensitization while sparing unwanted side effects associated with NMDAR antagonists. We examined the impact of small molecule PSD95-nNOS protein-protein interaction inhibitors (ZL006, IC87201) on both nociceptive behavior and formalin-evoked Fos protein expression within the lumbar spinal cord of rats. Comparisons were made with ZL007, an inactive analog of ZL006, and the NMDAR antagonist MK-801. IC87201 and ZL006, but not ZL007, suppressed phase 2 of formalin-evoked pain behavior and decreased the number of formalin-induced Fos-like immunoreactive cells in spinal dorsal horn regions associated with nociceptive processing. MK-801 suppressed Fos protein expression in both dorsal and ventral horns. MK-801 produced motor ataxia in the rotarod test whereas IC87201 and ZL006 failed to do so. ZL006 but not ZL007 suppressed paclitaxel-induced mechanical and cold allodynia in a model of chemotherapy-induced neuropathic pain. Co-immunoprecipitation experiments revealed the presence of the PSD95-nNOS complex in lumbar spinal cord of paclitaxel-treated rats, although ZL006 did not reliably disrupt the complex in all subjects. The present findings validate use of putative small molecule PSD95-nNOS protein-protein interaction inhibitors as novel analgesics and demonstrate, for the first time, that these inhibitors suppress inflammation-evoked neuronal activation at the level of the spinal dorsal horn.


Asunto(s)
Conducta Animal/efectos de los fármacos , Homólogo 4 de la Proteína Discs Large/antagonistas & inhibidores , Maleato de Dizocilpina/farmacología , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Nociceptores/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Animales , Formaldehído/farmacología , Masculino , Óxido Nítrico Sintasa de Tipo I/metabolismo , Nociceptores/metabolismo , Ratas Sprague-Dawley , Médula Espinal/metabolismo
11.
ACS Chem Neurosci ; 8(6): 1188-1203, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28103441

RESUMEN

The cannabinoid 1 receptor (CB1R) is one of the most widely expressed metabotropic G protein-coupled receptors in brain, and its participation in various (patho)physiological processes has made CB1R activation a viable therapeutic modality. Adverse psychotropic effects limit the clinical utility of CB1R orthosteric agonists and have promoted the search for CB1R positive allosteric modulators (PAMs) with the promise of improved drug-like pharmacology and enhanced safety over typical CB1R agonists. In this study, we describe the synthesis and in vitro and ex vivo pharmacology of the novel allosteric CB1R modulator GAT211 (racemic) and its resolved enantiomers, GAT228 (R) and GAT229 (S). GAT211 engages CB1R allosteric site(s), enhances the binding of the orthosteric full agonist [3H]CP55,490, and reduces the binding of the orthosteric antagonist/inverse agonist [3H]SR141716A. GAT211 displayed both PAM and agonist activity in HEK293A and Neuro2a cells expressing human recombinant CB1R (hCB1R) and in mouse-brain membranes rich in native CB1R. GAT211 also exhibited a strong PAM effect in isolated vas deferens endogenously expressing CB1R. Each resolved and crystallized GAT211 enantiomer showed a markedly distinctive pharmacology as a CB1R allosteric modulator. In all biological systems examined, GAT211's allosteric agonist activity resided with the R-(+)-enantiomer (GAT228), whereas its PAM activity resided with the S-(-)-enantiomer (GAT229), which lacked intrinsic activity. These results constitute the first demonstration of enantiomer-selective CB1R positive allosteric modulation and set a precedent whereby enantiomeric resolution can decisively define the molecular pharmacology of a CB1R allosteric ligand.


Asunto(s)
Agonistas de Receptores de Cannabinoides/química , Agonistas de Receptores de Cannabinoides/síntesis química , Agonistas de Receptores de Cannabinoides/farmacología , Indoles/química , Indoles/síntesis química , Indoles/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/efectos de los fármacos , Animales , Células HEK293 , Humanos , Isomerismo , Ratones
12.
ACS Chem Neurosci ; 7(6): 776-98, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27046127

RESUMEN

One of the most abundant G-protein coupled receptors (GPCRs) in brain, the cannabinoid 1 receptor (CB1R), is a tractable therapeutic target for treating diverse psychobehavioral and somatic disorders. Adverse on-target effects associated with small-molecule CB1R orthosteric agonists and inverse agonists/antagonists have plagued their translational potential. Allosteric CB1R modulators offer a potentially safer modality through which CB1R signaling may be directed for therapeutic benefit. Rational design of candidate, druglike CB1R allosteric modulators requires greater understanding of the architecture of the CB1R allosteric endodomain(s) and the capacity of CB1R allosteric ligands to tune the receptor's information output. We have recently reported the synthesis of a focused library of rationally designed, covalent analogues of Org27569 and PSNCBAM-1, two prototypic CB1R negative allosteric modulators (NAMs). Among the novel, pharmacologically active CB1R NAMs reported, the isothiocyanate GAT100 emerged as the lead by virtue of its exceptional potency in the [(35)S]GTPγS and ß-arrestin signaling assays and its ability to label CB1R as a covalent allosteric probe with significantly reduced inverse agonism in the [(35)S]GTPγS assay as compared to Org27569. We report here a comprehensive functional profiling of GAT100 across an array of important downstream cell-signaling pathways and analysis of its potential orthosteric probe-dependence and signaling bias. The results demonstrate that GAT100 is a NAM of the orthosteric CB1R agonist CP55,940 and the endocannabinoids 2-arachidonoylglycerol and anandamide for ß-arrestin1 recruitment, PLCß3 and ERK1/2 phosphorylation, cAMP accumulation, and CB1R internalization in HEK293A cells overexpressing CB1R and in Neuro2a and STHdh(Q7/Q7) cells endogenously expressing CB1R. Distinctively, GAT100 was a more potent and efficacious CB1R NAM than Org27569 and PSNCBAM-1 in all signaling assays and did not exhibit the inverse agonism associated with Org27569 and PSNCBAM-1. Computational docking studies implicate C7.38(382) as a key feature of GAT100 ligand-binding motif. These data help inform the engineering of newer-generation, druggable CB1R allosteric modulators and demonstrate the utility of GAT100 as a covalent probe for mapping structure-function correlates characteristic of the druggable CB1R allosteric space.


Asunto(s)
Sitio Alostérico/fisiología , Isotiocianatos/farmacología , Receptor Cannabinoide CB1/química , Transducción de Señal/efectos de los fármacos , Regulación Alostérica , Cannabinoides/farmacología , Células HEK293 , Humanos , Indoles/química , Indoles/farmacología , Isotiocianatos/química , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Piperidinas/química , Piperidinas/farmacología , Unión Proteica , Piridinas/química , Piridinas/farmacología , Receptor Cannabinoide CB1/metabolismo
13.
Behav Brain Res ; 305: 23-9, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26909849

RESUMEN

Limitations of preclinical models of human memory contribute to the pervasive view that rodent models do not adequately predict therapeutic efficacy in producing cognitive impairments or improvements in humans. We used a source-memory model (i.e., a representation of the origin of information) we developed for use in rats to evaluate possible drug-induced impairments of both spatial memory and higher order memory functions in the same task. Memory impairment represents a major barrier to use of NMDAR antagonists as pharmacotherapies. The scaffolding protein postsynaptic density 95kDa (PSD95) links NMDARs to the neuronal enzyme nitric oxide synthase (nNOS), which catalyzes production of the signaling molecule nitric oxide (NO). Therefore, interrupting PSD95-nNOS protein-protein interactions downstream of NMDARs represents a novel therapeutic strategy to interrupt NMDAR-dependent NO signaling while bypassing unwanted side effects of NMDAR antagonists. We hypothesized that the NMDAR antagonist MK-801 would impair source memory. We also hypothesized that PSD95-nNOS inhibitors (IC87201 and ZL006) would lack the profile of cognitive impairment associated with global NMDAR antagonists. IC87201 and ZL006 suppressed NMDA-stimulated formation of cGMP, a marker of NO production, in cultured hippocampal neurons. MK-801, at doses that did not impair motor function, impaired source memory under conditions in which spatial memory was spared. Thus, source memory was more vulnerable than spatial memory to impairment. By contrast, PSD95-nNOS inhibitors, IC87201 and ZL006, administered at doses that are behaviorally effective in rats, spared source memory, spatial memory, and motor function. Thus, PSD95-nNOS inhibitors are likely to exhibit favorable therapeutic ratios compared to NMDAR antagonists.


Asunto(s)
Maleato de Dizocilpina/toxicidad , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/toxicidad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/inducido químicamente , Óxido Nítrico Sintasa de Tipo I/metabolismo , Ácidos Aminosalicílicos/farmacología , Animales , Bencilaminas/farmacología , Células Cultivadas , Clorofenoles/farmacología , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Embrión de Mamíferos , Hipocampo/citología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Long-Evans , Triazoles/farmacología
14.
J Med Chem ; 59(1): 44-60, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26529344

RESUMEN

Undesirable side effects associated with orthosteric agonists/antagonists of cannabinoid 1 receptor (CB1R), a tractable target for treating several pathologies affecting humans, have greatly limited their translational potential. Recent discovery of CB1R negative allosteric modulators (NAMs) has renewed interest in CB1R by offering a potentially safer therapeutic avenue. To elucidate the CB1R allosteric binding motif and thereby facilitate rational drug discovery, we report the synthesis and biochemical characterization of first covalent ligands designed to bind irreversibly to the CB1R allosteric site. Either an electrophilic or a photoactivatable group was introduced at key positions of two classical CB1R NAMs: Org27569 (1) and PSNCBAM-1 (2). Among these, 20 (GAT100) emerged as the most potent NAM in functional assays, did not exhibit inverse agonism, and behaved as a robust positive allosteric modulator of binding of orthosteric agonist CP55,940. This novel covalent probe can serve as a useful tool for characterizing CB1R allosteric ligand-binding motifs.


Asunto(s)
Receptor Cannabinoide CB1/química , Receptor Cannabinoide CB1/efectos de los fármacos , Marcadores de Afinidad , Sitio Alostérico , Animales , Arrestinas/efectos de los fármacos , Arrestinas/metabolismo , Sitios de Unión/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/antagonistas & inhibidores , Ciclohexanoles/farmacología , Descubrimiento de Drogas/métodos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Indoles/farmacología , Ligandos , Modelos Moleculares , Compuestos de Fenilurea/farmacología , Piperidinas/farmacología , Piridinas/farmacología , Ensayo de Unión Radioligante , Ratas , Relación Estructura-Actividad
15.
Neuropharmacology ; 97: 464-75, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26071110

RESUMEN

Aberrant increases in NMDA receptor (NMDAR) signaling contributes to central nervous system sensitization and chronic pain by activating neuronal nitric oxide synthase (nNOS) and generating nitric oxide (NO). Because the scaffolding protein postsynaptic density 95kDA (PSD95) tethers nNOS to NMDARs, the PSD95-nNOS complex represents a therapeutic target. Small molecule inhibitors IC87201 (EC5O: 23.94 µM) and ZL006 (EC50: 12.88 µM) directly inhibited binding of purified PSD95 and nNOS proteins in AlphaScreen without altering binding of PSD95 to ErbB4. Both PSD95-nNOS inhibitors suppressed glutamate-induced cell death with efficacy comparable to MK-801. IC87201 and ZL006 preferentially suppressed phase 2A pain behavior in the formalin test and suppressed allodynia induced by intraplantar complete Freund's adjuvant administration. IC87201 and ZL006 suppressed mechanical and cold allodynia induced by the chemotherapeutic agent paclitaxel (ED50s: 2.47 and 0.93 mg/kg i.p. for IC87201 and ZL006, respectively). Efficacy of PSD95-nNOS disruptors was similar to MK-801. Motor ataxic effects were induced by MK-801 but not by ZL006 or IC87201. Finally, MK-801 produced hyperalgesia in the tail-flick test whereas IC87201 and ZL006 did not alter basal nociceptive thresholds. Our studies establish the utility of using AlphaScreen and purified protein pairs to establish and quantify disruption of protein-protein interactions. Our results demonstrate previously unrecognized antinociceptive efficacy of ZL006 and establish, using two small molecules, a broad application for PSD95-nNOS inhibitors in treating neuropathic and inflammatory pain. Collectively, our results demonstrate that disrupting PSD95-nNOS protein-protein interactions is effective in attenuating pathological pain without producing unwanted side effects (i.e. motor ataxia) associated with NMDAR antagonists.


Asunto(s)
Ácidos Aminosalicílicos/farmacología , Analgésicos/farmacología , Bencilaminas/farmacología , Clorofenoles/farmacología , Triazoles/farmacología , Animales , Ataxia/inducido químicamente , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Maleato de Dizocilpina/efectos adversos , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/metabolismo , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/fisiopatología , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA