Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Biochemistry ; 60(18): 1413-1419, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32930576

RESUMEN

This report describes the unique pharmacological profile of FBNTI, a potent DOR antagonist that acts as a MOR agonist via an allosteric mechanism. Binding of FBNTI to opioid receptors expressed in HEK 293 cells revealed a 190-fold greater affinity for DOR (Ki = 0.84 nM) over MOR (Ki = 160 nM). In mice, intrathecal FBNTI produced potent antinociception (ED50 = 46.9 pmol/mouse), which was antagonized by selective MOR antagonists (CTOP, ß-FNA). Autoantagonism of the MOR agonism by FBNTI was observed above the ED75 dose, suggesting antagonism of activated MOR. That FBNTI is devoid of agonism in DOR knockout mice is consistent with allosteric activation of the MOR protomer via FBNTI bound to within a MOR-DOR heteromer. This proposed mechanism is supported by calcium mobilization assays, which indicate that FBNTI selectively activates the MOR-DOR heteromer and functionally antagonizes the MOR protomer at >ED75. The unprecedented mode of MOR activation by FBNTI may be responsible for the lack of tolerance after intrathecal (i.t.) administration. FBNTI was highly effective upon topical administration to the ipsolateral hind paw in the Hargreaves assay (EC50 = 0.17 ± 0.08 µM) and without significant contralateral activity, suggesting a lack of systemic exposure.


Asunto(s)
Analgésicos Opioides/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Calcio/metabolismo , Células HEK293 , Humanos , Inyecciones Espinales , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Estructura Molecular , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo
2.
FASEB J ; 33(8): 9577-9587, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31162938

RESUMEN

Deregulation of innate immune TLR4 signaling contributes to various diseases including neuropathic pain and drug addiction. Naltrexone is one of the rare TLR4 antagonists with good blood-brain barrier permeability and showing no stereoselectivity for TLR4. By linking 2 naltrexone units through a rigid pyrrole spacer, the bivalent ligand norbinaltorphimine was formed. Interestingly, (+)-norbinaltorphimine [(+)-1] showed ∼25 times better TLR4 antagonist activity than naltrexone in microglial BV-2 cell line, whereas (-)-norbinaltorphimine [(-)-1] lost TLR4 activity. The enantioselectivity of norbinaltorphimine was further confirmed in primary microglia, astrocytes, and macrophages. The activities of meso isomer of norbinaltorphimine and the molecular dynamic simulation results demonstrate that the stereochemistry of (+)-1 is derived from the (+)-naltrexone pharmacophore. Moreover, (+)-1 significantly increased and prolonged morphine analgesia in vivo. The efficacy of (+)-1 is long lasting. This is the first report showing enantioselective modulation of the innate immune TLR signaling.-Zhang, X., Peng, Y., Grace, P. M., Metcalf, M. D., Kwilasz, A. J., Wang, Y., Zhang, T., Wu, S., Selfridge, B. R., Portoghese, P. S., Rice, K. C., Watkins, L. R., Hutchinson, M. R., Wang, X. Stereochemistry and innate immune recognition: (+)-norbinaltorphimine targets myeloid differentiation protein 2 and inhibits toll-like receptor 4 signaling.


Asunto(s)
Antígeno 96 de los Linfocitos/metabolismo , Naltrexona/análogos & derivados , Receptor Toll-Like 4/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Interleucina-1beta/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Naltrexona/química , Naltrexona/farmacología , Estructura Secundaria de Proteína , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(28): 11595-9, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798416

RESUMEN

The low effectiveness of morphine and related mu opioid analgesics for the treatment of chronic inflammatory pain is a result of opioid-induced release of proinflammatory cytokines and glutamate that lower the pain threshold. In this regard, the use of opioids with metabotropic glutamate-5 receptor (mGluR5) antagonist has been reported to increase the efficacy of morphine and prevent the establishment of adverse effects during chronic use. Given the presence of opioid receptors (MORs) and mGluR5 in glia and neurons, together with reports that suggest coexpressed MOR/mGluR5 receptors in cultured cells associate as a heteromer, the possibility that such a heteromer could be a target in vivo was addressed by the design and synthesis of a series of bivalent ligands that contain mu opioid agonist and mGluR5 antagonist pharmacophores linked through spacers of varying length (10-24 atoms). The series was evaluated for antinociception using the tail-flick and von Frey assays in mice pretreated with lipopolysaccharide (LPS) or in mice with bone cancer. In LPS-pretreated mice, MMG22 (4c, 22-atom spacer) was the most potent member of the series (intrathecal ED50 ∼9 fmol per mouse), whereas in untreated mice its ED50 was more than three orders of magnitude higher. As members of the series with shorter or longer spacers have ≥500-fold higher ED50s in LPS-treated mice, the exceptional potency of MMG22 may be a result of the optimal bridging of protomers in a putative MOR-mGluR5 heteromer. The finding that MMG22 possesses a >10(6) therapeutic ratio suggests that it may be an excellent candidate for treatment of chronic, intractable pain via spinal administration.


Asunto(s)
Inflamación/prevención & control , Dolor/prevención & control , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Animales , Inflamación/complicaciones , Ligandos , Ratones , Dolor/etiología , Unión Proteica , Receptor del Glutamato Metabotropico 5
4.
Proc Natl Acad Sci U S A ; 108(12): 5098-103, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21385944

RESUMEN

Numerous G protein-coupled receptors (GPCRs) have been shown to form heteromeric receptors in cell-based assays. Among the many heteromers reported in the opioid receptor family are µ/κ, κ/δ, and µ/δ. However, the in vivo physiological and behavioral relevance for the proposed heteromers have not yet been established. Here we report a unique example of a ligand, N-naphthoyl-ß-naltrexamine (NNTA) that selectively activates heteromeric µ/κ-opioid receptors in HEK-293 cells and induces potent antinociception in mice. NNTA was an exceptionally potent agonist in cells expressing µ/κ-opioid receptors. Intriguingly, it was found to be a potent antagonist in cells expressing only µ-receptors. In the mouse tail-flick assay, intrathecal (i.t.) NNTA produced antinociception that was ~100-fold greater than by intracerebroventricular (i.c.v.) administration. The κ-antagonist, norBNI, decreased the i.t. potency, and the activity was virtually abolished in µ-opioid receptor knockout mice. No tolerance was induced i.t., but marginal tolerance (3-fold) was observed via the i.c.v. route. Moreover, NNTA produced neither significant physical dependence nor place preference in the ED50 dose range. Taken together, this work provides an important pharmacologic tool for investigating the in vivo functional relevance of heteromeric µ/κ-opioid receptors and suggests an approach to potent analgesics with fewer deleterious side effects.


Asunto(s)
Analgésicos/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animales , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Naltrexona/efectos adversos , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/farmacología , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
5.
J Biol Chem ; 287(22): 18618-35, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22467877

RESUMEN

Allosteric binding pockets in peptide-binding G protein-coupled receptors create opportunities for the development of small molecule drugs with substantial benefits over orthosteric ligands. To gain insights into molecular determinants for this pocket within type 1 and 2 cholecystokinin receptors (CCK1R and CCK2R), we prepared a series of receptor constructs in which six distinct residues in TM2, -3, -6, and -7 were reversed. Two novel iodinated CCK1R- and CCK2R-selective 1,4-benzodiazepine antagonists, differing only in stereochemistry at C3, were used. When all six residues within CCK1R were mutated to corresponding CCK2R residues, benzodiazepine selectivity was reversed, yet peptide binding selectivity was unaffected. Detailed analysis, including observations of gain of function, demonstrated that residues 6.51, 6.52, and 7.39 were most important for binding the CCK1R-selective ligand, whereas residues 2.61 and 7.39 were most important for binding CCK2R-selective ligand, although the effect of substitution of residue 2.61 was likely indirect. Ligand-guided homology modeling was applied to wild type receptors and those reversing benzodiazepine binding selectivity. The models had high predictive power in enriching known receptor-selective ligands from related decoys, indicating a high degree of precision in pocket definition. The benzodiazepines docked in similar poses in both receptors, with C3 urea substituents pointing upward, whereas different stereochemistry at C3 directed the C5 phenyl rings and N1 methyl groups into opposite orientations. The geometry of the binding pockets and specific interactions predicted for ligand docking in these models provide a molecular framework for understanding ligand selectivity at these receptor subtypes. Furthermore, the strong predictive power of these models suggests their usefulness in the discovery of lead compounds and in drug development programs.


Asunto(s)
Benzodiazepinas/metabolismo , Receptores de Colecistoquinina/antagonistas & inhibidores , Sitio Alostérico , Secuencia de Aminoácidos , Animales , Benzodiazepinas/antagonistas & inhibidores , Células COS , Chlorocebus aethiops , Ligandos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
6.
Neuroscience ; 516: 54-61, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36805004

RESUMEN

MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores connected by a 22-atom linker. Intrathecal (i.t.) administration of MMG22 to inflamed mice has been reported to produce fmol-range antinociception in the reversal of LPS-induced hyperalgesia. MMG22 reduced hyperalgesia in the spared nerve injury (SNI) model of neuropathic pain at 10 days after injury but not at 30 days after injury, perhaps related to the inflammation that occurs early after injury but subsequently subsides. The present study determined the efficacy of MMG22 in cisplatin-treated male mice in order to provide data relating to the efficacy of MMG22 in the treatment of neuropathic pain that is associated with inflammation. Groups of eight mice each received daily intraperitoneal (i.p.) injections of cisplatin for seven days to produce robust mechanical allodynia defined by the decrease in withdrawal threshold using an electronic von Frey applied to the plantar surface of the hind paw. Intrathecal administration of MMG22 potently reduced mechanical hyperalgesia (ED50 0.04 fmol/mouse) without tolerance, whereas MMG10 was essentially inactive. Morphine was less potent than MMG22 by >5-orders of magnitude and displayed tolerance. Subcutaneous MMG22 was effective (ED50 = 2.41 mg/kg) and devoid of chronic tolerance. We propose that MMG22 induces the formation of a MOR-mGluR5 heteromer through selective interaction with the upregulated NR2B subunit of activated NMDAR, in view of the 4600-fold reduction of i.t. MMG22 antinociception by the selective NR2B antagonist, Ro25-6981. A possible explanation for the substantially reduced potency for MMG22 in the SNI model is discussed.


Asunto(s)
Hiperalgesia , Neuralgia , Ratones , Masculino , Animales , Hiperalgesia/tratamiento farmacológico , Cisplatino , Morfina/farmacología , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Inflamación , Modelos Animales de Enfermedad
7.
Mol Pharmacol ; 81(2): 264-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22072818

RESUMEN

The peripheral δ opioid receptor (DOR) is an attractive target for analgesic drug development. There is evidence that DOR can form heteromers with the κ-opioid receptor (KOR). As drug targets, heteromeric receptors offer an additional level of selectivity and, because of allosteric interactions between protomers, functionality. Here we report that selective KOR antagonists differentially altered the potency and/or efficacy of DOR agonists in primary cultures of adult rat peripheral sensory neurons and in a rat behavioral model of thermal allodynia. In vitro, the KOR antagonist nor-binaltorphimine (nor-BNI) enhanced the potency of [D-Pen(2,5)]-enkephalin (DPDPE), decreased the potency of [D-Ala(2),D-Leu(5)]-enkephalin (DADLE), and decreased the potency and efficacy of 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethylpiperazin-1-yl](3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC80) to inhibit prostaglandin E(2) (PGE(2))-stimulated adenylyl cyclase activity. In vivo, nor-BNI enhanced the effect of DPDPE and decreased the effect of SNC80 to inhibit PGE(2)-stimulated thermal allodynia. In contrast to nor-BNI, the KOR antagonist 5'-guanidinonaltrindole (5'-GNTI) reduced the response of DPDPE both in cultured neurons and in vivo. Evidence for DOR-KOR heteromers in peripheral sensory neurons included coimmunoprecipitation of DOR with KOR, a DOR-KOR heteromer selective antibody augmented the antinociceptive effect of DPDPE in vivo, and the DOR-KOR heteromer agonist 6'-GNTI inhibited adenylyl cyclase activity in vitro as well as PGE(2)-stimulated thermal allodynia in vivo. Taken together, these data suggest that DOR-KOR heteromers exist in rat primary sensory neurons and that KOR antagonists can act as modulators of DOR agonist responses most likely through allosteric interactions between the protomers of the DOR-KOR heteromer.


Asunto(s)
Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Células Receptoras Sensoriales/química , Regulación Alostérica , Animales , Células Cultivadas , Diseño de Fármacos , Hiperalgesia/etiología , Ligandos , Multimerización de Proteína , Ratas , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides kappa/agonistas
8.
Pain ; 161(9): 2041-2057, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32345918

RESUMEN

ABSTRACT: Functional interactions between the mu opioid receptor (MOR) and the metabotropic glutamate receptor 5 (mGluR5) in pain and analgesia have been well established. MMG22 is a bivalent ligand containing MOR agonist (oxymorphamine) and mGluR5 antagonist (MPEP) pharmacophores tethered by a 22-atom linker. MMG22 has been shown to produce potent analgesia in several models of chronic inflammatory and neuropathic pain (NP). This study assessed the efficacy of systemic administration of MMG22 at reducing pain behavior in the spared nerve injury (SNI) model of NP in mice, as well as its side-effect profile and abuse potential. MMG22 reduced mechanical hyperalgesia and spontaneous ongoing pain after SNI, with greater potency early (10 days) as compared to late (30 days) after injury. Systemic administration of MMG22 did not induce place preference in naive animals, suggesting absence of abuse liability when compared to traditional opioids. MMG22 also lacked the central locomotor, respiratory, and anxiolytic side effects of its monomeric pharmacophores. Evaluation of mRNA expression showed the transcripts for both receptors were colocalized in cells in the dorsal horn of the lumbar spinal cord and dorsal root ganglia. Thus, MMG22 reduces hyperalgesia after injury in the SNI model of NP without the typical centrally mediated side effects associated with traditional opioids.


Asunto(s)
Analgésicos Opioides , Neuralgia , Analgésicos Opioides/uso terapéutico , Animales , Hiperalgesia/tratamiento farmacológico , Ligandos , Ratones , Neuralgia/tratamiento farmacológico , Receptor del Glutamato Metabotropico 5 , Receptores Opioides mu/genética
9.
Bioorg Med Chem Lett ; 19(24): 6978-80, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19892550

RESUMEN

In an effort to develop antagonists for kappa-mu opioid receptor heterodimers, a series of bivalent ligands 3-6 containing kappa- and mu-antagonist pharmacophores were designed and synthesized. Evaluation of the series in HEK-293 cells revealed 4 (KMN-21) to selectively antagonize the activation of kappa-mu heterodimers, suggesting possible bridging of receptors when the bivalent ligand spacer contains 21 atoms.


Asunto(s)
Guanidinas/química , Naltrexona/análogos & derivados , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides mu/antagonistas & inhibidores , Línea Celular , Guanidinas/síntesis química , Guanidinas/farmacología , Humanos , Ligandos , Naltrexona/síntesis química , Naltrexona/química , Naltrexona/farmacología
10.
ACS Chem Neurosci ; 10(4): 2004-2011, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30110531

RESUMEN

Commonly prescribed opioid analgesics produce tolerance upon chronic use due in part to induction of hyperalgesia. Given that two reported bivalent ligands (MMG22 and MCC22) produce potent antinociception without tolerance only in inflamed mice, we have investigated the possible cellular and receptor targets of these ligands. The selective microglia inhibitors, minocycline and SB290157, antagonized intrathecal (i.t.) MCC22 antinociception orders of magnitude more potently than MMG22, suggesting that MCC22 selectively targets activated microglia. The astrocyte toxin, l-α-aminoadipic acid antagonized MMG22 antinociception 126-fold without reducing the potency of MCC22, indicating that activated astrocytes are targets of MMG22. MK-801 and Ro25-6981 antagonism of MMG22 antinociception, but not MCC22, is consistent with selective inhibition of activated NMDAR in astrocytes. The antinociception produced by i.t. MMG22 or MCC22 were both antagonized by the selective mu opioid receptor antagonist, ß-FNA, implicating interaction of these ligands with MOR in spinal afferent neurons. MCC22 antinociception was potently blocked by kainate or AMPA ion channel antagonists (LY382884; NBQX), in contrast to MMG22. It is concluded that i.t. MMG22 and MCC22 produce exceptional antinociception via potent inhibition of activated spinal glia, thereby leading to desensitization of spinal neurons and enhanced activation of neuronal MOR. Thus, the present study suggests a new approach to treatment of chronic inflammatory pain without tolerance through a single molecular entity that simultaneously inhibits activated glia and stimulates MOR in spinal neurons.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Tolerancia a Medicamentos , Neuroglía/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Dolor/tratamiento farmacológico , Receptores Opioides/agonistas , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Animales , Isoquinolinas/química , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Masculino , Ratones , Ratones Endogámicos ICR , Neuroglía/metabolismo , Dolor/metabolismo , Dimensión del Dolor/métodos , Piperidinas/química , Piperidinas/farmacología , Piperidinas/uso terapéutico , Receptores Opioides/metabolismo
11.
Neuropharmacology ; 158: 107598, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30970233

RESUMEN

Cisplatin and other widely employed platinum-based anticancer agents produce chemotherapy-induced peripheral neuropathy (CIPN) that often results in pain and hyperalgesia that are difficult to manage. We investigated the efficacy of a novel bivalent ligand, MCC22, for the treatment of pain arising from CIPN. MCC22 consists of mu opioid receptor (MOR) agonist and chemokine receptor 5 (CCR5) antagonist pharmacophores connected through a 22-atom spacer and was designed to target a putative MOR-CCR5 heteromer localized in pain processing areas. Mice received once daily intraperitoneal (i.p.) injections of cisplatin (1 mg/kg) for seven days and behavior testing began 7 days later. Cisplatin produced mechanical hyperalgesia that was decreased dose-dependently by MCC22 given by intrathecal (ED50 = 0.004 pmol) or i.p. (3.07 mg/kg) routes. The decrease in hyperalgesia was associated with decreased inflammatory response by microglia in the spinal cord. Unlike morphine, MCC22 given daily for nine days did not exhibit tolerance to its analgesic effect and its characteristic antihyperalgesic activity was fully retained in morphine-tolerant mice. Furthermore, MCC22 did not alter motor function and did not exhibit rewarding properties. Given the exceptional potency of MCC22 without tolerance or reward, MCC22 has the potential to vastly improve management of chronic pain due to CIPN. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Asunto(s)
Analgésicos Opioides/farmacología , Antineoplásicos/toxicidad , Antagonistas de los Receptores CCR5/farmacología , Cisplatino/toxicidad , Hiperalgesia/inducido químicamente , Isoquinolinas/farmacología , Neuralgia/inducido químicamente , Nocicepción/efectos de los fármacos , Piperidinas/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Receptores Opioides mu/agonistas
12.
Neuropharmacology ; 160: 107690, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31271770

RESUMEN

Pain is among the most common symptoms in cancer and approximately 90% of patients experience end-stage cancer pain. The management of cancer pain is challenging due to the significant side effects associated with opioids, and novel therapeutic approaches are needed. MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores joined by a 22-atom spacer. MMG22 exhibited extraordinary analgesia following intrathecal administration in a mouse model of bone cancer pain. Here, we assessed the effectiveness of systemic administration of MMG22 in reducing cancer pain and evaluated whether MMG22 displays side effects associated with opioids. Fibrosarcoma cells were injected into and around the calcaneus bone in C3H mice. Mechanical hyperalgesia was defined as an increase in the paw withdrawal frequencies (PWFs) evoked by application of a von Frey monofilament (3.9 mN bending force) applied to the plantar surface of the hind paw Subcutaneous (s.c.), intramuscular (i.m.), and oral (p.o.) administration of MMG22 produced robust dose-dependent antihyperalgesia, whose ED50 was orders of magnitude lower than morphine. Moreover, the ED50 for MMG22 decreased with disease progression. Importantly, s.c. administration of MMG22 did not produce acute (24 h) or long-term (9 days) tolerance, was not rewarding (conditioned place preference test), and did not produce naloxone-induced precipitated withdrawal or alter motor function. A possible mechanism of action of MMG22 is discussed in terms of inhibition of spinal NMDAR via antagonism of its co-receptor, mGluR5, and concomitant activation of neuronal MOR. We suggest that MMG22 may be a powerful alternative to traditional opioids for managing cancer pain. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Asunto(s)
Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/metabolismo , Receptores de Ácido Kaínico/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/metabolismo , Hiperalgesia/tratamiento farmacológico , Ligandos , Masculino , Ratones , Ratones Endogámicos C3H , Morfina/uso terapéutico , Receptores de Ácido Kaínico/administración & dosificación , Receptores Opioides mu/administración & dosificación
13.
J Med Chem ; 66(17): 11589-11590, 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37646574
14.
Arthritis Res Ther ; 20(1): 154, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30053832

RESUMEN

BACKGROUND: Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis. METHODS: MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance. RESULTS: MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore. CONCLUSIONS: MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use.


Asunto(s)
Analgésicos/farmacología , Artritis Experimental/patología , Artritis Reumatoide/patología , Isoquinolinas/farmacología , Dolor , Piperidinas/farmacología , Receptores CCR5/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Artritis Experimental/complicaciones , Artritis Reumatoide/complicaciones , Tolerancia a Medicamentos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dolor/etiología , Receptores CCR5/agonistas , Receptores CCR5/metabolismo
15.
Pain ; 159(7): 1382-1391, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29578946

RESUMEN

Sickle cell disease (SCD) is a chronic inflammatory disorder accompanied by chronic pain. In addition to ongoing pain and hyperalgesia, vaso-occlusive crises-induced pain can be chronic or episodic. Because analgesics typically used to treat pain are not very effective in SCD, opioids, including morphine, are a primary treatment for managing pain in SCD but are associated with many serious side effects, including constipation, tolerance, addiction, and respiratory depression. Thus, there is a need for the development of novel treatments for pain in SCD. In this study, we used the Townes transgenic mouse model of SCD to investigate the antinociceptive efficacy of the bivalent ligand, MCC22, and compared its effectiveness with morphine. MCC22 consists of a mu-opioid receptor agonist and a chemokine receptor-5 (CCR5) antagonist that are linked through a 22-atom spacer. Our results show that intraperitoneal administration of MCC22 produced exceptionally potent dose-dependent antihyperalgesia as compared to morphine, dramatically decreased evoked responses of nociceptive dorsal horn neurons, and decreased expression of proinflammatory cytokines in the spinal cord. Moreover, tolerance did not develop to its analgesic effects after repeated administration. In view of the extraordinary potency of MCC22 without tolerance, MCC22 and similar compounds may vastly improve the management of pain associated with SCD.


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos/farmacología , Anemia de Células Falciformes/fisiopatología , Hiperalgesia/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Analgésicos/uso terapéutico , Analgésicos Opioides/uso terapéutico , Animales , Modelos Animales de Enfermedad , Hiperalgesia/fisiopatología , Masculino , Ratones , Ratones Transgénicos
16.
J Med Chem ; 50(14): 3392-6, 2007 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-17555306

RESUMEN

Incorporation of a naphthalene-dialdehyde moiety into the delta antagonist, 6'-aminonaltrindole afforded a potent, selective, irreversible delta-agonist 1. However, flow cytometry studies revealed no time-dependent specific fluorescence, suggesting that both Lys214 and Cys216 at the recognition site are not involved in covalent binding. Molecular simulation studies suggest that compound 1 may form a Schiff base with the epsilon-amino group of Lys214, which could explain its irreversibility and transformation into a delta-agonist through a conformational change of TM5.


Asunto(s)
Marcadores de Afinidad , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/química , Receptores Opioides delta/metabolismo , Espectroscopía de Resonancia Magnética , Naltrexona/química , Espectrometría de Masa Bombardeada por Átomos Veloces
17.
Eur J Pharmacol ; 566(1-3): 75-82, 2007 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-17383633

RESUMEN

Treatment of pain with opioids is limited by their potential abuse liability. In an effort to develop analgesics without this side effect, a series of bivalent ligands containing a mu-opioid receptor agonist pharmacophore connected to a delta-opioid receptor antagonist pharmacophore through variable-length spacers (16-21 atoms) was synthesized. Members of this series [mu-opioid receptor (M)-delta-opioid receptor (D)-agonist (A)-antagonists (N): MDANs] are antinociceptive in the tail flick assay, but antinociceptive tolerance and physical dependence do not develop to ligands having spacers with 19-21 atoms. The current studies compared the rewarding properties of three bivalent ligands (MDAN-16, -19 and -21) and a mu-opioid receptor agonist (MA-19) to those of morphine in the conditioned place preference assay in mice after i.v. administration. Place preference developed to morphine and to MA-19, but not to the MDANs. The responses to MDAN-16 were highly variable, although place preference of borderline significance appeared to develop. Reinstatement was also evaluated after extinguishing morphine conditioned place preference; morphine and MA-19, but not the MDANs, reinstated morphine conditioned place preference. Taken together, these results suggest that the bivalents are less rewarding compared to morphine in opioid-naïve mice and do not induce reinstatement in previously morphine-preferring mice. The lack of a conditioned place preference response for MDAN-19 and -21, compared to the equivocal results with MDAN-16, suggests a minimum distance requirement between mu-opioid receptor and delta-opioid receptor recognition sites. This requirement may reflect the binding of MDAN-19 and -21 to mu-opioid receptor-delta-opioid receptor heterodimeric receptors that block reward but not antinociception.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Naltrexona/análogos & derivados , Oximorfona/análogos & derivados , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides mu/agonistas , Animales , Condicionamiento Psicológico/fisiología , Ligandos , Masculino , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Oximorfona/farmacología , Dolor/tratamiento farmacológico , Dolor/fisiopatología
18.
ACS Chem Neurosci ; 8(3): 426-428, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28139906

RESUMEN

It is proposed that two types of opioid receptor heteromers exist: a) those that are constitutive and b) those that are induced by bivalent ligands. Mu opioid agonists interact with constitutive MOR-DOR heteromer to mediate tolerance and dependence. Bivalent ligand, MDAN21, is devoid of these adverse effects by virtue of its DOR antagonist pharmacophore. We propose that bivalent ligands MMG22 and MCC22 induce colocalized receptors to form heteromers (MOR-mGluR5 and MOR-CCR5, respectively) that do not occur naturally, thereby promoting unique pharmacology. Heteromer induction with bivalent ligands offers a general approach to unique pharmacology that complements traditional SAR.


Asunto(s)
Analgésicos Opioides/farmacología , Multimerización de Proteína , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Modelos Químicos , Estructura Molecular , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Receptor del Glutamato Metabotropico 5/química , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores Opioides delta/química , Receptores Opioides mu/química , Relación Estructura-Actividad
19.
Pain ; 158(12): 2431-2441, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28891868

RESUMEN

The mu opioid receptor (MOR) and metabotropic glutamate receptor 5 (mGluR5) are well-established pharmacological targets in the management of chronic pain. Both receptors are expressed in the spinal cord. MMG22, a bivalent ligand containing 2 pharmacophores separated by 22 atoms, which simultaneously activates MOR and antagonizes mGluR5, has been shown to produce potent reversal of tactile hypersensitivity in rodent models of lipopolysaccharide (LPS)-and bone cancer-induced chronic pain. This study assessed whether intrathecal MMG22 also is effective in reducing pain of neuropathic origin. Furthermore, we theorized that MMG22 should reduce hyperalgesia in nerve-injured mice in a manner consistent with a synergistic interaction between MOR and mGluR5. Several weeks after spared nerve injury, tactile hypersensitivity was reversed in mice by the intrathecal injection of MMG22 (0.01-10 nmol) but also by its shorter spacer analog, MMG10, with similar potency. The potencies of the bivalent ligands were 10- to 14-fold higher than those of the compounds upon which the bivalent structure was based, the MOR agonist oxymorphone and the mGluR5 antagonist MPEP. Coadministration of oxymorphone and MPEP demonstrated analgesic synergism, an interaction confirmed by isobolographic analysis. This study indicates that in the spared nerve injury-induced model of neuropathic pain, the 2 pharmacophores of the bivalent ligands MMG22 and MMG10 target MOR and mGluR5 as separate receptor monomers. The observed increase in the potency of MMG22 and MMG10, compared with oxymorphone and MPEP, may reflect the synergistic interaction of the 2 pharmacophores of the bivalent ligand acting at their respective separate receptor monomers.


Asunto(s)
Analgésicos/uso terapéutico , Antagonistas de Narcóticos/farmacología , Neuralgia/tratamiento farmacológico , Receptor del Glutamato Metabotropico 5/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Hiperalgesia/tratamiento farmacológico , Inyecciones Espinales/métodos , Ligandos , Masculino , Ratones , Antagonistas de Narcóticos/administración & dosificación
20.
Pain ; 157(11): 2561-2570, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27437788

RESUMEN

Cold exposure and a variety of types of mild stress increase pain in patients with painful disorders such as fibromyalgia syndrome. Acutely, stress induces thermogenesis by increasing sympathetic activation of beta-3 (ß3) adrenergic receptors in brown adipose tissue. Chronic stress leads to the hypertrophy of brown adipose, a phenomenon termed adaptive thermogenesis. Based on the innervation of skeletal muscle by collaterals of nerves projecting to brown adipose, we theorized an association between brown adipose tissue activity and musculoskeletal hyperalgesia and tested this hypothesis in mice. Exposure to a cold swim or injection of BRL37344 (ß3 adrenergic agonist) each enhanced musculoskeletal hyperalgesia, as indicated by morphine-sensitive decreases in grip force responses, whereas SR59230A (ß3 adrenergic antagonist) attenuated swim-induced hyperalgesia. Chemical ablation of interscapular brown adipose, using Rose Bengal, attenuated the development of hyperalgesia in response to either swim stress or BRL37344. In addition, elimination of the gene expressing uncoupling protein-1 (UCP1), the enzyme responsible for thermogenesis, prevented musculoskeletal hyperalgesia in response to either a swim or BRL37344, as documented in UCP1-knockout (UCP1-KO) mice compared with wild-type controls. Together, these data provide a convergence of evidence suggesting that activation of brown adipose contributes to stress-induced musculoskeletal hyperalgesia.


Asunto(s)
Tejido Adiposo Pardo/patología , Hiperalgesia/etiología , Hiperalgesia/patología , Dolor Musculoesquelético/complicaciones , Tejido Adiposo Pardo/efectos de los fármacos , Agonistas Adrenérgicos beta/toxicidad , Animales , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Frío/efectos adversos , Modelos Animales de Enfermedad , Etanolaminas/toxicidad , Femenino , Hiperalgesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fuerza Muscular/efectos de los fármacos , Dolor Musculoesquelético/patología , Dolor Musculoesquelético/cirugía , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Natación/psicología , Cola (estructura animal)/inervación , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA