Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pain ; 16: 1744806920950866, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32811276

RESUMO

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) alleviate symptoms of experimental neuropathy, protect and stimulate regeneration of sensory neurons in animal models of neuropathic pain, and restore their functional activity. However, clinical development of GFL proteins is complicated by their poor pharmacokinetic properties and multiple effects mediated by several receptors. Previously, we have identified a small molecule that selectively activates the major signal transduction unit of the GFL receptor complex, receptor tyrosine kinase RET, as an alternative to GFLs, for the treatment of neuropathic pain. We then introduced a series of chemical changes to improve the biological activity of these compounds and tested an optimized compound named BT44 in a panel of biological assays. BT44 efficiently and selectively stimulated the GFL receptor RET and activated the intracellular mitogene-activated protein kinase/extracellular signal-regulated kinase pathway in immortalized cells. In cultured sensory neurons, BT44 stimulated neurite outgrowth with an efficacy comparable to that of GFLs. BT44 alleviated mechanical hypersensitivity in surgery- and diabetes-induced rat models of neuropathic pain. In addition, BT44 normalized, to a certain degree, the expression of nociception-related neuronal markers which were altered by spinal nerve ligation, the neuropathy model used in this study. Our results suggest that the GFL mimetic BT44 is a promising new lead for the development of novel disease-modifying agents for the treatment of neuropathy and neuropathic pain.


Assuntos
Biomimética/métodos , Neuralgia/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ret/agonistas , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nervos Espinhais/efeitos dos fármacos , Animais , Escala de Avaliação Comportamental , Linhagem Celular , Neuropatias Diabéticas/tratamento farmacológico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Nociceptividade/efeitos dos fármacos , Fosforilação , Ratos , Ratos Wistar , Células Receptoras Sensoriais/metabolismo , Nervos Espinhais/lesões
2.
Eur J Pharmacol ; 875: 173021, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32112778

RESUMO

Morphine-3-glucuronide (M3G), the main metabolite of morphine, has been implicated in the development of tolerance and of opioid-induced hyperalgesia, both limiting the analgesic use of morphine. We evaluated the acute and chronic effects of M3G and morphine as well as development of antinociceptive cross-tolerance between morphine and M3G after intrathecal administration and assessed the expression of pain-associated neurotransmitter substance P in the spinal cord. Sprague-Dawley rats received intrathecal M3G or morphine twice daily for 6 days. Nociception and tactile allodynia were measured with von Frey filaments after acute and chronic treatments. Substance P levels in the dorsal horn of the spinal cord were determined by immunohistochemistry after 4-day treatments. Acute morphine caused antinociception as expected, whereas acute M3G caused tactile allodynia, as did both chronic M3G and morphine. Chronic M3G also induced antinociceptive cross-tolerance to morphine. M3G and morphine increased substance P levels similarly in the nociceptive laminae of the spinal cord. This study shows that chronic intrathecal M3G sensitises animals to mechanical stimulation and elevates substance P levels in the nociceptive laminae of the spinal cord. Chronic M3G also induces antinociceptive cross-tolerance to morphine. Thus, chronic M3G exposure might contribute to morphine-induced tolerance and opioid-induced hyperalgesia.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Hiperalgesia/induzido quimicamente , Derivados da Morfina/farmacologia , Morfina/farmacologia , Nociceptividade/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Esquema de Medicação , Tolerância a Medicamentos , Humanos , Hiperalgesia/diagnóstico , Injeções Espinhais , Masculino , Morfina/metabolismo , Derivados da Morfina/metabolismo , Medição da Dor , Ratos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Substância P/metabolismo
3.
Neuroscience ; 375: 10-24, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29421434

RESUMO

Development of tolerance is a well known pharmacological characteristic of opioids and a major clinical problem. In addition to the known neuronal mechanisms of opioid tolerance, activation of glia has emerged as a potentially significant new mechanism. We studied activation of microglia and astrocytes in morphine tolerance and opioid-induced hyperalgesia in rats using immunohistochemistry, flow cytometry and RNA sequencing in spinal- and supraspinal regions. Chronic morphine treatment that induced tolerance and hyperalgesia also increased immunoreactivity of spinal microglia in the dorsal and ventral horns. Flow cytometry demonstrated that morphine treatment increased the proportion of M2-polarized spinal microglia, but failed to impact the number or the proportion of M1-polarized microglia. In the transcriptome of microglial cells isolated from the spinal cord (SC), morphine treatment increased transcripts related to cell activation and defense response. In the studied brain regions, no activation of microglia or astrocytes was detected by immunohistochemistry, except for a decrease in the number of microglial cells in the substantia nigra. In flow cytometry, morphine caused a decrease in the number of microglial cells in the medulla, but otherwise no change was detected for the count or the proportion of M1- and M2-polarized microglia in the medulla or sensory cortex. No evidence for the activation of glia in the brain was seen. Our results suggest that glial activation associated with opioid tolerance and opioid-induced hyperalgesia occurs mainly at the spinal level. The transcriptome data suggest that the microglial activation pattern after chronic morphine treatment has similarities with that of neuropathic pain.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Morfina/farmacologia , Neuroglia/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Tolerância a Medicamentos , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Modelos Animais , Neuroglia/metabolismo , Neuroglia/patologia , Dor Nociceptiva/tratamento farmacológico , Dor Nociceptiva/metabolismo , Dor Nociceptiva/patologia , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/patologia , Transcriptoma/efeitos dos fármacos
4.
Basic Clin Pharmacol Toxicol ; 122(5): 481-488, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29171155

RESUMO

Ketamine and its primary metabolite norketamine attenuate morphine tolerance by antagonising N-methyl-d-aspartate (NMDA) receptors. Ketamine is extensively metabolized to several other metabolites. The major secondary metabolite (2S,6S;2R,6R)-hydroxynorketamine (6-hydroxynorketamine) is not an NMDA antagonist. However, it may modulate nociception through negative allosteric modulation of α7 nicotinic acetylcholine receptors. We studied whether 6-hydroxynorketamine could affect nociception or the effects of morphine in acute or chronic administration settings. Male Sprague Dawley rats received subcutaneous 6-hydroxynorketamine or ketamine alone or in combination with morphine, as a cotreatment during induction of morphine tolerance, and after the development of tolerance induced by subcutaneous minipumps administering 9.6 mg morphine daily. Tail flick, hot plate, paw pressure and rotarod tests were used. Brain and serum drug concentrations were quantified with high-performance liquid chromatography-tandem mass spectrometry. Ketamine (10 mg/kg), but not 6-hydroxynorketamine (10 and 30 mg/kg), enhanced antinociception and decreased rotarod performance following acute administration either alone or combined with morphine. Ketamine efficiently attenuated morphine tolerance. Acutely administered 6-hydroxynorketamine increased the brain concentration of morphine (by 60%), and brain and serum concentrations of 6-hydroxynorketamine were doubled by morphine pre-treatment. This pharmacokinetic interaction did not, however, lead to altered morphine tolerance. Co-administration of 6-hydroxynorketamine 20 mg/kg twice daily did not influence development of morphine tolerance. Even though morphine and 6-hydroxynorketamine brain concentrations were increased after co-administration, the pharmacokinetic interaction had no effect on acute morphine nociception or tolerance. These results indicate that 6-hydroxynorketamine does not have antinociceptive properties or attenuate opioid tolerance in a similar way as ketamine.


Assuntos
Analgésicos Opioides/farmacologia , Anestésicos Dissociativos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Tolerância a Medicamentos , Ketamina/análogos & derivados , Morfina/farmacologia , Dor Nociceptiva/prevenção & controle , Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Anestésicos Dissociativos/sangue , Anestésicos Dissociativos/farmacocinética , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Interações Medicamentosas , Ketamina/sangue , Ketamina/farmacocinética , Ketamina/farmacologia , Masculino , Morfina/sangue , Morfina/farmacocinética , Atividade Motora/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/sangue , Dor Nociceptiva/fisiopatologia , Dor Nociceptiva/psicologia , Limiar da Dor/efeitos dos fármacos , Ratos Sprague-Dawley
5.
Front Pharmacol ; 8: 365, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28680400

RESUMO

Neuropathic pain caused by nerve damage is a common and severe class of chronic pain. Disease-modifying clinical therapies are needed as current treatments typically provide only symptomatic relief; show varying clinical efficacy; and most have significant adverse effects. One approach is targeting either neurotrophic factors or their receptors that normalize sensory neuron function and stimulate regeneration after nerve damage. Two candidate targets are glial cell line-derived neurotrophic factor (GDNF) and artemin (ARTN), as these GDNF family ligands (GFLs) show efficacy in animal models of neuropathic pain (Boucher et al., 2000; Gardell et al., 2003; Wang et al., 2008, 2014). As these protein ligands have poor drug-like properties and are expensive to produce for clinical use, we screened 18,400 drug-like compounds to develop small molecules that act similarly to GFLs (GDNF mimetics). This screening identified BT13 as a compound that selectively targeted GFL receptor RET to activate downstream signaling cascades. BT13 was similar to NGF and ARTN in selectively promoting neurite outgrowth from the peptidergic class of adult sensory neurons in culture, but was opposite to ARTN in causing neurite elongation without affecting initiation. When administered after spinal nerve ligation in a rat model of neuropathic pain, 20 and 25 mg/kg of BT13 decreased mechanical hypersensitivity and normalized expression of sensory neuron markers in dorsal root ganglia. In control rats, BT13 had no effect on baseline mechanical or thermal sensitivity, motor coordination, or weight gain. Thus, small molecule BT13 selectively activates RET and offers opportunities for developing novel disease-modifying medications to treat neuropathic pain.

6.
Basic Clin Pharmacol Toxicol ; 120(1): 38-45, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27312359

RESUMO

Spironolactone, eplerenone, chlorothiazide and furosemide are diuretics that have been suggested to have antinociceptive properties, for example via mineralocorticoid receptor antagonism. In co-administration, diuretics might enhance the antinociceptive effect of opioids via pharmacodynamic and pharmacokinetic mechanisms. Effects of spironolactone (100 mg/kg, i.p.), eplerenone (100 mg/kg, i.p.), chlorothiazide (50 mg/kg, i.p.) and furosemide (100 mg/kg, i.p.) were studied on acute oxycodone (0.75 mg/kg, s.c.)- and morphine (3 mg/kg, s.c.)-induced antinociception using tail-flick and hot plate tests in male Sprague Dawley rats. The diuretics were administered 30 min. before the opioids, and behavioural tests were performed 30 and 90 min. after the opioids. Concentrations of oxycodone, morphine and their major metabolites in plasma and brain were quantified by mass spectrometry. In the hot plate test at 30 and 90 min., spironolactone significantly enhanced the antinociceptive effect (% of maximum possible effect) of oxycodone from 10% to 78% and from 0% to 50%, respectively, and that of morphine from 12% to 73% and from 4% to 83%, respectively. The brain oxycodone and morphine concentrations were significantly increased at 30 min. (oxycodone, 46%) and at 90 min. (morphine, 190%). We did not detect any independent antinociceptive effects with the diuretics. Eplerenone and chlorothiazide did not enhance the antinociceptive effect of either opioid. The results suggest that spironolactone enhances the antinociceptive effect of both oxycodone and morphine by increasing their concentrations in the central nervous system.


Assuntos
Analgésicos/uso terapêutico , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Diuréticos/uso terapêutico , Neurônios/efeitos dos fármacos , Dor/prevenção & controle , Espironolactona/uso terapêutico , Analgésicos Opioides/sangue , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Clorotiazida/uso terapêutico , Interações Medicamentosas , Quimioterapia Combinada , Eplerenona , Furosemida/uso terapêutico , Masculino , Morfina/sangue , Morfina/metabolismo , Morfina/farmacocinética , Morfina/uso terapêutico , Neurônios/metabolismo , Oxicodona/sangue , Oxicodona/metabolismo , Oxicodona/farmacocinética , Oxicodona/uso terapêutico , Dor/sangue , Dor/metabolismo , Ratos Sprague-Dawley , Espironolactona/análogos & derivados , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...