Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.265
Filtrar
1.
Curr Pharm Des ; 28(32): 2639-2652, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35538798

RESUMO

BACKGROUND: Previous studies have reported that benzodiazepines (BZDs) seem to enhance euphoric and reinforcing properties of opioids in opioid users so that a direct effect on opioid receptors has been postulated, together with a possible synergistic induction of severe side effects due to co use of BDZs and opioids. This is particularly worrisome given the appearance on the market of designer benzodiazepines (DBZDs), whose activity/toxicity profiles are scarcely known. OBJECTIVES: This study aimed to evaluate, through computational studies, the binding affinity (or lack thereof) of 101 DBZDs identified online on the kappa, mu, and delta opioid receptors (K, M, DOR); and to assess whether their mechanism of action could include activation of the latter. METHODS: MOE® was used for the computational studies. Pharmacophore mapping based on strong opioids agonist binders' 3D chemical features was used to filter the DBZDs. Resultant DBZDs were docked into the crystallised 3D active conformation of KOR (PDB6B73), DOR (PDB6PT3) and MOR (PDB5C1M). Co-crystallised ligands and four strong agonists were used as reference compounds. A score (S, Kcal/mol) representative of the predicted binding affinity, and a description of ligand interactions were obtained from MOE®. RESULTS: The docking results, filtered for S < -8.0 and the interaction with the Asp residue, identified five DBZDs as putative binders of the three ORs : ciclotizolam, fluloprazolam, JQ1, Ro 48-6791, and Ro 48-8684. CONCLUSION: It may be inferred that at least some DBZDs may have the potential to activate opioid receptors. This could mediate/increase their anxiolytic, analgesic, and addiction potentials, as well as worsen the side effects associated with opioid co-use.


Assuntos
Analgésicos Opioides , Ansiolíticos , Benzodiazepinas , Drogas Desenhadas , Receptores Opioides , Humanos , Analgésicos , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Benzodiazepinas/efeitos adversos , Benzodiazepinas/química , Benzodiazepinas/farmacologia , Ligantes , Receptores Opioides/agonistas , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Drogas Desenhadas/efeitos adversos , Drogas Desenhadas/química , Drogas Desenhadas/farmacologia
2.
Neuropharmacology ; 206: 108938, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-34982972

RESUMO

Morphine tolerance (MT) caused by the long-term use of morphine is a major medical problem. The molecular mechanism of morphine tolerance remains elusive. Here, we established a morphine tolerance model in rats and verified whether the long noncoding RNA (lncRNA) MRAK159688 is involved in morphine tolerance and its specific molecular mechanism. We show the significant upregulation of MRAK159688 expression in the spinal cord of morphine-tolerant rats. Overexpression of MRAK159688 by a lentivirus reduces the analgesic efficacy of morphine and induces pain behavior. Downregulation of MRAK159688 using a small interfering RNA (siRNA) attenuates the formation of morphine tolerance, partially reverses the development of morphine tolerance and alleviates morphine-induced hyperalgesia. MRAK159688 is located in the nucleus and cytoplasm of neurons, and it colocalizes with repressor element-1 silencing transcription factor (REST) in the nucleus. MRAK159688 potentiates the expression and function of REST, thereby inhibiting the expression of mu opioid receptor (MOR) and subsequently inducing morphine tolerance. Moreover, REST overexpression blocks the effects of MRAK159688 siRNA on relieving morphine tolerance. In general, chronic morphine administration-mediated upregulation of MRAK159688 in the spinal cord contributes to morphine tolerance and hyperalgesia by promoting REST-mediated inhibition of MOR. MRAK159688 downregulation may represent a novel RNA-based therapy for morphine tolerance.


Assuntos
Tolerância a Medicamentos , Regulação da Expressão Gênica , Hiperalgesia , Morfina/farmacologia , Entorpecentes/farmacologia , Transtornos Relacionados ao Uso de Opioides , RNA Longo não Codificante , Receptores Opioides mu , Proteínas Repressoras/metabolismo , Medula Espinal , Animais , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , RNA Longo não Codificante/efeitos dos fármacos , RNA Longo não Codificante/metabolismo , Ratos , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Transtornos Relacionados ao Uso de Substâncias
3.
Cell Rep ; 38(4): 110253, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35081358

RESUMO

Acute myeloid leukemia (AML) is a genetically heterogeneous and frequently fatal malignancy. The ten-eleven translocation (TET)-mediated DNA demethylation is known to be critically associated with AML pathogenesis. Through chemical compound screening, we find that the opioid receptor agonist, loperamide hydrochloride (OPA1), significantly suppresses AML cell viability. The potential therapeutic effects of opioid receptor agonists, especially OPA1, are verified in AML cells in vitro and mouse and human AML models in vivo. OPA1-induced activation of OPRM1 signaling enhances the transcription of TET2 and thus activates both catalytic-dependent and -independent functions of TET2. Notably, AMLs with TET2 mutations or chemotherapy resistance are sensitive to OPA1 as well. Our results reveal the OPRM1-TET2 regulatory axis in AML and suggest that opioid agonists, particularly OPA1, a US Food and Drug Administration (FDA)-approved antidiarrheal drug, have therapeutic potential in AML, especially in TET2-mutated and chemotherapy-resistant AMLs, which have a poor prognosis.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Leucemia Mieloide Aguda/patologia , Receptores Opioides mu/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Leucemia Mieloide Aguda/metabolismo , Loperamida/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Opioides mu/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34755545

RESUMO

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Assuntos
Sistema Nervoso Entérico/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Xantonas/farmacologia , Analgésicos Opioides/farmacologia , Benzamidas/farmacologia , Colo/efeitos dos fármacos , Sistema Nervoso Entérico/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Humanos , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
5.
Int J Obes (Lond) ; 46(2): 400-407, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728775

RESUMO

BACKGROUND: Obesity is a pressing public health concern worldwide. Novel pharmacological means are urgently needed to combat the increase of obesity and accompanying type 2 diabetes (T2D). Although fully established obesity is associated with neuromolecular alterations and insulin resistance in the brain, potential obesity-promoting mechanisms in the central nervous system have remained elusive. In this triple-tracer positron emission tomography study, we investigated whether brain insulin signaling, µ-opioid receptors (MORs) and cannabinoid CB1 receptors (CB1Rs) are associated with risk for developing obesity. METHODS: Subjects were 41 young non-obese males with variable obesity risk profiles. Obesity risk was assessed by subjects' physical exercise habits, body mass index and familial risk factors, including parental obesity and T2D. Brain glucose uptake was quantified with [18F]FDG during hyperinsulinemic euglycemic clamp, MORs were quantified with [11C]carfentanil and CB1Rs with [18F]FMPEP-d2. RESULTS: Subjects with higher obesity risk had globally increased insulin-stimulated brain glucose uptake (19 high-risk subjects versus 19 low-risk subjects), and familial obesity risk factors were associated with increased brain glucose uptake (38 subjects) but decreased availability of MORs (41 subjects) and CB1Rs (36 subjects). CONCLUSIONS: These results suggest that the hereditary mechanisms promoting obesity may be partly mediated via insulin, opioid and endocannabinoid messaging systems in the brain.


Assuntos
Cérebro/metabolismo , Intolerância à Glucose/etiologia , Obesidade/diagnóstico , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Adulto , Índice de Massa Corporal , Cérebro/fisiopatologia , Feminino , Finlândia/epidemiologia , Intolerância à Glucose/epidemiologia , Intolerância à Glucose/metabolismo , Humanos , Modelos Lineares , Masculino , Obesidade/epidemiologia , Obesidade/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Tomografia por Emissão de Pósitrons/estatística & dados numéricos , Receptor CB1 de Canabinoide/metabolismo , Receptores Opioides mu/metabolismo , Fatores de Risco
6.
Med Clin North Am ; 106(1): 169-185, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34823729

RESUMO

Buprenorphine formulations (including buprenorphine/naloxone) are effective treatments of pain and opioid use disorder (OUD). Historically, perioperative management of patients prescribed buprenorphine involved abstinence from buprenorphine sufficient to allow for unrestricted mu-opioid receptor availability to full agonist opioid (FAO) treatment. Evidence is mounting that a multimodal analgesic strategy, including simultaneous administration of buprenorphine and FAO, nonopioid adjuncts such as acetaminophen and nonsteroidal anti-inflammatory drugs, and regional anesthesia, is a safe and effective perioperative strategy for the patient prescribed long-term buprenorphine treatment of OUD. This strategy will likely simplify management and more seamlessly provide continuous buprenorphine treatment of OUD after hospital discharge.


Assuntos
Analgésicos Opioides/farmacologia , Buprenorfina/farmacologia , Tratamento de Substituição de Opiáceos/métodos , Dor Pós-Operatória/prevenção & controle , Assistência Perioperatória/métodos , Acetaminofen/administração & dosagem , Acetaminofen/uso terapêutico , Idoso , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/uso terapêutico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/uso terapêutico , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/uso terapêutico , Artroplastia de Quadril/efeitos adversos , Artroplastia de Quadril/métodos , Buprenorfina/administração & dosagem , Buprenorfina/uso terapêutico , Terapia Combinada/métodos , Composição de Medicamentos/métodos , Feminino , Humanos , Comunicação Interdisciplinar , Masculino , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Dor/tratamento farmacológico , Manejo da Dor/métodos , Receptores Opioides mu/efeitos dos fármacos
7.
J Neurochem ; 160(4): 454-468, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34919270

RESUMO

There is increasing evidence for a daily rhythm of µ-opioid receptor (MOR) efficacy and the development of alcohol dependence. Previous studies show that ß-arrestin 2 (bArr2) has an impact on alcohol intake, at least partially mediated via modulation of MOR signaling, which in turn mediates the alcohol rewarding effects. Considering the interplay of circadian rhythms on MOR and alcohol dependence, we aimed to investigate bArr2 in alcohol dependence at different time points of the day/light cycle on the level of bArr2 mRNA (in situ hybridization), MOR availability (receptor autoradiography), and MOR signaling (Damgo-stimulated G-protein coupling) in the nucleus accumbens of alcohol-dependent and non-dependent Wistar rats. Using a microarray data set we found that bArr2, but not bArr1, shows a diurnal transcription pattern in the accumbens of naïve rats with higher expression levels during the active cycle. In 3-week abstinent rats, bArr2 is up-regulated in the accumbens at the beginning of the active cycle (ZT15), whereas no differences were found at the beginning of the inactive cycle (ZT3) compared with controls. This effect was accompanied by a specific down-regulation of MOR binding in the active cycle. Additionally, we detect a higher receptor coupling during the inactive cycle compared with the active cycle in alcohol-dependent animals. Together, we report daily rhythmicity for bArr2 expression linked to an inverse pattern of MOR, suggesting an involvement for bArr2 on circadian regulation of G-protein coupled receptors in alcohol dependence. The presented data may have implications for the development of novel bArr2-related treatment targets for alcoholism.


Assuntos
Alcoolismo/genética , Ritmo Circadiano/genética , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética , beta-Arrestina 2/genética , Alcoolismo/tratamento farmacológico , Animais , Regulação para Baixo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Masculino , Análise em Microsséries , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Recompensa
8.
Addict Biol ; 27(1): e13086, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34382313

RESUMO

Repeated morphine exposure has been shown to induce neuronal plasticity in reward-related areas of the brain. miR-132, a CREB-induced and activation-dependent microRNA, has been suggested to be involved in the neuronal plasticity by increasing neuronal dendritic branches and spinogenesis. However, it is still unclear whether miR-132 is related to morphine dependence. Here, we investigate whether miR-132 is involved in morphine dependence and whether it is related to the structural plasticity of the dentate gyrus (DG) neurons. Sprague-Dawley rats are treated with increasing doses of morphine injection for six consecutive days to develop morphine dependence. Our results show that dendritic branching and spinogenesis of the DG neurons of morphine dependent rats are increased. Morphine treatment (24 h) promotes the differentiation of N2a cells stably expressing µ-opioid receptor by up-regulating miR-132 expression. Moreover, inhibiting miR-132 3p (but not 5p) of the DG neurons can reverse the structural plasticity and disrupt the formation of morphine dependence in rats. These findings indicate that miR-132 in the DG neurons is involved in morphine dependence via modifying the neuronal plasticity.


Assuntos
Giro Denteado/efeitos dos fármacos , MicroRNAs/metabolismo , Dependência de Morfina/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos
9.
Biol Pharm Bull ; 44(11): 1746-1751, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34719650

RESUMO

Tramadol is a weak opioid that produces analgesic effect via both the µ-opioid receptor (MOR) and non-opioid targets. Constipation is the most common opioid-related side effect in patients with cancer and non-cancer pain. However, the contribution of MOR to tramadol-induced constipation is unclear. Therefore, we used naldemedine, a peripherally acting MOR antagonist, and MOR-knockout mice to investigate the involvement of peripheral MOR in tramadol-induced constipation using a small intestinal transit model. A single dose of tramadol (3-100 mg/kg, per os (p.o.)) inhibited small intestinal transit dose-dependently in rats. Naldemedine (0.01-10 mg/kg, p.o.) blocked the inhibition of small intestinal transit induced by tramadol (30 mg/kg, p.o.) in rats. The transition rate increased dose-dependently over the range of naldemedine 0.01-0.3 mg/kg, and complete recovery was observed at 0.3-10 m/kg. Additionally, tramadol (30 and 100 mg/kg, subcutaneously (s.c.)) inhibited small intestinal transit in wild-type mice but not in MOR-knockout mice. These results suggest that peripheral MOR participates in tramadol-induced constipation.


Assuntos
Analgésicos Opioides/efeitos adversos , Constipação Induzida por Opioides/etiologia , Receptores Opioides mu/efeitos dos fármacos , Tramadol/efeitos adversos , Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Animais , Intestino Delgado/efeitos dos fármacos , Masculino , Naltrexona/efeitos adversos , Naltrexona/análogos & derivados , Naltrexona/sangue , Naltrexona/farmacocinética , Nociceptividade/efeitos dos fármacos , Constipação Induzida por Opioides/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Opioides mu/metabolismo , Tramadol/sangue , Tramadol/farmacocinética
10.
Molecules ; 26(20)2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34684749

RESUMO

The current protocols for neuropathic pain management include µ-opioid receptor (MOR) analgesics alongside other drugs; however, there is debate on the effectiveness of opioids. Nevertheless, dose escalation is required to maintain their analgesia, which, in turn, contributes to a further increase in opioid side effects. Finding novel approaches to effectively control chronic pain, particularly neuropathic pain, is a great challenge clinically. Literature data related to pain transmission reveal that angiotensin and its receptors (the AT1R, AT2R, and MAS receptors) could affect the nociception both in the periphery and CNS. The MOR and angiotensin receptors or drugs interacting with these receptors have been independently investigated in relation to analgesia. However, the interaction between the MOR and angiotensin receptors has not been excessively studied in chronic pain, particularly neuropathy. This review aims to shed light on existing literature information in relation to the analgesic action of AT1R and AT2R or MASR ligands in neuropathic pain conditions. Finally, based on literature data, we can hypothesize that combining MOR agonists with AT1R or AT2R antagonists might improve analgesia.


Assuntos
Dor Crônica/tratamento farmacológico , Receptores de Angiotensina/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Humanos , Neuralgia/tratamento farmacológico , Nociceptividade/efeitos dos fármacos , Manejo da Dor/métodos , Proto-Oncogene Mas , Receptores de Angiotensina/metabolismo , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
11.
Molecules ; 26(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34299443

RESUMO

Although persistent pain is estimated to affect about 20% of the adult population, current treatments have poor results. Polypharmacology, which is the administration of more than one drug targeting on two or more different sites of action, represents a prominent therapeutic approach for the clinical management of persistent pain. Thus, in the drug discovery process the "one-molecule-multiple targets" strategy nowadays is highly recognized. Indeed, multitarget ligands displaying a better antinociceptive activity with fewer side effects, combined with favorable pharmacokinetic and pharmacodynamic characteristics, have already been shown. Multitarget ligands possessing non-opioid/opioid and opioid/opioid mechanisms of action are considered as potential drug candidates for the management of various pain conditions. In particular, dual-target MOPr (mu opioid peptide receptor)/DOPr (delta opioid peptide receptor) ligands exhibit an improved antinociceptive profile associated with a reduced tolerance-inducing capability. The benzomorphan-based compounds LP1 and LP2 belong to this class of dual-target MOPr/DOPr ligands. In the present manuscript, the structure-activity relationships and the pharmacological fingerprint of LP1 and LP2 compounds as suitable drug candidates for persistent pain relief is described.


Assuntos
Benzomorfanos/farmacologia , Dor/tratamento farmacológico , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Benzomorfanos/química , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Humanos , Ligantes , Dor/fisiopatologia , Manejo da Dor/métodos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Relação Estrutura-Atividade
12.
CNS Neurosci Ther ; 27(11): 1313-1326, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34255932

RESUMO

AIMS: Chronification of postoperative pain is a common clinical phenomenon following surgical operation, and it perplexes a great number of patients. Estrogen and its membrane receptor (G protein-coupled estrogen receptor, GPER) play a crucial role in pain regulation. Here, we explored the role of GPER in the rostral ventromedial medulla (RVM) during chronic postoperative pain and search for the possible mechanism. METHODS AND RESULTS: Postoperative pain was induced in mice or rats via a plantar incision surgery. Behavioral tests were conducted to detect both thermal and mechanical pain, showing a small part (16.2%) of mice developed into pain persisting state with consistent low pain threshold on 14 days after incision surgery compared with the pain recovery mice. Immunofluorescent staining assay revealed that the GPER-positive neurons in the RVM were significantly activated in pain persisting rats. In addition, RT-PCR and immunoblot analyses showed that the levels of GPER and phosphorylated µ-type opioid receptor (p-MOR) in the RVM of pain persisting mice were apparently increased on 14 days after incision surgery. Furthermore, chemogenetic activation of GPER-positive neurons in the RVM of Gper-Cre mice could reverse the pain threshold of pain recovery mice. Conversely, chemogenetic inhibition of GPER-positive neurons in the RVM could prevent mice from being in the pain persistent state. CONCLUSION: Our findings demonstrated that the GPER in the RVM was responsible for the chronification of postoperative pain and the downstream pathway might be involved in MOR phosphorylation.


Assuntos
Dor Crônica/genética , Bulbo/efeitos dos fármacos , Dor Pós-Operatória/genética , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Animais , Dor Crônica/fisiopatologia , Hiperalgesia/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medição da Dor , Dor Pós-Operatória/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética
13.
J Clin Pharm Ther ; 46(6): 1501-1504, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34111307

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Food and Drug Administration (FDA) risk evaluation and mitigation strategies (REMs) encourage emergency responders, paramedics, law enforcement agents, and even laypeople to be trained in the administration of naloxone with the intent of rescuing individuals from a known or suspected opioid overdose. COMMENT: Although naloxone is generally safe and effective at reversing respiratory depression caused by a conventional opioid such as morphine or heroin by competing with the opioid and displacing it from the µ-opioid receptor, questions increasingly are arising as to whether naloxone can adequately reverse opioid overdoses that may involve the potent opioids fentanyl and its analogues (F/FAs). In other words, as more and more opioid overdoses involve F/FAs, can naloxone keep up? WHAT IS NEW AND CONCLUSION: As a competitive antagonist at µ-opioid receptors, naloxone is often a life-saving agent in cases of overdose caused by conventional opioids, but it may not be versatile or powerful enough to combat the rising tide of overdoses due to fentanyl and its illicit analogues, or in cases of overdose involving combinations of opioids and non-opioids.


Assuntos
Fentanila/toxicidade , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Overdose de Opiáceos/tratamento farmacológico , Diafragma/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fentanila/farmacologia , Heroína/toxicidade , Humanos , Laringismo/induzido quimicamente , Rigidez Muscular/induzido quimicamente , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Parede Torácica/efeitos dos fármacos
14.
Eur J Pharmacol ; 903: 174132, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33933466

RESUMO

Opioid-induced constipation is the most prevalent adverse effect of opioid drugs. Peripherally acting mu opioid receptor antagonists (PAMORAs), including naloxegol, are indicated for the treatment of opioid-induced constipation. The aim of this study was the in vitro and in vivo pharmacological characterization of naloxegol in comparison with naloxone. In vitro experiments were performed to measure calcium mobilization in cells coexpressing opioid receptors and chimeric G proteins and mu receptor interaction with G protein and ß-arrestin 2 using bioluminescence resonance energy transfer. In vivo experiments were performed in mice to measure pain threshold using the tail withdrawal assay and colonic transit using the bead expulsion assay. In vitro, naloxegol behaved as a selective and competitive mu receptor antagonist similarly to naloxone, being 3-10-fold less potent. In vivo, naloxone was effective in blocking fentanyl actions when given subcutaneously (sc), but not per os (po). In contrast, naloxegol elicited very similar effects with sc or po administration counteracting in a dose dependent manner the constipating effects of fentanyl without interfering with the fentanyl mediated analgesia. Thus, a useful PAMORA action could be obtained with naloxegol both after po and sc administration.


Assuntos
Constipação Intestinal/tratamento farmacológico , Morfinanos/farmacologia , Antagonistas de Entorpecentes/farmacologia , Polietilenoglicóis/farmacologia , Administração Oral , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Células CHO , Cálcio/metabolismo , Constipação Intestinal/induzido quimicamente , Cricetulus , Fentanila/administração & dosagem , Fentanila/efeitos adversos , Fentanila/farmacologia , Injeções Subcutâneas , Masculino , Camundongos , Morfinanos/administração & dosagem , Morfina/farmacologia , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Dor/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/efeitos dos fármacos
15.
Eur J Pharmacol ; 901: 174089, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33826922

RESUMO

The participation of the peripheral opioid and cannabinoid endogenous systems in modulating muscle pain and inflammation has not been fully explored. Thus, the aim of this study was to investigate the involvement of these endogenous systems during muscular-tissue hyperalgesia induced by inflammation. Hyperalgesia was induced by carrageenan injection into the tibialis anterior muscles of male Wistar rats. We padronized an available Randal-Sellito test adaptation to evaluate nociceptive behavior elicited by mechanical insult in muscles. Western blot analysis was performed to evaluate the expression levels of opioid and cannabinoid receptors in the dorsal root ganglia. The non-selective opioid peptide receptor antagonist (naloxone) and the selective mu opioid receptor MOP (clocinnamox) and kappa opioid receptor KOP (nor-binaltorphimine) antagonists were able to intensify carrageenan-induced muscular hyperalgesia. On the other hand, the selective delta opioid receptor (DOP) antagonist (naltrindole) did not present any effect on nociceptive behavior. Moreover, the selective inhibitor of aminopeptidases (Bestatin) provoked considerable dose-dependent analgesia when intramuscularly injected into the hyperalgesic muscle. The CB1 receptor antagonist (AM251), but not the CB2 receptor antagonist (AM630), intensified muscle hyperalgesia. All irreversible inhibitors of anandamide hydrolase (MAFP), the inhibitor for monoacylglycerol lipase (JZL184) and the anandamide reuptake inhibitor (VDM11) decreased carrageenan-induced hyperalgesia in muscular tissue. Lastly, MOP, KOP and CB1 expression levels in DRG were baseline even after muscular injection with carrageenan. The endogenous opioid and cannabinoid systems participate in peripheral muscle pain control through the activation of MOP, KOP and CB1 receptors.


Assuntos
Mialgia/tratamento farmacológico , Receptores de Canabinoides/fisiologia , Receptores Opioides/fisiologia , Animais , Ácidos Araquidônicos/antagonistas & inibidores , Carragenina , Cinamatos/farmacologia , Endocanabinoides/antagonistas & inibidores , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/psicologia , Masculino , Monoacilglicerol Lipases/antagonistas & inibidores , Derivados da Morfina/farmacologia , Mialgia/induzido quimicamente , Mialgia/psicologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Medição da Dor/efeitos dos fármacos , Alcamidas Poli-Insaturadas/antagonistas & inibidores , Ratos , Ratos Wistar , Receptores de Canabinoides/efeitos dos fármacos , Receptores Opioides/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos
17.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33846240

RESUMO

Positive allosteric modulators (PAMs) of the mu-opioid receptor (MOR) have been hypothesized as potentially safer analgesics than traditional opioid drugs. This is based on the idea that PAMs will promote the action of endogenous opioid peptides while preserving their temporal and spatial release patterns and so have an improved therapeutic index. However, this hypothesis has never been tested. Here, we show that a mu-PAM, BMS-986122, enhances the ability of the endogenous opioid Methionine-enkephalin (Met-Enk) to stimulate G protein activity in mouse brain homogenates without activity on its own and to enhance G protein activation to a greater extent than ß-arrestin recruitment in Chinese hamster ovary (CHO) cells expressing human mu-opioid receptors. Moreover, BMS-986122 increases the potency of Met-Enk to inhibit GABA release in the periaqueductal gray, an important site for antinociception. We describe in vivo experiments demonstrating that the mu-PAM produces antinociception in mouse models of acute noxious heat pain as well as inflammatory pain. These effects are blocked by MOR antagonists and are consistent with the hypothesis that in vivo mu-PAMs enhance the activity of endogenous opioid peptides. Because BMS-986122 does not bind to the orthosteric site and has no inherent agonist action at endogenously expressed levels of MOR, it produces a reduced level of morphine-like side effects of constipation, reward as measured by conditioned place preference, and respiratory depression. These data provide a rationale for the further exploration of the action and safety of mu-PAMs as an innovative approach to pain management.


Assuntos
Regulação Alostérica/fisiologia , Dor/tratamento farmacológico , Receptores Opioides mu/metabolismo , Regulação Alostérica/efeitos dos fármacos , Analgesia/métodos , Analgésicos , Analgésicos Opioides/farmacologia , Animais , Células CHO , Cricetulus , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Morfina , Antagonistas de Entorpecentes , Manejo da Dor/métodos , Estudo de Prova de Conceito , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos
18.
Bioorg Med Chem Lett ; 41: 127953, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33766769

RESUMO

In the present work, we reported the application of a nitrogen-walk approach on developing a series of novel opioid ligands containing an azaindole moiety at the C6-position of the epoxymorphinan skeleton. In vitro study results showed that introducing a nitrogen atom around the indole moiety not only retained excellent binding affinity, but also led to significant functional switch at the mu opioid receptor (MOR). Further computational investigations provided corroborative evidence and plausible explanations of the results of the in vitro studies. Overall, our current work implemented a series of novel MOR ligands with high binding affinity and considerably low efficacy, which may shed light on rational design of low efficacy MOR ligands for opioid use disorder therapeutics.


Assuntos
Naltrexona/análogos & derivados , Nitrogênio/química , Receptores Opioides mu/efeitos dos fármacos , Sítios de Ligação , Humanos , Ligantes , Modelos Moleculares , Simulação de Acoplamento Molecular , Naltrexona/síntese química , Naltrexona/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Conformação Proteica
19.
Neural Plast ; 2021: 6682275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33688340

RESUMO

Chronic pain is considered an economic burden on society as it often results in disability, job loss, and early retirement. Opioids are the most common analgesics prescribed for the management of moderate to severe pain. However, chronic exposure to these drugs can result in opioid tolerance and opioid-induced hyperalgesia. On pain modulation strategies, exploiting the multitarget drugs with the ability of the superadditive or synergistic interactions attracts more attention. In the present report, we have reviewed the analgesic effects of different dopamine receptors, particularly D1 and D2 receptors, in different regions of the central nervous system, including the spinal cord, striatum, nucleus accumbens (NAc), and periaqueductal gray (PAG). According to the evidence, these regions are not only involved in pain modulation but also express a high density of DA receptors. The findings can be categorized as follows: (1) D2-like receptors may exert a higher analgesic potency, but D1-like receptors act in different manners across several mechanisms in the mentioned regions; (2) in the spinal cord and striatum, antinociception of DA is mainly mediated by D2-like receptors, while in the NAc and PAG, both D1- and D2-like receptors are involved as analgesic targets; and (3) D2-like receptor agonists can act as adjuvants of µ-opioid receptor agonists to potentiate analgesic effects and provide a better approach to pain relief.


Assuntos
Dor/tratamento farmacológico , Dor/fisiopatologia , Substância Cinzenta Periaquedutal/fisiopatologia , Receptores de Dopamina D2/agonistas , Analgésicos/farmacologia , Animais , Tolerância a Medicamentos/fisiologia , Humanos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiopatologia , Medição da Dor/métodos , Substância Cinzenta Periaquedutal/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo
20.
J Nat Prod ; 84(4): 1034-1043, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33635670

RESUMO

Ten indole and oxindole alkaloids (1-10) were isolated from the freshly collected leaves of Malaysian Mitragyna speciosa (Kratom). The chemical structures of these compounds were established on the basis of extensive 1D and 2D NMR and HRMS data analysis. The spectroscopic data of mitragynine oxindole B (4) are reported herein for the first time. The spatial configuration of mitragynine oxindole B (4) was confirmed by single-crystal X-ray diffraction. Simultaneous quantification of the isolated alkaloids in the M. speciosa leaf specimens collected from different locations in the northern region of Peninsular Malaysia was also performed using UPLC-MS/MS. The oxindole alkaloids (1-4) and the indole alkaloid (10) were assessed for binding affinity at opioid receptors. Corynoxine (1) showed high binding affinity to µ-opioid receptors with a Ki value of 16.4 nM. Further, corynoxine (1) was 1.8-fold more potent than morphine in rats subjected to a nociceptive hot plate assay. These findings have important implications for evaluating the combined effects of the minor oxindole alkaloids in the overall therapeutic activity of M. speciosa.


Assuntos
Analgésicos/farmacologia , Mitragyna/química , Oxindóis/farmacologia , Receptores Opioides mu/efeitos dos fármacos , Animais , Feminino , Humanos , Indóis , Malásia , Masculino , Estrutura Molecular , Folhas de Planta/química , Ratos , Ratos Sprague-Dawley , Alcaloides de Triptamina e Secologanina/farmacologia , Compostos de Espiro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...