Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29149605

RESUMEN

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Receptores Opioides mu/agonistas , Animales , Fentanilo/administración & dosificación , Proteínas de Unión al GTP/metabolismo , Ratones , Morfina/administración & dosificación , Receptores Opioides mu/química , Sistema Respiratorio/efectos de los fármacos , Transducción de Señal , beta-Arrestinas/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34819362

RESUMEN

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.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Bencimidazoles/farmacología , Proteínas de Unión al GTP/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Antagonistas de Narcóticos/farmacología , Piperidinas/farmacología , Receptores Acoplados a Proteínas G/fisiología , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiología , Transducción de Señal/fisiología , Arrestina beta 2/metabolismo
3.
J Am Chem Soc ; 139(16): 5865-5869, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28414442

RESUMEN

Stolonidiol, a marine natural product, has been reported to potentiate the activity of choline acetyltransferase (ChAT), the enzyme that produces the neurotransmitter acetylcholine. Here we report the total synthesis of stolonidiol starting from (R)-(+)-limonene. To identify the mechanism by which ChAT activity is increased, we sought to identify the biological target of stolonidiol. We show that stolonidiol binds to the phorbol ester binding site of protein kinase C (PKC), induces translocation of PKC to the cell membrane, and activates kinase activity. Furthermore, we confirmed the increase in ChAT activity observed upon treatment of cells with stolonidiol and show that this effect is mediated by PKC. Collectively, our data strongly suggest that PKC activation by stolonidiol is responsible for the resulting potentiation of ChAT activity.


Asunto(s)
Colina O-Acetiltransferasa/metabolismo , Diterpenos/farmacología , Cristalografía por Rayos X , Diterpenos/síntesis química , Diterpenos/química , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
4.
J Biol Chem ; 288(31): 22387-98, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23775075

RESUMEN

There is considerable evidence to suggest that drug actions at the κ-opioid receptor (KOR) may represent a means to control pain perception and modulate reward thresholds. As a G protein-coupled receptor (GPCR), the activation of KOR promotes Gαi/o protein coupling and the recruitment of ß-arrestins. It has become increasingly evident that GPCRs can transduce signals that originate independently via G protein pathways and ß-arrestin pathways; the ligand-dependent bifurcation of such signaling is referred to as "functional selectivity" or "signaling bias." Recently, a KOR agonist, 6'-guanidinonaltrindole (6'-GNTI), was shown to display bias toward the activation of G protein-mediated signaling over ß-arrestin2 recruitment. Therefore, we investigated whether such ligand bias was preserved in striatal neurons. Although the reference KOR agonist U69,593 induces the phosphorylation of ERK1/2 and Akt, 6'-GNTI only activates the Akt pathway in striatal neurons. Using pharmacological tools and ß-arrestin2 knock-out mice, we show that KOR-mediated ERK1/2 phosphorylation in striatal neurons requires ß-arrestin2, whereas Akt activation depends upon G protein signaling. These findings reveal a point of KOR signal bifurcation that can be observed in an endogenous neuronal setting and may prove to be an important indicator when developing biased agonists at the KOR.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Guanidinas/farmacología , Naltrexona/análogos & derivados , Neuronas/efectos de los fármacos , Receptores Opioides kappa/efectos de los fármacos , Animales , Células CHO , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Cricetinae , Cricetulus , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Naltrexona/farmacología , Neuronas/metabolismo , Fosforilación
5.
J Biol Chem ; 288(51): 36703-16, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24187130

RESUMEN

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.


Asunto(s)
Receptores Opioides kappa/agonistas , Animales , Arrestinas/metabolismo , Células CHO , Cricetinae , Cricetulus , Descubrimiento de Drogas , Proteínas de Unión al GTP/metabolismo , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Quinolonas/síntesis química , Quinolonas/farmacología , Receptores Opioides kappa/metabolismo , Transducción de Señal , Triazoles/síntesis química , Triazoles/farmacología , beta-Arrestinas
6.
Pharmacol Rev ; 63(4): 1001-19, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21873412

RESUMEN

Opioids are the most effective analgesic drugs for the management of moderate or severe pain, yet their clinical use is often limited because of the onset of adverse side effects. Drugs in this class produce most of their physiological effects through activation of the µ opioid receptor; however, an increasing number of studies demonstrate that different opioids, while presumably acting at this single receptor, can activate distinct downstream responses, a phenomenon termed functional selectivity. Functional selectivity of receptor-mediated events can manifest as a function of the drug used, the cellular or neuronal environment examined, or the signaling or behavioral measure recorded. This review summarizes both in vitro and in vivo work demonstrating functional selectivity at the µ opioid receptor in terms of G protein coupling, receptor phosphorylation, interactions with ß-arrestins, receptor desensitization, internalization and signaling, and details on how these differences may relate to the progression of analgesic tolerance after their extended use.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos/farmacología , Dolor/tratamiento farmacológico , Receptores Opioides mu/fisiología , Analgésicos/uso terapéutico , Analgésicos Opioides/uso terapéutico , Tolerancia a Medicamentos , Reguladores de Proteínas de Unión al GTP/efectos de los fármacos , Reguladores de Proteínas de Unión al GTP/fisiología , Humanos , Dolor/fisiopatología
7.
Crit Rev Biochem Mol Biol ; 45(6): 555-66, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20925600

RESUMEN

Serotonin receptors are the product of 15 distinct genes, 14 of which are G protein-coupled receptors. These receptors are expressed in a wide range of cell types, including distinct neuronal populations, and promote diverse functional responses in multiple organ systems. These receptors are important for mediating the in vivo effects of their cognate neurotransmitter, serotonin, as well as the endogenous tryptamines. In addition, the actions of many drugs are mediated, either directly or indirectly, through serotonin receptors, including antidepressants, antipsychotics, anxiolytics, sleep aids, migraine therapies, gastrointestinal therapeutics and hallucinogenic drugs. It is becoming increasingly evident that serotonin receptors can engage in differential signaling that is determined by the chemical nature of the ligand and that ligands that demonstrate a predilection for inducing a particular signaling cascade are considered to have "functional selectivity". The elucidation of the cellular signaling pathways that mediate the physiological responses to serotonin and other agonists is an active area of investigation and will be an onward-looking focal point for determining how to effectively and selectively promote beneficial serotonergic mimicry while avoiding unwanted clinical side effects. This review highlights the modulation of serotonin 2A, 2C, and four receptors by ß-arrestins, which may represent a fulcrum for biasing receptor responsiveness in vivo.


Asunto(s)
Arrestinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Serotonina/farmacología , Transducción de Señal , Animales , Ansiolíticos/farmacología , Antidepresivos/farmacología , Antipsicóticos/farmacología , Arrestinas/genética , Sistema Nervioso Central/metabolismo , Alucinógenos/farmacología , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Sistema Nervioso Periférico/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Serotonina/genética , Agonistas de Receptores de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , beta-Arrestinas
8.
Biomolecules ; 13(6)2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37371516

RESUMEN

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.


Asunto(s)
Morfina , Compuestos de Espiro , Ratones , Animales , Morfina/farmacología , Analgésicos Opioides/farmacología , Fentanilo/farmacología
9.
J Biol Chem ; 286(36): 31731-41, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21757712

RESUMEN

Morphine and other opiates mediate their effects through activation of the µ-opioid receptor (MOR), and regulation of the MOR has been shown to critically affect receptor responsiveness. Activation of the MOR results in receptor phosphorylation, ß-arrestin recruitment, and internalization. This classical regulatory process can differ, depending on the ligand occupying the receptor. There are two forms of ß-arrestin, ß-arrestin1 and ß-arrestin2 (also known as arrestin2 and arrestin3, respectively); however, most studies have focused on the consequences of recruiting ß-arrestin2 specifically. In this study, we examine the different contributions of ß-arrestin1- and ß-arrestin2-mediated regulation of the MOR by comparing MOR agonists in cells that lack expression of individual or both ß-arrestins. Here we show that morphine only recruits ß-arrestin2, whereas the MOR-selective enkephalin [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), recruits either ß-arrestin. We show that ß-arrestins are required for receptor internalization and that only ß-arrestin2 can rescue morphine-induced MOR internalization, whereas either ß-arrestin can rescue DAMGO-induced MOR internalization. DAMGO activation of the receptor promotes MOR ubiquitination over time. Interestingly, ß-arrestin1 proves to be critical for MOR ubiquitination as modification does not occur in the absence of ß-arrestin1 nor when morphine occupies the receptor. Moreover, the selective interactions between the MOR and ß-arrestin1 facilitate receptor dephosphorylation, which may play a role in the resensitization of the MOR and thereby contribute to overall development of opioid tolerance.


Asunto(s)
Arrestinas/agonistas , Arrestinas/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalinas , Ratones , Fosforilación , Transporte de Proteínas , Ubiquitinación , beta-Arrestinas
10.
J Neurosci ; 30(40): 13513-24, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926677

RESUMEN

Hallucinogens mediate many of their psychoactive effects by activating serotonin 2A receptors (5-HT(2A)R). Although serotonin is the cognate endogenous neurotransmitter and is not considered hallucinogenic, metabolites of serotonin also have high affinity at 5-HT(2A)R and can induce hallucinations in humans. Here we report that serotonin differs from the psychoactive N-methyltryptamines by its ability to engage a ß-arrestin2-mediated signaling cascade in the frontal cortex. Serotonin and 5-hydroxy-L-tryptophan (5-HTP) induce a head-twitch response in wild-type (WT) mice that is a behavioral proxy for 5-HT(2A)R activation. The response in ß-arrestin2 knock-out (ßarr2-KO) mice is greatly attenuated until the doses are elevated, at which point, ßarr2-KO mice display a head-twitch response that can exceed that of WT mice. Direct administration of N-methyltryptamines also produces a greater response in ßarr2-KO mice. Moreover, the inhibition of N-methyltransferase blocks 5-HTP-induced head twitches in ßarr2-KO mice, indicating that N-methyltryptamines, rather than serotonin, primarily mediate this response. Biochemical studies demonstrate that serotonin stimulates Akt phosphorylation in the frontal cortex and in primary cortical neurons through the activation of a ß-arrestin2/phosphoinositide 3-kinase/Src/Akt cascade, whereas N-methyltryptamines do not. Furthermore, disruption of any of the components of this cascade prevents 5-HTP-induced, but not N-methyltryptamine-induced, head twitches. We propose that there is a bifurcation of 5-HT(2A)R signaling that is neurotransmitter and ß-arrestin2 dependent. This demonstration of agonist-directed 5-HT(2A)R signaling in vivo may significantly impact drug discovery efforts for the treatment of disorders wherein hallucinations are part of the etiology, such as schizophrenia, or manifest as side effects of treatment, such as depression.


Asunto(s)
Arrestinas/fisiología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor de Serotonina 5-HT2A/metabolismo , Serotonina/fisiología , Transducción de Señal/fisiología , Triptaminas/fisiología , Familia-src Quinasas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Animales Recién Nacidos , Arrestinas/genética , Células Cultivadas , Lóbulo Frontal/citología , Lóbulo Frontal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/enzimología , Serotonina/metabolismo , Agonistas del Receptor de Serotonina 5-HT2 , Transmisión Sináptica/fisiología , Triptaminas/metabolismo , beta-Arrestinas
11.
Proc Natl Acad Sci U S A ; 105(3): 1079-84, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18195357

RESUMEN

Visual and auditory hallucinations accompany certain neuropsychiatric disorders, such as schizophrenia, and they also can be induced by the use or abuse of certain drugs. The heptahelical serotonin 2A receptors (5-HT2ARs) are molecular targets for drug-induced hallucinations. However, the cellular mechanisms by which the 5-HT2AR mediates these effects are not well understood. Drugs acting at the 5-HT2AR can trigger diverse signaling pathways that may be directed by the chemical properties of the drug. beta-arrestins are intracellular proteins that bind to heptahelical receptors and represent a point where such divergences in ligand-directed functional signaling could occur. Here we compare the endogenous agonist, serotonin, to a synthetic 5-HT2AR hallucinogenic agonist, 2,5-dimethoxy-4-iodoamphetamine (DOI), in mice lacking beta-arrestin-2, as well as in cells lacking beta-arrestins. In mice, we find that serotonin induces a head twitch response by a beta-arrestin-2-dependent mechanism. However, DOI invokes the behavior independent of beta-arrestin-2. The two structurally distinct agonists elicit different signal transduction and trafficking patterns upon activation of 5-HT2AR, which hinge on the presence of beta-arrestins. Our study suggests that the 5-HT2AR-beta-arrestin interaction may be particularly important in receptor function in response to endogenous serotonin levels, which could have major implications in drug development for treating neuropsychiatric disorders such as depression and schizophrenia.


Asunto(s)
Arrestinas/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Agonistas del Receptor de Serotonina 5-HT2 , Transducción de Señal/efectos de los fármacos , Animales , Arrestinas/deficiencia , Arrestinas/genética , Línea Celular , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Arrestina beta 2 , beta-Arrestinas
12.
Neuropharmacology ; 185: 108439, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33345829

RESUMEN

The mu opioid receptor-selective agonist, SR-17018, preferentially activates GTPγS binding over ßarrestin2 recruitment in cellular assays, thereby demonstrating signaling bias. In mice, SR-17018 stimulates GTPγS binding in brainstem and produces antinociception with potencies similar to morphine. However, it produces much less respiratory suppression and mice do not develop antinociceptive tolerance in the hot plate assay upon repeated dosing. Herein we evaluate the effects of acute and repeated dosing of SR-17018, oxycodone and morphine in additional models of pain-related behaviors. In the mouse warm water tail immersion assay, an assessment of spinal reflex to thermal nociception, repeated administration of SR-17018 produces tolerance as does morphine and oxycodone. SR-17018 retains efficacy in a formalin-induced inflammatory pain model upon repeated dosing, while oxycodone does not. In a chemotherapeutic-induced neuropathy pain model SR-17018 is more potent and efficacious than morphine or oxycodone, moreover, this efficacy is retained upon repeated dosing of SR-17018. These findings demonstrate that, with the exception of the tail flick test, SR-17018 retains efficacy upon chronic treatment across several pain models.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Bencimidazoles/administración & dosificación , Morfina/administración & dosificación , Neuralgia/tratamiento farmacológico , Oxicodona/administración & dosificación , Piperidinas/administración & dosificación , Receptores Opioides mu/agonistas , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Femenino , Bombas de Infusión Implantables , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Neuralgia/patología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Resultado del Tratamiento
13.
Pharmacol Ther ; 121(3): 285-93, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19100766

RESUMEN

G protein-coupled receptor-targeted drug discovery as well as "compound reassessment" requires the utilization of diverse screens to determine agonist efficacies and potencies beyond the scope of ligand binding and G protein coupling. Such efforts have arisen from extensive studies, both in cellular and animal models, demonstrating that these seven transmembrane domain-spanning, G protein-coupled receptors may engage in more diverse functions than their name suggests and particular focus is drawn to their interactions with beta-arrestins (betaarrestins). As regulators, betaarrestins are involved in dampening G protein-coupling pathways. betaArrestins can also play pro-signaling roles in receptor mediated events and the coupling of receptors to betaarrestins may be as important as their potential to couple to G proteins in the physiological setting. In the last decade, the development of betaarrestin deficient mouse models has allowed for the assessment of the contribution of individual betaarrestins to receptor function in vivo. This review will discuss the current literature that implicates betaarrestins in receptor function in respect to physiological and behavioral responses observed in the live animal model.


Asunto(s)
Arrestinas/fisiología , Animales , Arrestinas/genética , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , beta-Arrestinas
14.
J Pharmacol Exp Ther ; 334(1): 333-40, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20406855

RESUMEN

Lubiprostone activates ClC-2 chloride channels in epithelia. It is approved for treatment of chronic idiopathic constipation in adults and constipation-predominate irritable bowel syndrome in women. We tested a hypothesis that lubiprostone can reverse the constipating action of morphine and investigated the mechanism of action. Short-circuit current (Isc) was recorded in Ussing chambers as a marker for chloride secretion during pharmacological interactions between morphine and lubiprostone. Measurements of fecal wet weight were used to obtain information on morphine-lubiprostone interactions in conscious mice. Morphine decreased basal Isc, with an IC(50) of 96.1 nM. The action of dimethylphenylpiperazinium (DMPP), a nicotinic receptor agonist that stimulates neurogenic Isc, was suppressed by morphine. Lubiprostone applied after pretreatment with morphine reversed morphine suppression of both basal Isc and DMPP-evoked chloride secretion. Electrical field stimulation (EFS) of submucosal neurons evoked biphasic increases in Isc. Morphine abolished the first phase and marginally suppressed the second phase. Lubiprostone reversed, in concentration-dependent manner, the action of morphine on the first and second phases of the EFS-evoked responses. Subcutaneous lubiprostone increased fecal wet weight and numbers of pellets expelled. Morphine significantly reduced fecal wet weight and number of pellets. Injection of lubiprostone, 30-min after morphine, reversed morphine-induced suppression of fecal wet weight. We conclude that inhibitory action of morphine on chloride secretion reflects suppression of excitability of cholinergic secretomotor neurons in the enteric nervous system. Lubiprostone, which does not directly affect enteric neurons, bypasses the neurogenic constipating effects of morphine by directly opening chloride channels in the mucosal epithelium.


Asunto(s)
Alprostadil/análogos & derivados , Canales de Cloruro/metabolismo , Estreñimiento/prevención & control , Mucosa Intestinal/metabolismo , Morfina/efectos adversos , Alprostadil/administración & dosificación , Alprostadil/farmacología , Alprostadil/uso terapéutico , Animales , Canales de Cloruro CLC-2 , Canales de Cloruro/antagonistas & inhibidores , Cloro/metabolismo , Estreñimiento/inducido químicamente , Relación Dosis-Respuesta a Droga , Heces/química , Cobayas , Técnicas In Vitro , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Lubiprostona , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Neuronas/metabolismo , Neuronas/fisiología , Prostaglandinas/metabolismo
15.
Neuropsychopharmacology ; 45(2): 416-425, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31443104

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/metabolismo , Tolerancia a Medicamentos/fisiología , Dependencia de Morfina/metabolismo , Morfina/metabolismo , Receptores Opioides mu/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Bombas de Infusión Implantables , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Oxicodona/administración & dosificación , Oxicodona/metabolismo , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Receptores Opioides mu/agonistas , Síndrome de Abstinencia a Sustancias/prevención & control
16.
J Virol ; 82(2): 683-91, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17977961

RESUMEN

The retroviral Gag polyprotein directs virus particle assembly, resulting in the release of virions from the plasma membranes of infected cells. The earliest steps in assembly, those immediately following Gag synthesis, are very poorly understood. For Rous sarcoma virus (RSV), Gag proteins are synthesized in the cytoplasm and then undergo transient nuclear trafficking before returning to the cytoplasm for transport to the plasma membrane. Thus, RSV provides a useful model to study the initial steps in assembly because the early and later stages are spatially separated by the nuclear envelope. We previously described mutants of RSV Gag that are defective in nuclear export, thereby isolating these "trapped" Gag proteins at an early assembly step. Using the nuclear export mutants, we asked whether Gag protein-protein interactions occur within the nucleus. Complementation experiments revealed that the wild-type Gag protein could partially rescue export-defective Gag mutants into virus-like particles (VLPs). Additionally, the export mutants had a trans-dominant negative effect on wild-type Gag, interfering with its release into VLPs. Confocal imaging of wild-type and mutant Gag proteins bearing different fluorescent tags suggested that complementation between Gag proteins occurred in the nucleus. Additional evidence for nuclear Gag-Gag interactions was obtained using fluorescence resonance energy transfer, and we found that the formation of intranuclear Gag complexes was dependent on the NC domain. Bimolecular fluorescence complementation allowed the direct visualization of intranuclear Gag-Gag dimers. Together, these experimental results strongly suggest that RSV Gag proteins are capable of interacting within the nucleus.


Asunto(s)
Núcleo Celular/virología , Productos del Gen gag/metabolismo , Virus del Sarcoma de Rous/fisiología , Núcleo Celular/química , Transferencia Resonante de Energía de Fluorescencia , Productos del Gen gag/genética , Microscopía Confocal , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Coloración y Etiquetado/métodos
17.
Sci Signal ; 11(542)2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087177

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Neuronas/metabolismo , Receptores Opioides kappa/agonistas , Transducción de Señal , Animales , Animales Recién Nacidos , Bencenoacetamidas/farmacología , Células CHO , Línea Celular Tumoral , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ratones Noqueados , Pirrolidinas/farmacología , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
18.
Drug Alcohol Depend ; 191: 195-202, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30138791

RESUMEN

Previous reports assessing morphine effects in two bottle choice (TBC) paradigms often use taste adulterants such as sweeteners (e.g., saccharin) and/or bitterants (e.g., quinine) to demonstrate morphine preference with C57BL6 mice. The effect of these additional components on the morphine preference of C57BL6 remains poorly understood. Thus, we sought to elucidate the interrelationship of morphine and quinine in the TBC paradigm. As expected, when morphine was included in the opposite bottle from quinine, a preference for the morphine solution was observed. Conversely, when quinine was included in each bottle, or when fentanyl without quinine was used, no preference was observed. All opioid-drinking mice displayed withdrawal signs, and morphine was detectable in plasma and brain. When these results were compared to previous results via conversion to quinine preference scores, quinine was revealed to determine largely the measured morphine preference. Thus, quinine is effective to drive morphine consumption and engender dependence but may confound the ability to measure oral abuse liability of morphine. Together, these results suggest future TBC procedures should consider the effect of quinine upon measured preference for compounds in the opposite bottle, and that excessively high quinine concentrations appear to influence preference more so than the opposite solute when using C57BL6 mice. Alternative conditions to assess oral abuse liability may be necessary to complement and confirm results from TBC experiments utilizing morphine or other opioids.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Conducta de Elección/efectos de los fármacos , Quinina/administración & dosificación , Síndrome de Abstinencia a Sustancias/metabolismo , Analgésicos Opioides/farmacocinética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Conducta de Elección/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Quinina/farmacocinética , Sacarina/administración & dosificación , Síndrome de Abstinencia a Sustancias/diagnóstico , Gusto/efectos de los fármacos
19.
J Med Chem ; 61(19): 8895-8907, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30199635

RESUMEN

While mu opioid receptor (MOR) agonists are especially effective as broad-spectrum pain relievers, it has been exceptionally difficult to achieve a clear separation of analgesia from many problematic side effects. Recently, many groups have sought MOR agonists that induce minimal ßarrestin-mediated signaling because MOR agonist-treated ßarrestin2 knockout mice were found to display enhanced antinociceptive effects with significantly less respiratory depression and tachyphylaxis. Substantial data now exists to support the premise that G protein signaling biased MOR agonists can be effective analgesic agents. We recently showed that, within a chemical series, the degree of bias correlates linearly with the magnitude of the respiratory safety index. Herein we describe the synthesis and optimization of piperidine benzimidazolone MOR agonists that together display a wide range of bias (G/ßarr2). We identify structural features affecting potency and maximizing bias and show that many compounds have desirable properties, such as long half-lives and high brain penetration.


Asunto(s)
Analgésicos Opioides/farmacología , Barrera Hematoencefálica/metabolismo , Descubrimiento de Drogas/normas , Proteínas de Unión al GTP/metabolismo , Microsomas Hepáticos/metabolismo , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Barrera Hematoencefálica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/efectos de los fármacos , Conformación Proteica , Relación Estructura-Actividad , beta-Arrestinas/metabolismo
20.
ACS Cent Sci ; 3(12): 1329-1336, 2017 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-29296674

RESUMEN

Salvinorin A (SalA) is a plant metabolite that agonizes the human kappa-opioid receptor (κ-OR) with high affinity and high selectivity over mu- and delta-opioid receptors. Its therapeutic potential has stimulated extensive semisynthetic studies and total synthesis campaigns. However, structural modification of SalA has been complicated by its instability, and efficient total synthesis has been frustrated by its dense, complex architecture. Treatment of strategic bonds in SalA as dynamic and dependent on structural perturbation enabled the identification of an efficient retrosynthetic pathway. Here we show that deletion of C20 simultaneously stabilizes the SalA skeleton, simplifies its synthesis, and retains its high affinity and selectivity for the κ-OR. The resulting 10-step synthesis now opens the SalA scaffold to deep-seated property modification. Finally, we describe a workflow to identify structural changes that retain molecular complexity, but reduce synthetic complexity-two related, but independent ways of looking at complexity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA