Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
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
3.
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
4.
J Pharmacol Exp Ther ; 363(3): 394-401, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28970359

RESUMO

Diacylglycerol lipase (DAGL) α and ß, the major biosynthetic enzymes of the endogenous cannabinoid (endocannabinoid) 2-arachidonylglycerol (2-AG), are highly expressed in the nervous system and immune system, respectively. Genetic deletion or pharmacological inhibition of DAGL-ß protects against lipopolysaccharide (LPS)-induced inflammatory responses in mouse peritoneal macrophages and reverses LPS-induced allodynia in mice. To gain insight into the contribution of DAGL-α in LPS-induced allodynia, we tested global knockout mice as well as DO34, a dual DAGL-α/ß inhibitor. Intraperitoneal administration of DO34 (30 mg/kg) significantly decreased whole-brain levels of 2-AG (∼83%), anandamide (∼42%), and arachidonic acid (∼58%). DO34 dose-dependently reversed mechanical and cold allodynia, and these antinociceptive effects did not undergo tolerance after 6 days of repeated administration. In contrast, DO34 lacked acute thermal antinociceptive, motor, and hypothermal pharmacological effects in naive mice. As previously reported, DAGL-ß (-/-) mice displayed a protective phenotype from LPS-induced allodynia. However, DAGL-α (-/-) mice showed full allodynic responses, similar to their wild-type littermates. Interestingly, DO34 (30 mg/kg) fully reversed LPS-induced allodynia in DAGL-α (+/+) and (-/-) mice, but did not affect the antinociceptive phenotype of DAGL-ß (-/-) mice in this model, indicating a DAGL-α-independent site of action. These findings suggest that DAGL-α and DAGL-ß play distinct roles in LPS-induced nociception. Whereas DAGL-α appears to be dispensable for the development and expression of LPS-induced nociception, DAGL-ß inhibition represents a promising strategy to treat inflammatory pain.


Assuntos
Analgésicos/farmacologia , Lipopolissacarídeos/farmacologia , Lipase Lipoproteica/antagonistas & inibidores , Dor/enzimologia , Tiazóis/farmacologia , Ureia/análogos & derivados , Ureia/farmacologia , Analgésicos/uso terapêutico , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância a Medicamentos , Endocanabinoides/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/fisiopatologia , Inflamação/psicologia , Lipase Lipoproteica/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/fisiopatologia , Dor/psicologia , Tiazóis/uso terapêutico , Ureia/uso terapêutico
5.
J Pharmacol Exp Ther ; 357(1): 145-56, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26791602

RESUMO

Serious clinical liabilities associated with the prescription of opiates for pain control include constipation, respiratory depression, pruritus, tolerance, abuse, and addiction. A recognized strategy to circumvent these side effects is to combine opioids with other antinociceptive agents. The combination of opiates with the primary active constituent of cannabis (Δ(9)-tetrahydrocannabinol) produces enhanced antinociceptive actions, suggesting that cannabinoid receptor agonists can be opioid sparing. Here, we tested whether elevating the endogenous cannabinoid 2-arachidonoylglycerol through the inhibition of its primary hydrolytic enzyme monoacylglycerol lipase (MAGL), will produce opioid-sparing effects in the mouse chronic constriction injury (CCI) of the sciatic nerve model of neuropathic pain. The dose-response relationships of i.p. administration of morphine and the selective MAGL inhibitor 2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate (MJN110) were tested alone and in combination at equieffective doses for reversal of CCI-induced mechanical allodynia and thermal hyperalgesia. The respective ED50 doses (95% confidence interval) of morphine and MJN110 were 2.4 (1.9-3.0) mg/kg and 0.43 (0.23-0.79) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. Acute antinociceptive effects of the combination of morphine and MJN110 required µ-opioid, CB1, and CB2 receptors. This combination did not reduce gastric motility or produce subjective cannabimimetic effects in the drug discrimination assay. Importantly, combinations of MJN110 and morphine given repeatedly (i.e., twice a day for 6 days) continued to produce antiallodynic effects with no evidence of tolerance. Taken together, these findings suggest that MAGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects.


Assuntos
Analgésicos Opioides/uso terapêutico , Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Monoacilglicerol Lipases/antagonistas & inibidores , Neuralgia/tratamento farmacológico , Succinimidas/farmacologia , Animais , Ácidos Araquidônicos/metabolismo , Comportamento Animal/efeitos dos fármacos , Constrição Patológica/complicações , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/uso terapêutico , Neuralgia/induzido quimicamente , Neuralgia/psicologia , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB2 de Canabinoide/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
6.
J Pharmacol Exp Ther ; 354(2): 111-20, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25998048

RESUMO

Inhibition of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), the primary hydrolytic enzymes for the respective endocannabinoids N-arachidonoylethanolamine (AEA) and 2-arachidonylglycerol (2-AG), produces antinociception but with minimal cannabimimetic side effects. Although selective inhibitors of either enzyme often show partial efficacy in various nociceptive models, their combined blockade elicits augmented antinociceptive effects, but side effects emerge. Moreover, complete and prolonged MAGL blockade leads to cannabinoid receptor type 1 (CB1) receptor functional tolerance, which represents another challenge in this potential therapeutic strategy. Therefore, the present study tested whether full FAAH inhibition combined with partial MAGL inhibition would produce sustained antinociceptive effects with minimal cannabimimetic side effects. Accordingly, we tested a high dose of the FAAH inhibitor PF-3845 (N-​3-​pyridinyl-​4-​[[3-​[[5-​(trifluoromethyl)-​2-​pyridinyl]oxy]phenyl]methyl]-​1-​piperidinecarboxamide; 10 mg/kg) given in combination with a low dose of the MAGL inhibitor JZL184 [4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate] (4 mg/kg) in mouse models of inflammatory and neuropathic pain. This combination of inhibitors elicited profound increases in brain AEA levels (>10-fold) but only 2- to 3-fold increases in brain 2-AG levels. This combination produced significantly greater antinociceptive effects than single enzyme inhibition and did not elicit common cannabimimetic effects (e.g., catalepsy, hypomotility, hypothermia, and substitution for Δ(9)-tetrahydrocannabinol in the drug-discrimination assay), although these side effects emerged with high-dose JZL184 (i.e., 100 mg/kg). Finally, repeated administration of this combination did not lead to tolerance to its antiallodynic actions in the carrageenan assay or CB1 receptor functional tolerance. Thus, full FAAH inhibition combined with partial MAGL inhibition reduces neuropathic and inflammatory pain states with minimal cannabimimetic effects.


Assuntos
Amidoidrolases/antagonistas & inibidores , Analgésicos/administração & dosagem , Agonistas de Receptores de Canabinoides/administração & dosagem , Antagonistas de Receptores de Canabinoides/administração & dosagem , Monoacilglicerol Lipases/antagonistas & inibidores , Amidoidrolases/metabolismo , Animais , Benzodioxóis/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Quimioterapia Combinada , Hiperalgesia/tratamento farmacológico , Hiperalgesia/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monoacilglicerol Lipases/metabolismo , Piperidinas/administração & dosagem , Piridinas/administração & dosagem , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Fatores de Tempo , Resultado do Tratamento
7.
J Pharmacol Exp Ther ; 352(2): 195-207, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25398241

RESUMO

A growing body of evidence implicates endogenous cannabinoids as modulators of the mesolimbic dopamine system and motivated behavior. Paradoxically, the reinforcing effects of Δ(9)-tetrahydrocannabinol (THC), the primary psychoactive constituent of cannabis, have been difficult to detect in preclinical rodent models. In this study, we investigated the impact of THC and inhibitors of the endocannabinoid hydrolytic enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) on operant responding for electrical stimulation of the medial forebrain bundle [intracranial self-stimulation (ICSS)], which is known to activate the mesolimbic dopamine system. These drugs were also tested in assays of operant responding for food reinforcement and spontaneous locomotor activity. THC and the MAGL inhibitor JZL184 (4-[bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester) attenuated operant responding for ICSS and food, and also reduced spontaneous locomotor activity. In contrast, the FAAH inhibitor PF-3845 (N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide) was largely without effect in these assays. Consistent with previous studies showing that combined inhibition of FAAH and MAGL produces a substantially greater cannabimimetic profile than single enzyme inhibition, the dual FAAH-MAGL inhibitor SA-57 (4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester) produced a similar magnitude of ICSS depression as that produced by THC. ICSS attenuation by JZL184 was associated with increased brain levels of 2-arachidonoylglycerol (2-AG), whereas peak effects of SA-57 were associated with increased levels of both N-arachidonoylethanolamine (anandamide) and 2-AG. The cannabinoid receptor type 1 receptor antagonist rimonabant, but not the cannabinoid receptor type 2 receptor antagonist SR144528, blocked the attenuating effects of THC, JZL184, and SA-57 on ICSS. Thus, THC, MAGL inhibition, and dual FAAH-MAGL inhibition not only reduce ICSS, but also decrease other reinforced and nonreinforced behaviors.


Assuntos
Dronabinol/farmacologia , Endocanabinoides/metabolismo , Inibidores Enzimáticos/farmacologia , Feixe Prosencefálico Mediano/efeitos dos fármacos , Reforço Psicológico , Autoestimulação , Amidoidrolases/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Benzodioxóis/farmacologia , Compostos de Bifenilo/farmacologia , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estimulação Elétrica , Masculino , Feixe Prosencefálico Mediano/enzimologia , Feixe Prosencefálico Mediano/metabolismo , Camundongos Endogâmicos C57BL , Monoacilglicerol Lipases/antagonistas & inibidores , Atividade Motora/efeitos dos fármacos , Piperidinas/farmacologia , Compostos de Piridínio/farmacologia
8.
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
9.
Neuropharmacology ; 185: 108439, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33345829

RESUMO

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.


Assuntos
Analgésicos Opioides/administração & dosagem , Benzimidazóis/administração & dosagem , Morfina/administração & dosagem , Neuralgia/tratamento farmacológico , Oxicodona/administração & dosagem , Piperidinas/administração & dosagem , Receptores Opioides mu/agonistas , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Feminino , Bombas de Infusão Implantáveis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neuralgia/patologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Resultado do Tratamento
10.
J Med Chem ; 64(7): 3870-3884, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33761251

RESUMO

We report the development of novel cannabinergic probes that can stabilize the cannabinoid receptors (CBRs) through tight binding interactions. Ligand design involves the introduction of select groups at a judiciously chosen position within the classical hexahydrocannabinol template (monofunctionalized probes). Such groups include the electrophilic isothiocyanato, the photoactivatable azido, and the polar cyano moieties. These groups can also be combined to produce bifunctionalized probes potentially capable of interacting at two distinct sites within the CBR-binding domains. These novel compounds display remarkably high binding affinities for CBRs and are exceptionally potent agonists. A key ligand (27a, AM11245) exhibits exceptionally high potency in both in vitro and in vivo assays and was designated as "megagonist," a property attributed to its tight binding profile. By acting both centrally and peripherally, 27a distinguishes itself from our previously reported "megagonist" AM841, whose functions are restricted to the periphery.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Canabinoides/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Analgésicos/síntese química , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Células CHO , Agonistas de Receptores de Canabinoides/síntese química , Agonistas de Receptores de Canabinoides/metabolismo , Canabinoides/síntese química , Canabinoides/metabolismo , Cricetulus , Humanos , Ligantes , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Simulação de Acoplamento Molecular , Ratos
11.
Biol Psychiatry ; 87(1): 15-21, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31806082

RESUMO

The mu opioid receptor (MOR) is a diversely regulated target for the alleviation of pain in the clinical setting. However, untoward side effects such as tolerance, dependence, respiratory suppression, constipation, and abuse liability detract from the general activation of these receptors. Studies in genetically modified rodent models suggest that activating G protein signaling pathways while avoiding phosphorylation of the receptor or recruitment of ß-arrestin scaffolding proteins could preserve the analgesic properties of MOR agonists while avoiding certain side effects. With the development of novel MOR "biased" agonists, which lead to preferential activation of G protein pathways over receptor phosphorylation, internalization, or interaction with other effectors, this hypothesis can be tested in a native, physiological setting. Overall, it is clear that the MOR is not a simple on-off switch and that the diverse means by which the receptor can be regulated may present an opportunity to refine therapeutics for the treatment of pain.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Receptores Opioides mu/metabolismo , Transdução de Sinais , beta-Arrestinas/metabolismo
12.
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
13.
Neuropharmacology ; 150: 200-209, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30660628

RESUMO

One emerging strategy to address the opioid crisis is the development of mu opioid receptor (MOR) ligands that preferentially signal the G-protein vs. ß-arrestin pathway. The present study compared the relative potency and effectiveness of two G-protein biased (GPB)-MOR ligands TRV130 and SR-14968 to five unbiased MOR ligands (NAQ, nalbuphine, buprenorphine, morphine, and methadone) on therapeutic-related (e.g. antinociception) and abuse-related (e.g. discriminative stimulus effects) endpoints. Male and female rats were tested in a warm water tail-withdrawal procedure (50 °C) or trained to discriminate fentanyl (0.04 mg/kg, SC) from saline in a two-lever food-reinforced discrimination procedure. TRV130 and SR-14968 were approximately two-fold more potent to produce fentanyl stimulus effects vs. antinociception. Morphine, fentanyl, and methadone were significantly more potent in the fentanyl discrimination vs. tail withdrawal procedure. In addition, maximum antinociceptive and discriminative stimulus effects of fixed-proportion fentanyl/naltrexone mixtures (1:0.018, 1:0.054, 1:0.18, 1:0.3, and 1:0.54) were used to quantify 1) the relative in vivo efficacy of the two GPB-MOR agonists and five unbiased MOR ligands, and 2) potential species differences in MOR ligand effects between rats and monkeys. The efficacy-effect function generated from the fentanyl/naltrexone mixtures stratified the five unbiased ligands consistent with agonist-stimulated GTPγS binding (NAQ = nalbuphine < buprenorphine < morphine < methadone). However, TRV130 and SR-14968 produced greater antinociception and less fentanyl-like stimulus effects than was predicted. Furthermore, there was a significant positive correlation between rat and monkey antinociceptive effects. Overall, these results demonstrate GPB-MOR agonists produce undesirable abuse-related effects, albeit with slightly better potency and efficacy ratios compared to unbiased agonists. This article is part of the Special Issue entitled 'Opioid Neuropharmacology: Advances in treating pain and opioid addiction'.


Assuntos
Aprendizagem por Discriminação/efeitos dos fármacos , Fentanila/farmacologia , Nociceptividade/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Receptores Opioides mu/agonistas , Compostos de Espiro/farmacologia , Tiofenos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Feminino , Masculino , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley
14.
Drug Alcohol Depend ; 191: 195-202, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30138791

RESUMO

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.


Assuntos
Analgésicos Opioides/administração & dosagem , Comportamento de Escolha/efeitos dos fármacos , Quinina/administração & dosagem , Síndrome de Abstinência a Substâncias/metabolismo , Analgésicos Opioides/farmacocinética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Comportamento de Escolha/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Quinina/farmacocinética , Sacarina/administração & dosagem , Síndrome de Abstinência a Substâncias/diagnóstico , Paladar/efeitos dos fármacos
15.
Nat Commun ; 8: 13958, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28045021

RESUMO

The cannabinoid CB2 receptor (CB2R) represents a promising therapeutic target for various forms of tissue injury and inflammatory diseases. Although numerous compounds have been developed and widely used to target CB2R, their selectivity, molecular mode of action and pharmacokinetic properties have been poorly characterized. Here we report the most extensive characterization of the molecular pharmacology of the most widely used CB2R ligands to date. In a collaborative effort between multiple academic and industry laboratories, we identify marked differences in the ability of certain agonists to activate distinct signalling pathways and to cause off-target effects. We reach a consensus that HU910, HU308 and JWH133 are the recommended selective CB2R agonists to study the role of CB2R in biological and disease processes. We believe that our unique approach would be highly suitable for the characterization of other therapeutic targets in drug discovery research.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Neurônios/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Transdução de Sinais , Animais , Compostos Bicíclicos com Pontes/farmacologia , Células CHO , Canabinoides/farmacologia , Linhagem Celular Tumoral , Cricetulus , Expressão Gênica , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Cinética , Ligantes , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Ligação Proteica , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/genética
16.
J Basic Clin Physiol Pharmacol ; 27(3): 217-28, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-27149200

RESUMO

A series of in vivo and in vitro assays were conducted to characterize the pharmacological effects of the first generation abused synthetic cannabinoid CP47,497, a racemic bicyclic cannabinoid that is similar in structure to the potent, high-efficacy synthetic cannabinoid CP55,940. CP47,497 was less efficacious than CP55,940 in activating G-proteins and dose-dependently produced common CB1 receptor-dependent pharmacological effects (i.e. catalepsy, hypothermia, antinociception, and hypolocomotion). CP47,497 also substituted for Δ9-tetrahydrocannabinol (THC) in the mouse drug discrimination, indicating that both drugs elicited a similar interceptive stimulus. The pharmacological effects of CP47,497 underwent tolerance following repeated administration and showed cross-tolerance following repeated THC administration, further suggesting a common cannabimimetic mechanism of action. Finally, the CB1 receptor antagonist rimonabant precipitated similar magnitudes of somatic withdrawal responses in mice treated repeatedly with THC or CP47,497. Taken together, these data verify the acute cannabimimetic effects of CP47,497, and indicate tolerance and dependence following repeated administration. The assays used here provide a straightforward approach to characterize the emerging next generation of abused synthetic cannabinoids.


Assuntos
Canabinoides/farmacologia , Animais , Antagonistas de Receptores de Canabinoides/farmacologia , Dronabinol/farmacologia , Tolerância a Medicamentos , Endocanabinoides/farmacologia , Feminino , Proteínas de Ligação ao GTP/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto
17.
Drug Alcohol Depend ; 150: 31-7, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25772438

RESUMO

BACKGROUND: Synthetic cannabinoids have emerged as a significant public health concern. To increase the knowledge of how these molecules interact on brain reward processes, we investigated the effects of CP55,940, a high efficacy synthetic CB1 receptor agonist, in a frequency-rate intracranial self-stimulation (ICSS) procedure. METHODS: The impact of acute and repeated administration (seven days) of CP55,940 on operant responding for electrical brain stimulation of the medial forebrain bundle was investigated in C57BL/6J mice. RESULTS: CP55,940 attenuated ICSS in a dose-related fashion (ED50 (95% C.L.)=0.15 (0.12-0.18)mg/kg). This effect was blocked by the CB1 receptor antagonist rimonabant. Tolerance developed quickly, though not completely, to the rate-decreasing effects of CP55,940 (0.3mg/kg). Abrupt discontinuation of drug did not alter baseline responding for up to seven days. Moreover, rimonabant (10mg/kg) challenge did not alter ICSS responding in mice treated repeatedly with CP55,940. CONCLUSIONS: The finding that CP55,940 reduced ICSS in mice with no evidence of facilitation at any dose is consistent with synthetic cannabinoid effects on ICSS in rats. CP55,940-induced ICSS depression was mediated through a CB1 receptor mechanism. Additionally, tolerance and dependence following repeated CP55,940 administration were dissociable. Thus, CP55,940 does not produce reward-like effects in ICSS under these conditions.


Assuntos
Cicloexanóis/farmacologia , Receptor CB1 de Canabinoide/agonistas , Recompensa , Autoestimulação/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Catalepsia/induzido quimicamente , Condicionamento Operante/efeitos dos fármacos , Cicloexanóis/antagonistas & inibidores , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Masculino , Feixe Prosencefálico Mediano/fisiologia , Camundongos , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto
18.
Pharmacol Biochem Behav ; 124: 405-11, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25058512

RESUMO

UNLABELLED: Common pharmacological treatments of neuropathic and chronic inflammatory pain conditions generally lack efficacy and/or are associated with significant untoward side effects. However, recent preclinical data indicate that combined inhibition of cyclooxygenase (COX) and fatty acid amide hydrolase (FAAH), the primary catabolic enzyme of the endocannabinoid N-arachidonoylethanolamine (anandamide; AEA), produces enhanced antinociceptive effects in a variety of murine models of pain. Accordingly, the primary objective of the present study was to investigate the consequences of co-administration of the COX inhibitor diclofenac and the highly selective FAAH inhibitor PF-3845 in models of neuropathic pain (i.e., chronic constrictive injury of the sciatic nerve (CCI)) and inflammatory pain induced by an intraplantar injection of carrageenan. Here, we report that combined administration of subthreshold doses of these drugs produced enhanced antinociceptive effects in CCI and carrageenan pain models, the latter of which was demonstrated to require both CB1 and CB2 receptors. The combined administration of subthreshold doses of these drugs also increased AEA levels and decreased prostaglandin levels in whole brain. Together, these data add to the growing research that dual blockade of FAAH and COX represents a potential therapeutic strategy for the treatment of neuropathic and inflammatory pain states. PERSPECTIVE: Tandem inhibition of FAAH and COX attenuates inflammatory and neuropathic pain states, which may avoid potentially harmful side effects of other therapeutic options, such as NSAIDs or opioids.


Assuntos
Amidoidrolases/antagonistas & inibidores , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Inflamação/tratamento farmacológico , Neuralgia/tratamento farmacológico , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Animais , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa