Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Molecules ; 28(23)2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-38067494

RESUMEN

Opioid receptor agonists, particularly those that activate µ-opioid receptors (MORs), are essential analgesic agents for acute or chronic mild to severe pain treatment. However, their use has raised concerns including, among others, intestinal dysbiosis. In addition, growing data on constipation-evoked intestinal dysbiosis have been reported. Opioid-induced constipation (OIC) creates an obstacle to continuing treatment with opioid analgesics. When non-opioid therapies fail to overcome the OIC, opioid antagonists with peripheral, fast first-pass metabolism, and gastrointestinal localized effects remain the drug of choice for OIC, which are discussed here. At first glance, their use seems to only be restricted to constipation, however, recent data on OIC-related dysbiosis and its contribution to the appearance of several opioid side effects has garnered a great of attention from researchers. Peripheral MORs have also been considered as a future target for opioid analgesics with limited central side effects. The properties of MOR antagonists counteracting OIC, and with limited influence on central and possibly peripheral MOR-mediated antinociception, will be highlighted. A new concept is also proposed for developing gut-selective MOR antagonists to treat or restore OIC while keeping peripheral antinociception unaffected. The impact of opioid antagonists on OIC in relation to changes in the gut microbiome is included.


Asunto(s)
Antagonistas de Narcóticos , Estreñimiento Inducido por Opioides , Humanos , Antagonistas de Narcóticos/uso terapéutico , Analgésicos Opioides/efectos adversos , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico , Estreñimiento/metabolismo , Estreñimiento Inducido por Opioides/tratamiento farmacológico , Disbiosis/inducido químicamente , Disbiosis/tratamiento farmacológico , Receptores Opioides/metabolismo
2.
Molecules ; 28(12)2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37375318

RESUMEN

Opioids are considered the most effective analgesics for the treatment of moderate to severe acute and chronic pain. However, the inadequate benefit/risk ratio of currently available opioids, together with the current 'opioid crisis', warrant consideration on new opioid analgesic discovery strategies. Targeting peripheral opioid receptors as effective means of treating pain and avoiding the centrally mediated side effects represents a research area of substantial and continuous attention. Among clinically used analgesics, opioids from the class of morphinans (i.e., morphine and structurally related analogues) are of utmost clinical importance as analgesic drugs activating the mu-opioid receptor. In this review, we focus on peripheralization strategies applied to N-methylmorphinans to limit their ability to cross the blood-brain barrier, thus minimizing central exposure and the associated undesired side effects. Chemical modifications to the morphinan scaffold to increase hydrophilicity of known and new opioids, and nanocarrier-based approaches to selectively deliver opioids, such as morphine, to the peripheral tissue are discussed. The preclinical and clinical research activities have allowed for the characterization of a variety of compounds that show low central nervous system penetration, and therefore an improved side effect profile, yet maintaining the desired opioid-related antinociceptive activity. Such peripheral opioid analgesics may represent alternatives to presently available drugs for an efficient and safer pain therapy.


Asunto(s)
Analgésicos Opioides , Morfinanos , Humanos , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/química , Dolor/tratamiento farmacológico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Morfina/farmacología , Morfina/uso terapéutico , Receptores Opioides mu
3.
Molecules ; 26(20)2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34684749

RESUMEN

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


Asunto(s)
Dolor Crónico/tratamiento farmacológico , Receptores de Angiotensina/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Humanos , Neuralgia/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Manejo del Dolor/métodos , Proto-Oncogenes Mas , Receptores de Angiotensina/metabolismo , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
4.
Molecules ; 25(11)2020 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-32466522

RESUMEN

There is growing evidence on the role of peripheral µ-opioid receptors (MORs) in analgesia and analgesic tolerance. Opioid analgesics are the mainstay in the management of moderate to severe pain, and their efficacy in the alleviation of pain is well recognized. Unfortunately, chronic treatment with opioid analgesics induces central analgesic tolerance, thus limiting their clinical usefulness. Numerous molecular mechanisms, including receptor desensitization, G-protein decoupling, ß-arrestin recruitment, and alterations in the expression of peripheral MORs and microbiota have been postulated to contribute to the development of opioid analgesic tolerance. However, these studies are largely focused on central opioid analgesia and tolerance. Accumulated literature supports that peripheral MORs mediate analgesia, but controversial results on the development of peripheral opioid receptors-mediated analgesic tolerance are reported. In this review, we offer evidence on the consequence of the activation of peripheral MORs in analgesia and analgesic tolerance, as well as approaches that enhance analgesic efficacy and decrease the development of tolerance to opioids at the peripheral sites. We have also addressed the advantages and drawbacks of the activation of peripheral MORs on the sensory neurons and gut (leading to dysbiosis) on the development of central and peripheral analgesic tolerance.


Asunto(s)
Analgesia , Receptores Opioides mu/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Humanos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Manejo del Dolor/métodos
5.
Molecules ; 25(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192229

RESUMEN

The present work represents the in vitro (potency, affinity, efficacy) and in vivo (antinociception, constipation) opioid pharmacology of the novel compound 14-methoxycodeine-6-O-sulfate (14-OMeC6SU), compared to the reference compounds codeine-6-O-sulfate (C6SU), codeine and morphine. Based on in vitro tests (mouse and rat vas deferens, receptor binding and [35S]GTPγS activation assays), 14-OMeC6SU has µ-opioid receptor-mediated activity, displaying higher affinity, potency and efficacy than the parent compounds. In rats, 14-OMeC6SU showed stronger antinociceptive effect in the tail-flick assay than codeine and was equipotent to morphine, whereas C6SU was less efficacious after subcutaneous (s.c.) administration. Following intracerebroventricular injection, 14-OMeC6SU was more potent than morphine. In the Complete Freund's Adjuvant-induced inflammatory hyperalgesia, 14-OMeC6SU and C6SU in s.c. doses up to 6.1 and 13.2 µmol/kg, respectively, showed peripheral antihyperalgesic effect, because co-administered naloxone methiodide, a peripherally acting opioid receptor antagonist antagonized the measured antihyperalgesia. In addition, s.c. C6SU showed less pronounced inhibitory effect on the gastrointestinal transit than 14-OMeC6SU, codeine and morphine. This study provides first evidence that 14-OMeC6SU is more effective than codeine or C6SU in vitro and in vivo. Furthermore, despite C6SU peripheral antihyperalgesic effects with less gastrointestinal side effects the superiority of 14-OMeC6SU was obvious throughout the present study.


Asunto(s)
Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacología , Codeína/síntesis química , Codeína/farmacología , Analgésicos Opioides/química , Analgésicos Opioides/uso terapéutico , Animales , Unión Competitiva , Codeína/química , Codeína/uso terapéutico , Adyuvante de Freund , Tránsito Gastrointestinal/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inyecciones Intraventriculares , Masculino , Ratones , Naloxona/farmacología , Naloxona/uso terapéutico , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Ratas Wistar , Receptores Opioides mu/metabolismo
7.
Front Pharmacol ; 10: 347, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024314

RESUMEN

Reduction of the opioid analgesia in diabetic neuropathic pain (DNP) results from µ-opioid receptor (MOR) reserve reduction. Herein, we examined the antinociceptive and antiallodynic actions of a novel opioid agonist 14-O-methymorphine-6-O-sulfate (14-O-MeM6SU), fentanyl and morphine in rats with streptozocin-evoked DNP of 9-12 weeks following their systemic administration. The antinociceptive dose-response curve of morphine but not of 14-O-MeM6SU or fentanyl showed a significant right-shift in diabetic compared to non-diabetic rats. Only 14-O-MeM6SU produced antiallodynic effects in doses matching antinociceptive doses obtained in non-diabetic rats. Co-administered naloxone methiodide (NAL-M), a peripherally acting opioid receptor antagonist failed to alter the antiallodynic effect of test compounds, indicating the contribution of central opioid receptors. Reduction in spinal MOR binding sites and loss in MOR immunoreactivity of nerve terminals in the spinal cord and dorsal root ganglia in diabetic rats were observed. G-protein coupling assay revealed low efficacy character for morphine and high efficacy character for 14-O-MeM6SU or fentanyl at spinal or supraspinal levels (E max values). Furthermore, at the spinal level only 14-O-MeM6SU showed equal efficacy in G-protein activation in tissues of diabetic- and non-diabetic animals. Altogether, the reduction of spinal opioid receptors concomitant with reduced analgesic effect of morphine may be circumvented by using high efficacy opioids, which provide superior analgesia over morphine. In conclusion, the reduction in the analgesic action of opioids in DNP might be a consequence of MOR reduction, particularly in the spinal cord. Therefore, developing opioids of high efficacy might provide analgesia exceeding that of currently available opioids.

8.
Neurochem Res ; 43(6): 1250-1257, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29725918

RESUMEN

Opioid analgesics devoid of central side effects are unmet medical need in the treatment of acute pain (e.g. post-operative pain). Recently, we have reported on 14-O-methylmorphine-6-O-sulfate (14-O-MeM6SU), a novel opioid agonist of high efficacy producing peripheral antinociception in subchronic inflammatory pain in certain doses. The present study focused on the antinociceptive effect of 14-O-MeM6SU compared to morphine in formalin test of an early/acute (Phase I) and late/tonic (Phase II) pain phases. Subcutaneous 14-O-MeM6SU (253-1012 nmol/kg) and morphine (3884-31075 nmol/kg) dose dependently reduced the pain behaviors of both phases. Co-administered naloxone methiodide (NAL-M), a peripherally acting opioid antagonist, abolished the antinociceptive effect of 506 nmol/kg 14-O-MeM6SU. On the other hand, the effects of 14-O-MeM6SU (1012 nmol/kg) and morphine (15538 nmol/kg) were only partially affected by NAL-M, indicating the contribution of CNS to antinociception. Locally injected test compounds into formalin treated paws caused antinociception in both phases. Locally effective doses of test compounds were also injected into contralateral paws. Morphine showed effects in both phases, 14-O-MeM6SU in certain doses failed to produce antinociception in either phase. A NAL-M reversible systemic dose of 14-O-MeM6SU and the lowest systemic effective dose of morphine were evaluated for their sedative effects following isoflurane-induced sleeping (righting reflex). In contrast to morphine, 14-O-MeM6SU in certain antinociceptive doses showed no impact on sleeping time. These data highlight that high efficacy opioids of limited CNS penetration in certain doses mitigate somatic and inflammatory pain by targeting MOR at the periphery.


Asunto(s)
Dolor Agudo/tratamiento farmacológico , Analgésicos Opioides/administración & dosificación , Analgésicos/administración & dosificación , Codeína/análogos & derivados , Dimensión del Dolor/efectos de los fármacos , Dolor Agudo/metabolismo , Dolor Agudo/psicología , Analgésicos/química , Analgésicos Opioides/química , Animales , Codeína/administración & dosificación , Codeína/química , Relación Dosis-Respuesta a Droga , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inyecciones Subcutáneas , Masculino , Dimensión del Dolor/métodos , Ratas , Ratas Wistar
9.
Eur J Pharmacol ; 814: 264-273, 2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-28864212

RESUMEN

14-O-methyl (14-O-Me) group in morphine-6-O-sulfate (M6SU) or oxymorphone has been reported to be essential for enhanced affinity, potency and antinociceptive effect of these opioids. Herein we report on the pharmacological properties (potency, affinity and efficacy) of the new compound, 14-O-methylmorphine (14-O-MeM) in in vitro. Additionally, we also investigated the antinociceptive effect of the novel compound, as well as its inhibitory action on gastrointestinal transit in in vivo. The potency and efficacy of test compound were measured by [35S]GTPγS binding, isolated mouse vas deferens (MVD) and rat vas deferens (RVD) assays. The affinity of 14-O-MeM for opioid receptors was assessed by radioligand binding and MVD assays. The antinociceptive and gastrointestinal effects of the novel compound were evaluated in the rat tail-flick test and charcoal meal test, respectively. Morphine, DAMGO, Ile5,6 deltorphin II, deltorphin II and U-69593 were used as reference compounds. 14-O-MeM showed higher efficacy (Emax) and potency (EC50) than morphine in MVD, RVD or [35S]GTPγS binding. In addition, 14-O-MeM compared to morphine showed higher affinity for µ-opioid receptor (MOR). In vivo, in rat tail-flick test 14-O-MeM proved to be stronger antinociceptive agent than morphine after peripheral or central administration. Additionally, both compounds inhibited the gastrointestinal peristalsis. However, when the antinociceptive and antitransit doses for each test compound are compared, 14-O-MeM proved to have slightly more favorable pharmacological profile. Our results affirm that 14-O-MeM, an opioid of high efficacy and affinity for MOR can be considered as a novel analgesic agent of potential clinical value.


Asunto(s)
Analgésicos/metabolismo , Analgésicos/farmacología , Codeína/metabolismo , Codeína/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Animales , Unión Competitiva , Tránsito Gastrointestinal/efectos de los fármacos , Concentración 50 Inhibidora , Masculino , Ratones , Ratas , Especificidad por Sustrato , Conducto Deferente/efectos de los fármacos , Conducto Deferente/metabolismo
10.
Eur J Pharmacol ; 809: 111-121, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28502630

RESUMEN

Opioid antagonists, naloxone and naltrexone have long been used in clinical practice and research. In addition to their low selectivity, they easily pass through the blood-brain barrier. Quaternization of the amine group in these molecules, (e.g. methylnaltrexone) results in negligible CNS penetration. In addition, zwitterionic compounds have been reported to have limited CNS access. The current study, for the first time gives report on the synthesis and the in vitro [competition binding, G-protein activation, isolated mouse vas deferens (MVD) and mouse colon assay] pharmacology of the zwitterionic compound, naltrexone-14-O-sulfate. Naltrexone, naloxone, and its 14-O-sulfate analogue were used as reference compounds. In competition binding assays, naltrexone-14-O-sulfate showed lower affinity for µ, δ or κ opioid receptor than the parent molecule, naltrexone. However, the µ/κ opioid receptor selectivity ratio significantly improved, indicating better selectivity. Similar tendency was observed for naloxone-14-O-sulfate when compared to naloxone. Naltrexone-14-O-sulfate failed to activate [35S]GTPγS-binding but inhibit the activation evoked by opioid agonists (DAMGO, Ile5,6deltorphin II and U69593), similarly to the reference compounds. Schild plot constructed in MVD revealed that naltrexone-14-O-sulfate acts as a competitive antagonist. In mouse colon, naltrexone-14-O-sulfate antagonized the inhibitory effect of morphine with lower affinity compared to naltrexone and higher affinity when compared to naloxone or naloxone-14-O-sulfate. In vivo (mouse tail-flick test), subcutaneously injected naltrexone-14-O-sulfate antagonized morphine's antinociception in a dose-dependent manner, indicating it's CNS penetration, which was unexpected from such zwitter ionic structure. Future studies are needed to evaluate it's pharmacokinetic profile.


Asunto(s)
Analgésicos/síntesis química , Analgésicos/farmacología , Naltrexona/síntesis química , Naltrexona/farmacología , Antagonistas de Narcóticos/síntesis química , Antagonistas de Narcóticos/farmacología , Receptores Opioides/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Técnicas de Química Sintética , Colon/efectos de los fármacos , Colon/metabolismo , Masculino , Ratones , Morfina/farmacología , Naltrexona/análogos & derivados , Ratas , Ratas Wistar , Conducto Deferente/efectos de los fármacos , Conducto Deferente/metabolismo
11.
Neuropharmacology ; 111: 1-13, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27558347

RESUMEN

Glucocorticoids were long believed to primarily function through cytosolic glucocorticoid receptor (GR) activation and subsequent classical genomic pathways. Recently, however, evidence has emerged that suggests the presence of rapid non-genomic GR-dependent signaling pathways within the brain, though their existence in spinal and peripheral nociceptive neurons remains elusive. In this paper, we aim to systemically identify GR within the spinal cord and periphery, to verify their putative membrane location and to characterize possible G protein coupling and pain modulating properties. Double immunofluorescence confocal microscopy revealed that GR predominantly localized in peripheral peptidergic and non-peptidergic nociceptive C- and Aδ-neurons and existed only marginally in myelinated mechanoreceptive and proprioreceptive neurons. Within the spinal cord, GR predominantly localized in incoming presynaptic nociceptive neurons, in pre- and postsynaptic structures of the dorsal horn, as well as in microglia. GR saturation binding revealed that these receptors are linked to the cell membrane of sensory neurons and, upon activation, they trigger membrane targeted [35S]GTPγS binding, indicating G protein coupling to a putative receptor. Importantly, subcutaneous dexamethasone immediately and dose-dependently attenuated acute nociceptive behavior elicited in an animal model of formalin-induced pain hypersensitivity compared to naive rats. Overall, this study provides firm evidence for a novel neuronal mechanism of GR agonists that is rapid, non-genomic, dependent on membrane binding and G protein coupling, and acutely modulates nociceptive behavior, thus unraveling a yet unconsidered mechanism of pain relief.


Asunto(s)
Ganglios Espinales/metabolismo , Proteínas de la Membrana/metabolismo , Nociceptores/metabolismo , Dolor/metabolismo , Receptores de Glucocorticoides/metabolismo , Médula Espinal/metabolismo , Animales , Masculino , Mecanorreceptores , Neuroglía/metabolismo , Nocicepción/fisiología , Dolor/fisiopatología , Umbral del Dolor , Unión Proteica , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/fisiología , Nervio Ciático/metabolismo , Piel/metabolismo
12.
J Pharmacol Exp Ther ; 359(1): 171-81, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27435180

RESUMEN

Growing data support peripheral opioid antinociceptive effects, particularly in inflammatory pain models. Here, we examined the antinociceptive effects of subcutaneously administered, recently synthesized 14-O-methylmorphine-6-O-sulfate (14-O-MeM6SU) compared with morphine-6-O-sulfate (M6SU) in a rat model of inflammatory pain induced by an injection of complete Freund's adjuvant and in a mouse model of visceral pain evoked by acetic acid. Subcutaneous doses of 14-O-MeM6SU and M6SU up to 126 and 547 nmol/kg, respectively, produced significant and subcutaneous or intraplantar naloxone methiodide (NAL-M)-reversible antinociception in inflamed paws compared with noninflamed paws. Neither of these doses significantly affected thiobutabarbital-induced sleeping time or rat pulmonary parameters. However, the antinociceptive effects of higher doses were only partially reversed by NAL-M, indicating contribution of the central nervous system. In the mouse writhing test, 14-O-MeM6SU was more potent than M6SU after subcutaneous or intracerebroventricular injections. Both displayed high subcutaneous/intracerebroventricular ED50 ratios. The antinociceptive effects of subcutaneous 14-O-MeM6SU and M6SU up to 136 and 3043 nmol/kg, respectively, were fully antagonized by subcutaneous NAL-M. In addition, the test compounds inhibited mouse gastrointestinal transit in antinociceptive doses. Taken together, these findings suggest that systemic administration of the novel compound 14-O-MeM6SU similar to M6SU in specific dose ranges shows peripheral antinociception in rat and mouse inflammatory pain models without central adverse effects. These findings apply to male animals and must be confirmed in female animals. Therefore, titration of systemic doses of opioid compounds with limited access to the brain might offer peripheral antinociception of clinical importance.


Asunto(s)
Analgésicos/administración & dosificación , Analgésicos/farmacología , Morfina/administración & dosificación , Morfina/farmacología , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/fisiología , Masculino , Ratones , Morfina/química , Morfina/uso terapéutico , Dolor/tratamiento farmacológico , Ratas , Ratas Wistar , Respiración/efectos de los fármacos , Tiopental/análogos & derivados , Tiopental/farmacología
13.
Brain Res Bull ; 117: 32-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26235542

RESUMEN

The therapeutic use of opioids is limited by the development of tolerance to the analgesic effect and the cellular and molecular mechanisms underlying this phenomenon are still not completely understood. For this reason the search for new analgesic derivatives, endowed with lower tolerance, is always an active field. The newly synthesized 14-O-Methylmorphine-6-sulfate (14-O-MeM6SU) shows high efficacy in in vitro assays and a strong analgesic action in the rat tail flick test. The aim of present work was to investigate: the analgesic effect of 14-O-MeM6SU in mouse tail-flick test; the tolerance to analgesic effect of 14-O-MeM6SU compared to morphine in mice, the effects of test compounds on glutamatergic neurotransmission by measuring spontaneous excitatory postsynaptic currents (sEPSCs) of layer V pyramidal cells from rat prefrontal cortices; and the effect of acute and chronic 14-O-MeM6SU treatments on opioid receptor gene expression in SH-SY5Y neuroblastoma cells expressing µ-opioid (MOP) and nociceptin/opioid receptor-like 1 (NOP) receptors. 14-O-MeM6SU was 17 times more potent than morphine in analgesia and had long duration of action in analgesic dose equipotent to morphine. Mice were treated subcutaneously (s.c.) either with 200 µmol/kg morphine or with 14-O-MeM6SU (12 µmol/kg) twice daily for three days. The magnitude of tolerance or cross-tolerance indicated by the shift in antinociceptive ED50 measured was greater for morphine compared to 14-O-MeM6SU. Subsequent to behavioral testing, patch-clamp experiments in layer V pyramidal neurons of rat prefrontal cortical slices in the presence of bicuculline were performed. Both 14-O-MeM6SU (0.1 µM) and morphine (1 µM) decreased the frequency of sEPSCs, indicating reduction of glutamate release. The effect of the novel compound was reversed by the opioid receptor antagonist naloxone, indicating an opioid mediated action. In contrast, the amplitude was not affected. Finally, gene expression data showed a dose dependent down-regulation of MOP receptor after 24h and 48 h exposure to 14-O-MeM6SU. Interestingly, no changes were detected for NOP receptor gene expression. The specific lack of this effect could be related to the lower tolerance development to analgesic effect of 14-O-MeM6SU. Furthermore, 14-O-MeM6SU displayed high intrinsic efficacy possibly an important factor in the observed effects. Further, the observed inhibition of glutamatergic signaling might be attributed also to the reduction of opioid tolerance. Based on our results the development of a new clinically important, safe analgesic agent might be possible.


Asunto(s)
Analgésicos Opioides/farmacología , Codeína/análogos & derivados , Morfina/farmacología , Analgésicos Opioides/efectos adversos , Animales , Línea Celular Tumoral , Codeína/efectos adversos , Codeína/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Tolerancia a Medicamentos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Morfina/efectos adversos , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dolor Nociceptivo/tratamiento farmacológico , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas Wistar , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos , Receptor de Nociceptina
14.
Neuropharmacology ; 85: 142-50, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24863039

RESUMEN

Painful diabetic neuropathy is a disease of the peripheral sensory neuron with impaired opioid responsiveness. Since µ-opioid receptor (MOR) activation can inhibit the transient receptor potential vanilloid 1 (TRPV1) activity in peripherally sensory neurons, this study investigated the mechanisms of impaired opioid inhibitory effects on capsaicin-induced TRPV1 activity in painful diabetic neuropathy. Intravenous injection of streptozotocin (STZ, 45 mg/kg) in Wistar rats led to a degeneration of insulin producing pancreatic ß-cells, elevated blood glucose, and mechanical hypersensitivity (allodynia). In these animals, local morphine's inhibitory effects on capsaicin-induced nocifensive behavior as well as on capsaicin-induced TRPV1 current in dorsal root ganglion cells were significantly impaired. These changes were associated with a loss in MOR but not TRPV1 in peripheral sensory neurons. Intrathecal delivery of nerve growth factor in diabetic animals normalized sensory neuron MOR and subsequently rescued morphine's inhibitory effects on capsaicin-induced TRPV1 activity in vivo and in vitro. These findings identify a loss in functional MOR on sensory neurons as a contributing factor for the impaired opioid inhibitory effects on capsaicin-induced TRPV1 activity during advanced STZ-induced diabetes. Moreover, they support growing evidence of a distinct regulation of opioid responsiveness during various painful states of disease (e.g. arthritis, cancer, neuropathy) and may give novel therapeutic incentives.


Asunto(s)
Capsaicina/toxicidad , Neuropatías Diabéticas/fisiopatología , Factor de Crecimiento Nervioso/metabolismo , Neuralgia/fisiopatología , Receptores Opioides mu/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Glucemia , Diabetes Mellitus Experimental/fisiopatología , Ganglios Espinales/fisiopatología , Masculino , Potenciales de la Membrana/fisiología , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Médula Espinal/fisiopatología
15.
Eur J Pharmacol ; 713(1-3): 54-7, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23665110

RESUMEN

This study compared the peripheral analgesic effects of a novel opioid agonist 14-O-methylmorphine-6-O-sulfate (14-O-MeM6SU), to that of non-peptide (morphine, fentanyl) and peptide opioid agonists (Met-enkephalin; met-ENK and ß-endorphin; ß-END) in a model of localized inflammatory pain evoked by intraplantar (i.pl.) Freund's complete adjuvant (FCA). Nociceptive responses to local opioid agonists were measured by pressure paw-withdrawal procedures. In addition, the antinociceptive efficacy and potency of these test compounds in vivo was compared to that in vitro using the rat vas deferens (RVD) bioassay. Intraplantar 14-O-MeM6SU (0.32-2.53 nmol/rat), morphine (14.95-112.15 nmol/rat), fentanyl (0.19-2.36 nmol/rat), met-ENK (0.10-10 nmol/rat) and ß-END (0.77-5.00 nmol/rat) dose dependently increased paw pressure thresholds exclusively in inflamed hindpaws. At higher doses analgesic effects were also seen in noninflamed paws for 14-O-MeM6SU, morphine and fentanyl but not for met-ENK or ß-END. The maximal possible local analgesic effect (%) measured in inflamed paws was 50.6 ± 2.7, 18.23 ± 1.78, 37.44 ± 2.17, 36.00 ± 1.43, and 40.69 ± 0.91 for 14-O-MeM6SU, morphine, fentanyl, met-ENK and ß-END, respectively. Interestingly, i.pl. administered opioid peptides met-ENK and ß-END displayed a peripheral analgesic ceiling effect. This local antinociception was antagonized by co-administered opioid antagonist naloxone-methiodide (NAL-M). Similar to the analgesic testing, the RVD showed the following efficacy order of the test compounds: 14-O-MeM6SU>ß-END>fentanyl>met-ENK≫morphine. Taken together, 14-O-MeM6SU was more potent than morphine, fentanyl and met-ENK and ß-END and displayed superiority in the maximum antinociceptive effects. The superiority of local antinociceptive effects of 14-O-MeM6SU might be due to both pharmacodynamic and pharmacokinetic factors.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Codeína/análogos & derivados , Derivados de la Morfina/uso terapéutico , Dolor Nociceptivo/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Receptores Opioides/agonistas , Analgésicos Opioides/química , Animales , Codeína/química , Codeína/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Derivados de la Morfina/administración & dosificación , Derivados de la Morfina/química , Contracción Muscular/efectos de los fármacos , Dolor Nociceptivo/metabolismo , Péptidos Opioides/administración & dosificación , Péptidos Opioides/química , Ratas , Ratas Wistar , Conducto Deferente/efectos de los fármacos , Conducto Deferente/metabolismo
16.
J Pain ; 14(7): 720-30, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23623572

RESUMEN

UNLABELLED: This study investigated putative mechanisms of impaired spinal opioid antinociception such as a downregulation of mu-opioid receptor (MOR) number, coupling, and efficacy in rats with advanced (12 weeks) streptozotocin (STZ)-induced diabetes. Intravenous injection of STZ (45 mg/kg) in Wistar rats led to selective degeneration of insulin-producing pancreatic ß-cells, elevated blood glucose, and mechanical hyperalgesia. In these animals, dose-dependent and naloxone-reversible intrathecal fentanyl antinociception was significantly impaired and associated with a loss in MOR immunoreactivity of calcitonin gene-related peptide-immunoreactive (CGRP-IR) sensory nerve terminals, membrane-bound MOR binding sites, and MOR-stimulated G protein coupling within the dorsal horn of the spinal cord. Intrathecal delivery of nerve growth factor (NGF) in diabetic animals normalized spinal MOR number and G protein coupling and rescued spinal fentanyl-induced antinociception. These findings identify for the first time a loss in functional MOR on central terminals of sensory neurons as a contributing factor for the impaired spinal opioid responsiveness during advanced STZ-induced diabetes that can be reversed by NGF. Moreover, they support growing evidence of a distinct regulation of opioid responsiveness during various painful states of disease (eg, arthritis, cancer, neuropathy) and may give novel therapeutic incentives. PERSPECTIVE: In diabetic neuropathy a loss in sensory neuron mu-opioid receptor number and coupling contributes to impaired spinal opioid antinociception that can be reversed by NGF. These findings support growing evidence of a distinct regulation of opioid responsiveness during various painful diseases and may give novel therapeutic incentives.


Asunto(s)
Neuropatías Diabéticas/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Receptores Opioides mu/metabolismo , Médula Espinal/metabolismo , Animales , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/metabolismo , Inmunohistoquímica , Ratas , Ratas Wistar , Células Receptoras Sensoriales/metabolismo
17.
Curr Pharm Des ; 19(42): 7391-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23448473

RESUMEN

The well-known opioid agonists, oxycodone and oxymorphone, and the opioid antagonists, naloxone and naltrexone, are commonly used clinical agents and research tools in the opioid field. They belong to the class of morphinan-6-ones, and produce their pharmacological effects by interacting with opioid receptors, i.e. mu (MOR), delta (DOR) and kappa (KOR). The search for potent agonists and antagonists has continuously engaged the interest of pharmaceutical research, aiming for the identification of safer therapeutic agents or discovery of opioids with novel therapeutic properties and with lesser unwanted side effects. The chemically highly versatile carbonyl group in position 6 of mophinan-6-ones permits functionalization and modification leading to numerous opioid ligands. We have focused on representative examples of various derivatives and interesting approaches for the development of structurally distinct molecules with substitution at C6 (e.g. 6-methylene, 6-hydroxy, 6-amido, bifunctional ligands), as preclinically and clinically valuable opioids. In this work, the development of 6-amino and 6-guanidino substituted 14-alkoxymorphinans, including the synthesis and pharmacological investigations is presented. The new approach represented by the introduction of amino and guanidino groups into position 6 of the morphinan skeleton of 14-O-methyloxymorphone, led to compounds with high efficacy, MOR affinity and selectivity, which act as potent antinociceptive agents. Altogether, as a consequence of target drug design and synthetic efforts in the field of morphinan-6-ones, we achieve a better understanding of the function of the opioid system, and such efforts may open new avenues for further investigations.


Asunto(s)
Morfinanos/farmacología , Morfinanos/química
18.
Brain Res Bull ; 87(2-3): 238-43, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22079588

RESUMEN

This study describes the antinociceptive effects of µ-opioid agonists, d-Ala(2),N-Me-Phe(4),Gly(5)-ol-enkephalin (DAMGO) and morphine in a model of rat visceral pain in which nociceptive responses were triggered by 2% acetic acid intraperitoneal (i.p.) injections. DAMGO and morphine were administered i.p., to the same site where acetic acid was delivered or intracerebroventricularly (i.c.v.). The antinociceptive actions of i.p. versus i.c.v. administered DAMGO or morphine were evaluated in the late phase of permanent visceral nociceptive responses. Both compounds inhibited the nociceptive responses in a dose-dependent manner and exhibited more potent agonist activity after i.c.v. than i.p. administration. DAMGO and morphine showed comparable ED(50) values after i.p. injections. However, DAMGO was much stronger than morphine after central administration. Co-administration of the peripherally restricted opioid antagonist, naloxone methiodide (NAL-M), significantly attenuated the antinociceptive effects of i.p. DAMGO or morphine. On the other hand, i.c.v. injections of NAL-M partially antagonized the antinociceptive effect of i.p. morphine and failed to affect the antinociceptive action of i.p. DAMGO indicating the partial and pure peripheral antinociceptive effects of morphine and DAMGO, respectively. These results suggest the role of either central or peripheral µ-opioid receptors (MOR) in mediating antinociceptive effects of i.p. µ-opioid agonists in the rat late permanent visceral pain model which closely resembles the clinical situation.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Encefalina Ala(2)-MeFe(4)-Gli(5)/uso terapéutico , Morfina/uso terapéutico , Dolor Visceral/tratamiento farmacológico , Ácido Acético/toxicidad , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Interacciones Farmacológicas , Naloxona/administración & dosificación , Naloxona/análogos & derivados , Antagonistas de Narcóticos/administración & dosificación , Compuestos de Amonio Cuaternario/administración & dosificación , Ratas , Dolor Visceral/inducido químicamente
19.
J Med Chem ; 54(4): 980-8, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21235243

RESUMEN

The synthesis and the effect of a combination of 6-glycine and 14-phenylpropoxy substitutions in N-methyl- and N-cycloproplymethylmorphinans on biological activities are described. Binding studies revealed that all new 14-phenylpropoxymorphinans (11-18) displayed high affinity to opioid receptors. Replacement of the 14-methoxy group with a phenylpropoxy group led to an enhancement in affinity to all three opioid receptor types, with most pronounced increases in δ and κ activities, hence resulting in a loss of µ receptor selectivity. All compounds (11-18) showed potent and long-lasting antinociceptive effects in the tail-flick test in rats after subcutaneous administration. For the N-methyl derivatives 13 and 14, analgesic potencies were in the range of their 14-methoxy analogues 9 and 10, respectively. Even derivatives 15-18 with an N-cyclopropylmethyl substituent acted as potent antinociceptive agents, being several fold more potent than morphine. Subcutaneous administration of compounds 13 and 14 produced significant and prolonged antinociceptive effects mediated through peripheral opioid mechanisms in carrageenan-induced inflammatory hyperalgesia in rats.


Asunto(s)
Analgésicos/síntesis química , Analgésicos/farmacología , Morfinanos/síntesis química , Morfinanos/farmacología , Glicinas N-Sustituídas/síntesis química , Glicinas N-Sustituídas/farmacología , Receptores Opioides/metabolismo , Analgésicos/química , Animales , Unión Competitiva , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Espectroscopía de Resonancia Magnética , Morfinanos/química , Glicinas N-Sustituídas/química , Dolor/tratamiento farmacológico , Ratas , Espectrometría de Masa por Ionización de Electrospray , Espectroscopía Infrarroja por Transformada de Fourier , Relación Estructura-Actividad
20.
Brain Res Bull ; 84(1): 53-60, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20934489

RESUMEN

The aim of the present work was to further analyse the features of opioid dependence following chronic morphine treatment during pregnancy and lactation. Dams from the day of mating were treated either with saline or with morphine (10mg/kg) subcutaneously once daily. Physical and behavioural signs of morphine withdrawal were investigated both in the early postpartum period (maternal behaviour) and after weaning (physical signals, locomotion, anxiety-like behaviour). Maternal behaviour was evaluated after acute challenge with naloxone (3 mg/kgs.c.) or morphine (10 mg/kgs.c.) and morphine plus naloxone (10 mg/kgs.c. and 3 mg/kgs.c., respectively). After weaning sensitivity to the rewarding effect of morphine was measured by conditioned place preference and to the aversive effect of naloxone by conditioned place aversion tests. The intensity of physical and behavioural indices of dependence was also investigated by precipitation of withdrawal with naloxone (10 mg/kgs.c) after weaning. Naloxone impaired the maternal behaviour in morphine-treated dams but not in saline-ones. Acute challenge with morphine impaired maternal responsiveness both in saline and in morphine-treated dams, this effect of morphine, however could be completely antagonised by naloxone only in the saline-treated but not in the morphine-treated ones. Significantly increased sensitivity to the rewarding stimulus of morphine and more pronounced aversion to naloxone were observed in morphine-treated dams. Naloxone precipitated only moderate physical withdrawal signals in morphine-treated dams, while anxiety and locomotor activity after administration of naloxone (behavioural withdrawal) were not changed in them. In summary chronic, moderate dose morphine treatment during pregnancy and lactation resulted in only mild dependence, but it affected opioid-receptor sensitivity and presumably disrupted the functioning of endogenous opioid system.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Trastornos Relacionados con Opioides/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Operante , Femenino , Lactancia , Masculino , Conducta Materna/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Naloxona/farmacología , Pruebas Neuropsicológicas , Trastornos Relacionados con Opioides/psicología , Embarazo , Ratas , Ratas Wistar , Síndrome de Abstinencia a Sustancias/fisiopatología , Síndrome de Abstinencia a Sustancias/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...