Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pharmacol ; 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38769020

RESUMO

Opioid analgesics are widely used as a treatment option for pain management and relief. However, the misuse of opioid analgesics has contributed to the current opioid epidemic in the United States. Prescribed opioids such as morphine, codeine, oxycodone, and fentanyl are mu-opioid receptor (MOR) agonists primarily used in the clinic to treat pain or during medical procedures, but development of tolerance limits their utility for treatment of chronic pain. Here we explored the effects of biasing Gßγ-signaling on tolerance development following chronic morphine treatment in vivo We hypothesized that biasing Gßγ-signaling with gallein could prevent activation of regulatory signaling pathways that result in tolerance to antinociceptive effects of MOR agonists. Gallein has been shown to bind to Gßγ and inhibit interactions of Gßγ with PLCß3 or GRK2 but not GIRK channels. In mice, morphine-induced antinociception was evaluated in the 55{degree sign}C warm water tail withdrawal assay. We used two paradigms for gallein treatment: administration during and after 3x daily morphine administration. Our results show that gallein cotreatment during repeated administration of morphine decreased opioid tolerance development, and gallein treatment in an opioid-tolerant state enhanced the potency of morphine. Mechanistically, our data suggest that PLCß3 is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. These studies demonstrate that small molecules that target Gßγ-signaling could reduce the need for large doses of opioid analgesics to treat pain by producing an opioid-sparing effect. Significance Statement Biasing Gßγ-signaling prevents tolerance to repeated morphine administration in vivo and potentiates the antinociceptive effects of morphine in an opioid-tolerant state. Mechanistically, PLCß is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. This study identifies a novel treatment strategy to decrease the development of tolerance to the analgesic effects of MOR agonists, which are necessary to improve pain treatment and decrease the incidence of OUD.

2.
J Pharmacol Exp Ther ; 384(3): 343-352, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36456196

RESUMO

Major depressive disorder is a highly common disorder, with a lifetime prevalence in the United States of approximately 21%. Traditional antidepressant treatments are limited by a delayed onset of action and minimal efficacy in some patients. Ketamine is effective and fast-acting, but there are concerns over its abuse liability. Thus, there is a need for safe, fast-acting antidepressant drugs. The opioid buprenorphine shows promise but also has abuse liability due to its mu-agonist component. Preclinical evidence indicates that the delta-opioid system contributes to mood disorders, and delta-opioid agonists are effective in preclinical models of depression- and anxiety-like states. In this study, we test the hypothesis that the mu-opioid antagonist diprenorphine by virtue of its partial delta opioid agonist activity may offer a beneficial profile for an antidepressant medication without abuse liability. Diprenorphine was confirmed to bind with high affinity to all three opioid receptors, and functional experiments for G protein activation verified diprenorphine to be a partial agonist at delta- and kappa-opioid receptors and a mu-antagonist. Studies in C57BL/6 mice demonstrated that an acute dose of diprenorphine produced antidepressant-like effects in the tail suspension test and the novelty-induced hypophagia test that were inhibited in the presence of the delta-selective antagonist, naltrindole. Diprenorphine did not produce convulsions, a side effect of many delta agonists but rather inhibited convulsions caused by the full delta agonist SNC80; however, diprenorphine did potentiate pentylenetetrazole-induced convulsions. Diprenorphine, and compounds with a similar pharmacological profile, may provide efficient and safe rapidly acting antidepressants. SIGNIFICANCE STATEMENT: The management of major depressive disorder, particularly treatment-resistant depression, is a significant unmet medical need. Here we show that the opioid diprenorphine, a compound with mu-opioid receptor antagonist activity and delta- and kappa-opioid receptor partial agonist activities, has rapid onset antidepressant-like activity in animal models. Diprenorphine and compounds with a similar pharmacological profile to diprenorphine should be explored as novel antidepressant drugs.


Assuntos
Analgésicos Opioides , Transtorno Depressivo Maior , Diprenorfina , Animais , Camundongos , Analgésicos Opioides/farmacologia , Antidepressivos/farmacologia , Diprenorfina/farmacologia , Camundongos Endogâmicos C57BL , Receptores Opioides , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Convulsões/induzido quimicamente
3.
Mol Pharmacol ; 102(6): 269-279, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36116788

RESUMO

Pain management is an important problem worldwide. The current frontline approach for pain management is the use of opioid analgesics. The primary analgesic target of opioids is the µ-opioid receptor (MOR). Deletion of phospholipase Cß3 (PLCß3) or selective inhibition of Gßγ regulation of PLCß3 enhances the potency of the antinociceptive effects of morphine suggesting a novel strategy for achieving opioid-sparing effects. Here we investigated a potential mechanism for regulation of PLC signaling downstream of MOR in human embryonic kidney 293 cells and found that MOR alone could not stimulate PLC but rather required a coincident signal from a Gq-coupled receptor. Knockout of PLCß3 or pharmacological inhibition of its upstream regulators, Gßγ or Gq, ex vivo in periaqueductal gray slices increased the potency of the selective MOR agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin acetate salt in inhibiting presynaptic GABA release. Finally, inhibition of Gq- G protein-coupled receptor coupling in mice enhanced the antinociceptive effects of morphine. These data support a model where Gq and Gßγ-dependent signaling cooperatively regulate PLC activation to decrease MOR-dependent antinociceptive potency. Ultimately, this could lead to identification of new non-MOR targets that would allow for lower-dose utilization of opioid analgesics. SIGNIFICANCE STATEMENT: Previous work demonstrated that deletion of phospholipase Cß3 (PLCß3) in mice potentiates µ-opioid receptor (MOR)-dependent antinociception. How PLCß3 is regulated downstream of MOR had not been clearly defined. We show that PLC-dependent diacylglycerol generation is cooperatively regulated by MOR-Gßγ and Gq-coupled receptor signaling through PLCß3 and that blockade of either Gq-signaling or Gßγ signaling enhances the potency of opioids in ex vivo brain slices and in vivo. These results reveal potential novel strategies for improving opioid analgesic potency and safety.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Animais , Camundongos , Humanos , Analgésicos Opioides/farmacologia , Fosfolipase C beta , Camundongos Knockout , Receptores Opioides mu/fisiologia , Morfina/farmacologia , Analgésicos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia
4.
Front Syst Neurosci ; 16: 932546, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35993087

RESUMO

Enkephalin, an endogenous opioid peptide, is highly expressed in the reward pathway and may modulate neurotransmission to regulate reward-related behaviors, such as drug-taking and drug-seeking behaviors. Drugs of abuse also directly increase enkephalin in this pathway, yet it is unknown whether or not changes in the enkephalinergic system after drug administration mediate any specific behaviors. The use of animal models of substance use disorders (SUDs) concurrently with pharmacological, genetic, and molecular tools has allowed researchers to directly investigate the role of enkephalin in promoting these behaviors. In this review, we explore neurochemical mechanisms by which enkephalin levels and enkephalin-mediated signaling are altered by drug administration and interrogate the contribution of enkephalin systems to SUDs. Studies manipulating the receptors that enkephalin targets (e.g., mu and delta opioid receptors mainly) implicate the endogenous opioid peptide in drug-induced neuroadaptations and reward-related behaviors; however, further studies will need to confirm the role of enkephalin directly. Overall, these findings suggest that the enkephalinergic system is involved in multiple aspects of SUDs, such as the primary reinforcing properties of drugs, conditioned reinforcing effects, and sensitization. The idea of dopaminergic-opioidergic interactions in these behaviors remains relatively novel and warrants further research. Continuing work to elucidate the role of enkephalin in mediating neurotransmission in reward circuitry driving behaviors related to SUDs remains crucial.

5.
Mol Pharmacol ; 101(5): 371-380, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35236771

RESUMO

The synthetic cannabinoid WIN55,212-2 (WIN) is widely used as a pharmacological tool to study the biologic activity of cannabinoid receptors. In contrast to many other cannabinoid agonists, however, WIN also causes broad effects outside of neurons, such as reducing inflammatory responses, causing cell cycle arrest, and reducing general protein expression. How exactly WIN causes these broad effects is not known. Here we show that WIN partially disrupts the Golgi apparatus at nanomolar concentrations and fully disperses the Golgi apparatus in neuronal and non-neuronal cells at micromolar concentrations. WIN55,212-3, the enantiomer of WIN; JWH-018, a related alkylindole; or 2-arachidonoylglycerol, an endocannabinoid, did not cause Golgi disruption, suggesting that the effect was specific to the chirality of WIN. WIN treatment also perturbed the microtubule network. Importantly, WIN disrupted the Golgi in primary cortical neurons derived from mice where cannabinoid receptor-1 (CB1) was genetically knocked out, indicating that the effects were independent of CB1 signaling. The Golgi dispersion could not be explained by WIN's action on peroxisome proliferator-activated receptors. Our results show that WIN can disrupt the Golgi apparatus independent of CB1 in cultured cells. These effects could contribute to the unique physiologic effects that WIN exhibits in neuronal behavior, as well as its role as an antiproliferative and anti-inflammatory agent. SIGNIFICANCE STATEMENT: The synthetic cannabinoid WIN55,212-2 (WIN), widely used to investigate the cannabinoid system, also shows unique broader effects at cellular and organismal levels compared to endogenous cannabinoids. Our study shows that WIN can disrupt the Golgi apparatus and the microtubule network in multiple cell types, independent of cannabinoid receptors. These results could explain how WIN reduces surface levels of proteins and contributes to the unique physiological effects observed with WIN.


Assuntos
Benzoxazinas , Canabinoides , Animais , Benzoxazinas/farmacologia , Canabinoides/farmacologia , Complexo de Golgi , Camundongos , Morfolinas/farmacologia , Receptor CB1 de Canabinoide , Receptor CB2 de Canabinoide , Receptores de Canabinoides
7.
Biomedicines ; 10(2)2022 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-35203607

RESUMO

There is substantial evidence that cholinergic system function impairment plays a significant role in many central nervous system (CNS) disorders. During the past three decades, muscarinic receptors (mAChRs) have been implicated in various pathologies and have been prominent targets of drug-design efforts. However, due to the high sequence homology of the orthosteric binding site, many drug candidates resulted in limited clinical success. Although several advances in treating peripheral pathologies have been achieved, targeting CNS pathologies remains challenging for researchers. Nevertheless, significant progress has been made in recent years to develop functionally selective orthosteric and allosteric ligands targeting the mAChRs with limited side effect profiles. This review highlights past efforts and focuses on recent advances in drug design targeting these receptors for Alzheimer's disease (AD), schizophrenia (SZ), and depression.

9.
Behav Pharmacol ; 32(6): 505-514, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34320518

RESUMO

Food restriction promotes drug self-administration; however, the effects of food restriction on the conditioned reinforcing properties of drug-associated stimuli are less clear. We tested the extent to which food restriction modified the conditioned reinforcing properties of a remifentanil-associated stimulus following conditioning with 3.2 or 1.0 µg/kg/infusion of remifentanil. First, we provided restricted (20 g/day standard chow) or ad libitum access to standard chow to rats. Second, within each feeding condition, we exposed rats to 20 intravenous infusions of remifentanil and 20 stimulus presentations that were delivered response independently each day for 5 days. For the experimental group (paired Pavlovian), the remifentanil infusions and stimulus presentations were delivered concurrently. The control group (random control) received the same number of infusions and stimulus presentations, but were not paired. For 28 sessions, we tested the extent to which the stimulus functioned as a conditioned reinforcer by allowing rats to freely respond for presentations of the remifentanil-associated stimulus. Following conditioning with 3.2 µg/kg/infusion of remifentanil, we found that rats that in the Paired Pavlovian group responded for the remifentanil-associated stimulus significantly more than rats in the Random control group, regardless of feeding condition. Following conditioning with 1.0 µg/kg/infusion of remifentanil, the remifentanil-associated stimulus was not associated with conditioned reinforcing properties, regardless of feeding condition. These findings confirm previous research demonstrating that a remifentanil-associated stimulus takes on conditioned reinforcing properties in a dose-dependent manner.


Assuntos
Privação de Alimentos/fisiologia , Remifentanil/farmacologia , Analgésicos Opioides/farmacologia , Ração Animal , Animais , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Infusões Intravenosas , Modelos Animais , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Resultado do Tratamento
10.
J Pharmacol Exp Ther ; 378(3): 287-299, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34183434

RESUMO

There are no Food and Drug Administration-approved medications for cocaine use disorder, including relapse. The µ-opioid receptor (MOPr) partial agonist buprenorphine alone or in combination with naltrexone has been shown to reduce cocaine-positive urine tests and cocaine seeking in rodents. However, there are concerns over the abuse liability of buprenorphine. Buprenorphine's partial agonist and antagonist activity at the nociception receptor (NOPr) and κ-opioid receptor (KOPr), respectively, may contribute to its ability to inhibit cocaine seeking. Thus, we hypothesized that a buprenorphine derivative that exhibits antagonist activity at MOPr and KOPr with enhanced agonist activity at the NOPr could provide a more effective treatment. Here we compare the pharmacology of buprenorphine and two analogs, BU10119 and BU12004, in assays for antinociception and for cocaine- and stress-primed reinstatement in the conditioned place preference paradigm. In vitro and in vivo assays showed that BU10119 acts as an antagonist at MOPr, KOPr, and δ-opioid receptor (DOPr) and a partial agonist at NOPr, whereas BU12004 showed MOPr partial agonist activity and DOPr, KOPr, and NOPr antagonism. BU10119 and buprenorphine but not BU12004 lessened cocaine-primed reinstatement. In contrast, BU10119, BU12004, and buprenorphine blocked stress-primed reinstatement. The selective NOPr agonist SCH221510 but not naloxone decreased cocaine-primed reinstatement. Together, these findings are consistent with the concept that NOPr agonism contributes to the ability of BU10119 and buprenorphine to attenuate reinstatement of cocaine-conditioned place preference in mice. The findings support the development of buprenorphine analogs lacking MOPr agonism with increased NOPr agonism for relapse prevention to cocaine addiction. SIGNIFICANCE STATEMENT: There are no Food and Drug Administration-approved medications for cocaine use disorder. Buprenorphine has shown promise as a treatment for cocaine relapse prevention; however, there are concerns over the abuse liability of buprenorphine. Here we show a buprenorphine analogue, BU10119, which lacks µ-opioid receptor agonism and inhibits cocaine-primed and stress-primed reinstatement in a conditioned place-preference paradigm. The results suggest the development of BU10119 for the management of relapse to cocaine seeking.


Assuntos
Cocaína , Buprenorfina , Naltrexona , Receptores Opioides mu
13.
J Pharmacol Exp Ther ; 377(3): 336-345, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33712507

RESUMO

The cholinergic nervous system has been implicated in mood disorders, evident in the fast-onset antidepressant effects of scopolamine, a potent muscarinic antagonist, in clinical studies. One prominent disadvantage of the use of scopolamine in the treatment of depression is its detrimental effects on cognition, especially as such effects might aggravate cognitive deficits that occur with depression itself. Thus, the identification of antimuscarinic drugs that are free of such detrimental effects may provide an important avenue for the development of novel therapeutics for the management of depression. The present data in rats indicate that a historical muscarinic antagonist, L-687,306, and a muscarinic antagonist of our own design, CJ2100, were as or more effective than scopolamine in antagonizing both the bradycardic effects of the muscarinic agonist arecoline in cardiovascular studies and its discriminative stimulus and rate-decreasing effects in behavioral studies. Additionally, both novel muscarinic antagonists were as effective as scopolamine in decreasing immobility in the forced swim test, a preclinical indicator of potential antidepressant activity. However, at equieffective or even larger doses, they were considerably less disruptive than scopolamine in assays of cognition-related behavior. All three drugs displayed high specificity for the mAChRs with few off-target binding sites, and CJ2100 showed modest affinity across the mAChRs when compared with L-687,306 and scopolamine. These data emphasize the dissimilar pharmacological profiles that are evident across antimuscarinic compounds and the potential utility of novel antagonists for the improved treatment of depression. SIGNIFICANCE STATEMENT: Some clinical studies with the muscarinic antagonist scopolamine document its ability to produce antidepressant effects in patients with mood disorders; however, scopolamine also has well known adverse effects on both autonomic and centrally mediated physiological functions that limit its therapeutic use. This study characterizes the cardiovascular and discriminative stimulus effects of two novel muscarinic antagonists, L-687,306 and CJ2100, that produce antidepressant-like effects in a rodent model (forced swim test) without affecting touchscreen-based cognitive performance (titrating psychomotor vigilance and delayed matching-to-position).


Assuntos
Antagonistas Muscarínicos , Cognição , Escopolamina
14.
Exp Clin Psychopharmacol ; 29(6): 567-572, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32940488

RESUMO

Protein kinase C (PKC) is important for the mechanism of action of amphetamine (AMPH). Inhibiting PKC blocks AMPH-stimulated increases in extracellular dopamine levels and AMPH-stimulated locomotor activity. This study examined the effects of PKC inhibition on the reinforcing properties of AMPH. Male Sprague-Dawley rats were trained to respond for infusions of 0.032 mg/kg/infusion AMPH or for sucrose pellets under a progressive-ratio (PR) schedule of reinforcement. Number of infusions earned, breakpoints, and session duration were recorded over consecutive sessions. Once AMPH-maintained responding stabilized, rats were treated with 0, 10, or 30 pmol of enzastaurin, a PKCß-selective inhibitor, or 6 mg/kg 6c, a brain-permeable PKC inhibitor, 18 hr prior to a self-administration session. Pretreatment with 30 pmol enzastaurin or 6 mg/kg 6c decreased the number of AMPH infusions earned and breakpoints without altering sucrose-maintained behaviors. These data suggest that PKC inhibition decreases motivation for AMPH and, therefore, is worth pursuing as a potential treatment for AMPH-use disorder. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Assuntos
Anfetamina , Estimulantes do Sistema Nervoso Central , Anfetamina/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley , Esquema de Reforço , Reforço Psicológico , Autoadministração
15.
J Pharmacol Exp Ther ; 374(2): 319-330, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32467352

RESUMO

δ-Opioid receptor (δ-receptor) agonists produce antihyperalgesia, antidepressant-like effects, and convulsions in animals. However, the role of agonist efficacy in generating different δ-receptor-mediated behaviors has not been thoroughly investigated. To this end, efficacy requirements for δ-receptor-mediated antihyperalgesia, antidepressant-like effects, and convulsions were evaluated by comparing the effects of the partial agonist BU48 and the full agonist SNC80 and changes in the potency of SNC80 after δ-receptor elimination. Antihyperalgesia was measured in a nitroglycerin-induced thermal hyperalgesia assay. An antidepressant-like effect was evaluated in the forced swim test. Mice were observed for convulsions after treatment with SNC80 or the δ-opioid receptor partial agonist BU48. Ligand-induced G protein activation was measured by [35S]guanosine 5'-O-[γ-thio]triphosphate binding in mouse forebrain tissue, and δ-receptor number was measured by [3H]D-Pen2,5-enkephalin saturation binding. BU48 produced antidepressant-like effects and convulsions but antagonized SNC80-induced antihyperalgesia and G protein activation. The potency of SNC80 was shifted to the right in δ-receptor heterozygous knockout mice and naltrindole-5'-isothiocyanate-treated mice, and the magnitude of potency shift differed across assays, with the largest shift occurring in the thermal hyperalgesia assay, followed by the forced swim test and then convulsion observation. Naltrindole antagonized these SNC80-induced behaviors with similar potencies, suggesting that these effects are mediated by the same type of δ-receptor. These data suggest that δ-receptor-mediated behaviors display a rank order of efficacy requirement, with antihyperalgesia having the highest requirement, followed by antidepressant-like effects and then convulsions. These findings further our understanding of the pharmacological mechanisms mediating the in vivo effects of δ-opioid receptor agonists. SIGNIFICANCE STATEMENT: δ-Opioid receptor (δ-receptor) agonists produce antihyperalgesia, antidepressant-like effects, and convulsions in animal models. This study evaluates pharmacological properties, specifically the role of agonist efficacy and receptor reserve, underlying these δ-receptor-mediated behaviors. These data suggest that δ-receptor-mediated behaviors display a rank order of efficacy requirement, with antihyperalgesia having the highest requirement, followed by antidepressant-like effects and then convulsions.


Assuntos
Comportamento Animal/efeitos dos fármacos , Receptores Opioides delta/agonistas , Animais , Benzamidas/farmacologia , Buprenorfina/análogos & derivados , Buprenorfina/farmacologia , Agonismo Parcial de Drogas , Camundongos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Piperazinas/farmacologia
16.
Behav Pharmacol ; 31(2&3): 207-215, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32058349

RESUMO

Previous research demonstrated that a remifentanil-associated stimulus facilitated the acquisition of a previously unlearned response; however, it is unclear how long a remifentanil-associated stimulus maintains conditioned reinforcing properties under conditions of daily testing. To address this gap, we exposed adult male rats to response-independent stimulus presentations and deliveries of remifentanil (1.0, 3.2, or 10.0 µg/kg/infusion). Rats either received the stimulus presentations and remifentanil deliveries together (Paired Pavlovian conditioning) or according to separate clocks (Random control group). In the sessions following Pavlovian conditioning, we allowed rats to emit nose-poke responses for the presentation of the stimulus alone and measured the extent to which the stimulus facilitated and maintained a previously unlearned response. We tested responding for the stimulus presentations across 28 daily sessions to assess the Pavlovian extinction (degradation of the drug-stimulus association) of the conditioned reinforcing properties of the remifentanil-associated stimulus. We observed the highest and most persistent levels of responding in rats with a Paired Pavlovian conditioning history at 3.2 and 10.0 µg/kg/infusion. In addition, we included analyses of the variability in responding for each group, which revealed individual differences in the susceptibility of the remifentanil-associated stimulus acting as a conditioned reinforcer. These findings demonstrate that a remifentanil-associated stimulus has the ability to sustain drug-seeking behavior and underscores the importance of Pavlovian conditioning in promoting drug abuse.


Assuntos
Condicionamento Operante/efeitos dos fármacos , Remifentanil/farmacologia , Animais , Condicionamento Clássico/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Extinção Psicológica/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Remifentanil/metabolismo
17.
Behav Pharmacol ; 31(4): 359-367, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31922966

RESUMO

This study aimed to use central and peripheral assays to compare the effects of the muscarinic antagonist scopolamine with those of a novel muscarinic antagonist, L-687,306 [(3R,4R)-3-(3-cyclopropyl-1,2,4,oxadiazol[5-yl]-1-azabicyclo[2.2.1]heptane. Groups of rats were trained to discriminate the stimulus effects of the muscarinic agonist, arecoline (1.0 mg/kg); concomitant measures of response rate were recorded. Separate groups were prepared with telemetery devices for recording bradycardia induced by arecoline (10 mg/kg). Methyl arecoline and arecoline were nearly equally potent in producing a brief but profound bradycardia, indicative of an equivalent effect in the heart. L-687,306 and scopolamine were both able to block this peripheral effect of arecoline. L-687,306 produced a surmountable antagonism of both the discriminative and rate-suppressing effects of arecoline. Scopolamine, however, was unable to antagonize the rate-reducing effects of arecoline in the discrimination assay. This limited the number of rats that could respond to the discriminative stimulus effects of arecoline, as well as the amount of arecoline stimulus effects they were able to report. The data suggest that L-687,306 may be a more generally effective muscarinic antagonist than scopolamine and support earlier reports that this antagonist has less direct effect on behavior.


Assuntos
Bradicardia/fisiopatologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Aprendizagem por Discriminação/fisiologia , Oxidiazóis/farmacologia , Escopolamina/farmacologia , Animais , Arecolina/efeitos adversos , Arecolina/antagonistas & inibidores , Arecolina/farmacologia , Bradicardia/induzido quimicamente , Aprendizagem por Discriminação/efeitos dos fármacos , Masculino , Antagonistas Muscarínicos/farmacologia , Ratos
18.
J Med Chem ; 63(4): 1671-1683, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-31986033

RESUMO

We previously reported a novel SAR campaign that converted a metabolically unstable series of µ-opioid receptor (MOR) agonist/δ-opioid receptor (DOR) antagonist bicyclic core peptidomimetics with promising analgesic activity and reduced abuse liabilities into a more stable series of benzylic core analogues. Herein, we expanded the SAR of that campaign and determined that the incorporation of amines into the benzylic pendant produces enhanced MOR-efficacy in this series, whereas the reincorporation of an aromatic ring into the pendant enhanced MOR-potency. Two compounds, which contain a piperidine (14) or an isoindoline (17) pendant, retained the desired opioid profile in vitro, possessed metabolic half-lives of greater than 1 h in mouse liver microsomes (MLMs), and were active antinociceptive agents in the acetic acid stretch assay (AASA) at subcutaneous doses of 1 mg/kg.


Assuntos
Analgésicos Opioides/farmacologia , Peptidomiméticos/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/síntese química , Analgésicos Opioides/metabolismo , Animais , Células CHO , Cricetulus , Masculino , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Peptidomiméticos/síntese química , Peptidomiméticos/metabolismo , Relação Estrutura-Atividade
19.
Psychopharmacology (Berl) ; 236(11): 3243, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31270556

RESUMO

The middle initial of the author should be "A" instead of "C". The correct presentation of the author name is Colleen A. Carpenter.

20.
Psychopharmacology (Berl) ; 236(11): 3231-3242, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31134292

RESUMO

RATIONALE: Pathological amphetamine (AMPH) use is a serious public health concern with no pharmacological treatment options. Protein kinase Cß (PKCß) has been implicated in the mechanism of action of AMPH, such that inhibition of PKCß attenuates AMPH-stimulated dopamine efflux in vivo. With this in mind, inhibition of PKCß may be a viable therapeutic target for AMPH use disorder. OBJECTIVE: The purpose of this study is to demonstrate that selective pharmacological inhibition of PKCß alters AMPH-stimulated behaviors in rats. METHODS: Rats were administered intracerebroventricular (i.c.v.) injections of the PKCß-selective inhibitor enzastaurin 0.5, 3, 6, or 18 h before evaluating AMPH-stimulated locomotion (0.32-3.2 mg/kg). Rats were trained to make responses for different doses of AMPH infusions or sucrose under a fixed ratio 5 schedule of reinforcement, and the effects of enzastaurin pretreatment 3 or 18 h prior to a self-administration session were determined. Also, the effect of enzastaurin on AMPH-stimulated PKC activity in the ventral striatum was evaluated. RESULTS: A large dose of enzastaurin (1 nmol) decreased AMPH-stimulated locomotor activity 0.5 h following enzastaurin administration. Small doses of enzastaurin (10-30 pmol) attenuated AMPH-stimulated locomotor activity and shifted the AMPH dose-effect curve to the right following an 18-h pretreatment. Rats pretreated with enzastaurin 18 h, but not 3, prior to a self-administration session showed a decrease in the number of responses for AMPH, shifted the ascending limb of the amphetamine dose effect curve, and produced no change in responses for sucrose. AMPH-stimulated PKC activity was decreased following a 0.5- or 18-h pretreatment, but not a 3-h pretreatment of enzastaurin. CONCLUSIONS: These results demonstrate that inhibition of PKCß will decrease AMPH-stimulated behaviors and neurobiological changes and suggest that PKCß is potentially a viable target for AMPH use disorder.


Assuntos
Anfetamina/administração & dosagem , Comportamento Aditivo/prevenção & controle , Estimulantes do Sistema Nervoso Central/administração & dosagem , Indóis/farmacologia , Locomoção/efeitos dos fármacos , Proteína Quinase C beta/antagonistas & inibidores , Animais , Comportamento Aditivo/enzimologia , Comportamento Aditivo/psicologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Relação Dose-Resposta a Droga , Indóis/uso terapêutico , Locomoção/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Autoadministração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...