Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharm Pharmacol ; 74(1): 22-31, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34586411

RESUMEN

OBJECTIVES: 6-Hydroxykynurenic acid (6-HKA) is an organic acid component in extracts of Ginkgo biloba leaves and acts as a major contributor to neurorestorative effects, while its oral bioavailability was low. Therefore, using prodrug method to improve the bioavailability and brain content of 6-HKA is significant. METHODS: Three structural modified compounds of 6-HKA were synthesized, and ultra performance liquid chromatography-tandem mass spectrometry methods for quantification of these structural modified compounds in rat plasma and rat brain homogenate were established and comprehensively validated. The methods were effectively applied to investigate the effects of structural modification on apparent permeability coefficients in cells, the pharmacokinetics and the brain distribution in rats. KEY FINDINGS: The results illustrated that esterification can greatly improve the apparent permeability coefficient and bioavailability of 6-HKA. Comparing with direct oral administration of 6-HKA, the bioavailability of isopropyl ester was greatly improved (from 3.96 ± 1.45% to 41.8 ± 15.3%), and the contents of 6-HKA in rat brains (49.7 ± 9.2 ng/g brain) were significantly higher after oral administration. CONCLUSIONS: The bioavailability and the brain content of 6-HKA can be improved by the prodrug method. Among three structural modified compounds, isopropyl-esterified 6-HKA was the most promising treatment.


Asunto(s)
Disponibilidad Biológica , Encéfalo , Ginkgo biloba , Ácido Quinurénico/análogos & derivados , Administración Oral , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cromatografía Liquida/métodos , Ácido Quinurénico/administración & dosificación , Ácido Quinurénico/farmacocinética , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacocinética , Preparaciones de Plantas/administración & dosificación , Preparaciones de Plantas/farmacocinética , Profármacos/farmacología , Ratas , Relación Estructura-Actividad , Espectrometría de Masas en Tándem/métodos , Distribución Tisular
2.
Artículo en Inglés | MEDLINE | ID: mdl-30981181

RESUMEN

A sensitive and specific bioanalytical method of liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the quantification of 6-hydroxykynurenic acid (6-HKA) in a small quantity of rat plasma has been developed and comprehensively validated. Tolbutamide (Tol) was used as the internal standard (IS). An aliquot of 50 µL rat plasma sample was deproteinized by 150 µL methanol, and then 100 µL of its supernatant was mixed with 100 µL water after centrifugation. Chromatographic separation was performed on a ZORBAX Eclipse Plus C18 Rapid Resolution HD column (2.1 mm × 100 mm, 1.8 µm) with a gradient mobile phase consisting of water containing 2 mM ammonium formate and methanol at a flow rate of 0.2 mL/min over a total run time of 5.0 min. The mass spectrometer was operated under multiple reactions monitoring (MRM) mode with the transitions m/z 206.1 → 160.0 for 6-HKA and m/z 269.0 → 170.0 for Tol. The linear range was 2.5-250 ng/mL with the square regression coefficient (r2) of 0.997. The lower limit of quantification (LLOQ) was 2.5 ng/mL (Relative Error, RE +1.6% and RSD 4.8%, n = 5). The intra- and inter-day precision and accuracy was <13.3%. The mean recovery of 6-HKA in rat plasma was between the range of 99.3-103.4%. This method was successfully applied to study the pharmacokinetics of 6-HKA in rats after oral administration at a single dose of 20.0 mg/kg and after tail intravenous injection at a single dose of 2.0 mg/kg. Pharmacokinetic parameters bioavailability, Cmax, oral, Tmax, oral, AUC0-24h, oral, AUC0-∞, oral, AUC0-24h, iv and AUC0-∞, iv were 3.96%, 152.0 ±â€¯85.5 ng/mL, 0.4 ±â€¯0.1 h, 340.0 ±â€¯136.3 ng/mL ∗ h, 369.5 ±â€¯135.0 ng/mL ∗ h, 906.6 ±â€¯324.1 ng/mL*h and 932.9 ±â€¯336.5 ng/mL ∗ h, respectively.


Asunto(s)
Cromatografía Liquida/métodos , Ácido Quinurénico/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Animales , Femenino , Ácido Quinurénico/sangre , Ácido Quinurénico/química , Ácido Quinurénico/farmacocinética , Límite de Detección , Modelos Lineales , Masculino , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados
3.
Scand J Pain ; 17: 243-251, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29229209

RESUMEN

BACKGROUND AND AIMS: Neuropathic pain is a significant medical problem needing more effective treatments with fewer side effects. Overactive glutamatergic transmission via N-methyl-d-aspartate receptors (NMDARs) are known to play a role in central sensitization and neuropathic pain. Although ketamine, a NMDAR channel-blocking antagonist, is often used for neuropathic pain, its side-effect profile and abusive potential has prompted the search for a safer effective oral analgesic. A novel oral prodrug, AV-101 (l-4 chlorokynurenine), which, in the brain, is converted into one of the most potent and selective GlyB site antagonists of the NMDAR, has been demonstrated to be active in animal models of neuropathic pain. The two Phase 1 studies reported herein were designed to assess the safety and pharmacokinetics of AV-101, over a wide dose range, after daily dosing for 14-days. As secondary endpoints, AV-101 was evaluated in the capsaicin-induced pain model. METHODS: The Phase 1A study was a single-site, randomized, double-blind, placebo-controlled, single oral ascending dose (30-1800mg) study involving 36 normal healthy volunteers. The Phase 1B study was a single-site randomized, double-blind, placebo-controlled, study of multiple ascending doses (360, 1080, and 1440mg/day) of AV-101 involving 50 normal healthy volunteers, to whom AV-101 or placebo were administered orally daily for 14 consecutive days. Subjects underwent PK blood analyses, laboratory assessments, physical examination, 12-lead ECG, ophthalmological examination, and various neurocognitive assessments. The effect of AV-101 was evaluated in the intradermally capsaicin-induced pain model (ClinicalTrials.gov Identifier: NCT01483846). RESULTS: Two Phase 1, with an aggregate of 86 subjects, demonstrated that up to 14 days of oral AV-101, up to 1440mg per day, was safe and very well tolerated with AEs quantitively and qualitatively like those observed with placebo. Mean half-life values of AV-101 were consistent across doses, ranging with an average of 1.73h, with the highest Cmax (64.4µg/mL) and AUC0-t (196µgh/mL) values for AV-101 occurring in the 1440-mg dose group. In the capsaicin induce-pain model, there was no significant change in the area under the pain time curve (AUPC) for the spontaneous pain assessment between the treatment and the placebo groups on Day 1 or 14 (the primary endpoint). In contrast, there were consistent reductions at 60-180min on Day 1 after dosing for allodynia, mechanical hyperalgesia, heat hyperalgesia, and spontaneous pain, and on Day 14 after dosing for heat hyperalgesia. CONCLUSIONS: Although, AV-101 did not reach statistical significance in reducing pain, there were consistent reductions, for allodynia pain and mechanical and heat hyperalgesia. Given the excellent safety profile and PK characteristics demonstrated by this study, future clinical trials of AV-101 in neuropathic pain are justified. IMPLICATIONS: This article presents the safety and PK of AV-101, a novel oral prodrug producing a potent and selective GlyB site antagonist of the NMDA receptor. These data indicate that AV-101 has excellent safety and PK characteristics providing support for advancing AV-101 into Phase 2 studies in neuropathic pain, and even provides data suggesting that AV-101 may have a role in treating depression.


Asunto(s)
Voluntarios Sanos , Hiperalgesia , Ácido Quinurénico/análogos & derivados , Quinurenina/análogos & derivados , Neuralgia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacocinética , Adulto , Estudios Cruzados , Femenino , Humanos , Hiperalgesia/tratamiento farmacológico , Ácido Quinurénico/administración & dosificación , Ácido Quinurénico/sangre , Ácido Quinurénico/farmacocinética , Quinurenina/administración & dosificación , Quinurenina/sangre , Quinurenina/farmacocinética , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/sangre , Dimensión del Dolor
4.
J Neural Transm (Vienna) ; 124(1): 99-112, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27629500

RESUMEN

Kynurenic acid (KYNA) has well-established protective properties against glutamatergic neurotransmission, which plays an essential role in the activation and sensitization process during some primary headache disorders. The goal of this study was to compare the effects of two KYNA analogs, N-(2-N,N-dimethylaminoethyl)-4-oxo-1H-quinoline-2-carboxamide hydrochloride (KA-1) and N-(2-N-pyrrolidinylethyl)-4-oxo-1H-quinoline-2-carboxamide hydrochloride (KA-2), in the orofacial formalin test of trigeminal pain. Following pretreatment with KA-1 or KA-2, rats were injected with subcutaneous formalin solution in the right whisker pad. Thereafter, the rubbing activity and c-Fos immunoreactivity changes in the spinal trigeminal nucleus pars caudalis (TNC) were investigated. To obtain pharmacokinetic data, KA-1, KA-2 and KYNA concentrations were measured following KA-1 or KA-2 injection. Behavioral tests demonstrated that KA-2 induced larger amelioration of formalin-evoked alterations as compared with KA-1 and the assessment of c-Fos immunoreactivity in the TNC yielded similar results. Although KA-1 treatment resulted in approximately four times larger area under the curve values in the serum relative to KA-2, the latter resulted in a higher KYNA elevation than in the case of KA-1. With regard to TNC, the concentration of KA-1 was under the limit of detection, while that of KA-2 was quite small and there was no major difference in the approximately tenfold KYNA elevations. These findings indicate that the differences between the beneficial effects of KA-1 and KA-2 may be explained by the markedly higher peripheral KYNA levels following KA-2 pretreatment. Targeting the peripheral component of trigeminal pain processing would provide an option for drug design which might prove beneficial in headache conditions.


Asunto(s)
Analgésicos/farmacología , Dolor Facial/tratamiento farmacológico , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/farmacología , Núcleos del Trigémino/efectos de los fármacos , Analgésicos/sangre , Analgésicos/farmacocinética , Animales , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Dolor Facial/patología , Dolor Facial/fisiopatología , Formaldehído , Inmunohistoquímica , Ácido Quinurénico/sangre , Ácido Quinurénico/farmacocinética , Masculino , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Núcleos del Trigémino/metabolismo , Núcleos del Trigémino/patología , Vibrisas
5.
J Biomed Mater Res A ; 104(9): 2334-44, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27144507

RESUMEN

Dermal fibrosis, characterized by excessive extracellular matrix (ECM), is a pathological condition with limited effective therapeutic modalities. Lack of an antiscarring dressing further impedes the preventive measures for this condition. Here, we develop a new antiscarring dressing and investigate its potential as a slow-releasing vehicle for kynurenic acid (KynA), an antifibrotic agent. KynA was incorporated into polymethyl methacrylate (PMMA) nanofibers, containing increasing concentration of polyethylene glycol (PEG). Fibre morphology, water absorption capacity, surface hydrophilicity, in vitro drug release profile, and in vivo antifibrotic effects were investigated. Increasing concentrations of PEG (1-20%) significantly increased surface hydrophilicity, water absorption capacity, and drug release. Based on the obtained release profiles, PMMA + 10% PEG was the preferred formulation for sustained KynA release up to 120 hours. In vitro studies confirmed the preservation of KynA antifibrotic properties during electrospinning, indicated by fibroblasts proliferation suppression and ECM expression modulation. In vivo application of KynA-incorporated films significantly inhibited collagen (23.89 ± 4.79 vs. 6.99 ± 0.41, collagen-I/ß-actin mRNA expression, control vs. treated) and fibronectin expression (7.18 ± 1.09 vs. 2.31 ± 0.05, fibronectin/ß-actin mRNA expression, control vs. treated) and enhanced the production of an ECM-degrading enzyme (2.03 ± 0.88 vs. 11.88 ± 1.16 MMP-1/ß-actin mRNA expression, control vs. treated). The fabricated KynA-incorporated films can be exploited as antifibrotic wound dressings. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2334-2344, 2016.


Asunto(s)
Vendajes , Fibroblastos/metabolismo , Ácido Quinurénico , Nanofibras/química , Polimetil Metacrilato/química , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ácido Quinurénico/química , Ácido Quinurénico/farmacocinética , Ácido Quinurénico/farmacología , Masculino , Ratas Long-Evans
6.
Eur J Pharm Sci ; 86: 67-74, 2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-26924227

RESUMEN

Core-shell nanoparticles (CSNPs) were developed to get over therapeutic amount of kynurenic acid (KYNA) across the blood-brain barrier (BBB). Bovine serum albumin (BSA) was used as core for encapsulation of KYNA and the BSA/KYNA composite was finally encapsulated by poly(allylamine) hydrochloride (PAH) polymer as shell. In the interest of the optimization of the synthesis the BSA and KYNA interaction was studied by two-dimensional surface plasmon resonance (SPR) technique as well. The average size of d~100 nm was proven by dynamic light scattering (DLS) and transmission electron microscopy (TEM), while the structure of the composites was characterized by fluorescence (FL) and circular dichroism (CD) spectroscopy. The in vitro release properties of KYNA were investigated by a vertical diffusion cell at 25.0 °C and 37.5 °C and the kinetic of the release were discussed. The penetration capacity of the NPs into the central nervous system (CNS) was tested by an in vitro BBB model. The results demonstrated that the encapsulated KYNA had significantly higher permeability compared to free KYNA molecules. In the neurobiological serial of in vivo experiments the effects of peripherally administered KYNA with CSNPs were studied in comparison with untreated KYNA. These results clearly proved that KYNA in the CSNPs, administrated peripherally is suitable to cross the BBB and to induce electrophysiological effects within the CNS. As the neuroprotective properties of KYNA nowadays are proven, the importance of the results is obvious.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Portadores de Fármacos/administración & dosificación , Ácido Quinurénico/administración & dosificación , Nanopartículas/administración & dosificación , Poliaminas/administración & dosificación , Albúmina Sérica Bovina/administración & dosificación , Animales , Dicroismo Circular , Técnicas de Cocultivo , Portadores de Fármacos/química , Liberación de Fármacos , Células Endoteliales/metabolismo , Ácido Quinurénico/química , Ácido Quinurénico/farmacocinética , Nanopartículas/química , Neuroglía/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Pericitos/metabolismo , Poliaminas/química , Poliaminas/farmacocinética , Ratas Wistar , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacocinética , Espectrometría de Fluorescencia , Resonancia por Plasmón de Superficie
7.
Postepy Hig Med Dosw (Online) ; 66: 431-6, 2012 Jun 22.
Artículo en Polaco | MEDLINE | ID: mdl-22922142

RESUMEN

Atherosclerosis together with its cardiovascular consequences is the most common and significant cause of death, particularly in highly developed countries. The process of atherogenesis begins as soon as in childhood and depends on classical risk factors. Atherosclerosis also results from a chronic inflammatory-immune process which takes place in the vascular walls. Furthermore, it has been known for a number of years that the development of atherosclerotic lesions is closely connected with the concentration of homocysteine in serum. Homocysteine is a sulfur amino acid originating from methionine. An increased concentration of homocysteine in blood harmfully influences blood vessels, leading to a higher risk of ischemic heart disease and stroke. Since tackling classical atherosclerosis risk factors is not efficient enough when it comes to protecting the cardiovascular system from diseases, new substances possessing anti-atherogenic properties, especially endogenous ones, are sought. Recently, researchers have paid attention to a connection between homocysteine and an endogenous tryptophan derivative, kynurenic acid. Recently, it was revealed that kynurenic acid counteracts the harmful effects of homocysteine on endothelium cells in vitro. The hypothesis assuming homocysteine-kynurenate interplay suggests the existence of a new mechanism of atherogenesis and gives us an opportunity to use this knowledge in both prevention and treatment of cardiovascular diseases.


Asunto(s)
Hiperhomocisteinemia/tratamiento farmacológico , Ácido Quinurénico/uso terapéutico , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/complicaciones , Inflamación/complicaciones , Ácido Quinurénico/farmacocinética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control , Distribución Tisular
8.
Amino Acids ; 36(1): 75-80, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18231708

RESUMEN

Kynurenic acid (KYNA) is an endogenous antagonist of ionotropic glutamate receptors and the alpha 7 nicotinic acetylcholine receptor, showing anticonvulsant and neuroprotective activity. In this study, the presence of KYNA in food and honeybee products was investigated. KYNA was found in all 37 tested samples of food and honeybee products. The highest concentration of KYNA was obtained from honeybee products' samples, propolis (9.6 nmol/g), honey (1.0-4.8 nmol/g) and bee pollen (3.4 nmol/g). A high concentration was detected in fresh broccoli (2.2 nmol/g) and potato (0.7 nmol/g). Only traces of KYNA were found in some commercial baby products. KYNA administered intragastrically in rats was absorbed from the intestine into the blood stream and transported to the liver and to the kidney. In conclusion, we provide evidence that KYNA is a constituent of food and that it can be easily absorbed from the digestive system.


Asunto(s)
Abejas/química , Análisis de los Alimentos , Ácido Quinurénico/análisis , Animales , Ácido Quinurénico/farmacocinética , Masculino , Especificidad de Órganos/efectos de los fármacos , Ratas , Ratas Wistar
9.
Brain Res ; 1155: 42-8, 2007 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-17482581

RESUMEN

A very interesting and rapidly developing field of pain research is related to the roles of different endogenous ligands. This study determined the antinociceptive interactions of triple and quadruple combinations of different endogenous ligands (endomorphin-1, adenosine, agmatine and kynurenic acid) on carrageenan-induced inflammatory pain model at the spinal level. Intrathecal infusion (60 min) of these drugs alone, in double, triple or quadruple combinations, was followed by a 60-min observation period. During the infusion, antihyperalgesic effect of 0.3 microg/min endomorphin-1 was higher in the triple combinations than those in the double combinations. After cessation of drug administration, only the combination of 0.3 microg/min endomorphin-1, 1 microg/min agmatine, and 0.3 microg/min adenosine was more effective than the double combinations. In quadruple combinations, the antinociceptive effects of both 0.1 and 0.3 microg/min endomorphin-1 were significantly potentiated by the otherwise ineffective triple combination of adenosine, agmatine, and kynurenic acid. No side effects could be observed at these doses. These results demonstrate that triple and quadruple combinations of these endogenous ligands caused more effective antihyperalgesia compared with double combinations. Accordingly, the doses of these substances could be further reduced, thus, reinforcing the view that complex activation and/or inhibition of different systems can be sufficiently effective in blocking nociception without adverse effects. Because all of these drugs had effects on various receptors and systems, the possible types of these interactions were discussed.


Asunto(s)
Analgésicos/farmacología , Dolor , Médula Espinal/fisiología , Adenosina/administración & dosificación , Adenosina/farmacocinética , Adenosina/farmacología , Agmatina/administración & dosificación , Agmatina/farmacocinética , Agmatina/farmacología , Analgésicos/administración & dosificación , Analgésicos/farmacocinética , Animales , Área Bajo la Curva , Modelos Animales de Enfermedad , Combinación de Medicamentos , Inyecciones Espinales , Ácido Quinurénico/administración & dosificación , Ácido Quinurénico/farmacocinética , Ácido Quinurénico/farmacología , Ligandos , Masculino , Actividad Motora/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología , Receptores Opioides mu/efectos de los fármacos , Receptores Opioides mu/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología
10.
Eur J Neurosci ; 18(11): 2975-80, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14656293

RESUMEN

The current and calcium influx generated by NMDA receptors depend on the concentration of the coagonist glycine, or its analogue d-serine, in the synaptic cleft. If there is no release of glycine, the ionic stoichiometry of the glial GlyT1 glycine transporters expressed near NMDA receptors in the brain should be able to lower the extracellular glycine concentration to below the EC50 for coactivation of NMDA receptors. We examined whether changing the glycine or d-serine concentration in the superfusion solution altered the NMDA receptor mediated component of the synaptic current at the rat cerebellar mossy fibre to granule cell synapse. Adding up to 100 microM glycine or d-serine had no effect, implying that the glycine site is saturated. Using the competitive glycine site antagonist 7-chlorokynurenate, and plausible values for the kinetic parameters of NMDA receptors, we estimate that during activation of the mossy fibres the concentration of glycine or d-serine in the synaptic cleft is at least 4.6 microM or 1.5 microM, respectively, requiring active release of glycine or d-serine.


Asunto(s)
Cerebelo/citología , Glicina/metabolismo , Ácido Quinurénico/análogos & derivados , Fibras Nerviosas/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Animales , Animales Recién Nacidos , Unión Competitiva , Cerebelo/fisiología , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacocinética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Ácido Quinurénico/farmacocinética , Modelos Biológicos , Técnicas de Placa-Clamp/métodos , Ratas
11.
Eur J Pharmacol ; 471(2): 121-8, 2003 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-12818699

RESUMEN

Responding of rats was maintained under a 120-response fixed ratio (FR) schedule of food delivery, and animals received individual and combined injections of N-methyl-D-aspartic acid (NMDA), phencyclidine hydrochloride, (+)-MK-801 hydrogen maleate (MK-801), (+/-)-2-amino-5-phosphonopentanoic acid (AP5), 7-chlorokynurenic acid (7CK), ifenprodil tartrate, N(G)-nitro-L-arginine methyl ester hydorchloride (L-NAME), 7-nitroindazole, aminoguanidine hemisulfate, L-arginine, molsidomine, sodium nitroprusside, and 8-(diethylamino)octyl 3,4,5-trimethoxybenzoate hydrochloride (TMB-8). Behavioral suppression after NMDA was completely and dose-dependently reversed by MK-801, phencyclidine, AP5, and aminoguanidine; partially and dose-dependently attenuated by molsidomine, ifenprodil, and 7CK; and not attenuated at all by L-NAME, 7-nitroindazole, or TMB-8. These findings suggested that behavioral suppression after NMDA was associated with nitric oxide from the inducible synthase. In a second series of experiments, comparable behavioral suppression by 0.1 mg/kg MK-801, but not 3 mg/kg phencyclidine, was attenuated by nitroprusside, molsidomine, and L-arginine, suggesting that suppressions from MK-801 and phencyclidine were mediated by different final common pathways, and that behavioral suppression from MK-801, but not phencyclidine, may be associated with Ca(2+)-dependent nitric oxide.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Combinación de Medicamentos , Ácido Gálico/análogos & derivados , Ácido Quinurénico/análogos & derivados , Donantes de Óxido Nítrico/farmacocinética , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , 2-Amino-5-fosfonovalerato/antagonistas & inhibidores , Animales , Arginina/administración & dosificación , Arginina/farmacocinética , Maleato de Dizocilpina/administración & dosificación , Maleato de Dizocilpina/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ácido Gálico/administración & dosificación , Ácido Gálico/farmacocinética , Guanidinas/administración & dosificación , Guanidinas/farmacocinética , Indazoles/administración & dosificación , Indazoles/farmacocinética , Inyecciones Intraperitoneales , Ácido Quinurénico/administración & dosificación , Ácido Quinurénico/farmacocinética , Molsidomina/administración & dosificación , Molsidomina/farmacocinética , N-Metilaspartato/administración & dosificación , N-Metilaspartato/antagonistas & inhibidores , N-Metilaspartato/farmacocinética , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/farmacocinética , Donantes de Óxido Nítrico/administración & dosificación , Óxido Nítrico Sintasa/administración & dosificación , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/farmacocinética , Óxido Nítrico Sintasa de Tipo II , Nitroprusiato/administración & dosificación , Nitroprusiato/farmacocinética , Fenciclidina/administración & dosificación , Fenciclidina/farmacocinética , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos
12.
Eur J Pharmacol ; 445(1-2): 93-6, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-12065199

RESUMEN

Kynurenic acid as an endogenous ligand antagonizes all types of ionotropic glutamate receptors, with preferential affinity for the glycine-binding site of the N-methyl-D-aspartate (NMDA) receptor. The purpose of the present study was to investigate the antinociceptive potency of continuously administered kynurenic acid on carrageenan-induced thermal hyperalgesia by means of a paw withdrawal test in awake rats. The possible interaction between kynurenic acid and the endogenous mu-opioid receptor agonist peptide, endomorphin-1, was examined in the same set-up. Kynurenic acid at the higher doses (1-4 microg/min) significantly decreased the thermal hyperalgesia and increased the paw withdrawal latencies on the non-inflamed side. These doses were also associated with motor impairment on both sides. Low doses of kynurenic acid (0.01-0.1 microg/min) potentiated, but did not prolong, the antinociceptive effect of endomorphin-1 (0.1-1 microg/min) on the inflamed side. There was no sign of motor impairment during the combined treatment. These findings demonstrate that the combination of low doses of these two endogenous ligands provides effective and well-controlled antinociception without side effects.


Asunto(s)
Analgésicos/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Ácido Quinurénico/uso terapéutico , Oligopéptidos/uso terapéutico , Analgésicos/farmacocinética , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Sinergismo Farmacológico , Calor , Hiperalgesia/metabolismo , Bombas de Infusión , Inyecciones Espinales , Ácido Quinurénico/farmacocinética , Masculino , Oligopéptidos/farmacocinética , Ratas
13.
Br J Pharmacol ; 134(6): 1227-36, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11704642

RESUMEN

1. Magnesium (Mg)-deficient rats develop a mechanical hyperalgesia which is reversed by a N-Methyl-D-Aspartate (NMDA) receptor antagonist. Given that functioning of this receptor-channel is modulated by Mg, we wondered whether facilitated activation of NMDA receptors in Mg deficiency state may in turn trigger a cascade of specific intracellular events present in persistent pain. Hence, we tested several antagonists of NMDA and non-NMDA receptors as well as compounds interfering with the functioning of intracellular second messengers for effects on hyperalgesia in Mg-deficient rats. 2. Hyperalgesic Mg-deficient rats were administered intrathecally (10 microl) or intraperitoneally with different antagonists. After drug injection, pain sensitivity was evaluated by assessing the vocalization threshold in response to a mechanical stimulus (paw pressure test) over 2 h. 3. Intrathecal administration of MgSO4 (1.6, 3.2, 4.8, 6.6 micromol) as well as NMDA receptor antagonists such as MK-801 (0.6, 6.0, 60 nmol), AP-5 (10.2, 40.6, 162.3 nmol) and DCKA (0.97, 9.7, 97 nmol) dose-dependently reversed the hyperalgesia. Chelerythrine chloride, a protein kinase C (PKC) inhibitor (1, 10.4, 104.2 nmol) and 7-NI, a specific nitric oxide (NO) synthase inhibitor (37.5, 75, 150 micromol x kg(-1), i.p.) induced an anti-hyperalgesic effect in a dose-dependent manner. SR-140333 (0.15, 1.5, 15 nmol) and SR-48968 (0.17, 1.7, 17 nmol), antagonists of neurokinin receptors, produced a significant, but moderate, increase in vocalization threshold. 4. These results demonstrate that Mg-deficiency induces a sensitization of nociceptive pathways in the spinal cord which involves NMDA and non-NMDA receptors. Furthermore, the data is consistent with an active role of PKC, NO and, to a lesser extent substance P in the intracellular mechanisms leading to hyperalgesia.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacocinética , Ácido Quinurénico/análogos & derivados , Receptores de N-Metil-D-Aspartato/metabolismo , Columna Vertebral/metabolismo , 2-Amino-5-fosfonovalerato/farmacocinética , Alcaloides , Analgésicos/farmacocinética , Animales , Benzofenantridinas , Maleato de Dizocilpina/farmacocinética , Hiperalgesia/inducido químicamente , Indazoles/farmacocinética , Inyecciones Espinales , Ácido Quinurénico/farmacocinética , Sulfato de Magnesio/farmacología , Masculino , Neuronas/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Dimensión del Dolor , Fenantridinas/farmacocinética , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
14.
Neurosci Lett ; 304(3): 185-8, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11343833

RESUMEN

Peripheral administration of L-4-chlorokynurenine (4-Cl-KYN), which is enzymatically converted in astrocytes to form the glycine(B) receptor antagonist 7-chlorokynurenic acid (7-Cl-KYNA), has been shown to provide neuroprotection against excitotoxic damage. The present study was designed to examine the metabolic fate of 4-Cl-KYN after systemic injection, and to study its brain uptake and subsequent transamination during the acute phase following an excitotoxic insult. To this end, adult rats received intrastriatal injections of vehicle (1 microl) or quinolinic acid (QUIN) (240 nmol/1 microl), and were administered 50 mg/kg 4-Cl-KYN (intraperitoneally) immediately after surgery. After 90 and 180 min, 7-Cl-KYNA concentrations in the vehicle-injected striatum were 54+/-13 and 16+/-2 nM, respectively. The contralateral, QUIN-injected striatum contained 212+/-39 and 97+/-27 nM 7-Cl-KYNA, respectively. This injury-induced increase was accompanied by slightly higher 4-Cl-KYN levels in the QUIN-treated striatum, indicating that better pro-drug availability in part accounts for the enhanced 7-Cl-KYNA formation. These data demonstrate that systemic 4-Cl-KYN application, by targeting reactive glial cells during the early, reversible stage of excitotoxic neurodegeneration, produces disproportionately large amounts of the neuroprotectant 7-Cl-KYNA at the site of the emerging lesion.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/metabolismo , Quinurenina/análogos & derivados , Quinurenina/farmacología , Fármacos Neuroprotectores/farmacología , Neurotoxinas/farmacología , Profármacos/farmacología , Ácido Quinolínico/farmacología , Animales , Cuerpo Estriado/patología , Inyecciones Intraperitoneales , Ácido Quinurénico/farmacocinética , Masculino , Degeneración Nerviosa/metabolismo , Concentración Osmolar , Ratas , Ratas Sprague-Dawley
15.
J Drug Target ; 9(1): 23-37, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11378521

RESUMEN

In vitro studies have shown that conjugation of a model compound [p-di(hydroxyethyl)-amino-D-phenylalanine (D-MOD)] with L-Glu can improve D-MOD permeation through the bovine brain microvessel endothelial cell monolayers (Sakaeda et al., 2000). The transport of this D-MOD-L-Glu conjugate is facilitated by the L-Glu transport system. In this paper, we evaluate the in vivo brain delivery of model compounds (i.e. D-MOD, p-nitro-D-phenylalanine (p-nitro-D-Phe), 5,7-dichlorokynurenic acid (DCKA) and D-kyotorphin) and their L-Glu conjugates. DCKA was also conjugated with L-Asp and L-Gln amino acids. The analgesic activities of D-kyotorphin and its L-Glu conjugate were also evaluated. The results showed that the brain-to-plasma concentration ratio of D-MOD-L-Glu was higher than the D-MOD alone; however, the plasma concentration of both compounds were the same. The plasma concentration of p-nitro-D-Phe-L-Glu conjugate was higher than the parent p-nitro-D-Phe; however, the brain-to-plasma concentration ratio of p-nitro-D-Phe was higher than its conjugate. On the other hand, both DCKA and DCKA conjugates have a low brain-to-plasma concentration ratio due to their inability to cross the blood-brain barrier (BBB). The L-Asp and L-Glu conjugates of DCKA have elevated plasma concentrations relative to DCKA; however, the DCKA-L-Gln conjugate has the same plasma concentration as DCKA. For D-kyotorphin, both the parent and the L-Glu conjugate showed similar analgesic activity. In conclusion, conjugation of a non-permeable drug with L-Glu may improve the drug's brain delivery; however, this improvement may depend on the physicochemical and receptor binding properties of the conjugate.


Asunto(s)
Barrera Hematoencefálica , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos , Ácido Glutámico/farmacocinética , Profármacos/farmacocinética , Animales , Antineoplásicos Alquilantes/farmacocinética , Permeabilidad Capilar , Evaluación de Medicamentos , Endorfinas/farmacocinética , Ácido Glutámico/análogos & derivados , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/farmacocinética , Masculino , Melfalán/análogos & derivados , Melfalán/farmacocinética , Fenilalanina/análogos & derivados , Fenilalanina/farmacocinética , Ratas , Ratas Wistar
16.
Brain Res ; 860(1-2): 149-56, 2000 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-10727634

RESUMEN

We have synthesized D-glucose or D-galactose esters of 7-chlorokynurenic acid (7ClKynA) as prodrugs to facilitate the transport of 7ClKynA across the blood-brain barrier. Intraperitoneal (i.p.) administration of either 7ClKynA-D-glucopyranos-6'-ylester (7ClKynA/Glu6) or 7ClKynA-D-glucopyranos-3'-yl ester (7ClKynA/Glu3) was protective against seizures induced by N-methyl-D-aspartate (NMDA) in mice, with the former drug showing the highest anticonvulsive activity. Systemic injection of equal amounts of 7ClKynA-D-galactopyranos-6'-yl ester (7ClKynA/Gal6) or free 7ClKynA did not protect against NMDA seizures. Microdialysis in freely moving rats showed the presence of significant amounts of 7ClKynA/Glu6, as well as of 7ClKynA or KynA, in cortical perfusates after i.p. injections of 7ClKynA/Glu6. In contrast, only small amounts of 7ClKynA or KynA were detected after i.p. injection of unconjugated 7ClKynA. Prodrug metabolism has also been examined in mouse cortical cultures containing both neurons and astrocytes. 7ClKynA/Glu6 and 7ClKynA/Gal6 were rapidly metabolized into 7ClKynA and KynA, whereas 7ClKynA/Glu3 was metabolized with a slower kinetics. As a result of its conversion into 7ClKynA and KynA, 7ClKynA/Glu6 protected cortical neurons against NMDA toxicity. We conclude that sugar conjugates of 7ClKynA (and perhaps of other excitatory amino acid receptor antagonists) are prodrugs of potential interest in the experimental therapy of epilepsy and acute or chronic neurodegenerative disorders.


Asunto(s)
Anticonvulsivantes/farmacocinética , Epilepsias Parciales/tratamiento farmacológico , Epilepsia Tónico-Clónica/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacocinética , Glucosa/química , Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/metabolismo , Fármacos Neuroprotectores/farmacocinética , Profármacos/farmacocinética , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/síntesis química , Anticonvulsivantes/uso terapéutico , Astrocitos/efectos de los fármacos , Biotransformación , Barrera Hematoencefálica , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados , Epilepsias Parciales/inducido químicamente , Epilepsia Tónico-Clónica/inducido químicamente , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/síntesis química , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Galactosa/química , Inyecciones Intraperitoneales , Ácido Quinurénico/administración & dosificación , Ácido Quinurénico/química , Ácido Quinurénico/farmacocinética , Ácido Quinurénico/uso terapéutico , Masculino , Ratones , Microdiálisis , Estructura Molecular , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/uso terapéutico , Profármacos/síntesis química , Profármacos/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
17.
J Pharmacol Exp Ther ; 290(2): 496-504, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10411555

RESUMEN

Amantadine transport into renal proximal and distal tubules is bicarbonate dependent. In the present study, we addressed the effects of bicarbonate on renal clearance and urinary excretion of amantadine. Renal clearance of kynurenic acid was also studied to determine whether bicarbonate effects are specific for organic base transport by the kidney. After a moderate diuresis was established, animals received i.v. [(3)H]amantadine or [(3)H]kynurenic acid followed by an acute dose of sodium bicarbonate or physiological saline. Urine and blood samples were analyzed for [(3)H]amantadine or [(3)H]kynurenic acid, blood gases, and pH. Amantadine and kynurenic acid were excreted by the kidneys, and both compounds underwent renal tubular secretion. Amantadine metabolism occurred, and one metabolite was detected in the urine. In the bicarbonate-treated rats, the total amount of amantadine excreted in the urine was decreased, whereas the amount of metabolite recovered was similar in both groups. Bicarbonate treatment caused a sustained increase in blood bicarbonate levels, a mild increase in blood pH, and a decrease in amantadine renal clearance and in the amantadine/creatinine clearance ratio. Only a transient decrease in the renal clearance of kynurenic acid and the kynurenic acid/creatinine clearance ratio was observed. This study demonstrates that short-term changes in bicarbonate concentration may have significant effects on renal organic cation elimination. Coupled with our previous in vitro demonstration of bicarbonate-dependent organic cation transport, the present study suggests that bicarbonate inhibition of renal tubule organic cation secretion may explain the previous observation that bicarbonate dosing decreases amantadine excretion by the kidney.


Asunto(s)
Amantadina/orina , Bicarbonatos/metabolismo , Túbulos Renales/metabolismo , Riñón/metabolismo , Amantadina/farmacocinética , Animales , Transporte Biológico Activo , Análisis de los Gases de la Sangre , Cromatografía en Capa Delgada , Creatinina/orina , Concentración de Iones de Hidrógeno , Ácido Quinurénico/metabolismo , Ácido Quinurénico/farmacocinética , Masculino , Nefrectomía , Ratas , Ratas Sprague-Dawley
18.
Artículo en Inglés | MEDLINE | ID: mdl-10390731

RESUMEN

1. Kynurenic acid (KYNA) is a kynurenine metabolite and a broad spectrum excitatory amino acid antagonist that has been shown to be neuroprotective in models of cerebral ischemia, when administered exogenously. However, the actual concentration required in the CNS to evoke significant neuroprotection has never been assessed. 2. The purpose of this study was to address this question in the gerbil model of forebrain ischemia. KYNA (400-1600 mg/kg) or vehicle were administered i.p. 15 min before 5 min bilateral carotid occlusion. 3. Seven days after reperfusion, ischemia-induced hippocampal nerve cell loss (95% in vehicle-treated) was significantly lower in KYNA-treated gerbils (65% and 52% at 1000 and 1200 mg/Kg, respectively, P < 0.01). Treatment with 1000 mg/kg produced brain KYNA concentrations that were dramatically elevated (135.9 and 42.3 microM in CSF and whole brain, vs 0.032 and 0.16 microM in controls, at 15 min after ischemia), as measured in a separate group of transcardially-perfused gerbils. Cerebral KYNA concentrations tended to return to basal values 2 hours after reperfusion. 4. These results indicate that KYNA has a marked neuroprotective effect in a model of forebrain ischemia. This activity is associated with KYNA concentrations in the brain and CSF that are compatible with the in vitro affinity of the compound for ionotropic glutamate receptors.


Asunto(s)
Encéfalo/metabolismo , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/prevención & control , Ácido Quinurénico/farmacocinética , Ácido Quinurénico/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Gerbillinae , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Ataque Isquémico Transitorio/patología , Ácido Quinurénico/líquido cefalorraquídeo , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacocinética , Prosencéfalo , Reperfusión , Factores de Tiempo , Distribución Tisular
19.
Dev Neurosci ; 20(4-5): 408-16, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9778579

RESUMEN

Excitotoxic neuronal loss can be precipitated by defects in cerebral energy metabolism. Antagonists of excitatory amino acid receptors, such as the endogenous metabolite kynurenic acid, can effectively block excitotoxic lesions. Using brain tissue slices, the present study was designed to examine a potential link between metabolic status and cerebral kynurenic acid formation in adult rats. The results demonstrate that fluctuations in cerebral energy metabolism are closely associated with changes in kynurenic acid synthesis. Taken together, the data are compatible with the idea that the production of kynurenic acid in the brain is critically affected by the availability of pyruvate or other 2-oxoacids (i.e. co-substrates for the enzymatic transamination of the bioprecursor of kynurenic acid, L-kynurenine). Such metabolic control of kynurenic acid function may play a role in excitotoxic brain diseases


Asunto(s)
Encéfalo/metabolismo , Ácido Quinurénico/metabolismo , Animales , Encéfalo/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Electrofisiología , Metabolismo Energético/fisiología , Formazáns/metabolismo , Ácido Quinurénico/farmacocinética , Quinurenina/metabolismo , Masculino , Potasio/farmacología , Ácido Pirúvico/farmacología , Ácido Quinolínico/farmacología , Ratas , Ratas Sprague-Dawley , Sodio/fisiología , Especificidad por Sustrato , Sales de Tetrazolio/metabolismo , Veratridina/farmacología
20.
Pain ; 75(2-3): 247-55, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9583760

RESUMEN

Two classes of neurons with distinct responses to opioids have been identified in the rostral ventromedial medulla (RVM), a region with a well-documented role in nociceptive modulation. 'On-cells' are directly inhibited by opioids, and opioids can thus gain access to the modulatory circuitry of the RVM by an action on these neurons. 'Off-cells' are likely to exert a net inhibitory effect on nociceptive processing, and are activated by opioids. Because the opioid activation of off-cells is indirect, it has been proposed that on-cells function as inhibitory interneurons, and that opioid-induced suppression of on-cell firing in turn activates off-cells via disinhibition. The aim of the present study was to test this possibility. We had previously shown that excitatory amino acid (EAA) neurotransmission is crucial to the nocifensor reflex-related on-cell burst. We therefore infused the non-selective EAA receptor antagonist kynurenate (0.5-2 nmol, 200-500 nl) into the RVM while recording activity of on-, off- and neutral cells in lightly anesthetized rats. Kynurenate infusions produced a significant decrease in on-cell firing, with suppression of the on-cell burst. Off-cells nonetheless continued to display a tail flick-related pause in firing. Tail flick latency was used as an index of nociceptive responsiveness, and was unaffected by kynurenate infusions. These results demonstrate that a burst of on-cell firing is not required in order for the off-cell to exhibit a reflex-related pause in discharge, and do not support the proposed crucial role for on-cells as inhibitory interneurons within the RVM. In addition, preferential suppression of on-cell tiring was not associated with an increase in tail flick latency. This suggests that, under the conditions of these experiments, on-cell discharge is not a potent regulator of moment-to-moment variations in nociceptive responsiveness.


Asunto(s)
Aminoácidos Excitadores/metabolismo , Bulbo Raquídeo/metabolismo , Animales , Transporte Biológico/fisiología , Electrofisiología , Antagonistas de Aminoácidos Excitadores/farmacocinética , Ácido Quinurénico/farmacocinética , Masculino , Bulbo Raquídeo/citología , Bulbo Raquídeo/fisiología , Vías Nerviosas/fisiología , Neuronas/fisiología , Nociceptores/fisiología , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...