Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Food Funct ; 14(19): 8893-8902, 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37701930

RESUMEN

Increasing evidence suggests that dietary (poly)phenols and methylxanthines have neuroprotective effects; however, little is known about whether they can cross the blood-brain barrier (BBB) and exert direct effects on the brain. We investigated the presence of (poly)phenol and methylxanthine metabolites in plasma and cerebrospinal fluid (CSF) from 90 individuals at risk of dementia using liquid chromatography-mass spectrometry and predicted their mechanism of transport across the BBB using in silico modelling techniques. A total of 123 and 127 metabolites were detected in CSF and plasma, respectively. In silico analysis suggests that 5 of the 20 metabolites quantified in CSF can cross the BBB by passive diffusion, while at least 9 metabolites require the aid of cell transporters to cross the BBB. Our results showed that (poly)phenols and methylxanthines are bioavailable, can cross the BBB via passive diffusion or transport carriers, and can reach brain tissues to exert neuroprotective effects.


Asunto(s)
Barrera Hematoencefálica , Fármacos Neuroprotectores , Fenoles , Xantinas , Humanos , Barrera Hematoencefálica/metabolismo , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fármacos Neuroprotectores/metabolismo , Fenol , Fenoles/líquido cefalorraquídeo , Fenoles/metabolismo , Xantinas/líquido cefalorraquídeo , Xantinas/metabolismo
2.
Neurology ; 95(24): e3428-e3437, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32999056

RESUMEN

OBJECTIVE: To identify markers of resistance to developing Parkinson disease (PD) among LRRK2 mutation carriers (LRRK2+), we carried out metabolomic profiling in individuals with PD and unaffected controls (UC), with and without the LRRK2 mutation. METHODS: Plasma from 368 patients with PD and UC in the LRRK2 Cohort Consortium (LCC), comprising 118 LRRK2+/PD+, 115 LRRK2+/UC, 70 LRRK2-/PD+, and 65 LRRK2-/UC, and CSF available from 68 of them, were analyzed by liquid chromatography with mass spectrometry. For 282 analytes quantified in plasma and CSF, we assessed differences among the 4 groups and interactions between LRRK2 and PD status, using analysis of covariance models adjusted by age, study site cohort, and sex, with p value corrections for multiple comparisons. RESULTS: Plasma caffeine concentration was lower in patients with PD vs UC (p < 0.001), more so among LRRK2+ carriers (by 76%) than among LRRK2- participants (by 31%), with significant interaction between LRRK2 and PD status (p = 0.005). Similar results were found for caffeine metabolites (paraxanthine, theophylline, 1-methylxanthine) and a nonxanthine marker of coffee consumption (trigonelline) in plasma, and in the subset of corresponding CSF samples. Dietary caffeine was also lower in LRRK2+/PD+ compared to LRRK2+/UC with significant interaction effect with the LRRK2+ mutation (p < 0.001). CONCLUSIONS: Metabolomic analyses of the LCC samples identified caffeine, its demethylation metabolites, and trigonelline as prominent markers of resistance to PD linked to pathogenic LRRK2 mutations, more so than to idiopathic PD. Because these analytes are known both as correlates of coffee consumption and as neuroprotectants in animal PD models, the findings may reflect their avoidance by those predisposed to develop PD or their protective effects among LRRK2 mutation carriers.


Asunto(s)
Alcaloides/sangre , Cafeína/sangre , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Fármacos Neuroprotectores/sangre , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/genética , Anciano , Alcaloides/líquido cefalorraquídeo , Cafeína/líquido cefalorraquídeo , Cromatografía Liquida , Estudios de Cohortes , Femenino , Heterocigoto , Humanos , Masculino , Espectrometría de Masas , Metabolómica , Persona de Mediana Edad , Fármacos Neuroprotectores/líquido cefalorraquídeo , Enfermedad de Parkinson/líquido cefalorraquídeo , Teofilina/sangre , Teofilina/líquido cefalorraquídeo , Xantinas/sangre , Xantinas/líquido cefalorraquídeo
3.
J Clin Lipidol ; 14(5): 675-684.e2, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32758395

RESUMEN

BACKGROUND: The role of cholesterol homeostasis in neuroaxonal injury in multiple sclerosis is not known. OBJECTIVE: The objective of the study is to investigate the associations of cerebrospinal fluid (CSF) and serum neurofilament light chain levels (CSF-NfL and sNfL, respectively), which are biomarkers of neuroaxonal injury, with cholesterol biomarkers at the clinical onset of multiple sclerosis. METHODS: sNfL, serum cholesterol profile (total cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol), serum apolipoprotein (Apo) levels (ApoA-I, ApoA-II, ApoB, and ApoE), and albumin quotient were obtained for 133 patients (63% female, age: 29.9 ± 8.0 years) during the first demyelinating event. CSF-NfL was available for 103 (77%) patients. RESULTS: CSF-NfL and sNfL were negatively associated with serum ApoA-II (P = .005, P < .001) and positively associated with albumin quotient (P < .001, P < .0001). In addition, higher CSF-NfL was associated with lower serum ApoA-I (P = .009) levels and higher sNfL was associated with lower high-density lipoprotein cholesterol (P = .010). In stepwise regression, age (P = .045), serum ApoA-II (P = .022), and albumin quotient (P < .001) were associated with CSF-NfL; albumin quotient (P = .002) and ApoA-II (P = .001) were associated with sNfL. Path analysis identified parallel pathways from ApoA-II (P = .009) and albumin quotient (P < .001) to the sNfL outcome that were mediated by CSF-NfL (P < .001). The associations of CSF-NfL with ApoA-I (P = .014) and ApoA-II (P = .015) and sNfL with ApoA-II (P < .001) remained significant after adjusting for number of contrast-enhancing lesions and T2 lesion volume. CONCLUSION: Lower serum ApoA-II and ApoA-I levels are associated with greater neuroaxonal injury as measured by CSF-NfL.


Asunto(s)
Apolipoproteína A-II/sangre , Apolipoproteína A-I/sangre , Esclerosis Múltiple/sangre , Esclerosis Múltiple/líquido cefalorraquídeo , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Adulto , Femenino , Humanos , Estudios Longitudinales , Masculino , Esclerosis Múltiple/patología , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Pronóstico , Estudios Prospectivos
4.
Nutrients ; 9(10)2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28961195

RESUMEN

Choline is involved in relevant neurochemical processes. In particular, it is the precursor and metabolite of acetylcholine (ACh). Choline is an essential component of different membrane phospholipids that are involved in intraneuronal signal transduction. On the other hand, cholinergic precursors are involved in ACh release and carry out a neuroprotective effect based on an anti-inflammatory action. Based on these findings, the present study was designed to evaluate the effects of choline and choline precursor (Choline alphoscerate, GPC) in the modulation of inflammatory processes in the rat brain. Male Wistar rats were intraperitoneally treated with 87 mg of choline chloride/kg/day (65 mg/kg/day of choline), and at choline-equivalent doses of GPC (150 mg/kg/day) and vehicle for two weeks. The brains were dissected and used for immunochemical and immunohistochemical analysis. Inflammatory cytokines (Interleukin-1ß, IL-1ß; Interleukin-6 , IL-6 and Tumor Necrosis Factor-α, TNF-α) and endothelial adhesion molecules (Intercellular Adhesion Molecule, ICAM-1 and Vascular cell Adhesion Molecule, VCAM-1) were studied in the frontal cortex, hippocampus, and cerebellum. The results clearly demonstrated that treatment with choline or GPC did not affect the expression of the inflammatory markers in the different cerebral areas evaluated. Therefore, choline and GPC did not stimulate the inflammatory processes that we assessed in this study.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Corteza Cerebral/efectos de los fármacos , Colina/uso terapéutico , Encefalitis/prevención & control , Glicerilfosforilcolina/uso terapéutico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/efectos adversos , Biomarcadores/metabolismo , Cerebelo/efectos de los fármacos , Cerebelo/inmunología , Cerebelo/metabolismo , Cerebelo/patología , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Colina/administración & dosificación , Colina/efectos adversos , Citocinas/metabolismo , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalitis/patología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/inmunología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Glicerilfosforilcolina/administración & dosificación , Glicerilfosforilcolina/efectos adversos , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Inyecciones Intraperitoneales , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/líquido cefalorraquídeo , Ratas Wistar , Molécula 1 de Adhesión Celular Vascular/metabolismo
5.
Purinergic Signal ; 12(1): 149-59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26695181

RESUMEN

In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Guanosina/administración & dosificación , Guanosina/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Administración Intranasal , Animales , Conducta Animal , Isquemia Encefálica/psicología , Infarto Cerebral/patología , Infarto Cerebral/prevención & control , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Guanosina/líquido cefalorraquídeo , Guanosina/farmacocinética , Masculino , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fármacos Neuroprotectores/farmacocinética , Ratas , Ratas Wistar , Accidente Cerebrovascular/psicología
6.
Neurocrit Care ; 22(2): 306-19, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25231529

RESUMEN

BACKGROUND: Patients recovering from aneurysmal subarachnoid hemorrhage (SAH) are at risk for developing delayed cerebral ischemia (DCI). Experimental and human studies implicate the vasoconstrictor P450 eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE) in the pathogenesis of DCI. To date, no studies have evaluated the role of vasodilator epoxyeicosatrienoic acids (EETs) in DCI. METHODS: Using mass spectrometry, we measured P450 eicosanoids in cerebrospinal fluid (CSF) from 34 SAH patients from 1 to 14 days after admission. CSF eicosanoid levels were compared in patients who experienced DCI versus those who did not. We then studied the effect of EETs in a model of SAH using mice lacking the enzyme soluble epoxide hydrolase (sEH), which catabolizes EETs into their inactive diol. To assess changes in vessel morphology and cortical perfusion in the mouse brain, we used optical microangiography, a non-invasive coherence-based imaging technique. RESULTS: Along with increases in 20-HETE, we found that CSF levels of 14,15-EET were elevated in SAH patients compared to control CSF, and levels were significantly higher in patients who experienced DCI compared to those who did not. Mice lacking sEH had elevated 14,15-EET and were protected from the delayed decrease in microvascular cortical perfusion after SAH, compared to wild type mice. CONCLUSIONS: Our findings suggest that P450 eicosanoids play an important role in the pathogenesis of DCI. While 20-HETE may contribute to the development of DCI, 14,15-EET may afford protection against DCI. Strategies to enhance 14,15-EET, including sEH inhibition, should be considered as part of a comprehensive approach to prevent DCI.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Isquemia Encefálica/líquido cefalorraquídeo , Ácidos Hidroxieicosatetraenoicos/líquido cefalorraquídeo , Fármacos Neuroprotectores/líquido cefalorraquídeo , Hemorragia Subaracnoidea/líquido cefalorraquídeo , Ácido 8,11,14-Eicosatrienoico/líquido cefalorraquídeo , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad
7.
J Neurotrauma ; 30(17): 1484-9, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23540442

RESUMEN

Cyclosporin A (CsA), an immunosuppressive medication traditionally used in the prevention of post-transplant rejection, is a promising neuroprotective agent for traumatic brain injury (TBI). Preliminary studies in animals and humans describe the efficacy and safety of CsA when administered following neurotrauma. The objective of this study is to describe CsA exposure in adults with severe TBI by assessing concentrations in whole blood, cerebrospinal fluid (CSF), and brain extracellular fluid (ECF) dialysate as measured by brain microdialysis. Severe TBI patients were enrolled in a randomized controlled trial following the written informed consent of their legal guardians. Patients received either CsA 5 mg/kg as a continuous infusion over 24 h, or matching placebo. Noncompartmental exposure analyses were performed using CsA concentrations in whole blood, CSF, and ECF dialysate. There were 37 patients randomized to the CsA arm of the trial and included in this exposure analysis. CsA was detected in the ECF dialysate and CSF at a fraction of the whole blood concentration. Mean CsA maximum concentrations were achieved at 24 and 30 h from the start of the 24 h infusion, in the CSF and ECF dialysate, respectively. A correlation was found between ECF dialysate and CSF concentrations. CsA was detected in the blood, CSF, and brain ECF dialysate. CsA exposure characteristic differences exist for whole blood, CSF, and ECF dialysate in severe TBI patients when administered as a continuous intravenous infusion. These exposure characteristics should be used for safer CsA dose optimization to achieve target CsA concentrations for neuroprotection in future TBI studies.


Asunto(s)
Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Encéfalo/metabolismo , Ciclosporina/sangre , Ciclosporina/líquido cefalorraquídeo , Líquido Extracelular/metabolismo , Adolescente , Adulto , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Ciclosporina/administración & dosificación , Femenino , Humanos , Infusiones Intravenosas , Masculino , Microdiálisis/métodos , Persona de Mediana Edad , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Estudios Prospectivos , Ventriculostomía/métodos , Adulto Joven
8.
Eur J Pharm Sci ; 48(4-5): 595-603, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23354153

RESUMEN

The aim of this study was to develop novel multiple agents loaded poly (D,L-lactide-co-glycolide acid) (PLGA) nanoparticles (NPs) and evaluate their potential for brain delivery via inner ear administration. PLGA NPs loaded with salvianolic acid B (Sal B), tanshinone IIA (TS IIA) and panax notoginsenoside (PNS) were prepared by double emulsion/solvent evaporation method. It was observed that optimized NPs displayed satisfactory encapsulation efficiency and desired sustained-release characteristics. NPs following intratympanic administration (IT) in guinea pigs greatly improved drug distribution within the inner ear, cerebrospinal fluid (CSF) and brain tissues compared with intravenous administration (IV). Pharmacodynamic studies demonstrated that NPs following IT markedly inhibited oxidizing reactions and protected the brain from cerebral ischemia reperfusion (I/R) injury by upregulating superoxide dismutase (SOD) activity both in serum and brain tissues, simultaneously significantly reducing the levels of malondialdehyde (MDA) and nitric oxide synthase (NOS). Moreover intratympanic delivery did not cause injury of cochlear function by preliminary study on the toxicity. These findings suggested that PLGA NPs-based delivery system via inner ear administration was a promising candidate to brain delivery for the treatment of brain diseases.


Asunto(s)
Abietanos/administración & dosificación , Benzofuranos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Ginsenósidos/administración & dosificación , Nanopartículas/administración & dosificación , Daño por Reperfusión/tratamiento farmacológico , Abietanos/líquido cefalorraquídeo , Abietanos/farmacocinética , Animales , Benzofuranos/líquido cefalorraquídeo , Benzofuranos/farmacocinética , Encéfalo/metabolismo , Arterias Carótidas , Vías de Administración de Medicamentos , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Oído Interno , Ginsenósidos/líquido cefalorraquídeo , Ginsenósidos/farmacocinética , Cobayas , Ácido Láctico/administración & dosificación , Ácido Láctico/química , Masculino , Nanopartículas/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fármacos Neuroprotectores/farmacocinética , Panax notoginseng , Tamaño de la Partícula , Ácido Poliglicólico/administración & dosificación , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Daño por Reperfusión/metabolismo
9.
Leuk Res ; 35(11): 1467-71, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21846577

RESUMEN

Brain damage related to intrathecal methotrexate in children with acute lymphoblastic leukemia (ALL) is still unclear. Neuroinflammatory mechanisms and intracerebral production of specific biomarkers, play a key role in determining neuroprotective mechanisms after brain injury. To determine whether the CSF concentrations of neuron-specific enolase (NSE), neurotrophic factors and doublecortin (DCX) are influenced by repeated intrathecal methotrexate administrations, we prospectively collected CSF samples from 10 children with ALL and 10 controls. Our results showed an increased expression of the liquoral markers. This up-regulation could be interpreted as a neuroprotective response of the brain against the neuronal damages induced by MTX.


Asunto(s)
Lesiones Encefálicas/diagnóstico , Metotrexato/efectos adversos , Proteínas Asociadas a Microtúbulos/líquido cefalorraquídeo , Factores de Crecimiento Nervioso/líquido cefalorraquídeo , Neuropéptidos/líquido cefalorraquídeo , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fosfopiruvato Hidratasa/líquido cefalorraquídeo , Leucemia-Linfoma Linfoblástico de Células Precursoras/líquido cefalorraquídeo , Adolescente , Antimetabolitos Antineoplásicos/efectos adversos , Biomarcadores de Tumor/líquido cefalorraquídeo , Western Blotting , Lesiones Encefálicas/líquido cefalorraquídeo , Lesiones Encefálicas/inducido químicamente , Niño , Preescolar , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Estudios Prospectivos , Regulación hacia Arriba
10.
J Inorg Biochem ; 105(8): 1066-72, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21726769

RESUMEN

Alzheimer's disease is one of the most common forms of dementia in the elderly. One of its hallmarks is the abnormal aggregation and deposition of ß-amyloid (Aß). Endogenous and exogenous metal ions seem to influence ß-amyloid folding process, aggregation and deposition. Besides these variables other elements appear to affect ß-amyloid behavior, such as cholesterol. The physiological concentration of cholesterol in the cerebrospinal fluid (CSF) was used in order to determine the extent in which Aß and Aß-metal complexes in vitro aggregation and their toxicity on human neuroblastoma cell cultures is affected. Cholesterol did not appear to influence Aß and Aß-metal complexes aggregation, but it was effective in protecting neuroblastoma cells against Aß complexes' toxicity. The Aß-Al complex seemed to be the most effective in disrupting and damaging membrane external layer, and simultaneously it appears to increase its toxicity on cell cultures; both of these effects are preventable by cholesterol. The presence in physiological concentrations of cholesterol seemed to compensate membrane damage that occurred to neuroblastoma cells. These findings appear to contradict some data reported in literature. We believe that our results might shed some light on the role played by cholesterol at physiological concentrations in both cellular balance and membrane protection.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Colesterol/farmacología , Complejos de Coordinación/toxicidad , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Colesterol/líquido cefalorraquídeo , Complejos de Coordinación/metabolismo , Humanos , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fosfolípidos/metabolismo , Difracción de Rayos X
11.
J Neurovirol ; 17(3): 258-73, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21556959

RESUMEN

To begin to unravel the complexity of HIV-associated changes in the brain, broader, multifaceted analyses of cerebrospinal fluid (CSF) are needed that examine a wide range of proteins reflecting different functions. To provide the first broad profiles of protein changes in the CSF of HIV-infected patients, we used antibody arrays to measure 120 cytokines, chemokines, growth factors, and other proteins. CSF from HIV-infected patients with a range of cognitive deficits was compared to CSF from uninfected, cognitively normal patients to begin to identify protein changes associated with HIV infection and neurological disease progression. Uninfected patients showed relatively consistent patterns of protein expression. Highly expressed proteins in CSF included monocyte chemotactic protein-1, tissue inhibitors of metalloproteases, granulocyte colony-stimulating factor, adiponectin, soluble tumor necrosis factor receptor-1, urokinase-type plasminogen activator receptor, and insulin-like growth factor binding protein-2. Inflammatory and anti-inflammatory cytokines were expressed at low levels. HIV-infected patients showed increases in inflammatory proteins (interferon-gamma, tumor necrosis factor-alpha), anti-inflammatory proteins (IL-13), and chemokines but these correlated poorly with neurological status. The strongest correlation with increasing severity of neurological disease was a decline in growth factors, particularly, brain-derived neurotrophic factor and NT-3. These studies illustrate that HIV infection is associated with parallel changes in both inflammatory and neuroprotective proteins in the CSF. The inverse relationship between growth factors and neurological disease severity suggests that a loss of growth factor neuroprotection may contribute to the development of neural damage and may provide useful markers of disease progression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/líquido cefalorraquídeo , Trastornos del Conocimiento/psicología , Citocinas/líquido cefalorraquídeo , Fármacos Neuroprotectores/líquido cefalorraquídeo , Neurotrofina 3/líquido cefalorraquídeo , Encéfalo/patología , Encéfalo/virología , Trastornos del Conocimiento/líquido cefalorraquídeo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/virología , Femenino , Factor Estimulante de Colonias de Granulocitos/líquido cefalorraquídeo , VIH/fisiología , Infecciones por VIH/líquido cefalorraquídeo , Infecciones por VIH/patología , Infecciones por VIH/virología , Humanos , Inflamación/líquido cefalorraquídeo , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Análisis por Matrices de Proteínas , Receptores Tipo I de Factores de Necrosis Tumoral/líquido cefalorraquídeo , Índice de Severidad de la Enfermedad , Activador de Plasminógeno de Tipo Uroquinasa/líquido cefalorraquídeo
12.
J Cereb Blood Flow Metab ; 31(2): 439-47, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20628399

RESUMEN

The naturally occurring antagonist of interleukin-1, IL-1RA, is highly neuroprotective experimentally, shows few adverse effects, and inhibits the systemic acute phase response to stroke. A single regime pilot study showed slow penetration into cerebrospinal fluid (CSF) at experimentally therapeutic concentrations. Twenty-five patients with subarachnoid hemorrhage (SAH) and external ventricular drains were sequentially allocated to five administration regimes, using intravenous bolus doses of 100 to 500 mg and 4 hours intravenous infusions of IL-1RA ranging from 1 to 10 mg per kg per hour. Choice of regimes and timing of plasma and CSF sampling was informed by pharmacometric analysis of pilot study data. Data were analyzed using nonlinear mixed effects modeling. Plasma and CSF concentrations of IL-1RA in all regimes were within the predicted intervals. A 500-mg bolus followed by an intravenous infusion of IL-1RA at 10 mg per kg per hour achieved experimentally therapeutic CSF concentrations of IL-1RA within 45 minutes. Experimentally, neuroprotective CSF concentrations in patients with SAH can be safely achieved within a therapeutic time window. Pharmacokinetic analysis suggests that IL-1RA transport across the blood-CSF barrier in SAH is passive. Identification of the practicality of this delivery regime allows further studies of efficacy of IL-1RA in acute cerebrovascular disease.


Asunto(s)
Sistema Nervioso Central/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacocinética , Fármacos Neuroprotectores/farmacocinética , Hemorragia Subaracnoidea/metabolismo , Adulto , Anciano , Ventrículos Cerebrales/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Infusiones Intravenosas , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Proteína Antagonista del Receptor de Interleucina 1/líquido cefalorraquídeo , Interleucina-1/metabolismo , Masculino , Persona de Mediana Edad , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/líquido cefalorraquídeo , Dinámicas no Lineales , Proyectos Piloto
13.
J Neurochem ; 115(6): 1434-44, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21044072

RESUMEN

Brain injury caused by ischemia is a major cause of human mortality and physical/cognitive disability worldwide. Experimentally, brain ischemia can be induced surgically by permanent middle cerebral artery occlusion. Using this model, we studied the influence of transthyretin in ischemic stroke. Transthyretin (TTR) is normally responsible for the transport of thyroid hormones and retinol in the blood and CSF. We found that TTR null mice (TTR(-/-) ) did not show significant differences in cortical infarction 24 h after permanent middle cerebral artery occlusion compared with TTR(+/+) control littermates. However, TTR null mice, heterozygous for the heat-shock transcription factor 1 (TTR(-/-) HSF1(+/-) mice), which compromised the stress response, showed a significant increase in cortical infarction, cerebral edema and the microglial-leukocyte response compared with TTR(+/+) HSF1(+/-) mice. Unexpectedly, we observed novel TTR distribution throughout the infarct, localized to disintegrated ß-tubulin III(+) neurons and cell debris. Specific elimination of TTR synthesis in the liver by RNAi had no effect on TTR distribution in the infarct, indicating that the observed TTR infiltration derived from CSF and not from the serum. This finding is corroborated by results from 'in situ' hybridization and real time PCR that excluded the presence of transthyretin mRNA in the infarct and peri-infarct areas. Our data suggest that in conditions of a compromised heat-shock response, CSF TTR contributes to control neuronal cell death, edema and inflammation, thereby influencing the survival of endangered neurons in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/líquido cefalorraquídeo , Prealbúmina/líquido cefalorraquídeo , Animales , Biomarcadores/líquido cefalorraquídeo , Isquemia Encefálica/líquido cefalorraquídeo , Muerte Celular/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología
14.
Eur J Pharm Biopharm ; 70(3): 735-40, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18684400

RESUMEN

The estradiol(E(2))-loaded chitosan nanoparticles (CS-NPs) were prepared by ionic gelation of chitosan with tripolyphosphate anions (TPP). The CS-NPs had a mean size of (269.3+/-31.6)nm, a zeta potential of +25.4 mV, and loading capacity of E(2) CS-NPs suspension was 1.9 mg ml(-1), entrapment efficiency was 64.7% on average. Subsequently, this paper investigated the levels of E(2) in blood and the cerebrospinal fluid (CSF) in rats following intranasal administration of E(2) CS-NPs. E(2)-loaded CS-NPs were administered to male Wister rats either intranasally or intravenously at the dose of 0.48 mg kg(-1). The plasma levels achieved following intranasal administration (32.7+/-10.1 ng ml(-1); t(max) 28+/-4.5 min) were significantly lower than those after intravenous administration (151.4+/-28.2 ng ml(-1)), while CSF concentrations achieved after intranasal administration (76.4+/-14.0 ng ml(-1); t(max) 28+/-17.9 min) were significantly higher than those after intravenous administration (29.5+/-7.4 ng ml(-1)t(max) 60 min). The drug targeting index (DTI) of nasal route was 3.2, percent of drug targeting (DTP%) was 68.4%. These results showed that the E(2) must be directly transported from the nasal cavity into the CSF in rats. Finally, compared with E(2) inclusion complex, CS-NPs improved significantly E(2) being transported into central nervous system (CNS).


Asunto(s)
Encéfalo/metabolismo , Quitosano/química , Portadores de Fármacos , Estradiol/farmacocinética , Nanopartículas , Mucosa Nasal/metabolismo , Fármacos Neuroprotectores/farmacocinética , Administración Intranasal , Animales , Disponibilidad Biológica , Química Farmacéutica , Composición de Medicamentos , Estradiol/administración & dosificación , Estradiol/sangre , Estradiol/líquido cefalorraquídeo , Estradiol/química , Inyecciones Intravenosas , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fármacos Neuroprotectores/química , Tamaño de la Partícula , Permeabilidad , Ratas , Ratas Wistar
15.
J Cereb Blood Flow Metab ; 28(2): 387-94, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17684519

RESUMEN

The proinflammatory cytokine interleukin (IL)-1 mediates several forms of experimentally induced acute brain injury and has been implicated in chronic neurodegenerative disorders. The IL-1 receptor antagonist, IL-1RA, protects rodents against ischaemic brain injury, but its molecular mass (17 kDa) potentially limits the brain penetration of peripherally administered IL-1RA. We therefore sought to identify whether therapeutically effective concentrations of IL-1RA in the rat were also achieved in brain of patients with subarachnoid haemorrhage (SAH), using a peripheral administration regime that had proved to be safe and reduce peripheral inflammation in patients after stroke. An intravenous bolus of IL-1RA, followed by infusion, was administered to rats after induction of focal cerebral ischaemia. The effects of IL-1RA on brain ischaemia and the concentrations achieved in cerebrospinal fluid (CSF), were determined. Interleukin-1 receptor antagonist was similarly administered to patients with SAH, and CSF was sampled via external ventricular drains. In rats, IL-1RA significantly reduced brain injury induced by focal cerebral ischaemia. The plasma IL-1RA concentrations reached 12+/-2 microg/mL by 30 mins, and CSF concentrations were maintained between 91 and 232 ng/mL between 1 and 24 h of infusion. In patients with SAH, IL-1RA reached a steady-state plasma concentration of 22+/-4 microg/mL by 15 mins, and CSF concentrations were maintained at 78 to 558 ng/mL between 1 and 24 h. Intravenous delivery of IL-1RA leads to CSF concentrations in patients comparable to those that are neuroprotective in rats, and might therefore be of therapeutic benefit.


Asunto(s)
Encéfalo/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacocinética , Fármacos Neuroprotectores/farmacocinética , Adulto , Anciano , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Femenino , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Infusiones Intravenosas , Proteína Antagonista del Receptor de Interleucina 1/sangre , Proteína Antagonista del Receptor de Interleucina 1/líquido cefalorraquídeo , Masculino , Persona de Mediana Edad , Arteria Cerebral Media/fisiología , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo
16.
Neuropharmacology ; 53(6): 749-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17904590

RESUMEN

The current study describes the neuroprotective effects of an endogenous diketopiperazine, cyclo-glycyl-proline (cyclic GP), in rats with hypoxic-ischemic brain injury and the pre-clinical development of an analogue, cyclo-L-glycyl-L-2-allylproline (NNZ 2591), modified for improved bioavailability. The compounds were given either intracerebroventricularly or subcutaneously 2h after hypoxia-ischemia. Histology, immunohistochemistry and behavior were used to evaluate treatment effects. The central uptake of NNZ 2591 was also examined in normal and hypoxic-ischemic injured rats by HPLC-mass spectrometry. Central administration of cyclic GP or NNZ 2591 reduced the extent of brain damage in the lateral cortex, the hippocampus and the striatum (p<0.001), with NNZ 2591 being more potent. NNZ 2591 was stable in the plasma and crossed the blood-brain barrier independent of hypoxic-ischemic injury. The level of NNZ 2591 in the CSF was maintained for 2 h after a single subcutaneous dose, and modest neuroprotection was seen after a bolus subcutaneous administration (overall p<0.001). Treatment with NNZ 2591 for 5 d subcutaneously improved somatosensory-motor function (p<0.05) and long-term histological outcome (overall p<0.0001). NNZ 2591 treatment not only reduced both caspase-3 mediated apoptosis and microglial activation but also enhanced astrocytic reactivity, which may mediate its protective effect. The pharmacokinetic profile and potent long-term protective effects of NNZ 2591 suggests its utility for the treatment of ischemic brain injury and other neurological conditions requiring chronic intervention.


Asunto(s)
Lesión Encefálica Crónica/prevención & control , Encéfalo/efectos de los fármacos , Dicetopiperazinas/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Péptidos Cíclicos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Encéfalo/fisiopatología , Lesión Encefálica Crónica/etiología , Dicetopiperazinas/líquido cefalorraquídeo , Dicetopiperazinas/farmacocinética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/fisiopatología , Masculino , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fármacos Neuroprotectores/farmacocinética , Ratas , Ratas Wistar , Factores de Tiempo , Resultado del Tratamiento
17.
Pediatr Res ; 62(6): 680-3, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17957150

RESUMEN

Despite effective antibiotic treatment, neuronal injury is frequent among children and adults with bacterial meningitis resulting in a high rate of death and neurologic sequelae. The hematopoietic cytokine erythropoietin (EPO) provides neuroprotection in models of acute and chronic neurologic diseases. We studied whether recombinant EPO (rEPO) reduces neuronal damage in a rabbit model of Escherichia coli meningitis. Inflammation within the central nervous system (CNS) was monitored by measurement of bacterial load, pleocytosis, protein, and lactate in the cerebrospinal fluid (CSF). Neuronal damage was measured by quantification of the density of apoptotic neurons in the hippocampal dentate gyrus and the concentration of the global neuronal destruction marker neuron-specific enolase (NSE) in CSF. To increase clinical relevance, rEPO was applied as adjunctive therapy from the beginning of antibiotic therapy 12 h after infection. EPO treatment applied as an intravenous injection at a dose of 1000 IU/kg body weight resulted in plasma concentrations of 6993 +/- 1406 mIU/mL, CSF concentrations of 1291 +/- 568 mIU/mL, and a CSF-to-plasma ratio of 0.18 +/- 0.07 (mean +/- SD) 6 h after injection. Under these treatment conditions, no anti-inflammatory or neuroprotective effect of EPO was observed. "


Asunto(s)
Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Eritropoyetina/farmacología , Meningitis por Escherichia coli/tratamiento farmacológico , Enfermedades del Sistema Nervioso/prevención & control , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Antibacterianos/farmacología , Antiinflamatorios/administración & dosificación , Antiinflamatorios/sangre , Antiinflamatorios/líquido cefalorraquídeo , Giro Dentado/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Eritropoyetina/administración & dosificación , Eritropoyetina/sangre , Eritropoyetina/líquido cefalorraquídeo , Inyecciones Intravenosas , Meningitis por Escherichia coli/líquido cefalorraquídeo , Meningitis por Escherichia coli/complicaciones , Meningitis por Escherichia coli/patología , Enfermedades del Sistema Nervioso/líquido cefalorraquídeo , Enfermedades del Sistema Nervioso/microbiología , Enfermedades del Sistema Nervioso/patología , Neuronas/enzimología , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Fosfopiruvato Hidratasa/líquido cefalorraquídeo , Conejos , Proteínas Recombinantes , Índice de Severidad de la Enfermedad , Factores de Tiempo
18.
Neurobiol Dis ; 25(3): 514-25, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17239606

RESUMEN

Axonal destruction and neuronal loss occur early during multiple sclerosis, an autoimmune inflammatory CNS disease that frequently manifests with acute optic neuritis. Available therapies mainly target the inflammatory component of the disease but fail to prevent neurodegeneration. To investigate the effect of minocycline on the survival of retinal ganglion cells (RGCs), the neurons that form the axons of the optic nerve, we used a rat model of myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis. Optic neuritis in this model was diagnosed by recording visual evoked potentials and RGC function was monitored by measuring electroretinograms. Functional and histopathological data of RGCs and optic nerves revealed neuronal and axonal protection when minocycline treatment was started on the day of immunization. Furthermore, we demonstrate that minocycline-induced neuroprotection is related to a direct antagonism of multiple mechanisms leading to neuronal cell death such as the induction of anti-apoptotic intracellular signalling pathways and a decrease in glutamate excitotoxicity. From these observations, we conclude that minocycline exerts neuroprotective effects independent of its anti-inflammatory properties. This hypothesis was confirmed in a non-inflammatory disease model leading to degeneration of RGCs, the surgical transection of the optic nerve.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad Aguda , Animales , Antibacterianos/sangre , Antibacterianos/líquido cefalorraquídeo , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Potenciales Evocados Visuales , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Femenino , Ácido Glutámico/metabolismo , Minociclina/sangre , Minociclina/líquido cefalorraquídeo , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/líquido cefalorraquídeo , Nervio Óptico/inmunología , Nervio Óptico/patología , Nervio Óptico/fisiopatología , Neuritis Óptica/tratamiento farmacológico , Neuritis Óptica/inmunología , Neuritis Óptica/fisiopatología , Ratas , Ratas Endogámicas BN , Células Ganglionares de la Retina/inmunología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Índice de Severidad de la Enfermedad
20.
J Neuropathol Exp Neurol ; 65(8): 816-25, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16896315

RESUMEN

To investigate cytokine/chemokine changes in amyotrophic lateral sclerosis (ALS), we simultaneously measured 16 cytokine/chemokines (interleukin [IL]-1beta, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12 [p70], IL-13, IL-17, interferon-gamma, tumor necrosis factor-alpha, granulocyte colony stimulating factor [G-CSF], macrophage chemoattractant protein-1 [MCP-1], and macrophage inflammatory protein-1beta) in cerebrospinal fluid (CSF) and sera from 37 patients with sporadic ALS and 33 controls using a multiplexed fluorescent bead-based immunoassay. We also conducted immunohistochemical analyses from 8 autopsied ALS cases and 6 nonneurologic disease controls as well as cell culture analyses of relevant cytokines and their receptors. We found that concentrations of G-CSF and MCP-1 were significantly increased in ALS CSF compared with controls. In spinal cords, G-CSF was expressed in reactive astrocytes in ALS cases but not controls, whereas G-CSF receptor expression was significantly decreased in motor neurons of spinal cords from ALS cases. Biologically, G-CSF had a protective effect on the NSC34 cell line under conditions of both oxidative and nutritional stress. We suggested that G-CSF has potentially neuroprotective effects on motor neurons in ALS and that downregulation of its receptor might contribute to ALS pathogenesis. On the other hand, MCP-1 correlated with disease severity, which may aggravate motor neuron damage.


Asunto(s)
Esclerosis Amiotrófica Lateral/líquido cefalorraquídeo , Sistema Nervioso Central/metabolismo , Citoprotección/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/líquido cefalorraquídeo , Neuronas Motoras/metabolismo , Fármacos Neuroprotectores/líquido cefalorraquídeo , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/fisiopatología , Muerte Celular/inmunología , Línea Celular , Supervivencia Celular/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/fisiopatología , Líquido Cefalorraquídeo/inmunología , Líquido Cefalorraquídeo/metabolismo , Quimiocina CCL2/líquido cefalorraquídeo , Quimiocinas/líquido cefalorraquídeo , Citocinas/líquido cefalorraquídeo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/inmunología , Neuronas Motoras/patología , Degeneración Nerviosa/líquido cefalorraquídeo , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA