Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 30(5): 632-647.e10, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37146583

RESUMEN

Schwann cells (SCs) are the primary glia of the peripheral nervous system. SCs are involved in many debilitating disorders, including diabetic peripheral neuropathy (DPN). Here, we present a strategy for deriving SCs from human pluripotent stem cells (hPSCs) that enables comprehensive studies of SC development, physiology, and disease. hPSC-derived SCs recapitulate the molecular features of primary SCs and are capable of in vitro and in vivo myelination. We established a model of DPN that revealed the selective vulnerability of SCs to high glucose. We performed a high-throughput screen and found that an antidepressant drug, bupropion, counteracts glucotoxicity in SCs. Treatment of hyperglycemic mice with bupropion prevents their sensory dysfunction, SC death, and myelin damage. Further, our retrospective analysis of health records revealed that bupropion treatment is associated with a lower incidence of neuropathy among diabetic patients. These results highlight the power of this approach for identifying therapeutic candidates for DPN.


Asunto(s)
Diabetes Mellitus , Neuropatías Diabéticas , Ratones , Animales , Humanos , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/etiología , Bupropión/uso terapéutico , Estudios Retrospectivos , Nervio Ciático , Células de Schwann , Descubrimiento de Drogas
2.
Neuropharmacology ; 227: 109442, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36731721

RESUMEN

Illicitly manufactured fentanyl is driving the current opioid crisis, and various fentanyl analogs are appearing in recreational drug markets worldwide. To assess the potential health risks posed by fentanyl analogs, it is necessary to understand structure-activity relationships for these compounds. Here we compared the pharmacology of two structurally related fentanyl analogs implicated in opioid overdose: cyclopropylfentanyl and valerylfentanyl. Cyclopropylfentanyl has a three-carbon ring attached to the carbonyl group on the fentanyl scaffold, whereas valerylfentanyl has a four-carbon chain at the same position. In vitro assays examining µ-opioid receptor (MOR) coupling to G proteins in CHO cells showed that cyclopropylfentanyl is a full agonist (EC50 = 8.6 nM, %Emax = 113%), with potency and efficacy similar to fentanyl (EC50 = 10.3 nM, %Emax = 113%). By contrast, valerylfentanyl is a partial agonist at MOR (EC50 = 179.8 nM, %Emax = 60%). Similar results were found in assays assessing MOR-mediated ß-arrestin recruitment in HEK cells. In vivo studies in male CD-1 mice demonstrated that both fentanyl analogs induce naloxone-reversible antinociception and respiratory suppression, but cyclopropylfentanyl is 100-times more potent as an antinociceptive agent (ED50 = 0.04 mg/kg, s. c.) than valerylfentanyl (ED50 = 4.0 mg/kg, s. c.). Molecular simulation results revealed that the alkyl chain of valerylfentanyl cannot be well accommodated by the active state of MOR and may transition the receptor toward an inactive state, converting the fentanyl scaffold to a partial agonist. Taken together, our results suggest that cyclopropylfentanyl presents much greater risk of adverse effects when compared to valerylfentanyl. Moreover, the summed findings may provide clues to the design of therapeutic opioids with reduced adverse side effects.


Asunto(s)
Analgésicos Opioides , Fentanilo , Masculino , Ratones , Animales , Cricetinae , Cricetulus , Fentanilo/farmacología , Analgésicos Opioides/farmacología , Naloxona , Relación Estructura-Actividad , Receptores Opioides mu/agonistas
3.
J Med Chem ; 64(22): 16553-16572, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34783240

RESUMEN

The leaves of Mitragyna speciosa (kratom), a plant native to Southeast Asia, are increasingly used as a pain reliever and for attenuation of opioid withdrawal symptoms. Using the tools of natural products chemistry, chemical synthesis, and pharmacology, we provide a detailed in vitro and in vivo pharmacological characterization of the alkaloids in kratom. We report that metabolism of kratom's major alkaloid, mitragynine, in mice leads to formation of (a) a potent mu opioid receptor agonist antinociceptive agent, 7-hydroxymitragynine, through a CYP3A-mediated pathway, which exhibits reinforcing properties, inhibition of gastrointestinal (GI) transit and reduced hyperlocomotion, (b) a multifunctional mu agonist/delta-kappa antagonist, mitragynine pseudoindoxyl, through a CYP3A-mediated skeletal rearrangement, displaying reduced hyperlocomotion, inhibition of GI transit and reinforcing properties, and (c) a potentially toxic metabolite, 3-dehydromitragynine, through a non-CYP oxidation pathway. Our results indicate that the oxidative metabolism of the mitragynine template beyond 7-hydroxymitragynine may have implications in its overall pharmacology in vivo.


Asunto(s)
Alcaloides de Triptamina Secologanina/farmacología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Receptores Opioides mu
4.
Elife ; 102021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33555255

RESUMEN

Controlling receptor functional selectivity profiles for opioid receptors is a promising approach for discovering safer analgesics; however, the structural determinants conferring functional selectivity are not well understood. Here, we used crystal structures of opioid receptors, including the recently solved active state kappa opioid complex with MP1104, to rationally design novel mixed mu (MOR) and kappa (KOR) opioid receptor agonists with reduced arrestin signaling. Analysis of structure-activity relationships for new MP1104 analogs points to a region between transmembrane 5 (TM5) and extracellular loop (ECL2) as key for modulation of arrestin recruitment to both MOR and KOR. The lead compounds, MP1207 and MP1208, displayed MOR/KOR Gi-partial agonism with diminished arrestin signaling, showed efficient analgesia with attenuated liabilities, including respiratory depression and conditioned place preference and aversion in mice. The findings validate a novel structure-inspired paradigm for achieving beneficial in vivo profiles for analgesia through different mechanisms that include bias, partial agonism, and dual MOR/KOR agonism.


Asunto(s)
Morfinanos/química , Receptores Opioides kappa/química , Receptores Opioides mu/química , Secuencias de Aminoácidos , Analgésicos/química , Analgésicos/metabolismo , Animales , Sitios de Unión , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
5.
J Med Chem ; 63(22): 13618-13637, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33170687

RESUMEN

In this work, we studied a series of carfentanyl amide-based opioid derivatives targeting the mu opioid receptor (µOR) and the delta opioid receptor (δOR) heteromer as a credible novel target in pain management therapy. We identified a lead compound named MP135 that exhibits high G-protein activity at µ-δ heteromers compared to the homomeric δOR or µOR and low ß-arrestin2 recruitment activity at all three. Furthermore, MP135 exhibits distinct signaling profile, as compared to the previously identified agonist targeting µ-δ heteromers, CYM51010. Pharmacological characterization of MP135 supports the utility of this compound as a molecule that could be developed as an antinociceptive agent similar to morphine in rodents. In vivo characterization reveals that MP135 maintains untoward side effects such as respiratory depression and reward behavior; together, these results suggest that optimization of MP135 is necessary for the development of therapeutics that suppress the classical side effects associated with conventional clinical opioids.


Asunto(s)
Fentanilo/análogos & derivados , Receptores Opioides delta/agonistas , Analgésicos/síntesis química , Analgésicos/farmacología , Animales , Línea Celular , Fentanilo/síntesis química , Fentanilo/farmacología , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Long-Evans , Receptores Opioides delta/metabolismo
6.
ACS Cent Sci ; 5(6): 992-1001, 2019 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-31263758

RESUMEN

Mitragyna speciosa, more commonly known as kratom, is a plant native to Southeast Asia, the leaves of which have been used traditionally as a stimulant, analgesic, and treatment for opioid addiction. Recently, growing use of the plant in the United States and concerns that kratom represents an uncontrolled drug with potential abuse liability, have highlighted the need for more careful study of its pharmacological activity. The major active alkaloid found in kratom, mitragynine, has been reported to have opioid agonist and analgesic activity in vitro and in animal models, consistent with the purported effects of kratom leaf in humans. However, preliminary research has provided some evidence that mitragynine and related compounds may act as atypical opioid agonists, inducing therapeutic effects such as analgesia, while limiting the negative side effects typical of classical opioids. Here we report evidence that an active metabolite plays an important role in mediating the analgesic effects of mitragynine. We find that mitragynine is converted in vitro in both mouse and human liver preparations to the much more potent mu-opioid receptor agonist 7-hydroxymitragynine and that this conversion is mediated by cytochrome P450 3A isoforms. Further, we show that 7-hydroxymitragynine is formed from mitragynine in mice and that brain concentrations of this metabolite are sufficient to explain most or all of the opioid-receptor-mediated analgesic activity of mitragynine. At the same time, mitragynine is found in the brains of mice at very high concentrations relative to its opioid receptor binding affinity, suggesting that it does not directly activate opioid receptors. The results presented here provide a metabolism-dependent mechanism for the analgesic effects of mitragynine and clarify the importance of route of administration for determining the activity of this compound. Further, they raise important questions about the interpretation of existing data on mitragynine and highlight critical areas for further research in animals and humans.

7.
Mol Pharmacol ; 96(2): 247-258, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31243060

RESUMEN

Circular RNAs (circRNAs) are a distinct category of single-stranded, covalently closed RNAs formed by backsplicing. The functions of circRNAs are incompletely known and are under active investigation. Here, we report that in addition to traditional linear mRNAs (linRNA), mouse, rat, and human opioid receptor genes generate exonic circRNA isoforms. Using standard molecular biologic methods, Oprm1 circRNAs (circOprm1) were detected in RNAs of rodent and human brains and spinal cords, as well as human neuroblastoma cells, suggesting evolutionary conservation. Sequencing confirmed backsplicing using canonical splice sites. Oprm1 circRNAs were sense-stranded circRNAs resistant to RNase R digestion. The relative abundance of Oprm1 circRNA to linRNA determined by quantitative reverse transcription polymerase chain reaction varied among mouse brain regions, with circRNA isoforms predominating in rostral structures and less abundant in brain stem. Chronic morphine exposure in mice increased brain circOprm1e2.3 and circOprm1.e2.e3.e4(302) levels by 1.5- to 1.6-fold relative to linRNA. Sequence analysis predicted numerous microRNA binding sites within Oprm1 circRNA sequences, suggesting a potential role in microRNA sequestration through sponging. In addition, we observed that other opioid receptor genes including δ, κ, and nociceptin receptor genes produced similar circRNAs. In conclusion, all members of the opioid receptor gene family express circRNAs, with Oprm1 circRNA levels exceeding those of linear forms in some regions. SIGNIFICANCE STATEMENT: The modulation of Oprm1 circular RNA (circRNA) expression by morphine, coupled with the high abundance and existence of potential miRNA binding sites with circRNA sequences suggests the potential role of Oprm1 circRNAs in chronic opioid effects such as tolerance.


Asunto(s)
Encéfalo/metabolismo , Morfina/farmacología , Neuroblastoma/genética , ARN Circular/genética , Médula Espinal/metabolismo , Animales , Línea Celular Tumoral , Secuencia Conservada , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratas , Receptores Opioides mu/genética , Análisis de Secuencia de ARN
8.
Anesth Analg ; 128(2): 365-373, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29649035

RESUMEN

BACKGROUND: Levorphanol is a potent analgesic that has been used for decades. Most commonly used for acute and cancer pain, it also is effective against neuropathic pain. The recent appreciation of the importance of functional bias and the uncovering of multiple µ opioid receptor splice variants may help explain the variability of patient responses to different opioid drugs. METHODS: Here, we evaluate levorphanol in a variety of traditional in vitro receptor binding and functional assays. In vivo analgesia studies using the radiant heat tail flick assay explored the receptor selectivity of the responses through the use of knockout (KO) mice, selective antagonists, and viral rescue approaches. RESULTS: Receptor binding studies revealed high levorphanol affinity for all the µ, δ, and κ opioid receptors. In S-GTPγS binding assays, it was a full agonist at most µ receptor subtypes, with the exception of MOR-1O, but displayed little activity in ß-arrestin2 recruitment assays, indicating a preference for G-protein transduction mechanisms. A KO mouse and selective antagonists confirmed that levorphanol analgesia was mediated through classical µ receptors, but there was a contribution from 6 transmembrane targets, as illustrated by a lower response in an exon 11 KO mouse and its rescue with a virally transfected 6 transmembrane receptor splice variant. Compared to morphine, levorphanol had less respiratory depression at equianalgesic doses. CONCLUSIONS: While levorphanol shares many of the same properties as the classic opioid morphine, it displays subtle differences that may prove helpful in its clinical use. Its G-protein signaling bias is consistent with its diminished respiratory depression, while its incomplete cross tolerance with morphine suggests it may prove valuable clinically with opioid rotation.


Asunto(s)
Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Levorfanol/metabolismo , Levorfanol/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/fisiología , Receptores Acoplados a Proteínas G/agonistas , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
9.
Neuropharmacology ; 134(Pt A): 101-107, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28807672

RESUMEN

Novel synthetic opioids (NSO) are increasingly encountered in illicit heroin and counterfeit pain pills. Many NSO are resurrected from older biomedical literature or patent applications, so limited information is available about their biological effects. Here we examined the pharmacology of three structurally-distinct NSO found in the recreational drug market: N-(1-(2-phenylethyl)-4-piperidinyl)-N-phenylbutyramide (butyrylfentanyl), 3,4-dichloro-N-[(1R,2R)-2-(dimethylamino)cyclohexyl]-N-methylbenzamide (U-47700) and 1-cyclohexyl-4-(1,2-diphenylethyl)piperazine (MT-45). Radioligand binding and GTPγS functional assays were carried out in cells transfected with murine mu- (MOR-1), delta- (DOR-1) or kappa-opioid receptors (KOR-1). Antinociceptive effects were determined using the radiant heat tail flick technique in mice, and opioid specificity was assessed with the mu-opioid antagonist naloxone. Butyrylfentanyl, U-47700 and MT-45 displayed nM affinities at MOR-1, but were less potent than morphine, and had much weaker effects at DOR-1 and KOR-1. All NSO exhibited agonist actions at MOR-1 in the GTPγS assay. Butyrylfentanyl and U-47700 were 31- and 12-fold more potent than morphine in the tail flick assay, whereas MT-45 was equipotent with morphine. Analgesic effects were reversed by naloxone and absent in genetically-engineered mice lacking MOR-1. Our findings confirm that butyrylfentanyl, U-47700 and MT-45 are selective MOR-1 agonists with in vitro affinities less than morphine. However, analgesic potencies vary more than 30-fold across the compounds, and in vitro binding affinity does not predict in vivo potency. Taken together, our findings highlight the risks to humans who may unknowingly be exposed to these and other NSO when taking adulterated heroin or counterfeit pain medications. This article is part of the Special Issue entitled 'Designer Drugs and Legal Highs.'


Asunto(s)
Analgésicos Opioides/farmacología , Drogas Ilícitas/farmacología , Analgésicos Opioides/química , Animales , Benzamidas/farmacocinética , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Drogas Ilícitas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/farmacocinética , Dimensión del Dolor , Piperazinas/farmacocinética , Piperazinas/farmacología , Ensayo de Unión Radioligante , Ratas , Receptores Opioides/genética , Receptores Opioides/metabolismo , Isótopos de Azufre/farmacocinética , Factores de Tiempo , Transfección
10.
JCI Insight ; 2(22)2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-29202454

RESUMEN

W-18 (4-chloro-N-[1-[2-(4-nitrophenyl)ethyl]-2-piperidinylidene]-benzenesulfonamide) and W-15 (4-chloro-N-[1-(2-phenylethyl)-2-piperidinylidene]-benzenesulfonamide) represent two emerging drugs of abuse chemically related to the potent opioid agonist fentanyl (N-(1-(2-phenylethyl)-4-piperidinyl)-N-phenylpropanamide). Here, we describe the comprehensive pharmacological profiles of W-18 and W-15, as examination of their structural features predicted that they might lack opioid activity. We found W-18 and W-15 to be without detectible activity at µ, δ, κ, and nociception opioid receptors in a variety of assays. We also tested W-18 and W-15 for activity as allosteric modulators at opioid receptors and found them devoid of significant positive or negative allosteric modulatory activity. Comprehensive profiling at essentially all the druggable GPCRs in the human genome using the PRESTO-Tango platform revealed no significant activity. Weak activity at the sigma receptors and the peripheral benzodiazepine receptor was found for W-18 (Ki = 271 nM). W-18 showed no activity in either the radiant heat tail-flick or the writhing assays and also did not induce classical opioid behaviors. W-18 is extensively metabolized, but its metabolites also lack opioid activity. Thus, although W-18 and W-15 have been suggested to be potent opioid agonists, our results reveal no significant activity at these or other known targets for psychoactive drugs.


Asunto(s)
Drogas de Diseño/química , Drogas de Diseño/farmacología , Fentanilo/química , Fentanilo/farmacología , Analgésicos Opioides , Animales , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Drogas Ilícitas , Ratones , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB2/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos
11.
Medchemcomm ; 8(1): 152-157, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28603600

RESUMEN

It was previously reported that 6ß-aminomorphinan derivatives show high affinity for opiate receptors. Novel 6ß-heteroarylamidomorphinanes were designed based on the MOR selective antagonist NAP. The 6ß-aminomorphinanes were prepared with stereoselective Mitsunobu reaction and subsequently acylated with nicotinic acid and isonicotinic acid chloride hydrochlorides. The receptor binding and efficacy were determined in vitro and the analgesic activity was studied in vivo. The in vitro studies revealed moderate selectivity for MOR. At least two compounds in this series exhibited long-acting analgesic response when administered subcutaneously and intracerebroventricularly. When the substances were given intracerebroventricularly to mice, they showed analgesic potency comparable to morphine.

12.
Psychopharmacology (Berl) ; 234(12): 1891-1900, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28343361

RESUMEN

RATIONALE: Morphine is the prototypic mu opioid, producing its analgesic actions through traditional 7 transmembrane domain (7TM) G-protein-coupled receptors generated by the mu opioid receptor gene (Oprm1). However, the Oprm1 gene undergoes extensive alternative splicing to yield three structurally distinct sets of splice variants. In addition to the full-length 7TM receptors, it produces a set of truncated variants comprised of only 6 transmembrane domains (6TM). OBJECTIVES: This study explored the relative contributions of 7TM and 6TM variants in a range of morphine actions. METHODS: Groups of male and mixed-gender wild-type and exon 11 Oprm1 knockout mice were examined in a series of behavioral assays measuring analgesia, hyperalgesia, respiration, and reward in conditioned place preference assays. RESULTS: Loss of the 6TM variants in an exon 11 knockout (E11 KO) mouse did not affect morphine analgesia, reward, or respiratory depression. However, E11 KO mice lacking 6TM variants failed to show morphine-induced hyperalgesia, developed tolerance more slowly than wild-type mice, and did not display hyperlocomotion. CONCLUSIONS: Together, our findings confirm the established role of 7TM mu receptor variants in morphine analgesia, reward, and respiratory depression, but reveal an unexpected obligatory role for 6TM variants in morphine-induced hyperalgesia and a modulatory role in morphine tolerance and dependence.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Morfina/uso terapéutico , Variantes Farmacogenómicas/genética , Receptores Opioides mu/genética , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Analgesia/métodos , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos/genética , Femenino , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Variantes Farmacogenómicas/efectos de los fármacos , Receptores Opioides mu/deficiencia
13.
J Clin Invest ; 127(4): 1561-1573, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28319053

RESUMEN

Extensive 3' alternative splicing of the mu opioid receptor gene OPRM1 creates multiple C-terminal splice variants. However, their behavioral relevance remains unknown. The present study generated 3 mutant mouse models with truncated C termini in 2 different mouse strains, C57BL/6J (B6) and 129/SvEv (129). One mouse truncated all C termini downstream of Oprm1 exon 3 (mE3M mice), while the other two selectively truncated C-terminal tails encoded by either exon 4 (mE4M mice) or exon 7 (mE7M mice). Studies of these mice revealed divergent roles for the C termini in morphine-induced behaviors, highlighting the importance of C-terminal variants in complex morphine actions. In mE7M-B6 mice, the exon 7-associated truncation diminished morphine tolerance and reward without altering physical dependence, whereas the exon 4-associated truncation in mE4M-B6 mice facilitated morphine tolerance and reduced morphine dependence without affecting morphine reward. mE7M-B6 mutant mice lost morphine-induced receptor desensitization in the brain stem and hypothalamus, consistent with exon 7 involvement in morphine tolerance. In cell-based studies, exon 7-associated variants shifted the bias of several mu opioids toward ß-arrestin 2 over G protein activation compared with the exon 4-associated variant, suggesting an interaction of exon 7-associated C-terminal tails with ß-arrestin 2 in morphine-induced desensitization and tolerance. Together, the differential effects of C-terminal truncation illustrate the pharmacological importance of OPRM1 3' alternative splicing.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Receptores Opioides mu/metabolismo , Empalme Alternativo , Animales , Encéfalo/metabolismo , Codón sin Sentido , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Exones , Tránsito Gastrointestinal/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Dependencia de Morfina/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Opioides mu/genética
14.
J Med Chem ; 59(18): 8381-97, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27556704

RESUMEN

Natural products found in Mitragyna speciosa, commonly known as kratom, represent diverse scaffolds (indole, indolenine, and spiro pseudoindoxyl) with opioid activity, providing opportunities to better understand opioid pharmacology. Herein, we report the pharmacology and SAR studies both in vitro and in vivo of mitragynine pseudoindoxyl (3), an oxidative rearrangement product of the corynanthe alkaloid mitragynine. 3 and its corresponding corynantheidine analogs show promise as potent analgesics with a mechanism of action that includes mu opioid receptor agonism/delta opioid receptor antagonism. In vitro, 3 and its analogs were potent agonists in [(35)S]GTPγS assays at the mu opioid receptor but failed to recruit ß-arrestin-2, which is associated with opioid side effects. Additionally, 3 developed analgesic tolerance more slowly than morphine, showed limited physical dependence, respiratory depression, constipation, and displayed no reward or aversion in CPP/CPA assays, suggesting that analogs might represent a promising new generation of novel pain relievers.


Asunto(s)
Analgésicos Opioides/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Alcaloides de Triptamina Secologanina/farmacología , Arrestina beta 2/metabolismo , Analgésicos Opioides/química , Animales , Línea Celular , Humanos , Masculino , Ratones , Mitragyna/química , Simulación del Acoplamiento Molecular , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina Secologanina/química
15.
Proc Natl Acad Sci U S A ; 113(13): 3663-8, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976581

RESUMEN

The clinical management of severe pain depends heavily on opioids acting through mu opioid receptors encoded by the Oprm1 gene, which undergoes extensive alternative splicing. In addition to generating a series of prototypic seven transmembrane domain (7TM) G protein-coupled receptors (GPCRs), Oprm1 also produces a set of truncated splice variants containing only six transmembrane domains (6TM) through which selected opioids such as IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) mediate a potent analgesia without many undesirable effects. Although morphine analgesia is independent of these 6TM mu receptor isoforms, we now show that the selective loss of the 6TM variants in a knockout model eliminates the analgesic actions of delta and kappa opioids and of α2-adrenergic compounds, but not cannabinoid, neurotensin, or muscarinic drugs. These observations were confirmed by using antisense paradigms. Despite their role in analgesia, loss of the 6TM variants were not involved with delta opioid-induced seizure activity, aversion to the kappa drug U50, 488H, or α2-mediated hypolocomotion. These observations support the existence of parallel opioid and nonopioid pain modulatory systems and highlight the ability to dissociate unwanted delta, kappa1, and α2 actions from analgesia.


Asunto(s)
Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Empalme Alternativo , Analgesia , Analgésicos Opioides/farmacología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Manejo del Dolor , Dimensión del Dolor , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Receptores Opioides mu/deficiencia
16.
ACS Chem Neurosci ; 6(11): 1813-24, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26325040

RESUMEN

3-Iodobenzoyl naltrexamine (IBNtxA) is a potent analgesic belonging to the pharmacologically diverse 6ß-amidoepoxymorphinan group of opioids. We present the synthesis and pharmacological evaluation of five analogs of IBNtxA. The scaffold of IBNtxA was modified by removing the 14-hydroxy group, incorporating a 7,8 double bond and various N-17 alkyl substituents. The structural modifications resulted in analogs with picomolar affinities for opioid receptors. The lead compound (MP1104) was found to exhibit approximately 15-fold greater antinociceptive potency (ED50 = 0.33 mg/kg) compared with morphine, mediated through the activation of kappa- and delta-opioid receptors. Despite its kappa agonism, this lead derivative did not cause place aversion or preference in mice in a place-conditioning assay, even at doses 3 times the analgesic ED50. However, pretreatment with the lead compound prevented the reward behavior associated with cocaine in a conditioned place preference assay. Together, these results suggest the promise of dual acting kappa- and delta-opioid receptor agonists as analgesics and treatments for cocaine addiction.


Asunto(s)
Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacología , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Morfinanos/síntesis química , Morfinanos/farmacología , Analgésicos Opioides/química , Animales , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Trastornos Relacionados con Cocaína/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Evaluación Preclínica de Medicamentos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Morfinanos/química , Naltrexona/análogos & derivados , Naltrexona/química , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/farmacología , Distribución Aleatoria , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Recompensa , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
17.
ACS Chem Neurosci ; 6(9): 1570-7, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26148793

RESUMEN

We report a novel approach to synthesize carfentanil amide analogues utilizing the isocyanide-based four-component Ugi multicomponent reaction. A small library of bis-amide analogues of carfentanil was created using N-alkylpiperidones, aniline, propionic acid, and various aliphatic isocyanides. Our lead compound showed high affinity for mu (MOR) and delta opioid receptors (DOR) with no appreciable affinity for kappa (KOR) receptors in radioligand binding assays. The compound was found to be a mixed MOR agonist/partial DOR agonist in [(35)S]GTPγS functional assays, and it showed moderate analgesic potency in vivo. The compound showed no visible signs of physical dependence or constipation in mice. In addition, it produced less respiratory depression than morphine. Most mixed MOR/DOR opioids reported in the literature are peptides and thereby systemically inactive. Our approach utilizing a multicomponent reaction has the promise to deliver potent and efficacious small-molecule analgesics with potential clinical utility.


Asunto(s)
Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Amidas/síntesis química , Amidas/química , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/química , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Fentanilo/análogos & derivados , Fentanilo/síntesis química , Fentanilo/química , Masculino , Ratones , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Morfina/efectos adversos , Morfina/química , Morfina/farmacología , Dolor/tratamiento farmacológico , Respiración/efectos de los fármacos
18.
J Pharmacol Exp Ther ; 350(3): 710-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24970924

RESUMEN

IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) is a potent analgesic in mice lacking many traditional opioid side effects. In mice, it displays no respiratory depression, does not produce physical dependence with chronic administration, and shows no cross-tolerance to morphine. It has limited effects on gastrointestinal transit and shows no reward behavior. Biochemical studies indicate its actions are mediated through a set of µ-opioid receptor clone MOR-1 splice variants associated with exon 11 that lack exon 1 and contain only six transmembrane domains. Like the mouse and human, rats express exon 11-associated splice variants that also contain only six transmembrane domains, raising the question of whether IBNtxA would have a similar pharmacologic profile in rats. When given systemically, IBNtxA is a potent analgesic in rats, with an ED50 value of 0.89 mg/kg s.c., approximately 4-fold more potent than morphine. It shows no analgesic cross-tolerance in morphine-pelleted rats. IBNtxA displays no respiratory depression as measured by blood oxygen saturation. In contrast, oximetry shows that an equianalgesic dose of morphine lowers blood oxygen saturation values by 30%. IBNtxA binding is present in a number of brain regions, with the thalamus standing out with very high levels and the cerebellum with low levels. As in mice, IBNtxA is a potent analgesic in rats with a favorable pharmacologic profile and reduced side effects.


Asunto(s)
Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Naltrexona/análogos & derivados , Insuficiencia Respiratoria/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Naltrexona/metabolismo , Naltrexona/farmacología , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Unión Proteica/fisiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
19.
Eur J Med Chem ; 69: 786-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24103580

RESUMEN

Aminomorphinans are a relatively young class of opioid drugs among which substances of high in vitro efficacy and favorable in vivo action are found. We report the synthesis and pharmacological evaluation of novel 6ß-acylaminomorphinans. 6ß-Morphinamine and 6ß-codeinamine were stereoselectively synthesized by Mitsunobu reaction. The aminomorphinans were subsequently acylated with diversely substituted cinnamic acids. In vitro binding studies on cinnamoyl morphinamines showed moderate affinity for all opiate receptors with some selectivity for mu opioid receptors, while cinnamoyl codeinamines only showed affinity for mu opioid receptors. In vivo analgesia studies showed significant analgesic activity of 6ß-cinnamoylmorphinamine mediated by mu and delta receptors. The lead compound was found to be roughly equipotent to morphine (ED50 3.13 ± 1.09 mg/kg) but devoid of the dangerous side-effect respiratory depression, a major issue associated with traditional opioid therapy.


Asunto(s)
Analgésicos/farmacología , Morfinanos/farmacología , Umbral del Dolor/efectos de los fármacos , Analgésicos/síntesis química , Analgésicos/química , Animales , Relación Dosis-Respuesta a Droga , Calor , Ratones , Estructura Molecular , Morfinanos/síntesis química , Morfinanos/química , Morfina/efectos adversos , Morfina/farmacología , Antagonistas de Narcóticos , Dimensión del Dolor , Receptores Opioides/agonistas , Insuficiencia Respiratoria/inducido químicamente , Relación Estructura-Actividad , Factores de Tiempo
20.
J Inflamm Res ; 5: 99-109, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055766

RESUMEN

Neonates are known to exhibit increased susceptibility to bacterial and viral infections and increasing evidence demonstrates that the increased susceptibility is related to their attenuated immune response to infections. The lung is equipped with an innate defense system involving both cellular and humoral mediators. The present study was performed to characterize the expression of inflammatory mediators in the lung of neonatal rats in comparison with older animals. Rats at postnatal day 1 (P1), P21, and P70 were treated with saline or 0.25 mg/kg lipopolysaccharide (LPS) via intraperitoneal injection. Two hours later, animals were sacrificed and the transcriptional response of key inflammatory mediators and enzyme activity of myeloperoxidase (MPO) in the lung of these animals were examined. LPS-induced messenger RNA (mRNA) expression of pro-inflammatory cytokines, namely interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α, antiinflammatory cytokines, namely IL-10 and IL-1 receptor antagonist (IL-1ra), and chemokines, namely macrophage inflammatory protein (MIP)-1ß, MIP-2, and monocyte chemotactic protein-1, in P1 lung was much reduced compared to that in P21 and P70 animals at 2 hours postinjection. These data suggest that LPS-induced transcriptional response of cytokines and chemokines was much reduced in P1 lung even though the protein levels of these genes were not ascertained and mRNA levels of these genes may not reflect their final protein levels. MPO activity in LPS-treated P1 lung was also significantly attenuated compared to that in LPS-treated P70 lung, suggesting impaired neutrophil infiltration in P1 lung at 2 hours following LPS treatment. In parallel, the baseline mRNA expression of LPS-binding protein (LBP) in P1 lung was much lower than that in P21 and P70 lungs. While the protein level of LBP was not examined and the mRNA level of LBP may not reflect its final protein level, the reduced transcriptional response of cytokines and chemokines in P1 lung at 2 hours following LPS treatment may be attributed to lower LBP expression in P1 lung as compared to P21 and P70 lungs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA