Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 184(7): 1659-1660, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33798438

RESUMO

Partial agonism describes the relative efficacy of a drug compared to one that produces a greater response in a particular system; the designation is dependent upon the comparator and the system. In this issue of Cell, Huang et al. describe biophysical approaches to define the signature of GPCR partial agonists, providing direct measures of varying intrinsic efficacy.


Assuntos
Cafeína
2.
Cell ; 180(4): 655-665.e18, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32004463

RESUMO

Human endocannabinoid systems modulate multiple physiological processes mainly through the activation of cannabinoid receptors CB1 and CB2. Their high sequence similarity, low agonist selectivity, and lack of activation and G protein-coupling knowledge have hindered the development of therapeutic applications. Importantly, missing structural information has significantly held back the development of promising CB2-selective agonist drugs for treating inflammatory and neuropathic pain without the psychoactivity of CB1. Here, we report the cryoelectron microscopy structures of synthetic cannabinoid-bound CB2 and CB1 in complex with Gi, as well as agonist-bound CB2 crystal structure. Of important scientific and therapeutic benefit, our results reveal a diverse activation and signaling mechanism, the structural basis of CB2-selective agonists design, and the unexpected interaction of cholesterol with CB1, suggestive of its endogenous allosteric modulating role.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Receptor CB1 de Canabinoide/química , Receptor CB2 de Canabinoide/química , Transdução de Sinais , Regulação Alostérica , Sítio Alostérico , Animais , Células CHO , Agonistas de Receptores de Canabinoides/química , Canabinoides/química , Canabinoides/farmacologia , Linhagem Celular Tumoral , Colesterol/química , Colesterol/farmacologia , Cricetinae , Cricetulus , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Humanos , Simulação de Dinâmica Molecular , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Células Sf9 , Spodoptera
3.
Cell ; 167(3): 750-762.e14, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768894

RESUMO

Cannabinoid receptor 1 (CB1) is the principal target of Δ9-tetrahydrocannabinol (THC), a psychoactive chemical from Cannabis sativa with a wide range of therapeutic applications and a long history of recreational use. CB1 is activated by endocannabinoids and is a promising therapeutic target for pain management, inflammation, obesity, and substance abuse disorders. Here, we present the 2.8 Å crystal structure of human CB1 in complex with AM6538, a stabilizing antagonist, synthesized and characterized for this structural study. The structure of the CB1-AM6538 complex reveals key features of the receptor and critical interactions for antagonist binding. In combination with functional studies and molecular modeling, the structure provides insight into the binding mode of naturally occurring CB1 ligands, such as THC, and synthetic cannabinoids. This enhances our understanding of the molecular basis for the physiological functions of CB1 and provides new opportunities for the design of next-generation CB1-targeting pharmaceuticals.


Assuntos
Antagonistas de Receptores de Canabinoides/química , Morfolinas/química , Pirazóis/química , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/química , Sítios de Ligação , Canabinoides/farmacologia , Cannabis/química , Cristalografia por Raios X , Dronabinol/farmacologia , Endocanabinoides/farmacologia , Humanos , Ligantes , Morfolinas/síntese química , Ligação Proteica , Conformação Proteica em alfa-Hélice , Pirazóis/síntese química
4.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34819362

RESUMO

The ability of a ligand to preferentially promote engagement of one signaling pathway over another downstream of GPCR activation has been referred to as signaling bias, functional selectivity, and biased agonism. The presentation of ligand bias reflects selectivity between active states of the receptor, which may result in the display of preferential engagement with one signaling pathway over another. In this study, we provide evidence that the G protein-biased mu opioid receptor (MOR) agonists SR-17018 and SR-14968 stabilize the MOR in a wash-resistant yet antagonist-reversible G protein-signaling state. Furthermore, we demonstrate that these structurally related biased agonists are noncompetitive for radiolabeled MOR antagonist binding, and while they stimulate G protein signaling in mouse brains, partial agonists of this class do not compete with full agonist activation. Importantly, opioid antagonists can readily reverse their effects in vivo. Given that chronic treatment with SR-17018 does not lead to tolerance in several mouse pain models, this feature may be desirable for the development of long-lasting opioid analgesics that remain sensitive to antagonist reversal of respiratory suppression.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Benzimidazóis/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Entorpecentes/farmacologia , Piperidinas/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Transdução de Sinais/fisiologia , beta-Arrestina 2/metabolismo
5.
Biochemistry ; 61(18): 1923-1935, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34468132

RESUMO

In a recent report in Science Signaling (Gillis, A., et al. Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci. Signaling 2020, 13, eaaz3140 10.1126/scisignal.aaz3140), it was suggested that low intrinsic agonism, and not biased agonism, leads to an improvement in the separation of potency in opioid-induced respiratory suppression versus antinociception. Although many of the compounds that were tested have been shown to display G protein signaling bias in prior publications, the authors conclude that because they cannot detect biased agonism in their cellular signaling studies the compounds are therefore not biased agonists. Rather, they conclude that it is low intrinsic efficacy that leads to the therapeutic window improvement. Intrinsic efficacy is the extent to which an agonist can stimulate a G protein-coupled receptor response in a system, while biased agonism takes into consideration not only the intrinsic efficacy but also the potency of an agonist in an assay. Herein, we have reanalyzed the data presented in the published work (10.1126/scisignal.aaz3140) [including the recent Erratum (10.1126/scisignal.abf9803)] to derive intrinsic efficacy and bias factors as ΔΔlog(τ/KA) and ΔΔlog(Emax/EC50), respectively. On the basis of this reanalysis, the data support the conclusion that biased agonism, favoring G protein signaling, was observed. Moreover, a conservation of rank order intrinsic efficacy was not observed upon comparing responses in each assay, further suggesting that multiple active receptor states were present. These observations agree with prior studies in which oliceridine, PZM21, and SR-17018 were first described as biased agonists with improvement in antinociception over respiratory suppression in mice. Therefore, the data in the Science Signaling paper provide strong corroborating evidence that G protein signaling bias may be a means of improving opioid analgesia while avoiding certain undesirable side effects.


Assuntos
Analgésicos Opioides , Receptores Acoplados a Proteínas G , Analgésicos Opioides/efeitos adversos , Animais , Benzimidazóis , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Piperidinas , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/agonistas , Transdução de Sinais
6.
Mol Psychiatry ; 25(9): 2086-2100, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-30120413

RESUMO

The dopamine (DA) D2 receptor (D2R) is an important target for the treatment of neuropsychiatric disorders such as schizophrenia and Parkinson's disease. However, the development of improved therapeutic strategies has been hampered by our incomplete understanding of this receptor's downstream signaling processes in vivo and how these relate to the desired and undesired effects of drugs. D2R is a G protein-coupled receptor (GPCR) that activates G protein-dependent as well as non-canonical arrestin-dependent signaling pathways. Whether these effector pathways act alone or in concert to facilitate specific D2R-dependent behaviors is unclear. Here, we report on the development of a D2R mutant that recruits arrestin but is devoid of G protein activity. When expressed virally in "indirect pathway" medium spiny neurons (iMSNs) in the ventral striatum of D2R knockout mice, this mutant restored basal locomotor activity and cocaine-induced locomotor activity in a manner indistinguishable from wild-type D2R, indicating that arrestin recruitment can drive locomotion in the absence of D2R-mediated G protein signaling. In contrast, incentive motivation was enhanced only by wild-type D2R, signifying a dissociation in the mechanisms that underlie distinct D2R-dependent behaviors, and opening the door to more targeted therapeutics.


Assuntos
Arrestina , Locomoção , Motivação , Receptores de Dopamina D2 , Animais , Cocaína , Corpo Estriado/metabolismo , Camundongos , Camundongos Knockout , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo
7.
Mol Pharmacol ; 96(5): 619-628, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31515283

RESUMO

Cannabinoid receptor 1 (CB1) is a potential therapeutic target for the treatment of pain, obesity and obesity-related metabolic disorders, and addiction. The crystal structure of human CB1 has been determined in complex with the stabilizing antagonist AM6538. In the present study, we characterize AM6538 as a tight-binding/irreversible antagonist of CB1, as well as two derivatives of AM6538 (AM4112 and AM6542) as slowly dissociating CB1 antagonists across binding simulations and cellular signaling assays. The long-lasting nature of AM6538 was explored in vivo wherein AM6538 continues to block CP55,940-mediated behaviors in mice up to 5 days after a single injection. In contrast, the effects of SR141716A abate in mice 2 days after injection. These studies demonstrate the functional outcome of CB1 antagonist modification and open the path for development of long-lasting CB1 antagonists.


Assuntos
Antagonistas de Receptores de Canabinoides/metabolismo , Antagonistas de Receptores de Canabinoides/farmacologia , Nitratos/metabolismo , Nitratos/farmacologia , Piperidinas/metabolismo , Piperidinas/farmacologia , Pirazóis/metabolismo , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Receptor CB1 de Canabinoide/química
8.
Pharmacol Res ; 125(Pt B): 161-177, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28838808

RESUMO

The human cannabinoid subtype 1 receptor (hCB1R) is highly expressed in the CNS and serves as a therapeutic target for endogenous ligands as well as plant-derived and synthetic cannabinoids. Unfortunately, acute use of hCB1R agonists produces unwanted psychotropic effects and chronic administration results in development of tolerance and dependence, limiting the potential clinical use of these ligands. Studies in ß-arrestin knockout mice suggest that interaction of certain GPCRs, including µ-, δ-, κ-opioid and hCB1Rs, with ß-arrestins might be responsible for several adverse effects produced by agonists acting at these receptors. Indeed, agonists that bias opioid receptor activation toward G-protein, relative to ß-arrestin signaling, produce less severe adverse effects. These observations indicate that therapeutic utility of agonists acting at hCB1Rs might be improved by development of G-protein biased hCB1R agonists. Our laboratory recently reported a novel class of indole quinulidinone (IQD) compounds that bind cannabinoid receptors with relatively high affinity and act with varying efficacy. The purpose of this study was to determine whether agonists in this novel cannabinoid class exhibit ligand bias at hCB1 receptors. Our studies found that a novel IQD-derived hCB1 receptor agonist PNR-4-20 elicits robust G protein-dependent signaling, with transduction ratios similar to the non-biased hCB1R agonist CP-55,940. In marked contrast to CP-55,940, PNR-4-20 produces little to no ß-arrestin 2 recruitment. Quantitative calculation of bias factors indicates that PNR-4-20 exhibits from 5.4-fold to 29.5-fold bias for G protein, relative to ß-arrestin 2 signaling (when compared to G protein activation or inhibition of forskolin-stimulated cAMP accumulation, respectively). Importantly, as expected due to reduced ß-arrestin 2 recruitment, chronic exposure of cells to PNR-4-20 results in significantly less desensitization and down-regulation of hCB1Rs compared to similar treatment with CP-55,940. PNR-4-20 (i.p.) is active in the cannabinoid tetrad in mice and chronic treatment results in development of less persistent tolerance and no significant withdrawal signs when compared to animals repeatedly exposed to the non-biased full agoinst JWH-018 or Δ9-THC. Finally, studies of a structurally similar analog PNR- 4-02 show that it is also a G protein biased hCB1R agonist. It is predicted that cannabinoid agonists that bias hCB1R activation toward G protein, relative to ß-arrestin 2 signaling, will produce fewer and less severe adverse effects both acutely and chronically.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Quinuclidinas/farmacologia , Animais , Células CHO , Cricetulus , Cicloexanóis/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Naftalenos/farmacologia , Receptor CB1 de Canabinoide/metabolismo , beta-Arrestina 2/metabolismo
9.
Mol Pharmacol ; 87(5): 866-77, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25680753

RESUMO

Seven transmembrane receptors were originally named and characterized based on their ability to couple to heterotrimeric G proteins. The assortment of coupling partners for G protein-coupled receptors has subsequently expanded to include other effectors (most notably the ßarrestins). This diversity of partners available to the receptor has prompted the pursuit of ligands that selectively activate only a subset of the available partners. A biased or functionally selective ligand may be able to distinguish between different active states of the receptor, and this would result in the preferential activation of one signaling cascade more than another. Although application of the "standard" operational model for analyzing ligand bias is useful and suitable in most cases, there are limitations that arise when the biased agonist fails to induce a significant response in one of the assays being compared. In this article, we describe a quantitative method for measuring ligand bias that is particularly useful for such cases of extreme bias. Using simulations and experimental evidence from several κ opioid receptor agonists, we illustrate a "competitive" model for quantitating the degree and direction of bias. By comparing the results obtained from the competitive model with the standard model, we demonstrate that the competitive model expands the potential for evaluating the bias of very partial agonists. We conclude the competitive model provides a useful mechanism for analyzing the bias of partial agonists that exhibit extreme bias.


Assuntos
Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Animais , Arrestinas/metabolismo , Células CHO , Linhagem Celular , Cricetulus , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Ligantes , Receptores Opioides kappa/agonistas , Transdução de Sinais/efeitos dos fármacos
10.
J Biol Chem ; 288(51): 36703-16, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24187130

RESUMO

The kappa opioid receptor (KOR) is widely expressed in the CNS and can serve as a means to modulate pain perception, stress responses, and affective reward states. Therefore, the KOR has become a prominent drug discovery target toward treating pain, depression, and drug addiction. Agonists at KOR can promote G protein coupling and ßarrestin2 recruitment as well as multiple downstream signaling pathways, including ERK1/2 MAPK activation. It has been suggested that the physiological effects of KOR activation result from different signaling cascades, with analgesia being G protein-mediated and dysphoria being mediated through ßarrestin2 recruitment. Dysphoria associated with KOR activation limits the therapeutic potential in the use of KOR agonists as analgesics; therefore, it may be beneficial to develop KOR agonists that are biased toward G protein coupling and away from ßarrestin2 recruitment. Here, we describe two classes of biased KOR agonists that potently activate G protein coupling but weakly recruit ßarrestin2. These potent and functionally selective small molecule compounds may prove to be useful tools for refining the therapeutic potential of KOR-directed signaling in vivo.


Assuntos
Receptores Opioides kappa/agonistas , Animais , Arrestinas/metabolismo , Células CHO , Cricetinae , Cricetulus , Descoberta de Drogas , Proteínas de Ligação ao GTP/metabolismo , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinolonas/síntese química , Quinolonas/farmacologia , Receptores Opioides kappa/metabolismo , Transdução de Sinais , Triazóis/síntese química , Triazóis/farmacologia , beta-Arrestinas
11.
Eur J Med Chem ; 276: 116627, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38971050

RESUMO

Kappa opioid receptor (KOR) agonists represent promising therapeutics for pain relief due to their analgesic properties along with lower abuse potential than opioids that act at the mu opioid receptor. However, typical KOR agonists produce sedation and dysphoria. Previous studies have shown that G protein signaling-biased KOR agonists may present a means to untangle the desired analgesic properties from undesired side effects. In this paper, we report a new series of G protein signaling-biased KOR agonists entailing -S- → -CH2- replacement in a previously reported KOR agonist, triazole 1.1. With an optimized carbon linker in hand, further development of the scaffold was undertaken to investigate the appendages of the triazole core. The structure-activity relationship study of this series is described, including several analogues that display enhanced potency while maintaining G protein-signaling bias compared to triazole 1.1.

12.
ACS Cent Sci ; 9(8): 1567-1574, 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37637743

RESUMO

The salvinorins serve as templates for next generation analgesics, antipruritics, and dissociative hallucinogens via selective and potent agonism of the kappa-opioid receptor (KOR). In contrast to most opioids, the salvinorins lack basic amines and bind with high affinity and selectivity via complex polyoxygenated scaffolds that have frustrated deep-seated modification by synthesis. Here we describe a short asymmetric synthesis that relies on a sterically confined organocatalyst to dissociate acidity from reactivity and effect Robinson annulation of an unactivated nucleophile/unstable electrophile pair. Combined with a cobalt-catalyzed polarized diene-alkyne cycloaddition, the route allows divergent access to a focused library of salvinorins. We appraise the synthesis by its generation of multiple analogs that exceed the potency, selectivity, stability, and functional bias of salvinorin A itself.

13.
Biomolecules ; 13(6)2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37371516

RESUMO

Opioid analgesics such as morphine and fentanyl induce mu-opioid receptor (MOR)-mediated hyperactivity in mice. Herein, we show that morphine, fentanyl, SR-17018, and oliceridine have submaximal intrinsic efficacy in the mouse striatum using 35S-GTPγS binding assays. While all of the agonists act as partial agonists for stimulating G protein coupling in striatum, morphine, fentanyl, and oliceridine are fully efficacious in stimulating locomotor activity; meanwhile, the noncompetitive biased agonists SR-17018 and SR-15099 produce submaximal hyperactivity. Moreover, the combination of SR-17018 and morphine attenuates hyperactivity while antinociceptive efficacy is increased. The combination of oliceridine with morphine increases hyperactivity, which is maintained over time. These findings provide evidence that noncompetitive agonists at MOR can be used to suppress morphine-induced hyperactivity while enhancing antinociceptive efficacy; moreover, they demonstrate that intrinsic efficacy measured at the receptor level is not directly proportional to drug efficacy in the locomotor activity assay.


Assuntos
Morfina , Compostos de Espiro , Camundongos , Animais , Morfina/farmacologia , Analgésicos Opioides/farmacologia , Fentanila/farmacologia
14.
Mol Pharmacol ; 80(3): 378-88, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21602476

RESUMO

We have reported previously that amiodarone interacts with muscarinic receptors via a novel allosteric site. This study presents mechanistic details on the nature of that interaction. Amiodarone enhanced the maximal level of agonist-stimulated release of arachidonic acid (AA) from Chinese hamster ovary cells that expressed M3 muscarinic receptors; this enhancement was observed for acetylcholine and for the partial agonist pilocarpine. A similar effect of amiodarone was observed when pilocarpine was used to stimulate inositol phosphate (IP) metabolism, but not when acetylcholine was used. Subsequent studies showed that the IP response exhibited a much larger receptor reserve than the AA response, and reduction of that reserve by receptor alkylation unmasked amiodarone's enhancement of the maximal IP response to acetylcholine. Modulating the receptor reserve also revealed acetylcholine's greater affinity (K(A)) for the conformation of the receptor that mediates the AA response. The amiodarone analog N-ethylamiodarone (NEA) did not alter maximal agonist response but merely reduced agonist potency (that is, it appeared to be an antagonist). However, the action of NEA could be clearly distinguished from the action of the orthosteric antagonist NMS. Demonstration of this point was facilitated by an elaboration of Hall's allosteric two-state model; this new model represents a system composed of two ligands that compete with each other at the orthosteric site and two ligands that compete with each other at the allosteric site. In conclusion, amiodarone competes with NEA at a novel, extracellular, allosteric site to enhance the maximal stimulation evoked by acetylcholine and pilocarpine in two different responses.


Assuntos
Amiodarona/farmacologia , Receptor Muscarínico M3/agonistas , Regulação Alostérica , Amiodarona/análogos & derivados , Animais , Ácido Araquidônico/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Fosfatos de Inositol/metabolismo , Ensaio Radioligante
15.
J Pharmacol Exp Ther ; 334(1): 214-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20348203

RESUMO

Allosteric sites on muscarinic receptors may present superior therapeutic targets for several central nervous system disorders, due to the potential of allosteric ligands to provide more selective modulation and to preserve the spatiotemporal patterning that is characteristic of synaptic transmission. We have found that the antiarrhythmic drug amiodarone interacts allosterically with M(1) and M(5) muscarinic receptors. At both M(1) and M(5), amiodarone was only able to partially inhibit the binding of the orthosteric antagonist [(3)H]N-methylscopolamine (NMS). In addition, amiodarone was able to alter the rate of dissociation of [(3)H]NMS from M(1) and M(5) receptors. These findings suggest that NMS and amiodarone are able to bind to the receptor simultaneously. The pharmacology of the effect on NMS dissociation demonstrated that amiodarone was not interacting at the "common" site at which gallamine, obidoxime, and many other muscarinic allosteric ligands are known to bind. In functional studies, amiodarone enhanced the ability of acetylcholine (at EC(20)) to activate the M(5) receptor; however, under the same conditions, amiodarone did not enhance M(1) activation. More detailed studies at M(5) found that the effect of amiodarone was to enhance the efficacy of acetylcholine, without increasing its potency. This report describes the first demonstration of allosteric enhancement of efficacy at the M(5) receptor, and the first demonstration of enhancement of efficacy but not potency at any muscarinic receptor. In summary, amiodarone has been shown to be a novel positive allosteric modulator of muscarinic receptors that is selective for the M(5) subtype, relative to M(1).


Assuntos
Amiodarona/farmacologia , Antiarrítmicos/farmacologia , Membrana Celular/metabolismo , Receptor Muscarínico M5/metabolismo , Regulação Alostérica , Sítio Alostérico , Animais , Ligação Competitiva , Células CHO , Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Ligantes , Ensaio Radioligante , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M5/genética
16.
Neuropsychopharmacology ; 45(2): 416-425, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31443104

RESUMO

It has been demonstrated that opioid agonists that preferentially act at µ-opioid receptors to activate G protein signaling over ßarrestin2 recruitment produce antinociception with less respiratory suppression. However, most of the adverse effects associated with opioid therapeutics are realized after extended dosing. Therefore, we tested the onset of tolerance and dependence, and assessed for neurochemical changes associated with prolonged treatment with the biased agonist SR-17018. When chronically administered to mice, SR-17018 does not lead to hot plate antinociceptive tolerance, receptor desensitization in periaqueductal gray, nor a super-sensitization of adenylyl cyclase in the striatum, which are hallmarks of opioid neuronal adaptations that are seen with morphine. Interestingly, substitution with SR-17018 in morphine-tolerant mice restores morphine potency and efficacy, whereas the onset of opioid withdrawal is prevented. This is in contrast to buprenorphine, which can suppress withdrawal, but produces and maintains morphine antinociceptive tolerance. Biased agonists of this nature may therefore be useful for the treatment of opioid dependence while restoring opioid antinociceptive sensitivity.


Assuntos
Analgésicos Opioides/metabolismo , Tolerância a Medicamentos/fisiologia , Dependência de Morfina/metabolismo , Morfina/metabolismo , Receptores Opioides mu/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Bombas de Infusão Implantáveis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Oxicodona/metabolismo , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides mu/agonistas , Síndrome de Abstinência a Substâncias/prevenção & controle
17.
Methods Mol Biol ; 1957: 235-247, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30919358

RESUMO

G protein-coupled receptors (GPCRs) can interact with both G proteins and ß-arrestin proteins to propagate different signaling outputs. In some contexts, agonists may drive the receptor to preferentially engage one of these effectors over the other. Such "ligand bias" may present a means to impart pathway-selective signaling downstream of this class of receptors. In cases where physiological responses are mediated by diverse pathways, this could, in part, provide a means to refine GPCR therapeutics. Cell-based signaling assays are used to measure the potential for signaling bias in vitro, and these measures take into account potency, efficacy, and the overall capacity of the assay. However, narrow assay windows sometimes limit the confidence in estimating agonist activity, if a compound performs as a very weakly efficacious partial agonist. This lack of response in an assay hampers the ability to measure and compare potencies, and the degree of separation of an agonist's performance, between two assays. In this chapter, we describe in detail a method for the estimation of the relative activity of a partial agonist and provide a stepwise protocol for calculating bias when this case arises.


Assuntos
Bioensaio/métodos , Modelos Biológicos , Viés , Intervalos de Confiança , Ligantes , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo
18.
Life Sci ; 82(5-6): 324-30, 2008 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-18191952

RESUMO

Antigen challenge in sensitized guinea pig esophagus in vitro induces mast cell degranulation and histamine release. This study tests the hypothesis that antigen inhalation in vivo induces infiltration of the esophageal epithelium by mast cells and eosinophils via a histamine pathway. Actively sensitized guinea pigs were exposed to inhaled 0.1% ovalbumin. One or 24 h after inhalation exposure, the esophagus was processed for immunofluorescent staining of mast cell tryptase and eosinophil major basic protein (MBP). Additional animals were pretreated with thioperamide, a histamine H4/H3 receptor antagonist. Total tryptase- and MBP-labeled cells and percent of positive cells in the epithelial layer were counted. The total number of mast cells was unchanged after inhalation challenge, but the percentage in the epithelium increased 1 h after challenge. The total number of eosinophils increased 1 h after challenge, and the percentage migrating to the epithelium increased by 24 h after challenge. Mast cell migration into the mucosal epithelium preceded that of eosinophils. Thioperamide inhibited mast cell and eosinophil migration. In conclusion, antigen inhalation in sensitized animals induces mast cells and eosinophils to infiltrate in the esophageal epithelium via histamine-mediated mechanism.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Eosinófilos/imunologia , Esôfago/imunologia , Histamina/imunologia , Mastócitos/imunologia , Ovalbumina/imunologia , Administração por Inalação , Alérgenos/administração & dosagem , Alérgenos/imunologia , Animais , Antígenos/administração & dosagem , Antígenos/imunologia , Contagem de Células , Eosinófilos/metabolismo , Epitélio/imunologia , Epitélio/metabolismo , Esôfago/metabolismo , Cobaias , Masculino , Mastócitos/metabolismo , Ovalbumina/administração & dosagem , Receptores Histamínicos/imunologia , Receptores Histamínicos/metabolismo , Triptases/metabolismo
19.
Sci Signal ; 11(542)2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087177

RESUMO

Biased agonists of G protein-coupled receptors may present a means to refine receptor signaling in a way that separates side effects from therapeutic properties. Several studies have shown that agonists that activate the κ-opioid receptor (KOR) in a manner that favors G protein coupling over ß-arrestin2 recruitment in cell culture may represent a means to treat pain and itch while avoiding sedation and dysphoria. Although it is attractive to speculate that the bias between G protein signaling and ß-arrestin2 recruitment is the reason for these divergent behaviors, little evidence has emerged to show that these signaling pathways diverge in the neuronal environment. We further explored the influence of cellular context on biased agonism at KOR ligand-directed signaling toward G protein pathways over ß-arrestin-dependent pathways and found that this bias persists in striatal neurons. These findings advance our understanding of how a G protein-biased agonist signal differs between cell lines and primary neurons, demonstrate that measuring [35S]GTPγS binding and the regulation of adenylyl cyclase activity are not necessarily orthogonal assays in cell lines, and emphasize the contributions of the environment to assessing biased agonism.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Neurônios/metabolismo , Receptores Opioides kappa/agonistas , Transdução de Sinais , Animais , Animais Recém-Nascidos , Benzenoacetamidas/farmacologia , Células CHO , Linhagem Celular Tumoral , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Cricetinae , Cricetulus , Células HEK293 , Humanos , Camundongos Knockout , Pirrolidinas/farmacologia , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
20.
Methods Enzymol ; 593: 259-279, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28750807

RESUMO

G protein-coupled receptors, such as the cannabinoid type 1 receptor (CB1R), have been shown to interact with multiple binding partners to transmit signals. In both transfected cell systems and in endogenously expressing cell lines, CB1R signaling has been described as multifaceted. The question remains as to how this highly widely expressed receptor signals in a given cell at a given time in vivo. The concept of functional selectivity, or biased agonism, describes the ability of an agonist to engage the receptor in a manner that preferentially engages certain signaling interactions (e.g., G proteins) over others (e.g., ß-arrestins), presumably by stabilizing certain receptor conformations. There is growing interest in using such properties of ligands to direct signaling downstream of CB1R toward desirable therapeutic outcomes and to avoid adverse side effects. While it is not currently clear what pathways should be engaged and which should be avoided, the development of biased agonist tool compounds will aid in answering these questions. In this chapter, we discuss the approaches and caveats to assessing biased agonism at the CB1R.


Assuntos
Moduladores de Receptores de Canabinoides/farmacologia , Receptor CB1 de Canabinoide/fisiologia , Transdução de Sinais , Regulação Alostérica , Animais , Células Cultivadas , Humanos , Cinética , Ligantes , Processamento de Proteína Pós-Traducional , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA