Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 810
Filtrar
1.
J Clin Invest ; 133(18)2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37561576

RESUMO

The nucleus accumbens (NAc) is the most promising target for drug use disorder treatment. Deep brain stimulation (DBS) of NAc is effective for drug use disorder treatment. However, the mechanisms by which DBS produces its therapeutic effects remain enigmatic. Here, we define a behavioral cutoff criterion to distinguish depressive-like behaviors and non-depressive-like behaviors in mice after morphine withdrawal. We identified a basolateral amygdala (BLA) to NAc D1 medium spiny neuron (MSN) pathway that controls depressive-like behaviors after morphine withdrawal. Furthermore, the paraventricular nucleus of thalamus (PVT) to NAc D2 MSN pathway controls naloxone-induced acute withdrawal symptoms. Optogenetically induced long-term potentiation with κ-opioid receptor (KOR) antagonism enhanced BLA to NAc D1 MSN signaling and also altered the excitation/inhibition balance of NAc D2 MSN signaling. We also verified that a new 50 Hz DBS protocol reversed morphine withdrawal-evoked abnormal plasticity in NAc. Importantly, this refined DBS treatment effectively alleviated naloxone-induced withdrawal symptoms and depressive-like behaviors and prevented stress-induced reinstatement. Taken together, the results demonstrated that input- and cell type-specific synaptic plasticity underlies morphine withdrawal, which may lead to novel targets for the treatment of opioid use disorder.


Assuntos
Analgésicos Opioides , Síndrome de Abstinência a Substâncias , Camundongos , Animais , Analgésicos Opioides/farmacologia , Núcleo Accumbens/metabolismo , Receptores de Dopamina D2 , Morfina/efeitos adversos , Naloxona/farmacologia , Naloxona/metabolismo , Síndrome de Abstinência a Substâncias/terapia , Receptores de Dopamina D1/metabolismo , Camundongos Endogâmicos C57BL
2.
Int J Pharm ; 642: 123159, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37336303

RESUMO

NAL's hydrophilicity and the inherent lipophilic properties of the stratum corneum hinders its capacity for immediate delivery through skin in opioid rescue cases. In this study, we had sought to investigate the feasibility of using minimally invasive physical ablative techniques including sonophoresis, laser, dermaplaning, microneedles, and microdermabrasion for systemically delivering NAL via the skin. These techniques reduced lag time to NAL delivery to about 3-12 min from 71.22 ± 9.62 min seen for passive delivery. Also, they all significantly enhanced the amount of NAL delivered in 1 h and over 24 h period of evaluation as compared to the passive group (p < 0.05). Sonophoresis and laser showed the greatest delivery in 1 h, followed by dermaplaning. The cumulative amount of drug delivered by these approaches in 1 h were 1277.95 ± 387.06, 83.33 ± 11.11, 30.66 ± 5.67 µg/cm2, respectively. Though the most remarkable, inconsistencies in in vitro permeation profile of NAL were observed with the 1 MHz ultrasound frequency used. With proper optimization of the conditions of use and design, the different approaches explored in this study can be potentially applied for the systemic delivery of naloxone in opioid overdose emergencies and opioid disaccustoming purposes.


Assuntos
Técnicas de Ablação , Absorção Cutânea , Naloxona/metabolismo , Analgésicos Opioides/metabolismo , Administração Cutânea , Pele/metabolismo , Sistemas de Liberação de Medicamentos/métodos
3.
Artigo em Inglês | MEDLINE | ID: mdl-37127059

RESUMO

While pain results from the activation of nociceptors following noxious stimuli, mounting evidence links pain- and stress-related responses in mammals. In zebrafish, the activation of hypothalamic-pituitary-interrenal (HPI) axis may also regulate body pigmentation (the camouflage response). Here, we aimed to investigate a putative relationship between pain-, stress-, and camouflage-related parameters in adult zebrafish. To answer this question, we assessed whether intraperitoneal acetic acid injection can activate the HPI axis, measuring whole-body cortisol and the camouflage response as physiological endpoints in the presence or absence of morphine or naloxone, an opioid antagonist. Acetic acid induced a stereotypic circling behavior in the top of the tank, accompanied by abdominal writhing-like response, a specific phenotype that reflects local nociceptive effect. Both whole-body cortisol levels and camouflage response increased in the acetic acid group, while morphine prevented these responses, and naloxone antagonized morphine-induced effects. Moreover, we observed positive correlations between representative behavioral, physiological and skin coloration endpoints, and a "pain index" was proposed to summarize phenotypic profile of zebrafish under different pharmacological manipulations. Collectively, these findings suggest a coordinated activation of pain, camouflage- and stress-related pathways following acetic acid injection in zebrafish. Our data also support that camouflage response represents a novel and relevant biomarker for future probing pain and stress neurobiology, with a robust sensitivity to opioidergic drugs.


Assuntos
Ácido Acético , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Ácido Acético/toxicidade , Ácido Acético/metabolismo , Hidrocortisona/metabolismo , Naloxona/farmacologia , Naloxona/metabolismo , Morfina/toxicidade , Morfina/metabolismo , Dor , Fenótipo , Mamíferos/metabolismo
4.
J Chem Inf Model ; 63(7): 2196-2206, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36977188

RESUMO

The nation's opioid overdose deaths reached an all-time high in 2021. The majority of deaths are due to synthetic opioids represented by fentanyl. Naloxone, which is a FDA-approved reversal agent, antagonizes opioids through competitive binding at the µ-opioid receptor (mOR). Thus, knowledge of the opioid's residence time is important for assessing the effectiveness of naloxone. Here, we estimated the residence times (τ) of 15 fentanyl and 4 morphine analogs using metadynamics and compared them with the most recent measurement of the opioid kinetic, dissociation, and naloxone inhibitory constants (Mann et al. Clin. Pharmacol. Therapeut. 2022, 120, 1020-1232). Importantly, the microscopic simulations offered a glimpse at the common binding mechanism and molecular determinants of dissociation kinetics for fentanyl analogs. The insights inspired us to develop a machine learning approach to analyze the kinetic impact of fentanyl's substituents based on the interactions with mOR residues. This proof-of-concept approach is general; for example, it may be used to tune ligand residence times in computer-aided drug discovery.


Assuntos
Analgésicos Opioides , Naloxona , Analgésicos Opioides/farmacologia , Naloxona/farmacologia , Naloxona/metabolismo , Fentanila/metabolismo , Fentanila/farmacologia , Morfina/química , Receptores Opioides mu/metabolismo , Antagonistas de Entorpecentes
5.
Comb Chem High Throughput Screen ; 26(10): 1900-1906, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36372918

RESUMO

AIM: This study aimed to investigate how opioids affect phagocytosis and microglial nitrite and nitric oxide synthase (iNOS) production during inflammation. BACKGROUND: Opioids are a group of chemicals that are naturally found in the opium poppy plant and exert a variety of effects on the brain, including pain alleviation in some cases. They are commonly used in surgery and perioperative analgesia. However, research on the impact of opioids on microglial inflammatory factor production and phagocytosis is limited. OBJECTIVES: This study was designed to investigate the effects of opioids on inducible nitric oxide synthase (iNOS) activity and nitric oxide (NO) generation. Moreover, the influence of opioids on the engulfment of C8-B4 microglial cells after stimulation with LPS was also examined. METHODS: C8-B4 mouse microglial cells were exposed to various concentrations of opioids after stimulation with lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Nitrite production was assayed. The iNOS and Cox-2 were determined by Western blotting, and fluorescent immunostaining was performed to assess the percentage of microglia that engulfed fluorescent microspheres in total microglia cultivating with opioids after being activated by LPS. RESULTS: After LPS and IFN-γ stimulation, microglia produced lower amounts of nitric oxide (NO) production with buprenorphine, salvinorin A, and naloxone (P<0.05). When combined with naloxone, no significant differences were found than buprenorphine. It was observed that buprenorphine and salvinorin A could suppress iNOS expression activated by LPS and IFN-γ. Phagocytosis was greatly increased after LPS stimulation, and a significant increase was observed after adding salvinorin A. CONCLUSION: Buprenorphine and salvinorin A were found to reduce NO production and iNOS induction in microglial cells activated by LPS and IFN-γ. Salvinorin A promoted the phagocytosis of microglia cells treated by LPS.


Assuntos
Buprenorfina , Microglia , Camundongos , Animais , Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/farmacologia , Nitritos/metabolismo , Nitritos/farmacologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Lipopolissacarídeos/farmacologia , Óxido Nítrico , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interferon gama/metabolismo , Interferon gama/farmacologia , Naloxona/metabolismo , Naloxona/farmacologia , Fagocitose , Buprenorfina/metabolismo , Buprenorfina/farmacologia
6.
Sci Rep ; 12(1): 20234, 2022 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-36424418

RESUMO

The mu opioid receptor (MOR) and the orphan GPR151 receptor are inhibitory G protein coupled receptors that are enriched in the habenula, a small brain region involved in aversion processing, addiction and mood disorders. While MOR expression in the brain is widespread, GPR151 expression is restricted to the habenula. In a previous report, we created conditional ChrnB4-Cre × Oprm1fl/fl (so-called B4MOR) mice, where MORs are deleted specifically in Chrnb4-positive neurons restricted to the habenula, and shown a role for these receptors in naloxone aversion. Here we characterized the implication of habenular MORs in social behaviors. B4MOR-/- mice and B4MOR+/+ mice were compared in several social behavior measures, including the chronic social stress defeat (CSDS) paradigm, the social preference (SP) test and social conditioned place preference (sCPP). In the CSDS, B4MOR-/- mice showed lower preference for the social target (unfamiliar mouse of a different strain) at baseline, providing a first indication of deficient social interactions in mice lacking habenular MORs. In the SP test, B4MOR-/- mice further showed reduced sociability for an unfamiliar conspecific mouse. In the sCPP, B4MOR-/- mice also showed impaired place preference for their previous familiar littermates after social isolation. We next created and tested Gpr151-/- mice in the SP test, and also found reduced social preference compared to Gpr151+/+ mice. Altogether our results support the underexplored notion that the habenula regulates social behaviors. Also, our data suggest that the inhibitory habenular MOR and GPR151 receptors normally promote social reward, possibly by dampening the aversive habenula activity.


Assuntos
Habenula , Receptores Acoplados a Proteínas G , Receptores Opioides mu , Animais , Camundongos , Habenula/metabolismo , Naloxona/metabolismo , Neurônios/metabolismo , Receptores Opioides mu/metabolismo , Recompensa , Receptores Acoplados a Proteínas G/metabolismo
7.
Glia ; 70(7): 1289-1300, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35275429

RESUMO

Mitochondria are abundant in the fine processes of astrocytes, however, potential roles for astrocyte mitochondria remain poorly understood. In the present study, we performed a systematic examination of the effects of abnormal oxidative phosphorylation in astrocytes on several mouse behaviors. Impaired astrocyte oxidative phosphorylation was produced by astrocyte-specific deletion of the nuclear mitochondrial gene, Cox10, that encodes an accessory protein of complex IV, the protoheme:heme-O-farnesyl transferase. As expected, conditional deletion of the Cox10 gene in mice (cKO mice) significantly reduced expression of COX10 and Cytochrome c oxidase subunit I (MTCO1) of Complex IV, resulting in decreased oxidative phosphorylation without significantly affecting glycolysis. No effects of the deletion were observed on locomotor activity, anxiety-like behavior, nociception, or spontaneous alternation. Cox10 cKO female mice exhibited mildly impaired novel object recognition, while Cox10 cKO male mice were moderately deficient in trace fear conditioning. No group-related changes were observed in conditional place preference (CPP) that assessed effects of morphine on reward. In contrast to CPP, Cox10 cKO mice demonstrated significantly increased aversive behaviors produced by naloxone-precipitated withdrawal following chronic exposure to morphine, that is, jumping and avoidance behavior as assessed by conditional place aversion (CPA). Our study suggests that astrocyte oxidative phosphorylation may contribute to behaviors associated with greater cognitive load and/or aversive and stressful conditions.


Assuntos
Alquil e Aril Transferases , Dependência de Morfina , Síndrome de Abstinência a Substâncias , Alquil e Aril Transferases/metabolismo , Animais , Astrócitos/metabolismo , Medo , Feminino , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/metabolismo , Morfina/metabolismo , Morfina/farmacologia , Dependência de Morfina/metabolismo , Dependência de Morfina/psicologia , Naloxona/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/metabolismo , Antagonistas de Entorpecentes/farmacologia , Respiração , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/psicologia
8.
Int J Pharm ; 604: 120739, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-34048932

RESUMO

Naloxone (NAL) is administered parenterally or intranasally for treating opioid overdose. The short duration of action of NAL calls for frequent re-dosing which may be eliminated by the development of a transdermal system. This study aimed to assess the effect of microneedles on improving the skin permeation of NAL hydrochloride. In vitro permeation of NAL across intact and microneedle-treated (Dr. Pen™ Ultima A6) porcine skin was evaluated. The effect of microneedle length and application duration, and donor concentration on NAL permeation were investigated. In-vitro in-vivo correlation of the permeation results was done to predict the plasma concentration kinetics of NAL in patients. In vitro passive permeation of NAL after 6 h was observed to be 8.25±1.06 µg/cm2. A 56- and 37-fold enhancement was observed with 500 and 250 µm needles applied for 1 min, respectively. Application of 500 µm MNs for 2 min significantly reduced the lag time to ~ 8 min and increasing the donor concentration for the same treatment group doubled the permeation (p < 0.05). Modeling simulations demonstrated the attainment of pharmacokinetic profile of NAL comparable to those obtained with the FDA-approved intramuscular and intranasal devices. Microneedle-mediated transdermal delivery holds potential for rapid and sustained NAL delivery for opioid overdose treatment.


Assuntos
Agulhas , Overdose de Opiáceos , Administração Cutânea , Animais , Sistemas de Liberação de Medicamentos , Humanos , Naloxona/metabolismo , Pele/metabolismo , Absorção Cutânea , Suínos
9.
Biochim Biophys Acta Gen Subj ; 1865(3): 129838, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33373630

RESUMO

BACKGROUND: G protein-coupled receptors (GPCRs) comprise a family of membrane proteins that can be activated by a variety of external factors. The µ-opioid receptor (MOR), a class A GPCR, is the main target of morphine. Recently, enhanced sampling molecular dynamics simulations of a constitutively active mutant of MOR in its apo form allowed us to capture the novel intermediate states of activation, as well as the active state. This prompted us to apply the same techniques to wild type MOR in complex with ligands, in order to explore their contributions to the receptor conformational changes in the activation process. METHODS: MOR was modeled in complex with agonists (morphine, BU72), a partial agonist (naloxone benzoylhydrazone) and an antagonist (naloxone). Replica exchange with solute tempering (REST2) molecular dynamics simulations were carried out for all systems. Trajectory frames were clustered, and the activation state of each cluster was assessed by two different methods. RESULTS: Cluster sizes and activation indices show that while agonists stabilized structures in a higher activation state, the antagonist behaved oppositely. Morphine tends to drive the receptor towards increasing R165-T279 distances, while naloxone tends to increase the NPxxYA motif conformational change. CONCLUSIONS: Despite not observing a full transition between inactive and active states, an important conformational change of transmembrane helix 5 was observed and associated with a ligand-driven step of the process. GENERAL SIGNIFICANCE: The activation process of GPCRs is widely studied but still not fully understood. Here we carried out a step forward in the direction of gaining more details of this process.


Assuntos
Aminoácidos/química , Apoproteínas/química , Morfina/química , Fosfatidilcolinas/química , Receptores Opioides mu/química , Aminoácidos/metabolismo , Apoproteínas/metabolismo , Sítios de Ligação , Humanos , Ligantes , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Morfinanos/química , Morfinanos/metabolismo , Morfina/metabolismo , Naloxona/análogos & derivados , Naloxona/química , Naloxona/metabolismo , Fosfatidilcolinas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Pirróis/química , Pirróis/metabolismo , Receptores Opioides mu/metabolismo , Soluções , Água/química , Água/metabolismo
10.
Sci Rep ; 10(1): 833, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964994

RESUMO

While it is known that opioid receptors (ORs) are densely expressed in both the brain and periphery, it is widely accepted that hypoxic effects of opioids result solely from their direct action in the CNS. To examine the role of peripheral ORs in triggering brain hypoxia, we used oxygen sensors in freely moving rats to examine how naloxone-HCl and naloxone-methiodide, the latter which is commonly believed to be peripherally restricted, affect brain oxygen responses induced by intravenous heroin at low, human-relevant doses. Similar to naloxone-HCl, naloxone-methiodide at a relatively low dose (2 mg/kg) fully blocked heroin-induced decreases in brain oxygen levels. As measured by mass spectrometry, naloxone-methiodide was found to be ~40-fold less permeable than naloxone-HCl across the blood-brain barrier, thus acting as a selective blocker of peripheral ORs. Despite this selectivity, a low but detectable amount of naloxone was found in brain tissue after naloxone-methiodide administration, potentially influencing our results. Therefore, we examined the effects of naloxone-methiodide at a very low dose (0.2 mg/kg; at which naloxone was undetectable in brain tissue) and found that this drug still powerfully attenuates heroin-induced brain oxygen responses. These data demonstrate the role of peripheral ORs in triggering heroin-induced respiratory depression and subsequent brain hypoxia.


Assuntos
Heroína/efeitos adversos , Hipóxia Encefálica/etiologia , Receptores Opioides/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Hipóxia Encefálica/tratamento farmacológico , Naloxona/administração & dosagem , Naloxona/análogos & derivados , Naloxona/metabolismo , Naloxona/farmacologia , Oxigênio/metabolismo , Compostos de Amônio Quaternário/administração & dosagem , Compostos de Amônio Quaternário/metabolismo , Compostos de Amônio Quaternário/farmacologia , Ratos , Receptores Opioides/metabolismo
11.
J Pharmacol Exp Ther ; 371(2): 453-475, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31492824

RESUMO

In December 2018, the Centers for Disease Control declared fentanyl the deadliest drug in America. Opioid overdose is the single greatest cause of death in the United States adult population (ages 18-50), and fentanyl and its analogs [fentanyl/fentanyl analogs (F/FAs)] are currently involved in >50% of these deaths. Anesthesiologists in the United States were introduced to fentanyl in the early 1970s when it revolutionized surgical anesthesia by combining profound analgesia with hemodynamic stability. However, they quickly had to master its unique side effect. F/FAs can produce profound rigidity in the diaphragm, chest wall and upper airway within an extremely narrow dosing range. This clinical effect was called wooden chest syndrome (WCS) by anesthesiologists and is not commonly known outside of anesthesiology or to clinicians or researchers in addiction research/medicine. WCS is almost routinely fatal without expert airway management. This review provides relevant clinical human pharmacology and animal data demonstrating that the significant increase in the number of F/FA-induced deaths may involve α-adrenergic and cholinergic receptor-mediated mechanical failure of the respiratory and cardiovascular systems with rapid development of rigidity and airway closure. Although morphine and its prodrug, heroin, can cause mild rigidity in abdominal muscles at high doses, neither presents with the distinct and rapid respiratory failure seen with F/FA-induced WCS, separating F/FA overdose from the slower onset of respiratory depression caused by morphine-derived alkaloids. This distinction has significant consequences for the design and implementation of new pharmacologic strategies to effectively prevent F/FA-induced death. SIGNIFICANCE STATEMENT: Deaths from fentanyl and F/FAs are increasing in spite of availability and awareness of the opioid reversal drug naloxone. This article reviews literature suggesting that naloxone may be ineffective against centrally mediated noradrenergic and cholinergic effects of F/FAs, which clinically manifest as severe muscle rigidity and airway compromise (e.g., wooden chest syndrome) that is rapid and distinct from respiratory depression seen with morphine-derived alkaloids. A physiologic model is proposed and implications for new drug development and treatment are discussed.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Analgésicos Opioides/efeitos adversos , Fentanila/efeitos adversos , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Epidemia de Opioides/prevenção & controle , Neurônios Adrenérgicos/metabolismo , Analgésicos Opioides/metabolismo , Overdose de Drogas/metabolismo , Overdose de Drogas/prevenção & controle , Fentanila/metabolismo , Humanos , Rigidez Muscular/induzido quimicamente , Rigidez Muscular/tratamento farmacológico , Rigidez Muscular/metabolismo , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Epidemia de Opioides/tendências , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/metabolismo , Tempo para o Tratamento/tendências
12.
Eur J Pharm Sci ; 138: 105017, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31356868

RESUMO

Development of opioid analgesics with minimal side effects requires substantial knowledge on structure-kinetic and -thermodynamic relationship of opioid-receptor interactions. Here, combined kinetics and thermodynamics of opioid agonist binding to human µ-opioid receptor (h-µOR) was investigated using real-time label-free surface plasmon resonance (SPR)-based method. The N-terminal end truncated and C-terminal 6His-tagged h-µOR was constructed and expressed in E. coli. Receptor was purified, detergent-solubilized and characterized by circular dichroism. The uniform immobilization of h-µOR on Ni-NTA chips was achieved using hybrid capture-coupling approach followed by reconstitution in lipid bilayer. Thermodynamic equilibrium affinities of opioids were in narrow nanomolar range and in near quantitative agreement with their Ki values. However, they did not correlate with their in vitro EC50 values, indicating that they might not have thermodynamic selectivity. Contrary, on and off rates exhibited much larger dispersion and well correlated with EC50 values, indicating that opioids might exhibit kinetic-selectivity towards their target. Temperature-dependent SPR assays provided access to rate and equilibrium thermodynamic data, which demonstrated binding of morphine and naloxone to µOR was exothermic and essentially enthalpy driven. This work suggests that kinetic-based structure-activity of opioids in drug design and incorporation into the pharmacokinetics-pharmacodynamics predictions may have more value than thermodynamic equilibrium constants alone.


Assuntos
Analgésicos Opioides/metabolismo , Receptores Opioides mu/metabolismo , Técnicas Biossensoriais/métodos , Escherichia coli/metabolismo , Humanos , Cinética , Morfina/metabolismo , Naloxona/metabolismo , Termodinâmica
13.
Chem Biol Drug Des ; 91(4): 902-914, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29222847

RESUMO

Novel substituted amino acid tethered norsufentanil derivatives were synthesized by the four-component Ugi reaction. Norsufentanil was reacted with succinic anhydride to produce the corresponding carboxylic acid. The resulting carboxylic acid has undergone a multicomponent reaction with different aldehydes, amines, and isocyanides to produce a library of the desired compounds. In all cases, amide bond rotation was observed in the NMR spectra. In vivo analgesic activity of the synthesized compounds was evaluated by a tail flick test. Very encouraging results were obtained for a number of the synthesized products. Some of the synthesized compounds such as 5a, 5b, 5h, 5j, and 5r were found to be more potent than sufentanil, sufentanil citrate, and norsufentanil. Binding modes between the compounds and mu and delta-opioid receptors were studied by molecular docking method. The relationship between the molecular structural features and the analgesic activity was investigated by a quantitative structure-activity relationship model. The results of the molecular modeling studies and the in vivo analgesic activity suggested that the majority of the synthesized compounds were more potent than sufentanil and norsufentanil.


Assuntos
Analgésicos/síntese química , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade , Sufentanil/análogos & derivados , Dor Aguda/tratamento farmacológico , Analgésicos/metabolismo , Analgésicos/uso terapêutico , Animais , Sítios de Ligação , Masculino , Camundongos , Naloxona/química , Naloxona/metabolismo , Estrutura Terciária de Proteína , Receptores Opioides delta/química , Receptores Opioides delta/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Sufentanil/química , Sufentanil/metabolismo , Sufentanil/uso terapêutico
14.
Bull Exp Biol Med ; 163(6): 737-741, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29063329

RESUMO

We compared samples of microencapsulated naloxone prepared by using spray drying technique. 2-Hydroxypropyl-ß-cyclodextrin, sodium alginate, polycaprolactone, and carboxymethyl cellulose were used as the carriers. It was found that the combination of naloxone with sodium alginate was characterized by the highest naloxone content in the matrix and the lowest release rate (100% release time was 60 min). Using the model of respiratory disturbances caused by 10 ED50 fentanyl (anesthetic effect), we studied the effects of naloxone-sodium alginate complex on the dynamics of CO2 concentration in the expired air. It was shown that treatment with the developed microencapsulated naloxone after fentanyl injection allowed reducing the therapeutic dose of the antagonist by more than 2 times and eliminated the necessity of repeated injections.


Assuntos
Portadores de Fármacos , Fentanila/envenenamento , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/envenenamento , 2-Hidroxipropil-beta-Ciclodextrina/química , Alginatos/química , Animais , Animais não Endogâmicos , Carboximetilcelulose Sódica/química , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Fentanila/antagonistas & inibidores , Fentanila/toxicidade , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Cinética , Masculino , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Entorpecentes/toxicidade , Poliésteres/química , Ratos , Respiração/efeitos dos fármacos
15.
Proc Natl Acad Sci U S A ; 114(20): 5247-5252, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461466

RESUMO

To provide new preclinical evidence toward improving the efficacy of oxytocin (OT) in treating social dysfunction, we tested the benefit of administering OT under simultaneously induced opioid antagonism during dyadic gaze interactions in monkeys. OT coadministered with a µ-opioid receptor antagonist, naloxone, invoked a supralinear enhancement of prolonged and selective social attention, producing a stronger effect than the summed effects of each administered separately. These effects were consistently observed when averaging over entire sessions, as well as specifically following events of particular social importance, including mutual eye contact and mutual reward receipt. Furthermore, attention to various facial regions was differentially modulated depending on social context. Using the Allen Institute's transcriptional atlas, we further established the colocalization of µ-opioid and κ-opioid receptor genes and OT genes at the OT-releasing sites in the human brain. These data across monkeys and humans support a regulatory relationship between the OT and opioid systems and suggest that administering OT under opioid antagonism may boost the therapeutic efficacy of OT for enhancing social cognition.


Assuntos
Fixação Ocular/efeitos dos fármacos , Ocitocina/metabolismo , Ocitocina/farmacologia , Analgésicos Opioides/antagonistas & inibidores , Animais , Atenção/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Feminino , Macaca mulatta/fisiologia , Masculino , Naloxona/metabolismo , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides kappa , Receptores Opioides mu/efeitos dos fármacos , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Comportamento Social
16.
Behav Pharmacol ; 28(5): 401-404, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28418942

RESUMO

Studies in animal models suggest that sugar deprivation following excessive intake elicits some opioid-like withdrawal signs. In the present study, opioid-like effects of excessive sucrose intake were further characterized in C57BL/6 mice by comparing the effects of the opioid antagonist naloxone on food-reinforced responding before and during sucrose availability and, in parallel experiments, following chronic morphine administration. Results show that naloxone produced time-dependent and dose-dependent decreases in operant response rates after 4 weeks of excessive sucrose consumption, and that these effects were comparable with those produced by chronic morphine injections. These findings extend the observation that excessive sucrose consumption may produce opioid-like withdrawal signs, and suggest that operant assays of withdrawal-suppressed behaviors may be useful for further study of excessive sucrose consumption.


Assuntos
Naloxona/farmacologia , Tempo de Reação/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Dependência de Morfina , Naloxona/metabolismo , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/antagonistas & inibidores , Esquema de Reforço , Síndrome de Abstinência a Substâncias , Sacarose/administração & dosagem , Sacarose/metabolismo , Sacarose/farmacologia
17.
Biochim Biophys Acta Proteins Proteom ; 1865(3): 336-343, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28012872

RESUMO

Naloxone is an alkaloid antagonist that acts as an antidote to opioids through the mu-opioid receptor (MOR), a G protein-coupled receptor. However, its binding site on the MOR remains unknown. To investigate the binding interfaces necessary for naloxone and MOR, available structural information was combined with a cell-based photocrosslinking approach. Computer prediction revealed that four binding sites on MOR were required for naloxone binding. In addition, in the photocrosslinking approach, an amber stop codon was used to replace the sense codon of the MOR at 266 selected individual positions, in order to introduce the photoreactive amino acid p-benzoyl-l-phenylalanine (BzF) into MOR to evaluate the results of the computer analysis. The BzF-incorporated MOR mutant genes were expressed in CHO cells, in which MOR retained the ability to interact with its ligands, such as morphine, and exhibited MOR-dependent activation of ERK signaling following morphine stimulation. Notably, after treatment with tritium-labeled naloxone and exposure to UV light, we observed naloxone crosslinking with BzF replacement at hydrophobic residues and some polar/uncharged residues in the computer-predicted sites 1 and 3, indicating that these two sites in the MOR interact with naloxone. In conclusion, these results indicate that MOR has two naloxone binding sites and that the hydrophobic and polar/uncharged residues within these sites are important for naloxone binding.


Assuntos
Reagentes de Ligações Cruzadas/metabolismo , Naloxona/metabolismo , Receptores Opioides mu/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Cricetulus , Ligantes , Sistema de Sinalização das MAP Quinases/fisiologia , Transdução de Sinais/fisiologia
18.
Biomed Chromatogr ; 31(2)2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27390058

RESUMO

A rapid and sensitive LC-MS/MS method was developed and validated for the simultaneous determination of buprenorphine and its three metabolites (buprenorphine glucuronide, norbuprenorphine and norbuprenorphine glucuronide) as well as naloxone and its metabolite naloxone glucuronide in the rat plasma. A hydrophilic interaction chromatography column and a mobile phase containing acetonitrile and ammonium formate buffer (pH 3.5) were used for the chromatographic separation. Mass spectrometric detection was achieved by an electrospray ionization source in the positive mode coupled to a triple quadrupole mass analyzer. The calibration curves for the six analytes displayed good linearity over the concentration range 1.0 or 5.0-1000 ng/mL. The intra and inter-day precision (CV) ranged from 2.68 to 16.4% and from 9.02 to 14.5%, respectively. The intra- and inter-day accuracy (bias) ranged from -14.2 to 15.2% and from -9.00 to 4.80%, respectively. The extraction recoveries for all the analytes ranged from 55 to 86.9%. The LC-MS/MS method was successfully applied to a pharmacokinetic study of buprenorphine-naloxone combination in rats.


Assuntos
Buprenorfina/análogos & derivados , Cromatografia Líquida de Alta Pressão/métodos , Glucuronídeos/sangue , Naloxona/sangue , Antagonistas de Entorpecentes/sangue , Espectrometria de Massas em Tandem/métodos , Animais , Buprenorfina/sangue , Buprenorfina/metabolismo , Linhagem Celular Tumoral , Glucuronídeos/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Limite de Detecção , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Ratos
19.
Sci Rep ; 6: 39271, 2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-27982096

RESUMO

Opioids are considered the gold standard therapy for pain. However, TLR-dependent negative effects in analgesia have highlighted the complexities in the pharmacodynamics of opioids. While successive studies have reported that morphine and Morphine-3-glucuronide (M3G) activate the TLR4 pathway, the structural details of this mechanism are lacking. Here, we have utilized various computational tools to reveal the structural dynamics of the opioid-bound TLR4/MD2 complex, and have proposed a potential TLR4 activation mechanism. Our results support previous findings, and include the novel insight that the stable binding of morphine and naloxone, but not M3G, in the MD2 cavity, is TLR4 dependent. Morphine interacts with MD2 near its Phe126 loop to induce the active conformation (MD2C); however, this binding is likely reversible, and the complex gains stability upon interaction with TLR4. M3G also induces the MD2C state, with both the Phe126 loop and the H1 loop being involved in MD2-M3G complex stability. Remarkably, naloxone, which requires TLR4 interaction for complex stability, switches the conformation of the gating loop to the inactive state (MD2°). Cumulatively, our findings suggest that ligand binding and receptor clustering occur successively in opioid-induced TLR4 signaling, and that MD2 plasticity and pocket hydrophobicity are crucial for the recognition and accommodation of ligands.


Assuntos
Analgésicos Opioides/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/metabolismo , Animais , Sítios de Ligação , Ligação de Hidrogênio , Ligantes , Antígeno 96 de Linfócito/química , Antígeno 96 de Linfócito/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , Morfina/química , Morfina/metabolismo , Morfina/farmacologia , Derivados da Morfina/química , Derivados da Morfina/metabolismo , Derivados da Morfina/farmacologia , Naloxona/química , Naloxona/metabolismo , Naloxona/farmacologia , Análise de Componente Principal , Ligação Proteica , Estrutura Terciária de Proteína , Termodinâmica , Receptor 4 Toll-Like/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...