Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Microvasc Res ; 114: 19-25, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28546077

RESUMEN

The kynurenine pathway is a cascade of enzymatic steps generating biologically active compounds. l-kynurenine (l-KYN) is a central metabolite of tryptophan degradation. In the mammalian brain, l-KYN is partly converted to kynurenic acid (KYNA), which exerts multiple effects on neurotransmission. Recently, l-KYN or one of its derivatives were attributed a direct role in the regulation of the systemic circulation. l-KYN dilates arterial blood vessels during sepsis in rats, while it increases cerebral blood flow (CBF) in awake rabbits. Therefore, we hypothesized that acute elevation of systemic l-KYN concentration may exert potential effects on mean arterial blood pressure (MABP) and on resting CBF in the mouse brain. C57Bl/6 male mice were anesthetized with isoflurane, and MABP was monitored in the femoral artery, while CBF was assessed through the intact parietal bone with the aid of laser speckle contrast imaging. l-KYN sulfate (l-KYNs) (300mg/kg, i.p.) or vehicle was administered intraperitoneally. Subsequently, MABP and CBF were continuously monitored for 2.5h. In the control group, MABP and CBF were stable (69±4mmHg and 100±5%, respectively) throughout the entire data acquisition period. In the l-KYNs-treated group, MABP was similar to that, of control group (73±6mmHg), while hypoperfusion transients of 22±6%, lasting 7±3min occurred in the cerebral cortex over the first 60-120min following drug administration. In conclusion, the systemic high-dose of l-KYNs treatment destabilizes resting CBF by inducing a number of transient hypoperfusion events. This observation indicates the careful consideration of the dose of l-KYN administration by interpreting the effect of kynurenergic manipulation on brain function. By planning clinical trials basing on kynurenergic manipulation possible vascular side effects should also be considered.


Asunto(s)
Corteza Cerebral/irrigación sanguínea , Circulación Cerebrovascular/efectos de los fármacos , Trastornos Cerebrovasculares/inducido químicamente , Quinurenina/toxicidad , Sulfatos/toxicidad , Animales , Presión Arterial , Velocidad del Flujo Sanguíneo , Trastornos Cerebrovasculares/fisiopatología , Inyecciones Intraperitoneales , Quinurenina/administración & dosificación , Quinurenina/análogos & derivados , Flujometría por Láser-Doppler , Masculino , Ratones Endogámicos C57BL , Sulfatos/administración & dosificación , Factores de Tiempo
2.
Oxid Med Cell Longev ; 2015: 498401, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26301042

RESUMEN

Impaired function of certain mitochondrial respiratory complexes has long been linked to the pathogenesis of chronic neurodegenerative disorders such as Parkinson's and Huntington's diseases. Furthermore, genetic alterations of mitochondrial genome or nuclear genes encoding proteins playing essential roles in maintaining proper mitochondrial function can lead to the development of severe systemic diseases associated with neurodegeneration and vacuolar myelinopathy. At present, all of these diseases lack effective disease modifying therapy. Following a brief commemoration of Professor Albert Szent-Györgyi, a Nobel Prize laureate who pioneered in the field of cellular respiration, antioxidant processes, and the roles of free radicals in health and disease, the present paper overviews the current knowledge on the involvement of mitochondrial dysfunction in central nervous system diseases associated with neurodegeneration including Parkinson's and Huntington's disease as well as mitochondrial encephalopathies. The review puts special focus on the involvement and the potential therapeutic relevance of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α), a nuclear-encoded master regulator of mitochondrial biogenesis and antioxidant responses in these disorders, the transcriptional activation of which may hold novel therapeutic value as a more system-based approach aiming to restore mitochondrial functions in neurodegenerative processes.


Asunto(s)
Transporte de Electrón/fisiología , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Transporte de Electrón/genética , Metabolismo Energético , Humanos
3.
Nat Rev Drug Discov ; 12(1): 64-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23237916

RESUMEN

Various pathologies of the central nervous system (CNS) are accompanied by alterations in tryptophan metabolism. The main metabolic route of tryptophan degradation is the kynurenine pathway; its metabolites are responsible for a broad spectrum of effects, including the endogenous regulation of neuronal excitability and the initiation of immune tolerance. This Review highlights the involvement of the kynurenine system in the pathology of neurodegenerative disorders, pain syndromes and autoimmune diseases through a detailed discussion of its potential implications in Huntington's disease, migraine and multiple sclerosis. The most effective preclinical drug candidates are discussed and attention is paid to currently under-investigated roles of the kynurenine pathway in the CNS, where modulation of kynurenine metabolism might be of therapeutic value.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Quinurenina/metabolismo , Triptófano/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/fisiopatología , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Tolerancia Inmunológica , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Dolor/tratamiento farmacológico , Dolor/fisiopatología
4.
Neuropharmacology ; 61(5-6): 1026-32, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21781978

RESUMEN

Postconditioning can be induced by a broad range of stimuli within minutes to days after an ischemic cerebral insult. A special form is elicited by pharmacological intervention called second pathophysiological stress. The present study aimed to evaluate the effects of low-dose (5 mg/kg) kainate postconditioning with onsets 0, 24 and 48 h after the ischemic insult on the hippocampal synaptic plasticity in a 2-vessel occlusion model in rat. The hippocampal function was tested by LTP measurements of Schaffer collateral-CA1 pyramidal cell synapses in acute slices and the changes in density of Golgi-Cox-stained apical dendritic spines. Postconditioning 0 and 24 h after ischemia was not protective, whereas 48-h-onset postconditioning resulted in the reappearance of a normal spine density (>100,000 spines) 3 days after ischemia, in parallel with the long-term restoration of the damaged LTP function. Similar, but somewhat less effects were observed after 10 days. Our data clearly demonstrate the onset dependence of postconditioning elicited by a subconvulsant dose of kainate treatment in global ischemia, with restoration of the structural plasticity and hippocampal function.


Asunto(s)
Región CA1 Hipocampal/fisiología , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Animales , Región CA1 Hipocampal/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Electroencefalografía , Fenómenos Electrofisiológicos/efectos de los fármacos , Fenómenos Electrofisiológicos/fisiología , Ataque Isquémico Transitorio/mortalidad , Ataque Isquémico Transitorio/fisiopatología , Potenciación a Largo Plazo/fisiología , Masculino , Terapia Molecular Dirigida , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Distribución Aleatoria , Ratas , Ratas Wistar , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/fisiología
5.
J Pharm Biomed Anal ; 55(3): 540-3, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21392919

RESUMEN

The changes in concentration of kynurenic acid (KYNA) in different biological samples are of great interest in the pathomechanism and medication of several disorders, and especially those affecting the nervous system. Besides the recent pharmaceutical advances targeting the kynurenine pathway, there is a constant need for further drug development through the synthesis of novel analogs. Reliable analytical methods should be set up to monitor the metabolism and effects of these analogs in both preclinical experiments and human studies. Following a sample preparation procedure based on protein precipitation, new high-performance liquid chromatographic methods with fluorescence and mass spectrometric detection were developed for the determination of KYNA and a novel KYNA analog (N-(2-N,N-dimethylaminoethyl)-4-oxo-1H-quinoline-2-carboxamide hydrochloride; KYNA amide) in mouse serum samples. The analytical parameters obtained in the validation procedure suggest that the developed method with mass spectrometric detection is simple, fast, accurate and suitable for the measurement of KYNA and its analogs. The results reveal the good in vivo stability of the novel KYNA amide.


Asunto(s)
Ácido Quinurénico/análogos & derivados , Ácido Quinurénico/sangre , Animales , Calibración , Cromatografía Líquida de Alta Presión , Evaluación Preclínica de Medicamentos , Femenino , Inyecciones Intraperitoneales , Ácido Quinurénico/administración & dosificación , Límite de Detección , Masculino , Ratones , Ratones Endogámicos C57BL , Estándares de Referencia , Reproducibilidad de los Resultados , Factores de Tiempo
6.
Cell Mol Neurobiol ; 29(6-7): 827-35, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19259807

RESUMEN

A traumatic brain injury or a focal brain lesion is followed by acute excitotoxicity caused by the presence of abnormally high glutamate (Glu) levels in the cerebrospinal and interstitial fluids. It has recently been demonstrated that this excess Glu in the brain can be eliminated into the blood following the intravenous administration of oxaloacetate (OxAc), which, by scavenging the blood Glu, induces an enhanced and neuroprotective brain-to-blood Glu efflux. In this study, we subjected rats to a photothrombotic lesion and treated them after the illumination with a single 30-min-long administration of OxAc (1.2 mg/100 g, i.v.). Following induction of the lesion, we measured the infarct size and the amplitudes of the somatosensory evoked potentials (SEPs) as recorded from the skull surface. The photothrombotic lesion resulted in appreciably decreased amplitudes of the evoked potentials, but OxAc administration significantly attenuated this reduction, and also the infarct size assessed histologically. We suggest that the neuroprotective effects of OxAc are due to its blood Glu-scavenging activity, which, by increasing the brain-to-blood Glu efflux, reduces the excess Glu responsible for the anatomical and functional correlates of the ischemia, as evaluated by electrophysiological evoked potential (EP) measurements.


Asunto(s)
Infarto Cerebral/tratamiento farmacológico , Potenciales Evocados Somatosensoriales/fisiología , Ácido Oxaloacético/uso terapéutico , Corteza Somatosensorial/patología , Corteza Somatosensorial/fisiopatología , Animales , Infarto Cerebral/inducido químicamente , Infarto Cerebral/patología , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Fluoresceínas , Masculino , Compuestos Orgánicos , Ratas , Rosa Bengala/toxicidad , Corteza Somatosensorial/irrigación sanguínea
7.
Cell Mol Neurobiol ; 26(7-8): 1505-19, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16758322

RESUMEN

1. Dehydroepiandrosterone (DHEA) and its sulfate (DHEAS) are sex hormone precursors which exert marked neurotrophic and/or neuroprotective activity in the central nervous system (CNS). 2. In the present electrophysiological experiments, we studied the effects of peripherally administered DHEAS on responses of the primary somatosensory (SSI) and motor cortices (MI) of (i) anesthetized controls and (ii) MI focal cold-lesioned rats. (iii) The effects of DHEAS on the field excitatory postsynaptic potentials (fEPSPs) were also studied in vitro brain slices. DHEAS (50 mg/kg) was injected subcutaneously 12 h before and immediately after cold lesion induction. The anesthetized rats were fixed in a stereotaxic frame, the SSI and MI were exposed, and control SSI and MI responses were evoked by contralateral whisker pad stimulation. After registration of the evoked responses for a 35-min period, a copper cylinder (2 mm in diameter) cooled with a mixture of acetone and dry ice (-78 degrees C) was applied to produce a lesion in the MI and the registration of the evoked responses was then continued for an additional 360 min. 3. In the controls, DHEAS administration resulted in slight increases in amplitude of both the SSI and the MI responses. After focal cold lesion induction, the most significant reduction in amplitude was observed at the focus of the lesion in the primary MI, but the amplitudes of the SSI responses were also decreased. After 3-5 h of lesion induction, the amplitudes started to increase around the injury in the primary MI, while the SSI response had already started to recover 2 h after induction of the MI lesion. In the course of the postlesion recovery period, the MI responses peripherally to the center of the lesion frequently exhibited extremely high and low amplitudes. The paired-pulse paradigm revealed changing, but basically high levels of disinhibition and facilitation in extended cortical areas after focal cortical cold lesion induction. The deviations (e.g., the extremely augmented responses) in cortical functioning of the anesthetized rats were unambiguously diminished by DHEAS administration, and the period required for the cortical responses to recover was significantly shorter after the steroid treatment. In the in vitro studies, however, DHEAS administration resulted in an enhanced level of disinhibition in extended cortical areas of both the hemispheres. 4. This observation draws attention to the possible differences between the results obtained in different models (in vitro vs. in situ). Nevertheless, all the presented data suggest that DHEAS treatment might have neuroprotective effect on the neocortex at least at a short-time scale.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Sulfato de Deshidroepiandrosterona/farmacología , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Corteza Motora/efectos de los fármacos , Animales , Lesiones Encefálicas/tratamiento farmacológico , Sulfato de Deshidroepiandrosterona/administración & dosificación , Evaluación Preclínica de Medicamentos , Inyecciones Subcutáneas , Masculino , Modelos Biológicos , Corteza Motora/fisiología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Corteza Somatosensorial/efectos de los fármacos
8.
Endocrinology ; 147(2): 683-6, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16293666

RESUMEN

Dehydroepiandrosterone and its sulfate (DHEAS) are sex hormone precursors that exert marked neurotrophic and/or neuroprotective activity in the central nervous system. The present study evaluated the effects of DHEAS and 17beta-estradiol (E2) in a focal cortical cold lesion model, in which DHEAS (50 mg/kg, sc) and E2 (35 mg/kg, sc) were administered either as pretreatment (two subsequent injections 1 d and 1 h before lesion induction) or posttreatment (immediately after lesion induction). The focal cortical cold lesion was induced in the primary motor cortex by means of a cooled copper cylinder placed directly onto the cortical surface. One hour later, the animals were killed, the brains cut into 0.4-mm-thick slices, and the sections stained with 1% triphenyltetrazolium chloride. The volume of the hemispheric lesion was calculated for each animal. The results demonstrated that the lesion area was significantly attenuated in both the DHEAS- and E2- pre- and posttreated groups and that in the presence of letrozole, a nonsteroidal aromatase inhibitor, no neuroprotection was observed, suggesting that the beneficial effect of DHEAS on the cold injury might depend on the conversion of DHEAS to E2 within the brain. It is concluded that even a single posttraumatic administration of DHEAS may be of substantial therapeutic benefit in the treatment of focal brain injury with vasogenic edema.


Asunto(s)
Lesiones Encefálicas/prevención & control , Frío , Deshidroepiandrosterona/administración & dosificación , Estradiol/fisiología , Corteza Motora/lesiones , Fármacos Neuroprotectores/administración & dosificación , Animales , Lesiones Encefálicas/etiología , Deshidroepiandrosterona/metabolismo , Esquema de Medicación , Estradiol/administración & dosificación , Masculino , Modelos Animales , Corteza Motora/efectos de los fármacos , Corteza Motora/metabolismo , Fármacos Neuroprotectores/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA