Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.361
Filtrar
1.
J Med Chem ; 67(9): 7603-7619, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38687204

RESUMEN

The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.


Asunto(s)
Receptores de Neuropéptido , Receptores Opioides mu , Animales , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Ratones , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Masculino , Analgésicos/farmacología , Analgésicos/química , Analgésicos/uso terapéutico , Analgésicos/síntesis química , Humanos , Relación Estructura-Actividad , Analgésicos Opioides/farmacología , Analgésicos Opioides/química
2.
Kaohsiung J Med Sci ; 40(5): 456-466, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38446546

RESUMEN

Transcutaneous electrical nerve stimulation (TENS) activates various pathways to induce antinociceptive effects, based on the frequencies used. This study evaluates the preemptive analgesic effects and their duration of low- (LT: 4 Hz) and high-frequency TENS (HT: 100 Hz) using a rat model of acute inflammatory pain. Acute inflammation was induced by injecting 1% formalin into the hind paws of rats. LT or HT was applied for 30 min before formalin injection. Pain-related behaviors, such as licking, flinching, and lifting, were recorded for 60 min postinjection. Immunohistochemistry was used to assess the number of phosphorylated extracellular signal-regulated kinase (pERK)- and c-fos-positive cells in the spinal cord. Naloxone, a µ-opioid receptors (MORs) antagonist, and naltrindole, a δ-opioid receptors (DORs) antagonist, were administered before TENS application. Pain behavior duration and pERK- and c-fos-positive cell expression were then measured. LT and HT pretreatment significantly reduced both pain behaviors and the number of pERK- and c-fos-positive cells postformalin injection. Naloxone and naltrindole partially reversed the effects of LT and HT, respectively. Notably, HT's analgesic effect lasted up to 120 min whereas that of LT persisted for 90 min. LT and HT effectively exerted their preemptive analgesic effects on acute inflammatory pain by inhibiting pERK and c-fos expression in the spinal cord. HT presented a longer-lasting effect compared to LT. MOR and DOR activation may contribute to LT and HT's analgesic mechanisms, respectively.


Asunto(s)
Inflamación , Naloxona , Proteínas Proto-Oncogénicas c-fos , Ratas Sprague-Dawley , Estimulación Eléctrica Transcutánea del Nervio , Animales , Estimulación Eléctrica Transcutánea del Nervio/métodos , Masculino , Naloxona/farmacología , Ratas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Dolor Agudo/terapia , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Antagonistas de Narcóticos/farmacología , Naltrexona/farmacología , Naltrexona/análogos & derivados , Médula Espinal/metabolismo , Médula Espinal/efectos de los fármacos , Receptores Opioides mu/metabolismo , Receptores Opioides mu/antagonistas & inhibidores , Manejo del Dolor/métodos , Fosforilación/efectos de los fármacos , Modelos Animales de Enfermedad
3.
Bioorg Chem ; 130: 106257, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36375349

RESUMEN

Ten new indole alkaloids (1-10) as well as eleven known analogs (11-21) were isolated from the stems and hooks of Uncaria rhynchophylla. Their structure elucidation was based on extensive NMR studies, MS and ECD data, with the essential aid of DFT prediction of ECD spectra. Compound 1 was determined as a 17,19-seco-cadambine-type alkaloid, and compound 3 was confirmed to be a 3,4-seco-tricyclic monoterpene indole alkaloid, which are the first seco-alkaloids possessing such cleavage positions from U. rhynchophylla. All the isolated compounds were evaluated for their bioactivities on dopamine D2 and Mu opioid receptors for discovering natural therapeutic drugs targeting central nervous system (CNS) diseases. Compounds 1, 2, 4, 5, 20 and 21 showed antagonistic bioactivities on the D2 receptor (IC50 0.678-15.200 µM), and compounds 1, 3, 6, 9, 10, 13, 18, 19 and 21 exhibited antagonistic effects on the Mu receptor (IC50 2.243-32.200 µM). Among them, compounds 1 and 21 displayed dual-target activities. Compound 1 showed conspicuous antagonistic activity on D2 and Mu receptors with the IC50 values of 0.678 ± 0.182 µM and 13.520 ± 2.480 µM, respectively. Compound 21 displayed moderate antagonistic activity on the two receptors with the IC50 values at 15.200 ± 1.764 µM and 32.200 ± 5.695 µM, respectively.


Asunto(s)
Antagonistas de los Receptores de Dopamina D2 , Alcaloides Indólicos , Uncaria , Alcaloides/química , Alcaloides/farmacología , Dopamina/metabolismo , Alcaloides Indólicos/farmacología , Alcaloides Indólicos/química , Receptores Opioides mu/antagonistas & inhibidores , Uncaria/química , Antagonistas de los Receptores de Dopamina D2/química , Antagonistas de los Receptores de Dopamina D2/farmacología , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/farmacología
4.
Eur J Med Chem ; 241: 114649, 2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-35961067

RESUMEN

To develop safer and potent analgesics, we designed, synthesized, and evaluated a new series of benzylaminofentanyl derivates as bifunctional µ opioid receptor (MOR) and σ1 receptor (σ1R) ligands. Compound 68 (Tao-191) showed desirable MOR agonism (Ki = 6.5 nΜ; EC50 = 48.5 nΜ, Emax = 66.3%) and σ1R antagonism (Ki = 35.7 nM) in vitro, and exerted powerful analgesic effects in the abdominal constriction test (ED50 = 0.32 mg/kg, in mice), formalin-induced pain test (phase II, ED50 = 2.26 mg/kg, in rats), and paclitaxel-induced neuropathic pain model (ED50 = 0.30 mg/kg, in mice). The contributions of MOR and σ1R to its antinociceptive effect were verified by combined administration with the MOR antagonist naloxone and the σ1R agonist PRE-084, respectively. At equianalgesic doses, compound 68 induced fewer MOR-related side effects-including physical and psychological dependence, respiratory depression, constipation, and acute hyperlocomotion-than fentanyl. The results provide a rationale for further exploration of the action and safety of dual MOR/σ1R ligands as a promising avenue for the development of potent and safe analgesics.


Asunto(s)
Analgésicos , Neuralgia , Receptores Opioides mu , Receptores sigma , Analgésicos/farmacología , Animales , Ligandos , Ratones , Naloxona/farmacología , Ratas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores sigma/agonistas , Receptores sigma/antagonistas & inhibidores , Receptor Sigma-1
5.
Acta Neurobiol Exp (Wars) ; 82(2): 217-225, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35833821

RESUMEN

The lateral hypothalamus (LH) sends neural pathways to structures involved on predator­related defensive behaviours, escape and antinociception. The aim of this study was to investigate the role played by µ-opioid receptors located on LH neurons in defensive behaviour and unconditioned fear­induced antinociception elicited by electric stimulation of LH. To achieve the goals, the µ1-opioid receptor selective antagonist naloxonazine was administered at different concentrations in the LH, and the defensive behaviour and fear­induced antinociception elicited by electrical stimulation of LH were evaluated. The electrical stimulation of LH caused escape behaviour followed by defensive antinociception. Microinjections of naloxonazine in a concentration of 5.0 µg/0.2 µL in the LH decreased the aversive stimulus­induced escape behaviour thresholds, but diminished defensive antinociception. These findings suggest that µ-opioid receptors of LH can be critical to panic attack­related symptoms and facilitate the unconditioned fear­induced antinociception produced by LH neurons activation.


Asunto(s)
Conducta Animal , Área Hipotalámica Lateral , Trastorno de Pánico , Receptores Opioides mu , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Bicuculina/farmacología , Miedo/fisiología , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Naloxona/análogos & derivados , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Nocicepción , Pánico/fisiología , Trastorno de Pánico/metabolismo , Trastorno de Pánico/psicología , Ratas , Ratas Wistar , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo
6.
Nature ; 606(7916): 917-921, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35551513

RESUMEN

Ingestion of alkaloid metabolites from the bark of Galbulimima (GB) sp. leads to psychotropic and excitatory effects in humans1-4. Limited, variable supply of GB alkaloids5, however, has impeded their biological exploration and clinical development6. Here we report a solution to the supply of GB18, a structural outlier and putative psychotropic principle of Galbulimima bark. Efficient access to its challenging tetrahedral attached-ring motif required the development of a ligand-controlled endo-selective cross-electrophile coupling and a diastereoselective hydrogenation of a rotationally dynamic pyridine. Reliable, gram-scale access to GB18 enabled its assignment as a potent antagonist of κ- and µ-opioid receptors-the first new targets in 35 years-and lays the foundation to navigate and understand the biological activity of Galbulimima metabolites.


Asunto(s)
Alcaloides , Magnoliopsida , Alcaloides/síntesis química , Alcaloides/farmacología , Técnicas de Química Sintética , Humanos , Hidrogenación , Ligandos , Magnoliopsida/química , Corteza de la Planta/química , Piridinas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores
7.
J Pharm Pharmacol ; 74(1): 139-146, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34355768

RESUMEN

BACKGROUND: Xanthomicrol is one of the methoxylated flavones and a promising cancer chemopreventive agent, but its anti-migration and anti-invasion ability on human hepatocellular carcinoma (HCC) remains unknown. OBJECTIVES: This study aims to explore Xanthomicrol's effects on migration and invasion ability of the human HCC Huh7 cell line. METHODS: Viability of Huh7 cells was measured by cell counting kit-8 (CCK8) assay. Cell apoptosis was assayed with flow cytometry analysis. The ability of migration and invasion of Huh7 cells was then detected through Transwell assays. Epithelial-mesenchymal transition (EMT)-related proteins were also detected through Western blot. KEY FINDINGS: Xanthomicrol inhibits the migration and invasion of Huh7 cells. The overexpression of Μu-opioid receptor (MOR) increases Huh7 cells' proliferation and enhances migration and invasion ability, while xanthomicrol treatment decreases the expression of MOR. Moreover, xanthomicrol can reverse migration, invasion and EMT-related protein expression by overexpressed MOR. CONCLUSIONS: These results suggest that xanthomicrol is a potential MOR antagonist, and it possesses potent anti-migration and anti-invasion ability on Huh7 cells.


Asunto(s)
Carcinoma Hepatocelular , Movimiento Celular/efectos de los fármacos , Flavonas/farmacología , Neoplasias Hepáticas , Invasividad Neoplásica/prevención & control , Receptores Opioides mu/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología
8.
Biochem Pharmacol ; 195: 114805, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673011

RESUMEN

Opioid-related fatalities involving synthetic opioids have reached unprecedented levels. This study evaluated the respiratory depressant effects of seven fentanyl analogs that have either emerged in the illicit drug supply or been identified in toxicological analyses following fatal or non-fatal intoxications. Adult male Swiss Webster mice were administered fentanyl analogs (isobutyrylfentanyl, crotonylfentanyl, para-methoxyfentanyl, para-methoxybutyrylfentanyl, 3-furanylfentanyl, thiophenefentanyl, and benzodioxolefentanyl) and their effects on minute volume as compared to mu-opioid receptor (MOR) agonist standards (fentanyl, morphine, and buprenorphine) were measured using whole body plethysmography (WBP). All drugs elicited significant (p ≤ 0.05) hypoventilation relative to vehicle for at least one dose tested: morphine (1, 3.2, 10, 32 mg/kg), buprenorphine, (0.032, 0.1, 0.32, 1, 3.2 mg/kg), fentanyl (0.0032, 0.01, 0.032, 0.1, 1, 32 mg/kg), isobutyrylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), crotonylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), para-methoxyfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), para-methoxybutyrylfentanyl (0.32, 1, 3.2, 10 mg/kg), 3-furanylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), thiophenefentanyl (1, 3.2, 10, 32, 100 mg/kg), and benzodioxolefentanyl (3.2, 10, 32, 100 mg/kg). The ED50 values for hypoventilation showed a rank order of potency as follows: fentanyl (ED50 = 0.96 mg/kg) > 3-furanylfentanyl (ED50 = 2.60 mg/kg) > crotonylfentanyl (ED50 = 2.72 mg/kg) > para-methoxyfentanyl (ED50 = 3.31 mg/kg) > buprenorphine (ED50 = 10.8 mg/kg) > isobutyrylfentanyl (ED50 = 13.5 mg/kg) > para-methoxybutyrylfentanyl (ED50 = 16.1 mg/kg) > thiophenefentanyl (ED50 = 18.0 mg/kg) > morphine (ED50 = 55.3 mg/kg) > benzodioxolefentanyl (ED50 = 10,168 mg/kg). A naloxone pretreatment (10 mg/kg) attenuated the hypoventilatory effects of all drugs. These results establish that the respiratory depressant effects of these fentanyl analogs are at least in part mediated by the MOR.


Asunto(s)
Fentanilo/análogos & derivados , Hipoventilación/prevención & control , Naloxona/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Animales , Fentanilo/química , Fentanilo/toxicidad , Hipoventilación/inducido químicamente , Hipoventilación/fisiopatología , Masculino , Ratones , Estructura Molecular , Antagonistas de Narcóticos/farmacología , Pletismografía/métodos , Receptores Opioides mu/fisiología , Mecánica Respiratoria/efectos de los fármacos , Mecánica Respiratoria/fisiología
9.
Bioorg Med Chem ; 51: 116509, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34798381

RESUMEN

A new series of enkephalin-like tetrapeptide analogs modified at the C-terminus by an N-(3,4-dichlorophenyl)-N-(piperidin-4-yl)propionamide (DPP) moiety were designed, synthesized, and tested for their binding affinities at opioid receptors and monoamine transporters to evaluate their potential multifunctional activity for the treatment of chronic pain. Most ligands exhibited high binding affinities in the nanomolar range at the opioid receptors with a slight delta-opioid receptor (DOR) selectivity over mu-opioid receptor (MOR) and kappa-opioid receptor (KOR) and low binding affinities in the micromolar range at the monoamine transporters, SERT and NET. Ligands of which the positions 1 and 4 were substituted by Dmt and Phe(4-X) residues, respectively, showed the excellent binding affinities at three opioid receptors. Among them, Dmt-d-Tic-Gly-Phe(4-F)-DPP was the most promising considering its excellent opioid affinities, particularly unexpected high binding affinity (Ki = 0.13 nM) at the KOR, and moderate interactions with serotonin/norepinephrine reuptake inhibitors (SNRIs). Docking studies revealed that the ligand was a good fit for the KOR binding pocket (binding score = 8,750).


Asunto(s)
Amidas/farmacología , Oligopéptidos/farmacología , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Amidas/química , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Oligopéptidos/síntesis química , Oligopéptidos/química , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
10.
Life Sci ; 285: 119996, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34597607

RESUMEN

AIMS: Dezocine and pentazocine, widely prescribed in China for postoperative pain, were initially considered as mixed agonist/antagonist targeting µ-opioid receptors (MORs) and κ-opioid receptors (KORs). However, dezocine has been revealed to alleviate chronic neuropathic pain through MOR activation and norepinephrine reuptake inhibition (NRI). This study investigated dezocine- and pentazocine-induced antinociception and physical dependence development, compared to the typical MOR-NRI opioid tapentadol. MAIN METHODS: Calcium mobilization assay was conducted to assess the potency of the drugs while hot-plate test was performed to compare the antinociception. Physical dependence development was compared with morphine. KEY FINDINGS: Treatment with dezocine, pentazocine and tapentadol stimulated calcium mobilization in HEK293 cells stably expressed MORs but not KORs, whereas dezocine and pentazocine inhibited KOR activities. Subcutaneously injected dezocine-, tapentadol- and pentazocine-induced antinociception dose-dependently, in hot-plate test. Intrathecally injected MOR antagonist CTAP, norepinephrine depletor 6-OHDA and α2-adrenoceptor (α2-AR) antagonist yohimbine partially antagonized dezocine, pentazocine and tapentadol antinociception. Whereas specific KOR antagonist GNTI did not alter their antinociception, the putative inverse KOR agonist nor-BNI reduced dezocine and pentazocine antinociception. Moreover, combined CTAP and 6-OHDA or yohimbine blocked dezocine and tapentadol antinociception but displayed the same partial inhibition on pentazocine antinociception as CTAP alone. Furthermore, compared to morphine and pentazocine, long-term treatment with dezocine and tapentadol produced much less physical dependence-related withdrawal signs, which were restored by spinal 6-OHDA or yohimbine treatment. SIGNIFICANCE: Our findings illustrated that dezocine and tapentadol, but not pentazocine, exert remarkable antinociception in nociceptive pain with less abuse liability via dual mechanisms of MOR activation and NRI.


Asunto(s)
Analgésicos Opioides/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Pentazocina/farmacología , Receptores Opioides mu/agonistas , Tapentadol/farmacología , Tetrahidronaftalenos/farmacología , Inhibidores de Captación Adrenérgica/química , Inhibidores de Captación Adrenérgica/farmacología , Analgésicos Opioides/química , Analgésicos Opioides/uso terapéutico , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Agonismo de Drogas , Antagonismo de Drogas , Células HEK293 , Humanos , Ratones , Pentazocina/química , Pentazocina/uso terapéutico , Receptores Adrenérgicos/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Tapentadol/química , Tapentadol/uso terapéutico , Tetrahidronaftalenos/química , Tetrahidronaftalenos/uso terapéutico
11.
Pharmacol Res Perspect ; 9(6): e00887, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34713624

RESUMEN

Opioid overdose is a leading cause of death in the United States. The only treatment available currently is the competitive antagonist, naloxone (Narcan® ). Although naloxone is very effective and has saved many lives, as a competitive antagonist it has limitations. Due to the short half-life of naloxone, renarcotization can occur if the ingested opioid agonist remains in the body longer. Moreover, because antagonism by naloxone is surmountable, renarcotization can also occur in the presence of naloxone if a relatively larger dose of opioid agonist is taken. In such circumstances, a long-lasting, non-surmountable antagonist would offer an improvement in overdose treatment. Methocinnamox (MCAM) has been reported to have a long duration of antagonist action at mu opioid receptors in vivo. In HEK cells expressing the human mu opioid receptor, MCAM antagonism of mu agonist-inhibition of cAMP production was time-dependent, non-surmountable and non-reversible, consistent with (pseudo)-irreversible binding. In vivo, MCAM injected locally into the rat hindpaw antagonized mu agonist-mediated inhibition of thermal allodynia for up to 96 h. By contrast, antagonism by MCAM of delta or kappa agonists in HEK cells and in vivo was consistent with simple competitive antagonism. Surprisingly, MCAM also shifted the concentration-response curves of mu agonists in HEK cells in the absence of receptor reserve in a ligand-dependent manner. The shift in the [D-Ala2 ,N-MePhe4 ,Gly-ol5 ]-enkephalin (DAMGO) concentration-response curve by MCAM was insensitive to naloxone, suggesting that in addition to (pseudo)-irreversible orthosteric antagonism, MCAM acts allosterically to alter the affinity and/or intrinsic efficacy of mu agonists.


Asunto(s)
Cinamatos/farmacología , Derivados de la Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Animales , AMP Cíclico/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Células HEK293 , Humanos , Ligandos , Masculino , Naloxona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Factores de Tiempo
12.
Curr Oncol Rep ; 23(10): 111, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34342720

RESUMEN

PURPOSE OF REVIEW: Opioids are still the most effective and widely used treatments for acute and chronic pain in cancer patients. This review focuses on the impact of opioids and mu-opioid receptors (MOR) on tumor progression and providing new ideas for targeting the MOR in cancer treatment. RECENT FINDINGS: Studies estimated that opioids facilitate tumor progression and are related to the worse prognosis in cancer patients. As the primary receptor of opioids, MOR is involved in the regulation of malignant transformation of tumors and participating in proliferation, invasion, metastasis, and angiogenesis. MOR may be a new molecular marker of malignant tumors and thus become a new target for cancer therapy, which may be beneficial to the outcomes of cancer patients.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Receptores Opioides mu/metabolismo , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/uso terapéutico , Animales , Progresión de la Enfermedad , Humanos , Inmunidad , Inflamación , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores
13.
Biol Pharm Bull ; 44(8): 1081-1087, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34334493

RESUMEN

Naldemedine (NAL), a peripherally acting µ-opioid receptor antagonist, is effective for opioid-induced constipation (OIC). However, diarrhea is the most common adverse event. We investigated the incidence of NAL-induced diarrhea in patients who started NAL at Nagasaki University Hospital between June 2017 and March 2019. Predictors of NAL-induced diarrhea were analyzed using a multivariate logistic regression model. Two hundred and forty-two patients were included in the present study, and NAL-induced diarrhea was observed in 17.8% (43 patients). The results of multiple logistic regression analyses identified the administration of opioid analgesics for 8 d or longer before the initiation of NAL (odds ratio (OR): 2.20, 95% confidence interval (95% CI): 1.04-4.64, p = 0.039), the combination of a laxative (OR: 2.22, 95%CI: 1.03-4.81, p = 0.042), and the combination of CYP3A4 inhibitors (strong/moderate) (OR: 2.80, 95%CI: 1.02-7.67, p = 0.045) as risk factors. Therefore, the development of diarrhea needs to be considered in patients with these risk factors. Furthermore, diarrhea may be controlled by the initiation of NAL within 7 d of opioid analgesics and, where possible, the discontinuation of or change in the combination of moderate or strong CYP3A4 inhibitors.


Asunto(s)
Estreñimiento/tratamiento farmacológico , Diarrea/inducido químicamente , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/efectos adversos , Anciano , Analgésicos Opioides/efectos adversos , Estreñimiento/inducido químicamente , Inhibidores del Citocromo P-450 CYP3A/efectos adversos , Femenino , Humanos , Laxativos/efectos adversos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Naltrexona/administración & dosificación , Naltrexona/efectos adversos , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/uso terapéutico , Oportunidad Relativa , Receptores Opioides mu/antagonistas & inhibidores , Factores de Riesgo
14.
Front Immunol ; 12: 687806, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326841

RESUMEN

Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating multi-systemic chronic condition of unknown aetiology classified as an immune dysfunction syndrome and neurological disorder. The discovery of the widely expressed Transient Receptor Potential Melastatin 3 (TRPM3) as a nociceptor channel substantially targeted by certain opioid receptors, and its implication in calcium (Ca2+)-dependent Natural Killer (NK) cell immune functions has raised the possibility that TRPM3 may be pharmacologically targeted to treat characteristic symptoms of ME/CFS. Naltrexone hydrochloride (NTX) acts as an antagonist to the mu (µ)-opioid receptor thus negating its inhibitory function on TRPM3. Based on the benefits reported by patients on their symptoms, low dose NTX (LDN, 3.0-5.0 mg/day) treatment seems to offer some potential benefit suggesting that its effect may be targeted towards the pathomechanism of ME/CFS. As there is no literature confirming the efficacy of LDN for ME/CFS patients in vitro, this study investigates the potential therapeutic effect of LDN in ME/CFS patients. TRPM3 ion channel activity was measured after modulation with Pregnenolone sulfate (PregS) and ononetin in NK cells on 9 ME/CFS patients taking LDN and 9 age- and sex-matched healthy controls using whole-cell patch-clamp technique. We report that ME/CFS patients taking LDN have restored TRPM3-like ionic currents in NK cells. Small ionic currents with a typical TRPM3-like outward rectification were measured after application of PregS, a TRPM3-agonist, in NK cells from patients taking LDN. Additionally, PregS-evoked ionic currents through TRPM3 were significantly modulated by ononetin, a TRPM3-antagonist, in NK cells from ME/CFS patients taking LDN. These data support the hypothesis that LDN may have potential as a treatment for ME/CFS by characterising the underlying regulatory mechanisms of LDN treatment involving TRPM3 and opioid receptors in NK cells. Finally, this study may serve for the repurpose of marketed drugs, as well as support the approval of prospective randomized clinical studies on the role and dose of NTX in treating ME/CFS patients.


Asunto(s)
Síndrome de Fatiga Crónica/tratamiento farmacológico , Células Asesinas Naturales/efectos de los fármacos , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Receptores Opioides mu/antagonistas & inhibidores , Canales Catiónicos TRPM/efectos de los fármacos , Adulto , Señalización del Calcio/efectos de los fármacos , Estudios de Casos y Controles , Reposicionamiento de Medicamentos , Síndrome de Fatiga Crónica/inmunología , Síndrome de Fatiga Crónica/metabolismo , Síndrome de Fatiga Crónica/fisiopatología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Persona de Mediana Edad , Naltrexona/efectos adversos , Antagonistas de Narcóticos/efectos adversos , Receptores Opioides mu/metabolismo , Canales Catiónicos TRPM/metabolismo , Resultado del Tratamiento
15.
J Cell Physiol ; 236(11): 7698-7710, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34038587

RESUMEN

The Mu-opioid receptor (MOR) has been implicated in tumorigenesis and metastasis. Methylnaltrexone (MNTX), an antagonist of MOR, has shown to inhibit tumor growth and metastasis in lung cancer cell lines. The effect of MNTX on other cell lines such as head and neck squamous cell carcinoma (HNSCC) has not been investigated. We measured the expression and activity of the receptor in different HNSCC cell lines. Then, we evaluated the impact of modulating the expression MOR and the effect of MNTX on the proliferation, clonogenic activity, invasion, and migration of two HNSCC (FaDu and MDA686Tu) cell lines expressing MOR and one cell line (UMSCC47) not expressing the receptor. We also evaluated the impact of MNTX on tumor growth and metastasis formation in vivo. Activation of the receptor with [d-Ala2,N-Me-Phe4, Gly5-ol] (DAMGO) caused a significant reduction in cyclic adenosine monophosphate levels in FaDu cells. Knockdown of MOR inhibited in vitro aggressive cell behaviors on FaDu and MDA686Tu cells and correlated with a reduction in markers of epithelial-mesenchymal transition. In vitro studies showed that MNTX strongly inhibited the proliferation, clonogenic activity, invasion, and migration of FaDu and MDA686Tu cells but has no effect on UMSCC47 cells. In vivo experiments demonstrated that MNTX suppresses tumor growth in HNSCC cell tumor-bearing mice. Our studies indicate that MOR could be considered as a therapeutic target to treat HNSCC.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Naltrexona/farmacología , Invasividad Neoplásica , Compuestos de Amonio Cuaternario/farmacología , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
CNS Neurosci Ther ; 27(7): 831-841, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34018697

RESUMEN

AIMS: Naltrexone is a mu opioid receptor (MOR) antagonist used to treat drug dependence in patients. Previous reports indicated that MOR antagonists reduced neurodegeneration and inflammation after brain injury. The purpose of this study was to evaluate the neuroprotective effect of naltrexone in cell culture and a mouse model of traumatic brain injury (TBI). METHODS: The neuroprotective effect of naltrexone was examined in primary cortical neurons co-cultured with BV2 microglia. Controlled cortical impact (CCI) was delivered to the left cerebral cortex of adult male MOR wild-type (WT) and knockout (KO) mice. Naltrexone was given daily for 4 days, starting from day 2 after lesioning. Locomotor activity was evaluated on day 5 after the CCI. Brain tissues were collected for immunostaining, Western, and qPCR analysis. RESULTS: Glutamate reduced MAP2 immunoreactivity (-ir), while increased IBA1-ir in neuron/BV2 co-culture; both responses were antagonized by naltrexone. TBI significantly reduced locomotor activity and increased the expression of IBA1, iNOS, and CD4 in the lesioned cortex. Naltrexone significantly and equally antagonized the motor deficits and expression of IBA1 and iNOS in WT and KO mice. TBI-mediated CD4 protein production was attenuated by naltrexone in WT mice, but not in KO mice. CONCLUSION: Naltrexone reduced TBI-mediated neurodegeneration and inflammation in MOR WT and KO mice. The protective effect of naltrexone involves non-MOR and MOR mechanisms.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Receptores Opioides mu/deficiencia , Animales , Técnicas de Cocultivo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Fármacos Neuroprotectores/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/genética
17.
Eur J Pharmacol ; 903: 174132, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33933466

RESUMEN

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.


Asunto(s)
Estreñimiento/tratamiento farmacológico , Morfinanos/farmacología , Antagonistas de Narcóticos/farmacología , Polietilenglicoles/farmacología , Administración Oral , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Conducta Animal/efectos de los fármacos , Células CHO , Calcio/metabolismo , Estreñimiento/inducido químicamente , Cricetulus , Fentanilo/administración & dosificación , Fentanilo/efectos adversos , Fentanilo/farmacología , Inyecciones Subcutáneas , Masculino , Ratones , Morfinanos/administración & dosificación , Morfina/farmacología , Naloxona/administración & dosificación , Naloxona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Dolor/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/efectos de los fármacos
18.
Peptides ; 141: 170543, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33794284

RESUMEN

Endomorphin analogs containing unnatural amino acids have demonstrated potent analgesic effects in our previous studies. In the present study, the differences in antinociception and the mechanisms thereof for analogs 1-3 administered intracerebroventricularly and intrathecally were explored. All analogs at different routes of administration produced potent analgesia compared to the parent peptide endomorphin-1. Multiple antagonists and antibodies were used to explore the mechanisms of action of these analogs, and it was inferred that analogs 1-3 stimulated the µ opioid receptor to induce antinociception. Moreover, the antibody data suggested that analog 2 may induce the release of immunoreactive [Leu5]-enkephaline and [Met5]-enkephaline to produce a secondary component of antinociception at the spinal level and analog 3 may stimulate the the release of immunoreactive [Met5]-enkephaline at the spinal level. Finally, analogs 2 and 3 produced no acute tolerance in the spinal cord. We hypothesize that the unique characteristics of the endomorphin analogs result from their capacities to stimulate the release of endogenous antinociceptive substances.


Asunto(s)
Analgésicos/farmacología , Encefalinas/metabolismo , Oligopéptidos/química , Receptores Opioides mu/metabolismo , Médula Espinal/efectos de los fármacos , Analgésicos/administración & dosificación , Animales , Dolor Crónico/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Dinorfinas/metabolismo , Encefalina Metionina/metabolismo , Inyecciones Intraventriculares , Ratones , Naloxona/análogos & derivados , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Oligopéptidos/farmacología , Receptores Opioides mu/antagonistas & inhibidores
19.
Curr Treat Options Oncol ; 22(3): 26, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33635493

RESUMEN

OPINION STATEMENT: Management of chronic pain is crucial to improve the quality of life of cancer and palliative care patients. Opioid-based treatments used to control pain can be prolonged over time. Unfortunately, constipation is one of the most disturbing adverse effects of long-term use of opioids. Opioid-induced constipation (OIC) occurs when opioids bind to the specific receptors present in the gastrointestinal (GI) tract, and can affect any patients receiving chronic opioid therapy, including cancer patients. The limited efficacy of laxatives to treat OIC symptoms prompted the search for new therapeutic strategies. Peripherally acting µ-opioid receptor antagonists (PAMORAs) have recently emerged as new effective drugs for OIC management due to their specific binding to enteric µ-receptors. Little information is available on the use of PAMORAs in real-life practice for OIC treatment in cancer patients. In this paper, a panel of experts specializing in cancer and palliative care pools their clinical experience with PAMORAs in cancer patients presenting OIC and highlights the importance of timing and choice of therapy in achieving prompt OIC management and benefitting patients.


Asunto(s)
Dolor en Cáncer/tratamiento farmacológico , Oncología Médica , Antagonistas de Narcóticos/uso terapéutico , Receptores Opioides mu/antagonistas & inhibidores , Factores de Edad , Dolor en Cáncer/etiología , Toma de Decisiones Clínicas , Comorbilidad , Manejo de la Enfermedad , Interacciones Farmacológicas , Quimioterapia Combinada , Medicina Basada en la Evidencia , Humanos , Oncología Médica/métodos , Antagonistas de Narcóticos/farmacología , Neoplasias/complicaciones , Pautas de la Práctica en Medicina , Nivel de Atención , Resultado del Tratamiento
20.
Commun Biol ; 4(1): 238, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619305

RESUMEN

Antibodies represent powerful tools to examine signal transduction pathways. Here, we present a strategy integrating multiple state-of-the-art methods to produce, validate, and utilize antibodies. Focusing on understudied synaptic proteins, we generated 137 recombinant antibodies. We used yeast display antibody libraries from the B cells of immunized rabbits, followed by FACS sorting under stringent conditions to identify high affinity antibodies. The antibodies were validated by high-throughput functional screening, and genome editing. Next, we explored the temporal dynamics of signaling in single cells. A subset of antibodies targeting opioid receptors were used to examine the effect of treatment with opiates that have played central roles in the worsening of the 'opioid epidemic.' We show that morphine and fentanyl exhibit differential temporal dynamics of receptor phosphorylation. In summary, high-throughput approaches can lead to the identification of antibody-based tools required for an in-depth understanding of the temporal dynamics of opioid signaling.


Asunto(s)
Anticuerpos/farmacología , Especificidad de Anticuerpos , Ensayos Analíticos de Alto Rendimiento , Proteína Quinasa C/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Anticuerpos/inmunología , Línea Celular Tumoral , Activación Enzimática , Fentanilo/farmacología , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Morfina/farmacología , Fosforilación , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Conejos , Receptores Opioides mu/inmunología , Receptores Opioides mu/metabolismo , Transducción de Señal , Sinapsis/inmunología , Sinapsis/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...