Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 446: 102-112, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32858141

RESUMEN

It was recently shown that local injection, systemic administration or topical application of the peripherally-restricted mu-opioid receptor (MOR) agonist loperamide (Lo) and the delta-opioid receptor (DOR) agonist oxymorphindole (OMI) synergized to produce highly potent anti-hyperalgesia that was dependent on both MOR and DOR located in the periphery. We assessed peripheral mechanisms by which this Lo/OMI combination produces analgesia in mice expressing the light-sensitive protein channelrhodopsin2 (ChR2) in neurons that express NaV1.8 voltage-gated sodium channels. These mice (NaV1.8-ChR2+) enabled us to selectively target and record electrophysiological activity from these neurons (the majority of which are nociceptive) using blue light stimulation of the hind paw. We assessed the effect of Lo/OMI on nociceptor activity in both naïve mice and mice treated with complete Freund's adjuvant (CFA) to induce chronic inflammation of the hind paw. Teased fiber recording of tibial nerve fibers innervating the plantar hind paw revealed that the Lo/OMI combination reduced responses to light stimulation in naïve mice and attenuated spontaneous activity (SA) as well as responses to light and mechanical stimuli in CFA-treated mice. These results show that Lo/OMI reduces activity of C-fiber nociceptors that express NaV1.8 and corroborate recent behavioral studies demonstrating the potent analgesic effects of this drug combination. Because of its peripheral site of action, Lo/OMI might produce effective analgesia without the side effects associated with activation of opioid receptors in the central nervous system.


Asunto(s)
Loperamida , Nociceptores , Animales , Hiperalgesia/tratamiento farmacológico , Inflamación , Loperamida/farmacología , Ratones , Morfolinas , Fibras Nerviosas Amielínicas , Receptores Opioides delta , Receptores Opioides mu
2.
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
3.
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
4.
Neurosci Lett ; 617: 82-7, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-26861198

RESUMEN

In previous studies we have reported that spinal nerve ligation (SNL), a model of neuropathic pain, results in the loss of over 20% of neurons in the rostral portion of the ventromedial medulla (RVM) in rats, 10 days after SNL. The RVM is involved in pain modulation and we have proposed that loss of pain inhibition from the RVM, including loss of RVM serotonin neurons, contributes to the increased hypersensitivity observed after SNL. In the present study we examined whether RVM neuronal loss occurs in two other models of neuropathic pain, chronic constriction injury (CCI) and spared nerve injury (SNI). We found no evidence for neuronal loss 10 days after either nerve injury, a time when robust tactile hypersensitivity is present in both CCI and SNI. We conclude that loss of RVM neurons appears not to be required for expression of tactile hypersensitivity in these models of neuropathic pain.


Asunto(s)
Bulbo Raquídeo/patología , Neuralgia/patología , Neuroglía/patología , Neuronas/patología , Traumatismos de los Nervios Periféricos/patología , Nervio Ciático/lesiones , Animales , Recuento de Células , Enfermedad Crónica , Constricción Patológica , Modelos Animales de Enfermedad , Miembro Posterior/inervación , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Masculino , Neuralgia/fisiopatología , Traumatismos de los Nervios Periféricos/fisiopatología , Estimulación Física , Ratas Sprague-Dawley , Factores de Tiempo , Tacto
5.
Pain ; 155(7): 1229-1237, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657450

RESUMEN

VGF (nonacronymic) is a granin-like protein that is packaged and proteolytically processed within the regulated secretory pathway. VGF and peptides derived from its processing have been implicated in neuroplasticity associated with learning, memory, depression, and chronic pain. In sensory neurons, VGF is rapidly increased following peripheral nerve injury and inflammation. Several bioactive peptides generated from the C-terminus of VGF have pronociceptive spinal effects. The goal of the present study was to examine the spinal effects of the peptide TLQP-21 and determine whether it participates in spinal mechanisms of persistent pain. Application of exogenous TLQP-21 induced dose-dependent thermal hyperalgesia in the warm-water immersion tail-withdrawal test. This hyperalgesia was inhibited by a p38 mitogen-activated protein kinase inhibitor, as well as inhibitors of cyclooxygenase and lipoxygenase. We used immunoneutralization of TLQP-21 to determine the function of the endogenous peptide in mechanisms underlying persistent pain. In mice injected intradermally with complete Freund adjuvant, intrathecal treatment with anti-TLQP-21 immediately prior to or 5hours after induction of inflammation dose-dependently inhibited tactile hypersensitivity and thermal hyperalgesia. Intrathecal anti-TL21 administration also attenuated the development and maintenance of tactile hypersensitivity in the spared nerve injury model of neuropathic pain. These results provide evidence that endogenous TLQP-21 peptide contributes to the mechanisms of spinal neuroplasticity after inflammation and nerve injury.


Asunto(s)
Hiperalgesia/metabolismo , Inflamación/metabolismo , Neuralgia/metabolismo , Neuropéptidos/metabolismo , Nocicepción/fisiología , Fragmentos de Péptidos/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Modelos Animales de Enfermedad , Adyuvante de Freund/envenenamiento , Calor , Hiperalgesia/inducido químicamente , Inflamación/inducido químicamente , Inyecciones Espinales , Inhibidores de la Lipooxigenasa/farmacología , Ratones , Factores de Crecimiento Nervioso , Nocicepción/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Nervio Peroneo/lesiones , Piel/efectos de los fármacos , Nervio Tibial/lesiones , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA