Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 492
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Behav Brain Res ; 461: 114832, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38142860

RESUMEN

Popular medicine has been using oleoresin from several species of copaíba tree for the treatment of various diseases and its clinical administration potentially causes antinociception. Electrical stimulation of ventrolateral (vlPAG) and dorsolateral (dlPAG) columns of the periaqueductal gray matter also causes antinociception. The aim this study was to verify the antinociceptive effect of oleoresin extracted from Copaifera langsdorffii tree and to test the hypothesis that oleoresin-induced antinociception is mediated by µ1- and κ-opioid receptors in the vlPAG and dlPAG. Nociceptive thresholds were determined by the tail-flick test in Wistar rats. The copaíba tree oleoresin was administered at different doses (50, 100 and 200 mg/kg) through the gavage technique. After the specification of the most effective dose of copaíba tree oleoresin (200 mg/kg), rats were pretreated with either the µ1-opioid receptor selective antagonist naloxonazine (at 0.05, 0.5 and 5 µg/ 0.2 µl in vlPAG, and 5 µg/ 0.2 µl in dlPAG) or the κ-opioid receptor selective antagonist nor-binaltorphimine (at 1, 3 and 9 nmol/ 0.2 µl in vlPAG, and 9 nmol/ 0.2 µl in dlPAG). The blockade of µ1 and κ opioid receptors of vlPAG decreased the antinociception produced by copaíba tree oleoresin. However, the blockade of these receptors in dlPAG did not alter copaíba tree oleoresin-induced antinociception. These data suggest that vlPAG µ1 and κ opioid receptors are critically recruited in the antinociceptive effect produced by oleoresin extracted from Copaifera langsdorffii.


Asunto(s)
Sustancia Gris Periacueductal , Extractos Vegetales , Receptores Opioides kappa , Ratas , Animales , Ratas Wistar , Árboles , Antagonistas de Narcóticos/farmacología , Analgésicos/farmacología , Receptores Opioides mu
2.
Molecules ; 28(21)2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37959790

RESUMEN

Kratom (Mitragyna speciosa Korth. Havil) has been considered a narcotic drug for years, barred by the law in many parts of the world, while extensive research over the past few decades proves its several beneficial effects, some of which are still in ambiguity. In many countries, including Thailand, the indiscriminate use and abuse of kratom have led to the loss of life. Nonetheless, researchers have isolated almost fifty pure compounds from kratom, most of which are alkaloids. The most prevalent compounds, mitragynine and 7-hydroxy mitragynine, are reported to display agonist morphine-like effects on human µ-opioid receptors and antagonists at κ- and δ-opioid receptors with multimodal effects at other central receptors. Mitragynine is also credited to be one of the modulatory molecules for the Keap1-Nrf2 pathway and SOD, CAT, GST, and associated genes' upregulatory cascades, leading it to play a pivotal role in neuroprotective actions while evidently causing neuronal disorders at high doses. Additionally, its anti-inflammatory, antioxidative, antibacterial, and gastroprotective effects are well-cited. In this context, this review focuses on the research gap to resolve ambiguities about the neuronal effects of kratom and demonstrate its prospects as a therapeutic target for neurological disorders associated with other pharmacological effects.


Asunto(s)
Mitragyna , Alcaloides de Triptamina Secologanina , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2 , Morfina , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Receptores Opioides mu , Alcaloides de Triptamina Secologanina/farmacología
3.
J Chem Neuroanat ; 132: 102324, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37557929

RESUMEN

Advances have been made in recent years in using opioid receptor antagonists as an adjunct therapy to psychotropic medication to reduce debilitating weight gain and metabolic adverse effects associated with in particular second generation antipsychotics. However, it is unknown whether second generation antipsychotics produce a change in opioid receptor expression in the brain. The present study investigated early changes in opioid receptor expression in the female rat hypothalamus, a master controller of hunger and metabolic regulation, after acute treatment with olanzapine, a commonly used second generation antipsychotic. Using quantitative spatial in situ hybridization and receptor autoradiography, expression levels of the three opioid receptors; kappa, mu and delta, were determined at mRNA and protein level, respectively, in the five hypothalamic areas: paraventricular nucleus, arcuate nucleus, ventromedial nucleus, dorsomedial nucleus and lateral hypothalamus. After 48 h of olanzapine treatment at clinically relevant plasma concentration weight gain and food intake changes, and increased plasma glucose were observed in female rats. Olanzapine treatment also led to a significant increase in mu opioid receptor availability in the arcuate nucleus, which contains both satiety and hunger controlling neurons. No other areas showed any opioid receptor expressional changes with olanzapine treatment on neither at mRNA nor protein level. Technical difficulties made it impossible to analyze mRNA levels in the lateral hypothalamus and overall binding of delta opioid receptors. Thus, the present study provided insights in to how olanzapine at clinically relevant plasma levels already at an early stage modulated the opioid system in the hypothalamus.


Asunto(s)
Antipsicóticos , Receptores Opioides mu , Ratas , Femenino , Animales , Olanzapina/farmacología , Olanzapina/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Antipsicóticos/farmacología , Hipotálamo/metabolismo , Receptores Opioides/metabolismo , Conducta Alimentaria , Aumento de Peso , ARN Mensajero
4.
Transl Psychiatry ; 13(1): 78, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869037

RESUMEN

Disrupted sleep is a symptom of many psychiatric disorders, including substance use disorders. Most drugs of abuse, including opioids, disrupt sleep. However, the extent and consequence of opioid-induced sleep disturbance, especially during chronic drug exposure, is understudied. We have previously shown that sleep disturbance alters voluntary morphine intake. Here, we examine the effects of acute and chronic morphine exposure on sleep. Using an oral self-administration paradigm, we show that morphine disrupts sleep, most significantly during the dark cycle in chronic morphine, with a concomitant sustained increase in neural activity in the Paraventricular Nucleus of the Thalamus (PVT). Morphine binds primarily to Mu Opioid Receptors (MORs), which are highly expressed in the PVT. Translating Ribosome Affinity Purification (TRAP)-Sequencing of PVT neurons that express MORs showed significant enrichment of the circadian entrainment pathway. To determine whether MOR + cells in the PVT mediate morphine-induced sleep/wake properties, we inhibited these neurons during the dark cycle while mice were self-administering morphine. This inhibition decreased morphine-induced wakefulness but not general wakefulness, indicating that MORs in the PVT contribute to opioid-specific wake alterations. Overall, our results suggest an important role for PVT neurons that express MORs in mediating morphine-induced sleep disturbance.


Asunto(s)
Morfina , Trastornos del Sueño-Vigilia , Animales , Ratones , Analgésicos Opioides , Receptores Opioides mu , Neuronas , Tálamo
5.
Pharmacol Rep ; 75(3): 634-646, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36637684

RESUMEN

BACKGROUND: Strong opioid analgesics such as morphine alleviate moderate to severe acute nociceptive pain (e.g. post-surgical or post-trauma pain) as well as chronic cancer pain. However, they evoke many adverse effects and so there is an unmet need for opioid analgesics with improved tolerability. Recently, a prominent hypothesis has been that opioid-related adverse effects are mediated by ß-arrestin2 recruitment at the µ-opioid (MOP) receptor and this stimulated research on discovery of G-protein biassed opioid analgesics. In other efforts, opioids with MOP agonist and δ-opioid (DOP) receptor antagonist profiles are promising for reducing side effects c.f. morphine. Herein, we report on the in vivo pharmacology of a novel opioid peptide (CYX-5) that is a G-protein biassed MOP receptor agonist, DOP receptor antagonist and kappa opioid (KOP) receptor agonist. METHODS: Male Sprague-Dawley received intracerebroventricular bolus doses of CYX-5 (3, 10, 20 nmol), morphine (100 nmol) or vehicle, and antinociception (tail flick) was assessed relative to constipation (charcoal meal and castor oil-induced diarrhoea tests) and respiratory depression (whole body plethysmography). RESULTS: CYX-5 evoked naloxone-sensitive, moderate antinociception, at the highest dose tested. Although CYX-5 did not inhibit gastrointestinal motility, it reduced stool output markedly in the castor oil-induced diarrhoea test. In contrast to morphine that evoked respiratory depression, CYX-5 increased tidal volume, thereby stimulating respiration. CONCLUSION: Despite its lack of recruitment of ß-arrestin2 at MOP, DOP and KOP receptors, CYX-5 evoked constipation, implicating a mechanism other than ß-arrestin2 recruitment at MOP, DOP and KOP receptors, mediating constipation evoked by CYX-5 and potentially other opioid ligands.


Asunto(s)
Estreñimiento , Morfina , Receptores Opioides delta , Insuficiencia Respiratoria , Animales , Masculino , Ratas , Analgésicos Opioides/efectos adversos , Aceite de Ricino/efectos adversos , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico , Diarrea/tratamiento farmacológico , Proteínas de Unión al GTP , Morfina/efectos adversos , Antagonistas de Narcóticos/farmacología , Ratas Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Insuficiencia Respiratoria/inducido químicamente
6.
Biomolecules ; 12(10)2022 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-36291680

RESUMEN

Receptor transporter protein 4 (RTP4), one of the receptor chaperone proteins, contributes to the maturation and membrane trafficking of opioid receptor heteromers consisting of mu (MOPr) and delta (DOPr) opioid receptors (MOPr-DOPr). Although MOPr-DOPr is known to mediate the development of morphine tolerance, the extent to which RTP4 plays a role in this process has not been elucidated. Given that RTP4 can be upregulated by repeated administration of morphine, especially in the hypothalamus, here we investigated the effect of hypothalamus-selective ablation of RTP4 on the development of antinociceptive tolerance to morphine. In this study, we generated RTP4flox mice and selectively knocked-out RTP4 using local injection of adeno-associated virus expressing Cre recombinase (AAV-Cre) into the hypothalamus. The AAV-Cre injection partially, but significantly, decreased the level of RTP4 expression, and suppressed the development of antinociceptive tolerance to morphine. Next, we examined the mechanism of regulation of RTP4 and found that, in neuronal cells, Rtp4 induction is via Gi and MAPK activation, while, in microglial cells, the induction is via Toll-like receptor 4. Together, these studies highlight the role of MOR activity in regulating RTP4, which, in turn, plays an important role in modulating morphine effects in vivo.


Asunto(s)
Morfina , Receptor Toll-Like 4 , Ratones , Animales , Morfina/farmacología , Receptor Toll-Like 4/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Receptores Opioides/metabolismo , Hipotálamo/metabolismo , Chaperonas Moleculares/metabolismo
7.
J Neurosci ; 42(16): 3316-3328, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35256532

RESUMEN

Opioid tolerance (OT) leads to dose escalation and serious side effects, including opioid-induced hyperalgesia (OIH). We sought to better understand the mechanisms underlying this event in the gastrointestinal tract. Chronic in vivo administration of morphine by intraperitoneal injection in male C57BL/6 mice evoked tolerance and evidence of OIH in an assay of colonic afferent nerve mechanosensitivity; this was inhibited by the δ-opioid receptor (DOPr) antagonist naltrindole when intraperitoneally injected in previous morphine administration. Patch-clamp studies of DRG neurons following overnight incubation with high concentrations of morphine, the µ-opioid receptors (MOPr) agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-Enkephalin (DAMGO) or the DOPr agonist [D-Ala2, D-Leu5]-Enkephalin evoked hyperexcitability. The pronociceptive actions of these opioids were blocked by the DOPr antagonist SDM25N but not the MOPr antagonist D-Pen-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 The hyperexcitability induced by DAMGO was reversed after a 1 h washout, but reapplication of low concentrations of DAMGO or [D-Ala2, D-Leu5]-Enkephalin restored the hyperexcitability, an effect mediated by protein kinase C. DOPr-dependent DRG neuron hyperexcitability was blocked by the endocytosis inhibitor Pitstop 2, and the weakly internalizing DOPr agonist ARM390 did not cause hyperexcitability. Bioluminescence resonance energy transfer studies in HEK cells showed no evidence of switching of G-protein signaling from Gi to a Gs pathway in response to either high concentrations or overnight incubation of opioids. Thus, chronic high-dose opioid exposure leads to opioid tolerance and features of OIH in the colon. This action is mediated by DOPr signaling and is dependent on receptor endocytosis and downstream protein kinase C signaling.SIGNIFICANCE STATEMENT Opioids are effective in the treatment of abdominal pain, but escalating doses can lead to opioid tolerance and potentially opioid-induced hyperalgesia. We found that δ-opioid receptor (DOPr) plays a central role in the development of opioid tolerance and opioid-induced hyperalgesia in colonic afferent nociceptors following prolonged exposure to high concentrations of MOPr or DOPr agonists. Furthermore, the role of DOPr was dependent on OPr internalization and activation of a protein kinase C signaling pathway. Thus, targeting DOPr or key components of the downstream signaling pathway could mitigate adverse side effects by opioids.


Asunto(s)
Analgésicos Opioides , Morfina , Analgésicos Opioides/efectos adversos , Animales , Tolerancia a Medicamentos , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/uso terapéutico , Tracto Gastrointestinal , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/farmacología , Morfina/uso terapéutico , Antagonistas de Narcóticos/farmacología , Proteína Quinasa C , Receptores Opioides , Receptores Opioides mu , Transducción de Señal
8.
Front Immunol ; 12: 689453, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616393

RESUMEN

Evidence concerning the role of alcohol-induced neuroinflammation in alcohol intake and relapse has increased in the last few years. It is also proven that mu-opioid receptors (MORs) mediate the reinforcing properties of alcohol and, interestingly, previous research suggests that neuroinflammation and MORs could be related. Our objective is to study neuroinflammatory states and microglial activation, together with adaptations on MOR expression in the mesocorticolimbic system (MCLS) during the abstinence and relapse phases. To do so, we have used a sex-dependent rat model of complete Freund's adjuvant (CFA)-induced alcohol deprivation effect (ADE). Firstly, our results confirm that only CFA-treated female rats, the only experimental group that showed relapse-like behavior, exhibited specific alterations in the expression of phosphorylated NFκB, iNOS, and COX2 in the PFC and VTA. More interestingly, the analysis of the IBA1 expression revealed a decrease of the microglial activation in PFC during abstinence and an increase of its expression in the relapse phase, together with an augmentation of this activation in the NAc in both phases that only occur in female CFA-treated rats. Additionally, the expression of IL1ß also evidenced these dynamic changes through these two phases following similar expression patterns in both areas. Furthermore, the expression of the cytokine IL10 showed a different profile than that of IL1ß, indicating anti-inflammatory processes occurring only during abstinence in the PFC of CFA-female rats but neither during the reintroduction phase in PFC nor in the NAc. These data indicate a downregulation of microglial activation and pro-inflammatory processes during abstinence in the PFC, whereas an upregulation can be observed in the NAc during abstinence that is maintained during the reintroduction phase only in CFA-female rats. Secondly, our data reveal a correlation between the alterations observed in IL1ß, IBA1 levels, and MOR levels in the PFC and NAc of CFA-treated female rats. Although premature, our data suggest that neuroinflammatory processes, together with neural adaptations involving MOR, might play an important role in alcohol relapse in female rats, so further investigations are warranted.


Asunto(s)
Alcoholismo/metabolismo , Sistema Límbico/metabolismo , Microglía/metabolismo , Neuroinmunomodulación , Dolor/metabolismo , Corteza Prefrontal/metabolismo , Receptores Opioides mu/metabolismo , Abstinencia de Alcohol , Alcoholismo/inmunología , Alcoholismo/fisiopatología , Animales , Proteínas de Unión al Calcio/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Femenino , Adyuvante de Freund , Mediadores de Inflamación/metabolismo , Sistema Límbico/inmunología , Sistema Límbico/fisiopatología , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/inmunología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Dolor/inducido químicamente , Dolor/inmunología , Dolor/fisiopatología , Fosforilación , Corteza Prefrontal/inmunología , Corteza Prefrontal/fisiopatología , Ratas Sprague-Dawley , Recurrencia , Factores Sexuales
9.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34576074

RESUMEN

Masticatory myofascial pain (MMP) is one of the most common causes of chronic orofacial pain in patients with temporomandibular disorders. To explore the antinociceptive effects of ultra-low frequency transcutaneous electrical nerve stimulation (ULF-TENS) on alterations of pain-related biochemicals, electrophysiology and jaw-opening movement in an animal model with MMP, a total of 40 rats were randomly and equally assigned to four groups; i.e., animals with MMP receiving either ULF-TENS or sham treatment, as well as those with sham-MMP receiving either ULF-TENS or sham treatment. MMP was induced by electrically stimulated repetitive tetanic contraction of masticatory muscle for 14 days. ULF-TENS was then performed at myofascial trigger points of masticatory muscles for seven days. Measurable outcomes included maximum jaw-opening distance, prevalence of endplate noise (EPN), and immunohistochemistry for substance P (SP) and µ-opiate receptors (MOR) in parabrachial nucleus and c-Fos in rostral ventromedial medulla. There were significant improvements in maximum jaw-opening distance and EPN prevalence after ULF-TENS in animals with MMP. ULF-TENS also significantly reduced SP overexpression, increased MOR expression in parabrachial nucleus, and increased c-Fos expression in rostral ventromedial medulla. ULF-TENS may represent a novel and applicable therapeutic approach for improvement of orofacial pain induced by MMP.


Asunto(s)
Dolor Crónico/complicaciones , Dolor Crónico/terapia , Trastornos de la Articulación Temporomandibular/complicaciones , Trastornos de la Articulación Temporomandibular/fisiopatología , Estimulación Eléctrica Transcutánea del Nervio , Animales , Dolor Crónico/fisiopatología , Modelos Animales de Enfermedad , Electromiografía , Fenómenos Electrofisiológicos , Músculos Masticadores/fisiopatología , Placa Motora/fisiopatología , Síndromes del Dolor Miofascial/complicaciones , Síndromes del Dolor Miofascial/fisiopatología , Síndromes del Dolor Miofascial/terapia , Núcleos Parabraquiales/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores Opioides mu/metabolismo , Sustancia P/metabolismo
10.
Endocrinology ; 162(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34270714

RESUMEN

Energetic status often affects reproductive function, glucose homeostasis, and feeding in mammals. Malnutrition suppresses pulsatile release of the gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) and increases gluconeogenesis and feeding. The present study aims to examine whether ß-endorphin-µ-opioid receptor (MOR) signaling mediates the suppression of pulsatile GnRH/LH release and an increase in gluconeogenesis/feeding induced by malnutrition. Ovariectomized female rats treated with a negative feedback level of estradiol-17ß (OVX + low E2) receiving 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, intravenously (iv) were used as a malnutrition model. An administration of D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), a selective MOR antagonist, into the third ventricle blocked the suppression of the LH pulse and increase in gluconeogenesis/feeding induced by iv 2DG administration. Histological analysis revealed that arcuate Kiss1 (kisspeptin gene)-expressing cells and preoptic Gnrh1 (GnRH gene)-expressing cells co-expressed little Oprm1 (MOR gene), while around 10% of arcuate Slc17a6 (glutamatergic marker gene)-expressing cells co-expressed Oprm1. Further, the CTOP treatment decreased the number of fos-positive cells in the paraventricular nucleus (PVN) in OVX + low E2 rats treated with iv 2DG but failed to affect the number of arcuate fos-expressing Slc17a6-positive cells. Taken together, these results suggest that the central ß-endorphin-MOR signaling mediates the suppression of pulsatile LH release and that the ß-endorphin may indirectly suppress the arcuate kisspeptin neurons, a master regulator for GnRH/LH pulses during malnutrition. Furthermore, the current study suggests that central ß-endorphin-MOR signaling is also involved in gluconeogenesis and an increase in food intake by directly or indirectly acting on the PVN neurons during malnutrition in female rats.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Antagonistas de Narcóticos/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores Opioides mu/metabolismo , betaendorfina/metabolismo , Animales , Glucemia/análisis , Femenino , Gluconeogénesis , Hipotálamo , Kisspeptinas/metabolismo , Ratas , Ratas Wistar , Receptores Opioides mu/biosíntesis , Transducción de Señal , Proteína 2 de Transporte Vesicular de Glutamato/biosíntesis
11.
Brain Struct Funct ; 226(7): 2431-2458, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34318365

RESUMEN

The lateral hypothalamus is a major integrative hub with a complex architecture characterized by intricate and overlapping cellular populations expressing a large variety of neuro-mediators. In rats, the subfornical lateral hypothalamus (LHsf) was identified as a discrete area with very specific outputs, receiving a strong input from the nucleus incertus, and involved in defensive and foraging behaviors. We identified in the mouse lateral hypothalamus a discrete subfornical region where a conspicuous cluster of neurons express the mu opioid receptor. We thus examined the inputs and outputs of this LHsf region in mice using retrograde tracing with the cholera toxin B subunit and anterograde tracing with biotin dextran amine, respectively. We identified a connectivity profile largely similar, although not identical, to what has been described in rats. Indeed, the mouse LHsf has strong reciprocal connections with the lateral septum, the ventromedial hypothalamic nucleus and the dorsal pre-mammillary nucleus, in addition to a dense output to the lateral habenula. However, the light input from the nucleus incertus and the moderate bidirectional connectivity with nucleus accumbens are specific to the mouse LHsf. A preliminary neurochemical study showed that LHsf neurons expressing mu opioid receptors also co-express calcitonin gene-related peptide or somatostatin and that the reciprocal connection between the LHsf and the lateral septum may be functionally modulated by enkephalins acting on mu opioid receptors. These results suggest that the mouse LHsf may be hodologically and functionally comparable to its rat counterpart, but more atypical connections also suggest a role in consummatory behaviors.


Asunto(s)
Área Hipotalámica Lateral , Animales , Habénula , Hipotálamo , Ratones , Vías Nerviosas , Neuronas , Núcleos del Rafe , Receptores Opioides mu
12.
Nat Commun ; 12(1): 3858, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158473

RESUMEN

Mitragynine (MG) is the most abundant alkaloid component of the psychoactive plant material "kratom", which according to numerous anecdotal reports shows efficacy in self-medication for pain syndromes, depression, anxiety, and substance use disorders. We have developed a synthetic method for selective functionalization of the unexplored C11 position of the MG scaffold (C6 position in indole numbering) via the use of an indole-ethylene glycol adduct and subsequent iridium-catalyzed borylation. Through this work we discover that C11 represents a key locant for fine-tuning opioid receptor signaling efficacy. 7-Hydroxymitragynine (7OH), the parent compound with low efficacy on par with buprenorphine, is transformed to an even lower efficacy agonist by introducing a fluorine substituent in this position (11-F-7OH), as demonstrated in vitro at both mouse and human mu opioid receptors (mMOR/hMOR) and in vivo in mouse analgesia tests. Low efficacy opioid agonists are of high interest as candidates for generating safer opioid medications with mitigated adverse effects.


Asunto(s)
Mitragyna/química , Extractos Vegetales/farmacología , Receptores Opioides mu/agonistas , Alcaloides de Triptamina Secologanina/farmacología , Analgésicos/química , Analgésicos/farmacología , Animales , Glicol de Etileno/química , Humanos , Ratones Noqueados , Modelos Químicos , Estructura Molecular , Extractos Vegetales/química , Unión Proteica , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina Secologanina/química
13.
Alcohol ; 95: 1-6, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34022405

RESUMEN

Clinical studies have postulated that ß-endorphin deficiency generates excessive alcohol consumption, and it has been shown that the reduction of ß-endorphin neurons increases alcohol intake in animal models. The ß-endorphin produce their rewarding effect when they act mainly on the µ-opioid receptors (MOR) located in mesolimbic structures. Thus, it is possible that individual differences in these components of the endogenous opioid system are related to different levels of alcohol consumption. The present study thus examines the relation between two levels of alcohol consumption and intrinsic characteristics of the components of the opioid system in outbred Wistar rats that were not genetically selected. We analyzed the number of ß-endorphin-positive neurons in the arcuate nucleus (ArN) and the expression of µ-opioid receptors (MOR) in regions of the reward system, such as the nucleus accumbens (NAc), amygdala (Amy), and ventral tegmental area (VTA) in outbred rats with low (LC) or high (HC) voluntary alcohol consumption. Findings showed that the HC rats had a lower number of ß-endorphin-positive neurons in the hypothalamic ArN and a higher expression of MOR in the NAc and VTA, compared to the LC rats. No changes in the expression of MOR in the Amy were observed between the two groups. Results suggest that intrinsic variability in the number of ß-endorphin neurons and in the expression of MOR in the LC and HC rats could explain their different patterns for alcohol intake.


Asunto(s)
Endorfinas , Consumo de Bebidas Alcohólicas , Animales , Hipotálamo/metabolismo , Neuronas/metabolismo , Ratas , Ratas Wistar , Receptores Opioides mu/genética
14.
Physiol Rep ; 9(5): e14788, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33661571

RESUMEN

Anorexia nervosa (AN) has a lifetime prevalence of up to 4% and a high mortality rate (~5-10%), yet little is known regarding the etiology of this disease. In an attempt to fill the gaps in knowledge, activity-based anorexia (ABA) in rodents has been a widely used model as it mimics several key features of AN including severely restricted food intake and excessive exercise. Using this model, a role for the hypothalamic proopiomelanocortin (POMC) system has been implicated in the development of ABA as Pomc mRNA is elevated in female rats undergoing the ABA paradigm. Since the Pomc gene product α-MSH potently inhibits food intake, it could be that elevated α-MSH might promote ABA. However, the α-MSH receptor antagonist SHU9119 does not protect against the development of ABA. Interestingly, it has also been shown that female mice lacking the mu opioid receptor (MOR), the primary receptor activated by the Pomc-gene-derived opioid ß-endorphin, display blunted food anticipatory behavior (FAA), a key feature of ABA. Thus, we hypothesized that the elevation in Pomc mRNA observed during ABA may lead to increased ß-endorphin concentrations and MOR activation to promote ABA. Further, given the known sex differences in AN and ABA, we hypothesized that MORs may contribute differentially in male and female mice. Using wild-type and MOR knockout mice of both sexes, a MOR antagonist and careful analysis of food anticipatory behavior and ß-endorphin levels, we found 1) increased Pomc mRNA levels in both female and male mice that underwent ABA, 2) increased ß-endorphin in female mice that underwent ABA, and 3) blunted FAA in both sexes in response to MOR genetic deletion yet blunted FAA only in males in response to MOR antagonism. The results presented provide support for both hypotheses and suggest that it may be the ß-endorphin resulting from increased Pomc transcription that supports the development of some features of ABA.


Asunto(s)
Anorexia/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , betaendorfina/farmacología , Animales , Anorexia/metabolismo , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Proopiomelanocortina/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
15.
Mol Pain ; 17: 1744806921992628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33599155

RESUMEN

Electroconvulsive therapy (ECT) has been applied for chronic pain for decades. The amounts of opioids to treat pain are sometimes reduced after a series of ECT. The effect of ECT on morphine-induced analgesia and its mechanism underlying the reduction of morphine requirement has yet to be clarified. Therefore, we administered electroconvulsive shocks (ECS) to mice and investigated the antinociceptive effect of morphine in a hot plate test. We examined the expression level of µ-opioid receptor in the thalami of mice 25 h after administration of ECS compared to the thalami of mice without ECS administration using western blotting. ECS disturbed the development of a decrease in the percentage of maximal possible effect (%MPE), which was observed 24 h after a morphine injection, when ECS was applied 25, 23, 21, and 12 h before the second administration of morphine. We also examined the effect of ECS on the dose-response curve of %MPE to morphine-antinociception. Twenty-five hours after ECS, the dose-response curve was shifted to the left, and the EC50 of morphine given to ECS-pretreated mice decreased by 30.1% compared to the mice that were not pretreated with ECS. We also found that the expression level of µ-opioid receptors was significantly increased after ECS administration. These results confirm previous clinical reports showing that ECT decreased the required dose of opioids in neuropathic pain patients and suggest the hypothesis that this effect of ECT works through the thalamus.


Asunto(s)
Electrochoque , Morfina/farmacología , Nocicepción/fisiología , Animales , Masculino , Ratones Endogámicos C57BL , Nocicepción/efectos de los fármacos , Receptores Opioides mu/metabolismo , Tálamo/efectos de los fármacos , Tálamo/metabolismo
16.
Biol Pharm Bull ; 44(2): 159-161, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33518669

RESUMEN

The agonistic activity of fluorinated and nonfluorinated fentanyl analogs on µ-opioid receptor was investigated using a cell-based assay system. Based on the activity, fentanyl analogs were ranked as follows: fentanyl > isobutyrylfentanyl ≈ butyrylfentanyl ≈ methoxyacetylfentanyl > acetylfentanyl. However, among the fentanyl analogs fluorinated on the N-phenyl ring, 2-fluoro analogs and 3-fluoro analogs showed the strongest and weakest activities, respectively. These results suggest that the 2-fluorinated isomers of fentanyl analogs are more likely to cause poisoning.


Asunto(s)
Analgésicos Opioides/farmacología , Fentanilo/farmacología , Receptores Opioides mu/agonistas , Animales , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Fentanilo/análogos & derivados
17.
J Ethnopharmacol ; 270: 113872, 2021 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-33485984

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Mu opioid receptor (MOR) is mainly a drug target for analgesia. Opioid-like agonists such as morphine have been clinically used for analgesia but have potential adverse effects. MOR antagonists have been demonstrated to alleviate these side effects. Plants (Carthamus tinctorius L, Cynanchum otophyllum C. K. Schneid., Coffea arabica L., Prinsepia utilis Royle and Lepidium meyenii Walp.) and Ganoderma fungi (Ganoderma hainanense J. D. Zhao, Ganoderma capense (Lloyd) Teng, Ganoderma cochlear (Blume et Nees) Bres., Ganoderma resinaceum Boud and Ganoderma applanatum (Pers.) Pat.) are traditional medicines with beneficial effects on immunoregulation, analgesia and the nervous system, but whether MORs are engaged in their effects remains unknown. AIM OF THE STUDY: This work aimed to identify MOR ligands among compounds isolated from the above-mentioned 10 species, and to investigate selectivity against four opioid receptor subtypes. By analyzing the structure-activity relationship and off-target effects, we could provide a new direction for the future development of MOR drugs. MATERIALS AND METHODS: Four opioid receptor subtype models, including MOR, delta (DOR), kappa (KOR) and nop (NOR), were established with a label-free phenotypic dynamic mass redistribution assay to systematically profile the pharmacological properties of known ligands. Then, 82 natural compounds derived from the 10 species were screened against MOR to identify new ligands. The selectivity of the new ligands was characterized against the four subtypes, and off-target effects were also investigated on eight G protein-coupled receptors (GPCRs). RESULTS: The pharmacological properties of known ligands on transfected HEK293T-MOR, HEK293-DOR, HEK293-KOR and HEK293-NOR cell lines were characterized. Seven compounds purified from Ganoderma cochlear (Blume et Nees) Bres. and Carthamus tinctorius L were MOR antagonists with micromolar potency. Among them, compound 35 showed the strongest antagonistic activity on MOR with an IC50 value of 10.0 ± 3.0 µM. To a certain extent, these seven new antagonists, exhibited antagonistic activity on the other opioid receptor subtypes, and they had almost no effect on other GPCRs, including CB1, CB2, M2 and beta2AR. Additionally, a compound from Lepidium meyenii Walp. displayed MOR agonistic activity. CONCLUSIONS: The established screening models opened new avenues for the discovery and evaluation of opioid receptor ligand selectivity. Together, the novel MOR antagonists and agonists will enrich the inventory of MOR ligands and benefit related therapies.


Asunto(s)
Productos Biológicos/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides/metabolismo , Productos Biológicos/química , Técnicas Biosensibles/métodos , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Humanos , Ligandos , Receptores Opioides/agonistas , Receptores Opioides/genética , Relación Estructura-Actividad
18.
Cell Mol Neurobiol ; 41(5): 1131-1143, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33433723

RESUMEN

Chronic administration of opioids produces physical dependence and opioid-induced hyperalgesia. Users claim the Thai traditional tea "kratom" and component alkaloid mitragynine ameliorate opioid withdrawal without increased sensitivity to pain. Testing these claims, we assessed the combined kratom alkaloid extract (KAE) and two individual alkaloids, mitragynine (MG) and the analog mitragynine pseudoindoxyl (MP), evaluating their ability to produce physical dependence and induce hyperalgesia after chronic administration, and as treatments for withdrawal in morphine-dependent subjects. C57BL/6J mice (n = 10/drug) were administered repeated saline, or graded, escalating doses of morphine (intraperitoneal; i.p.), kratom alkaloid extract (orally, p.o.), mitragynine (p.o.), or MP (subcutaneously, s.c.) for 5 days. Mice treated chronically with morphine, KAE, or mitragynine demonstrated significant drug-induced hyperalgesia by day 5 in a 48 °C warm-water tail-withdrawal test. Mice were then administered naloxone (10 mg/kg, s.c.) and tested for opioid withdrawal signs. Kratom alkaloid extract and the two individual alkaloids demonstrated significantly fewer naloxone-precipitated withdrawal signs than morphine-treated mice. Additional C57BL/6J mice made physically dependent on morphine were then used to test the therapeutic potential of combined KAE, mitragynine, or MP given twice daily over the next 3 days at either a fixed dose or in graded, tapering descending doses. When administered naloxone, mice treated with KAE, mitragynine, or MP under either regimen demonstrated significantly fewer signs of precipitated withdrawal than control mice that continued to receive morphine. In conclusion, while retaining some liabilities, kratom, mitragynine, and mitragynine pseudoindoxyl produced significantly less physical dependence and ameliorated precipitated withdrawal in morphine-dependent animals, suggesting some clinical value.


Asunto(s)
Analgésicos Opioides/efectos adversos , Mitragyna , Dependencia de Morfina/prevención & control , Alcaloides de Triptamina Secologanina/administración & dosificación , Alcaloides de Triptamina Secologanina/síntesis química , Síndrome de Abstinencia a Sustancias/prevención & control , Analgésicos Opioides/administración & dosificación , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Dependencia de Morfina/metabolismo , Dependencia de Morfina/psicología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Alcaloides de Triptamina Secologanina/efectos adversos , Alcaloides de Triptamina Secologanina/aislamiento & purificación , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/psicología
19.
J Nat Prod ; 84(1): 71-80, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33326237

RESUMEN

The seeds of the akuamma tree (Picralima nitida) have been used as a traditional treatment for pain and fever. Previous studies have attributed these effects to a series of indole alkaloids found within the seed extracts; however, these pharmacological studies were significantly limited in scope. Herein, an isolation protocol employing pH-zone-refining countercurrent chromatography was developed to provide six of the akuamma alkaloids in high purity and quantities sufficient for more extensive biological evaluation. Five of these alkaloids, akuammine (1), pseudo-akuammigine (3), akuammicine (4), akuammiline (5), and picraline (6), were evaluated against a panel of >40 central nervous system receptors to identify that their primary targets are the opioid receptors. Detailed in vitro investigations revealed 4 to be a potent kappa opioid receptor agonist, and three alkaloids (1-3) were shown to have micromolar activity at the mu opioid receptor. The mu opioid receptor agonists were further evaluated for analgesic properties but demonstrated limited efficacy in assays of thermal nociception. These findings contradict previous reports of the antinociceptive properties of the P. nitida alkaloids and the traditional use of akuamma seeds as analgesics. Nevertheless, their opioid-preferring activity does suggest the akuamma alkaloids provide distinct scaffolds from which novel opioids with unique pharmacologic properties and therapeutic utility can be developed.


Asunto(s)
Alcaloides/farmacología , Analgésicos/uso terapéutico , Apocynaceae/química , Indoles/farmacología , Receptores Opioides mu/uso terapéutico , Terpenos/farmacología , Alcaloides/química , Alcaloides/aislamiento & purificación , Analgésicos/química , Animales , Indoles/química , Indoles/aislamiento & purificación , Receptores Opioides kappa , Receptores Opioides mu/agonistas , Receptores Opioides mu/análisis , Alcaloides de Triptamina Secologanina/química , Semillas/química , Terpenos/química , Terpenos/aislamiento & purificación
20.
Cell Mol Neurobiol ; 41(5): 949-960, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32107752

RESUMEN

OBJECTIVE: Pharmacological evaluation of the mu-opioid receptor (MOR) agonist properties of NKTR-181 in rodent models. METHODS: Graded noxious stimulus intensities were used in rats to establish the antinociceptive potency and efficacy of NKTR-181 relative to morphine, fentanyl, and oxycodone. Characteristics of MOR agonist actions, as measured by antinociceptive tolerance and cross-tolerance, as well as opioid-induced hyperalgesia (OIH) and naloxone-precipitated withdrawal in NKTR-181- and morphine-dependent in mice, were compared. RESULTS: NKTR-181 showed dose- and time-related antinociception with similar maximal effects to morphine in the rat and mouse hot-water tail-flick test. No sex or species differences were observed in NKTR-181 or morphine antinociception. Rats treated with NKTR-181 and morphine exhibited decreases in both potency and maximal efficacy as nociceptive stimulus intensity was increased from a water temperature of 50 °C to 54 °C. Evaluation of antinociception at a high stimulus intensity revealed that oxycodone and fentanyl exhibited greater efficacy than either NKTR-181 or morphine. The relative potency difference between NKTR-181 and morphine across all tail-flick studies was determined to be 7.6-fold (90% confidence interval, 2.6, 21.5). The peak antinociceptive effect of NKTR-181 was delayed compared to that of the other opioids and cumulative drug effects were not observed. Repeated treatment with escalating, approximately equi-analgesic doses of NKTR-181 or morphine, produced antinociceptive tolerance and cross-tolerance. Under these pharmacological conditions, OIH and naloxone-precipitated physical dependence were similar for NKTR-181 and morphine. CONCLUSIONS: NKTR-181 had a slower onset, but similar efficacy, to morphine in the models studied supporting reduced abuse potential while maintaining analgesic effect in comparison with current opioids.


Asunto(s)
Analgésicos Opioides/farmacología , Morfinanos/farmacología , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Masculino , Ratones , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/fisiología , Roedores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA