Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Kidney Int ; 100(3): 570-584, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34051264

RESUMEN

Ischemia-reperfusion injury is a major cause of acute kidney injury. Recent studies on the pathophysiology of ischemia-reperfusion-induced acute kidney injury showed that immunologic responses significantly affect kidney ischemia-reperfusion injury and repair. Nuclear factor (NF)-ĸB signaling, which controls cytokine production and cell survival, is significantly involved in ischemia-reperfusion-induced acute kidney injury, and its inhibition can ameliorate ischemic acute kidney injury. Using EXPLOR, a novel, optogenetically engineered exosome technology, we successfully delivered the exosomal super-repressor inhibitor of NF-ĸB (Exo-srIĸB) into B6 wild type mice before/after kidney ischemia-reperfusion surgery, and compared outcomes with those of a control exosome (Exo-Naïve)-injected group. Exo-srIĸB treatment resulted in lower levels of serum blood urea nitrogen, creatinine, and neutrophil gelatinase-associated lipocalin in post-ischemic mice than in the Exo-Naïve treatment group. Systemic delivery of Exo-srIĸB decreased NF-ĸB activity in post-ischemic kidneys and reduced apoptosis. Post-ischemic kidneys showed decreased gene expression of pro-inflammatory cytokines and adhesion molecules with Exo-srIĸB treatment as compared with the control. Intravital imaging confirmed the uptake of exosomes in neutrophils and macrophages. Exo-srIĸB treatment also significantly affected post-ischemic kidney immune cell populations, lowering neutrophil, monocyte/macrophage, and T cell frequencies than those in the control. Thus, modulation of NF-ĸB signaling through exosomal delivery can be used as a novel therapeutic method for ischemia-reperfusion-induced acute kidney injury.


Asunto(s)
Lesión Renal Aguda , Exosomas , Daño por Reperfusión , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Animales , Riñón , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Daño por Reperfusión/prevención & control
2.
J Neurosci Res ; 93(8): 1267-78, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25711470

RESUMEN

Monoamine oxidase B (MAO-B) is well known as a therapeutic target for Parkinson's disease (PD). MAO-B inhibitors retain antiparkinsonism abilities to improve motor function and prevent neuronal loss by decreasing dopamine metabolism and oxidative stress in the brain. From the study to find novel antiparkinsonism drugs that can inhibit MAO-B activity, neuronal loss, and behavioral deficits in the mouse model of PD, we identified that 1-[2-(4-benzyloxyphenoxy)ethyl]imidazole (BPEI) or safinamide strongly and selectively inhibited MAO-B activities in a dose-dependent manner (IC50 of BPEI and safinamide for MAO-B were 0.016 and 0.0021 µM and for MAO-A were 70.0 and 370 µM, respectively). In ex vivo studies after an administration (30 mg/kg, i.p.) of BPEI or safinamide to normal mice, the MAO-B activity in the brain was reduced by up to 90.6% or 82.4% at 1.0 hr. BPEI (20 mg/kg, i.p.) or safinamide (20 mg/kg, i.p.) significantly reversed the behavioral impairments, dopamine levels in the striatum, and neuronal loss in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice compared with the MPTP-alone-treated group. In the 6-hydroxydopamine-induced PD rat model, behavioral improvement by levodopa sparing activity was observed in the BPEI- or safinamide-treated (20 mg/kg, i.p.) rats. Moreover, BPEI revealed additional curative activities for nonmotor symptoms of PD such as pain, anxiety, epilepsy, and depression in rodent disease models. Therefore, BPEI has broad therapeutic potential for treating motor symptoms via strong and selective inhibitory effects on MAO-B, with additional benefits for comorbid symptoms in PD.


Asunto(s)
Modelos Animales de Enfermedad , Imidazoles/uso terapéutico , Intoxicación por MPTP/enzimología , Intoxicación por MPTP/prevención & control , Inhibidores de la Monoaminooxidasa/uso terapéutico , Neuronas/enzimología , Animales , Recuento de Células , Imidazoles/farmacología , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/prevención & control , Ratas , Ratas Sprague-Dawley
3.
Am J Reprod Immunol ; 85(2): e13329, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32846024

RESUMEN

The extracellular vesicles (EVs) research area has grown rapidly because of their pivotal roles in intercellular communications and maintaining homeostasis of individual organism. As a subtype of EVs, exosomes are made via unique biogenesis pathway and exhibit disparate functional and phenotypic characteristics. Functionally, exosomes transfer biological messages from donor cell to recipient cell, which makes exosomes as a novel therapeutic platform delivering therapeutic materials to the target tissue/cell. Currently, both academia and industry try to develop exosome platform-based therapeutics for disease management, some of which are already in clinical trials. In this review, we will discuss focusing on therapeutic values of exosomes, recent advances in therapeutic exosome platform development, and late development of exosome therapeutics in diverse therapeutic areas.


Asunto(s)
Terapia Biológica/métodos , Exosomas/metabolismo , Animales , Comunicación Celular , Sistemas de Liberación de Medicamentos , Vesículas Extracelulares , Humanos
4.
J Neurochem ; 109(5): 1483-96, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19457162

RESUMEN

Although there is known to be a marked concentration of reactive microglia in the substantia nigra pars compacta (SNpc) of patients with Parkinson's disease (PD), a disorder in which alpha-synuclein plays a key pathogenic role, the specific roles of alpha-synuclein and microglia remains poorly understood. In this study, we investigated the effects of alpha-synuclein and the mechanisms of invasive microglial migration into the SNpc. We show that alpha-synuclein up-regulates the expressions of the cell adhesion molecule CD44 and the cell surface protease membrane-type 1 matrix metalloproteinase through the extracellular regulated kinases 1/2 pathway. These concurrent inductions increased the generation of soluble CD44 to liberate microglia from the surrounding extracellular matrix for migration. The effects of alpha-synuclein were identical in BV-2 murine microglial cells subjected to cDNA transfection and extracellular treatment. These inductions in primary microglial cultures of C57Bl/6 mice were identical to those in BV-2 cells. alpha-Synuclein-induced microglial migration into the SNpc was confirmed in vivo using a 6-hydroxydopamine mouse model of PD. Our data demonstrate a correlation between alpha-synuclein-induced phenotypic changes and microglial migration. With the recruitment of the microglial population into the SNpc during dopaminergic neurodegeneration, alpha-synuclein may play a role in accelerating the pathogenesis of PD.


Asunto(s)
Movimiento Celular/fisiología , Receptores de Hialuranos/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Microglía/fisiología , Regulación hacia Arriba/fisiología , alfa-Sinucleína/fisiología , Animales , Encéfalo/citología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Trasplante de Células/métodos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Receptores de Hialuranos/genética , Metaloproteinasa 14 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/cirugía , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sustancia Negra/metabolismo , Sustancia Negra/cirugía , Transfección , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , alfa-Sinucleína/genética
5.
J Neurosci Res ; 87(1): 260-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18712849

RESUMEN

In Oriental medicine, roots of Polygala tenuifolia Willdenow have been known to be an important herb that exhibits sedative effects in insomnia, palpitation with anxiety, restlessness, and disorientation in humans. We previously reported that BT-11, extracted from those roots, improved scopolamine-induced amnesia in rats and inhibited acetylcholinesterase activities in vitro. Therefore, we proposed that BT-11 could remedy stress-induced memory deficits in rats. In this study, the stress-induced memory impairments in rats were significantly reversed almost to the control level by BT-11 treatment. To seek an active component of BT-11 that plays an important role in antipsychotic effects, we compared BT-11 with 3,4,5-trimethoxycinnamic acid (TMCA), which is a constituent of those root extracts. However, the effects of TMCA were less or were not consistent with those of BT-11 in some of tests. In particular, BT-11 reversed the stress-induced reduction of glucose utilization by [(18)fluorodeoxyglucose]FDG-PET and the levels of neural cell adhesion molecule (NCAM) in rat brains to the control levels, whereas TMCA did not. Therefore, BT-11 improved stress-induced memory impairments through increment of glucose utilization and total NCAM levels in rat brains. In conclusion, BT-11 may be strongly effective against stress-induced amnesia in rats, through the combined effects of TMCA and other active components of BT-11.


Asunto(s)
Encéfalo/metabolismo , Glucosa/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Fitoterapia/métodos , Polygala/química , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Ciclohexilaminas/uso terapéutico , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Fluorodesoxiglucosa F18/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Tomografía de Emisión de Positrones/métodos , Ratas , Ratas Wistar , Estrés Psicológico/complicaciones
6.
Mol Cell Biol ; 26(11): 4327-38, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705182

RESUMEN

Amyloid precursor protein (APP) has eight potential phosphorylation sites in its cytoplasmic domain. Recently, it has demonstrated that the constitutive phosphorylation of APP at T668 (APP695 isoform numbering) was observed specifically in the brain. Neuron-specific phosphorylation of APP at T668 is thought to be important for neuronal functions of APP, although its exact physiological significance remains to be clarified. In this study, we show that the phosphorylation of the APP intracellular domain (AICD) at T668 is essential for its binding to Fe65 and its nuclear translocation and affects the resultant neurotoxicity, possibly mediated through the induction of glycogen synthase kinase 3beta and tau phosphorylation by enhancing the formation of a ternary complex with Fe65 and CP2 transcription factor. Taken together, these results suggest that the phosphorylation of AICD at T668 contributes to the neuronal degeneration in Alzheimer's disease (AD) by regulating its translocation into the nucleus and then affects neurodegeneration; therefore, the specific inhibitor of T668 phosphorylation might be the target of AD therapy.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Núcleo Celular/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Treonina/metabolismo , Transporte Activo de Núcleo Celular , Enfermedad de Alzheimer/patología , Animales , Encéfalo/citología , Encéfalo/patología , Muerte Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas Nucleares/metabolismo , Células PC12 , Fosforilación , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Proteínas tau/metabolismo
7.
J Ethnopharmacol ; 232: 119-129, 2019 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-30576772

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Polygala tenuifolia Willdenow root extract (BT-11) has beneficial effects on central nervous system disorders in human. The safety of BT-11 should be elucidated in younger person further. AIM OF THE STUDY: To evaluate the safety of BT-11 in human aged from 9 to 19 years old. MATERIAL AND METHODS: The safety was evaluated in randomly assigned subjects who received the test products (61 subjects in BT-11 300 mg daily or 60 subjects in matching placebo) for 12 weeks. Adverse reactions were analyzed by the incidence rate, type, and severity. The clinical examination included hematology and blood chemistry tests, urinalysis, vital signs, body weight, and electrocardiogram (ECG). RESULTS: Eleven adverse reactions were observed in ten subjects receiving BT-11 while seven adverse reactions in six subjects receiving placebo. There were no statistical differences in the incidence of adverse reactions between the two groups. Serious adverse reactions such as acute appendicitis and acute viral gastroenteritis were observed in the BT-11 group4 and the placebo group, respectively. However, it was confirmed that they were not associated with the test product. All other adverse reactions observed during the test period were resolved completely without special treatment. No statistical difference was also observed in safety laboratory tests, vital signs, and ECG between two groups. CONCLUSIONS: This study demonstrates the safety of BT-11 in the adolescent by showing no apparent adverse reactions related to it.


Asunto(s)
Extractos Vegetales/administración & dosificación , Polygala , Adolescente , Adulto , Niño , Método Doble Ciego , Femenino , Humanos , Masculino , Raíces de Plantas , Polvos , Adulto Joven
8.
Neuropsychopharmacology ; 32(11): 2393-404, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17406652

RESUMEN

Minocycline is a semi-synthetic tetracycline antibiotic that effectively crosses the blood-brain barrier. Minocycline has been reported to have significant neuroprotective effects in models of cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, and Huntington's and Parkinson's diseases. In this study, we demonstrate that minocycline has neuroprotective effects in in vitro and in vivo Alzheimer's disease models. Minocycline was found to attenuate the increases in the phosphorylation of double-stranded RNA-dependent serine/threonine protein kinase, eukaryotic translation initiation factor-2 alpha and caspase 12 activation induced by amyloid beta peptide1-42 treatment in NGF-differentiated PC 12 cells. In addition, increases in the phosphorylation of eukaryotic translation initiation factor-2 alpha were attenuated by administration of minocycline in Tg2576 mice, which harbor mutated human APP695 gene including the Swedish double mutation and amyloid beta peptide(1-42)-infused rats. We found that minocycline administration attenuated deficits in learning and memory in amyloid beta peptide(1-42)-infused rats. Increased phosphorylated state of eukaryotic translation initiation factor-2 alpha is observed in Alzheimer's disease patients' brains and may result in impairment of cognitive functions in Alzheimer's disease patients by decreasing the efficacy of de novo protein synthesis required for synaptic plasticity. On the basis of these results, minocycline may prove to be a good candidate as an effective therapeutic agent for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento , Minociclina/uso terapéutico , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/patología , Estudios de Casos y Controles , Muerte Celular/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Minociclina/farmacología , Factor de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Células PC12/efectos de los fármacos , Fragmentos de Péptidos , Ratas , Ratas Wistar , Transfección
9.
FASEB J ; 20(6): 729-31, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16467370

RESUMEN

Although chronic stress is known to be linked with memory and other neurological disorders, little is known about the relationship between chronic stress and the onset or development of Alzheimer's disease (AD). In this study, we investigated the effects of long-term stress on the onset and severity of cognitive deficits and pathological changes in APPV717I-CT100 mice overexpressing human APP-CT100 containing the London mutation (V717I) after exposure to immobilization stress. We found that chronic immobilization stress accelerated cognitive impairments, as accessed by the Passive avoidance and the Social Transfer of Food Preference (STFP) tests. Moreover, the numbers and densities of vascular and extracellular deposits containing amyloid beta peptide (Abeta) and carboxyl-terminal fragments of amyloid precursor protein (APP-CTFs), which are pathologic markers of AD, were significantly elevated in stressed animals, especially in the hippocampus. Moreover, stressed animals, also showed highly elevated levels of neurodegeneration and tau phosphorylation and increased intraneuronal Abeta and APP-CTFs immunoreactivities in the hippocampus and in the entorhinal and piriform cortex. This study provides the first evidence that chronic stress accelerates the onset and severity of cognitive deficits and that these are highly correlated with pathological changes, which thus indicates that chronic stress may be an important contributor to the onset and development of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Trastornos de la Memoria/metabolismo , Estrés Psicológico/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Corteza Cerebral/metabolismo , Enfermedad Crónica , Corticosterona/sangre , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Restricción Física/efectos adversos , Estrés Psicológico/genética
10.
Int J Dev Neurosci ; 24(1): 81-5, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16289942

RESUMEN

Huntington's disease (HD) is a dominant neurodegenerative disorder caused by the expansion of a CAG repeat in the gene encoding huntingtin. Moreover, the nuclear targeting of mutant huntingtin increases cellular toxicity, whereas normal huntingtin resides mainly in the cytoplasm, and is associated with membranes or microtubules. Huntingtin is enriched in neurons and its expression is increased during neural development. The inactivation of the HD gene results in embryonic lethality before nervous system development. Thus, huntingtin is critical during early embryonic development. Nevertheless, the function of huntingtin at this stage is unknown, even the distribution of the protein has not been described. The present study was undertaken to elucidate the distribution of huntingtin during the early developmental period in the mouse embryo. At the preimplantation stage, huntingtin was detected in nuclei up to 2.5 days post coitum (dpc), but disappeared from nuclei during the blastocyst stage (3.5 dpc). Following this stage, huntingtin was mainly localized in the cytoplasm and co-localized with mitotic spindles. These data suggest that the nuclear targeting of normal huntingtin is required during early embryo development in mice.


Asunto(s)
Blastocisto/metabolismo , Núcleo Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Blastocisto/citología , Humanos , Proteína Huntingtina , Enfermedad de Huntington , Inmunohistoquímica , Ratones , Oocitos/citología , Oocitos/metabolismo
11.
CNS Neurol Disord Drug Targets ; 15(8): 935-944, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27577574

RESUMEN

We previously demonstrated that dehydroevodiamine•HCl (DHED), which was purified from Evodia rutaecarpa Bentham (Rutaceae), has beneficial effects on memory impairment and neuronal damage in three disease models. To investigate the preventive action of DHED in Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory decline, amyloid-ß (Aß) protein-containing neuritic plaques and neurofibrillary tangles, in this study, we proposed that DHED may be therapeutically effective against the memory impairment and disease-related neurochemical changes that occur in Tg2576 (Tg) mice. DHED (0.5 mg/kg) was intraperitoneally administered to 7-month-old Tg and wild type mice for 4 months. In passive avoidance and water maze tests, DHED improved memory impairment of Tg mice after 4 months of administration. DHED also reduced cortical levels of soluble Aß40, soluble Aß42 and total Aß peptides in the Tg mice. Additionally, we investigated whether DHED may be a ß-secretase inhibitor that affects the production of Aß related to the formation of neuritic plaques. DHED directly inhibited ß-secretase activity in a concentrationdependent manner. The concentration required for 50 % enzyme inhibition (IC50) was 40.96 µM, and DHED may act as a competitive inhibitor of ß-secretase. Moreover, DHED interacted strongly with BACE1 (ß-secretase 2QP8), as demonstrated in the analysis of the binding mode of DHED in the active site of human BACE1. In conclusion, DHED may exhibit therapeutic effects for AD as a ß-secretase inhibitor.


Asunto(s)
Alcaloides/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Modelos Moleculares , Mutación/genética , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética
12.
FASEB J ; 17(11): 1556-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12824278

RESUMEN

Many environmental factors during the pre- or postnatal period can affect an individual's cognitive function and neural development throughout life. Little is known, however, about the combined effects of the pre- and postnatal environments on cognitive function of adult offspring and structural alterations in the adult brain. In this study, we confirmed that pre- or postnatal stress impaired learning and memory performance of rats. Conversely, pre- or postnatal enriched housing improved behavioral performance. These experience-dependent behavioral alterations were consistent with changes in 5-bromo-2'-deoxyuridine-labeled cell number in the granule cell layer of the hippocampus and in the expression level of synaptic markers such as neuronal cell adhesion molecule and synaptophysin, and expression of a neurotrophic factor, brain-derived neurotrophic factor. Postnatal stress appeared to have no influence on cell proliferation, however. We did find that postnatal environment could attenuate prenatal effects partly via a longitudinal cross-housing study, in which pups born to mothers housed under enriched conditions were reared under stressful conditions and vice versa. These results suggest that postnatal environmental manipulations can counteract the cognitive alterations in early adulthood and the structural changes in the young adult brain induced by prenatal experience.


Asunto(s)
Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Cognición , Plasticidad Neuronal , Animales , Animales Recién Nacidos , Encéfalo/fisiología , División Celular , Giro Dentado/citología , Giro Dentado/embriología , Giro Dentado/crecimiento & desarrollo , Exposición a Riesgos Ambientales , Estudios Longitudinales , Masculino , Memoria , Modelos Neurológicos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Sinapsis/fisiología
13.
FASEB J ; 18(13): 1615-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15289452

RESUMEN

Though the etiology of Parkinson's disease (PD) remains unclear, alpha-synuclein (alpha-SN) is regarded as a major causative agent of PD. Several lines of evidence indicate that immunological abnormalities are associated with PD for unknown reasons. The present study was performed to assess whether peripheral blood mononuclear cells (PBMCs) show altered alpha-SN expression in PD patients and to identify its functions, which may be related to peripheral immune abnormalities in PD. alpha-SN was found to be expressed more in 151 idiopathic PD (IPD) patients than in 101 healthy controls, who nevertheless showed as age-dependent increases. By in vitro transfection, alpha-SN expression was shown to be correlated with glucocorticoid sensitive apoptosis, possibly caused by the enhanced expression of glucocorticoid receptor (GR), caspase activations (caspase-8, caspase-9), CD95 up-regulation, and reactive oxygen species (ROS) production. An understanding of the correlation between alpha-SN levels and apoptosis in the presence of the coordinated involvement of multiple processes would provide an insight into the molecular basis of the disease. The present study provides a clue that the alpha-SN may be one of the primary causes of the immune abnormalities observed in PD and offers new targets for pharmacotherapeutic intervention.


Asunto(s)
Apoptosis , Regulación de la Expresión Génica , Linfocitos/inmunología , Linfocitos/patología , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/metabolismo , Estudios de Casos y Controles , Caspasas/metabolismo , Células Cultivadas , Activación Enzimática , Humanos , Linfocitos/enzimología , Linfocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Glucocorticoides/metabolismo , Sinucleínas , alfa-Sinucleína , Receptor fas/genética , Receptor fas/metabolismo
14.
FASEB J ; 17(13): 1951-3, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12923068

RESUMEN

The AICD (amyloid precursor protein [APP] intracellular domain) and C31, the caspase-cleaved C-terminal fragment of APP, have been found in the brains of patients with Alzheimer's disease (AD). Here, we demonstrate for the first time that the C-terminal fragments of APP (AICD [C57, C59] and C31) exert neurotoxicity on differentiated PC 12 cells and rat primary cortical neurons by inducing the expression of glycogen synthase kinase 3beta, forming a ternary complex with Fe65 and CP2/LSF/LBP1 in the nucleus, whereas deletion mutants and a point mutant with Y682G of the YENPTY domain, a Fe65 binding domain, do not. Moreover, expression of APP770 and Swedish mutant form of APP increased the levels of C-terminal fragments of APP (APP-CTs) in neuronal cells and also induced the up-regulation of glycogen synthase kinase-3beta at both the mRNA and the protein levels. In addition, we show that CP2/LSF/LBP1 binding site (nt +0 to approximately +10) in human glycogen synthase kinase 3beta promoter region is essential for the induction of the gene transcription by APP-CTs. The neurotoxicities induced by APP-CTs (AICD and C31) were accompanied by an increase in the active form of glycogen synthase kinase-3beta, and by the induction of tau phosphorylation and a reduction in nuclear beta-catenin levels, and led to apoptosis.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Activación Transcripcional , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3 beta , Modelos Genéticos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Células PC12 , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Proteínas de Unión al ARN , Ratas , Factores de Transcripción/metabolismo , Transfección
15.
FASEB J ; 16(13): 1826-8, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12223445

RESUMEN

Alpha-synuclein (alpha-SN) is a ubiquitous protein that is especially abundant in the brain and has been postulated to play a central role in the pathogenesis of Parkinson's disease, Alzheimer's disease, and other neurodegenerative disorders. However, little is known about the neuronal functions of alpha-SN and the molecular and cellular mechanisms underlying neuronal loss. Here, we show that alpha-SN plays dual roles of neuroprotection and neurotoxicity depending on its concentration or level of expression. At nanomolar concentrations, a-SN protected neurons against serum deprivation, oxidative stress, and excitotoxicity through the PI3/Akt signaling pathway, and its protective effect was increased by Bcl-2 overexpression. Conversely, at both low micromolar and overexpressed levels in the cell, alpha-SN resulted in cytotoxicity. This might be related to decreased Bcl-xL expression and increased bax expression, which is subsequently followed by cytochrome c release and caspase activation and also by microglia-mediated inflammatory responses via the NFkappaB and mitogen-activated protein kinase pathways.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Morfolinas/farmacología , NG-Nitroarginina Metil Éster/farmacología , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Células PC12 , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/genética , Piridinas/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Sinucleínas , Factores de Tiempo , Células Tumorales Cultivadas , alfa-Sinucleína
16.
Neuroreport ; 13(15): 1989-93, 2002 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-12395106

RESUMEN

Amyloid beta peptide (Abeta) is a neurotoxic metabolic product of the amyloid precursor protein (APP). Abeta is strongly implicated in the pathology of Alzheimer's disease (AD) and can be formed intracellularly. In this study, we show that the addition of Abeta to isolated mouse brain mitochondria can directly induce cytochrome c (Cyt c) release and mitochondrial swelling, which were partially inhibited by cyclosporin A (CsA). These results suggest that the Abetaaccumulated intracellularly by APP processing might exert neurotoxicity by interacting with mitochondria and inducing mitochondrial swelling and release of Cyt c, which activates caspase-3 and finally can lead to apoptosis in neuronal cells and to neurodegeneration in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Péptidos beta-Amiloides/metabolismo , Encéfalo/enzimología , Grupo Citocromo c/metabolismo , Mitocondrias/enzimología , Neuronas/enzimología , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Células Cultivadas , Ciclosporina/farmacología , Grupo Citocromo c/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/toxicidad
17.
Neurosci Lett ; 557 Pt B: 95-100, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24176882

RESUMEN

Medium spiny neurons (MSNs) are the major type of neurons found in the striatum. The dendritic spines on these cells contain glutamatergic synaptic contacts between the cortex (or the thalamus) and the striatum. The complexity of the dendritic structure of MSNs may therefore reflect the functional status of the basal ganglia because the striatum is the major input structure in which signals from different regions are integrated. We examined the structural alterations in the dendrites of striatal MSNs in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease (PD). Acute MPTP treatment rapidly damaged dopaminergic neurons and their terminals within the striatum and caused behavioral impairments. However, mice injected with MPTP spontaneously recovered from these behavioral impairments within one week. This recovery was accompanied by the restoration of dendritic structures on MSNs, but the damage to dopaminergic neurons remained extensive. Furthermore, we demonstrated that rasagiline, a monoamine oxidase-B (MAO-B) inhibitor that has been shown to be efficacious for PD, could enhance the dendritic complexity of cultured MSNs. The effect of rasagiline on the spine-like structures of dendrites, however, appears not to require DA availability because the small protrusions of dendrites in cultured MSNs without major source of DA input was similarly changed by rasagiline. Our data suggest that the dendritic structures of striatal MSNs change dynamically, reflecting the progression of motor-related symptoms in PD, and the restoration of functional synapses in the MSNs of PD patients may constitute a clinical target for symptomatic alleviation.


Asunto(s)
Cuerpo Estriado/patología , Dendritas/ultraestructura , Trastornos Parkinsonianos/patología , Animales , Conducta Animal , Células Cultivadas , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Recuperación de la Función
18.
PLoS One ; 7(9): e44259, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028510

RESUMEN

The p53 and NF-κB pathways play important roles in diverse cellular functions, including cell growth, apoptosis, and tumorigenesis. Mutations that inactivate the p53 gene and constitutive NF-κB pathway activation are common occurrences in human cancers. Although many drugs are being developed that selectively activate p53 or inhibit NF-κB, there are few drug candidates that can do both. Simultaneous activation of p53 and inhibition of the NF-κB pathway is therefore a prime target for new cancer drug development. This study is the first report of a high-throughput approach with mass compounds that concurrently target both pathways. Using a cell-based screening assay and a library of 200,000 synthetic compounds, we identified 9 small molecules that simultaneously inhibit NF-κB and activate p53. One of these compounds, N-2, increased the expression of p53 target genes, including p21 and GADD45a. In addition, N-2 inhibited the transcriptional activity of NF-κB, concomitantly repressing interleukin-6 and monocyte chemotactic protein-1 (MCP-1) expression. When cell lines derived from a diverse range of cancers were treated in vitro with N-2, we observed increased cell death. N-2 also significantly inhibited allograft growth in murine models of melanoma and lung carcinoma. Our findings suggest that N-2 may act as a bivalent anti-cancer agent through simultaneous modulation of NF-κB and p53 activities.


Asunto(s)
Antineoplásicos/farmacología , FN-kappa B/antagonistas & inhibidores , Fenantridinas/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/clasificación , Línea Celular Tumoral , Quimiocina CCL2/genética , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Ensayos Analíticos de Alto Rendimiento , Humanos , Interleucina-6/genética , Dosificación Letal Mediana , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Fenantridinas/química , Fenantridinas/clasificación , Piridinas/química , Piridinas/clasificación , Bibliotecas de Moléculas Pequeñas , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Transcriptoma , Proteína p53 Supresora de Tumor/genética
19.
J Neurosci Res ; 85(11): 2500-11, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17600377

RESUMEN

To develop a novel and effective drug that could enhance cognitive function and neuroprotection, we newly synthesized maltolyl p-coumarate by the esterification of maltol and p-coumaric acid. In the present study, we investigated whether maltolyl p-coumarate could improve cognitive decline in scopolamine-injected rats and in amyloid beta peptide(1-42)-infused rats. Maltolyl p-coumarate was found to attenuate cognitive deficits in both rat models using passive avoidance test and to reduce apoptotic cell death observed in the hippocampus of the amyloid beta peptide(1-42)-infused rats. We also examined the neuroprotective effects of maltolyl p-coumarate in vitro using SH-SY5Y cells. Cells were pretreated with maltolyl p-coumarate, before exposed to amyloid beta peptide(1-42), glutamate or H2O2. We found that maltolyl p-coumarate significantly decreased apoptotic cell death and reduced reactive oxygen species, cytochrome c release, and caspase 3 activation. Taking these in vitro and in vivo results together, our study suggests that maltolyl p-coumarate is a potentially effective candidate against Alzheimer's disease that is characterized by wide spread neuronal death and progressive decline of cognitive function.


Asunto(s)
Ácidos Cumáricos/farmacología , Demencia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Pironas/farmacología , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/efectos de los fármacos , Cognición/efectos de los fármacos , Citocromos c/efectos de los fármacos , Demencia/inducido químicamente , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Antagonistas Muscarínicos/toxicidad , Fármacos Neuroprotectores/química , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Escopolamina/toxicidad
20.
Mol Pharmacol ; 69(1): 76-84, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16223958

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) exert anti-inflammatory, analgesic, and antipyretic activities and suppress prostaglandin synthesis by inhibiting cyclooxygenase, an enzyme that catalyzes the formation of prostaglandin precursors from arachidonic acid. Epidemiological observations indicate that the long-term treatment of patients suffering from rheumatoid arthritis with NSAIDs results in reduced risk and delayed onset of Alzheimer's disease. In this study, we investigated the therapeutic potential for Alzheimer's disease of mefenamic acid, a commonly used NSAID that is a cyclooxygenase-1 and 2 inhibitor with only moderate anti-inflammatory properties. We found that mefenamic acid attenuates the neurotoxicities induced by amyloid beta peptide (Abeta)(1-42) treatment and the expression of a Swedish double mutation (KM595/596NL) of amyloid precursor protein (Swe-APP) or the C-terminal fragments of APP (APP-CTs) in neuronal cells. We also show that mefenamic acid decreases the production of the free radical nitric oxide and reduces cytochrome c release from mitochondria induced by Abeta(1-42), Swe-APP, or APP-CTs in neuronal cells. In addition, mefenamic acid up-regulates expression of the antiapoptotic protein Bcl-X(L). Moreover, our study demonstrates for the first time that mefenamic acid improves learning and memory impairment in an Abeta(1-42)-infused Alzheimer's disease rat model. Taking these in vitro and in vivo results together, our study suggests that mefenamic acid could be used as a therapeutic agent in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Antiinflamatorios no Esteroideos/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Ácido Mefenámico/farmacología , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Caspasa 3 , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Activación Enzimática , Masculino , Ácido Mefenámico/uso terapéutico , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/uso terapéutico , Células PC12 , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA