Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 33(3): 3758-3771, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30481490

RESUMEN

Accumulating evidence suggests that the abnormal aggregation of amyloid-ß (Αß) peptide in Alzheimer's disease (AD) begins intraneuronally, within vesicles of the endosomal-lysosomal pathway where Aß is both generated and degraded. Metalloproteases, including endothelin-converting enzyme (ECE)-1 and -2, reside within these vesicles and normally limit the accumulation of intraneuronally produced Aß. In this study, we determined whether disruption of Aß catabolism could trigger Aß aggregation within neurons and increase the amount of Aß associated with exosomes, small extracellular vesicles derived from endosomal multivesicular bodies. Using cultured cell lines, primary neurons, and organotypic brain slices from an AD mouse model, we found that pharmacological inhibition of the ECE family of metalloproteases increased intracellular and extracellular Aß levels and promoted the intracellular formation of Aß oligomers, a process that did not require internalization of secreted Aß. In vivo, the accumulation of intraneuronal Aß aggregates was accompanied by increased levels of both extracellular and exosome-associated Aß, including oligomeric species. Neuronal exosomes were found to contain both ECE-1 and -2 activities, suggesting that multivesicular bodies are intracellular sites of Aß degradation by these enzymes. ECE dysfunction could lead to the accumulation of intraneuronal Aß aggregates and their subsequent release into the extracellular space via exosomes.-Pacheco-Quinto, J., Clausen, D., Pérez-González, R., Peng, H., Meszaros, A., Eckman, C. B., Levy, E., Eckman, E. A. Intracellular metalloprotease activity controls intraneuronal Aß aggregation and limits secretion of Aß via exosomes.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Exosomas/metabolismo , Metaloendopeptidasas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Endosomas/metabolismo , Enzimas Convertidoras de Endotelina/metabolismo , Espacio Extracelular/metabolismo , Femenino , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Cuerpos Multivesiculares/metabolismo , Neuronas/metabolismo , Proteolisis
3.
Hepatology ; 53(4): 1294-305, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21480332

RESUMEN

UNLABELLED: Mechanisms of brain edema in acute liver failure (ALF) are not completely understood. We recently demonstrated that matrix metalloproteinase 9 (MMP-9) induces significant alterations to occludin in brain endothelial cells in vitro and in brains of mice with experimental ALF (Hepatology 2009;50:1914). In this study we show that MMP-9-induced transactivation of epidermal growth factor receptor (EGFR) and p38 MAPK/NFκB (mitogen-activated protein kinase/nuclear factor-kappa B) signals participate in regulating brain endothelial occludin level. Mouse brain endothelial bEnd3 cells were exposed to MMP-9 or p38 MAPK up-regulation in the presence and absence of EGFR inhibitor, p38 MAPK inhibitor, NFκB inhibitor, and/or appropriate small interfering RNA. Reverse-transcription polymerase chain reaction (RT-PCR) and western blotting were used for messenger RNA and protein expression analyses. Immunohistochemical staining and confocal microscopy were used to demonstrate cellular EGFR activation. Intraperitoneal azoxymethane was use to induce ALF in mice. Brains of comatose ALF mice were processed for histological and biochemical analyses. When bEnd3 cells were exposed to MMP-9, EGFR was significantly transactivated, followed by p38 MAPK activation, I-kappa B alpha (IκBα) degradation, NFκB activation, and suppression of occludin synthesis and expression. Similar EGFR activation and p38 MAPK/NFκB activation were found in the brains of ALF mice, and these changes were attenuated with GM6001 treatment. CONCLUSION: EGFR activation with p38 MAPK/NFκB signaling contributes to the regulation of tight junction integrity in ALF. EGFR activation may thus play an important role in vasogenic brain edema in ALF.


Asunto(s)
Receptores ErbB/fisiología , Fallo Hepático Agudo/metabolismo , Proteínas de la Membrana/fisiología , Animales , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , FN-kappa B/metabolismo , Ocludina , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
J Surg Res ; 178(2): 907-14, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22591919

RESUMEN

BACKGROUND: Orthotopic liver transplantation (OLT) models in rats have been investigated in many studies. The reconstruction of hepatic artery is required for reliable OLT and also requires advanced skills. METHODS: The hepatic artery reconstructions by a hand-suture technique and a new method using a micro T-tube were investigated in rats with a whole-liver syngeneic graft. Operative time and postoperative patency were compared between the hand-suture and micro T-tube techniques. RESULTS: Our technique using the micro T-tube shortened the operative time of recipient surgery compared with the hand-suture technique and prolonged the operative time for the donor. The patency ratio was maintained at 24h after OLT with hand suturing but was significantly reduced with the micro T-tube, which had a patency ratio of 0.83 only up to 6h after OLT. CONCLUSION: The micro T-tube technique may have potential usefulness in the rat OLT model but requires further modification.


Asunto(s)
Arteria Hepática/cirugía , Trasplante de Hígado/métodos , Animales , Apoptosis , Tempo Operativo , Ratas , Ratas Endogámicas Lew , Grado de Desobstrucción Vascular
5.
J Neurosci ; 30(16): 5489-97, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20410102

RESUMEN

Globoid cell leukodystrophy (GLD) (Krabbe disease) is an autosomal recessive, degenerative, lysosomal storage disease caused by a severe loss of galactocerebrosidase (GALC) enzymatic activity. Of the >70 disease-causing mutations in the GALC gene, most are located outside of the catalytic domain of the enzyme. To determine how GALC mutations impair enzymatic activity, we investigated the impact of multiple disease-causing mutations on GALC processing, localization, and enzymatic activity. Studies in mammalian cells revealed dramatic decreases in GALC activity and a lack of appropriate protein processing into an N-terminal GALC fragment for each of the mutants examined. Consistent with this, we observed significantly less GALC localized to the lysosome and impairment in either the secretion or reuptake of mutant GALC. Notably, the D528N mutation was found to induce hyperglycosylation and protein misfolding. Reversal of these conditions resulted in an increase in proper processing and GALC activity, suggesting that glycosylation may play a critical role in the disease process in patients with this mutation. Recent studies have shown that enzyme inhibitors can sometimes "chaperone" misfolded polypeptides to their appropriate target organelle, bypassing the normal cellular quality control machinery and resulting in enhanced activity. To determine whether this may also work for GLD, we examined the effect of alpha-lobeline, an inhibitor of GALC, on D528N mutant cells. After treatment, GALC activity was significantly increased. This study suggests that mutations in GALC can cause GLD by impairing protein processing and/or folding and that pharmacological chaperones may be potential therapeutic agents for patients carrying certain mutations.


Asunto(s)
Galactosilceramidasa/genética , Leucodistrofia de Células Globoides/tratamiento farmacológico , Leucodistrofia de Células Globoides/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/uso terapéutico , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Galactosilceramidasa/antagonistas & inhibidores , Galactosilceramidasa/metabolismo , Humanos , Leucodistrofia de Células Globoides/enzimología , Chaperonas Moleculares/farmacología , Mutagénesis Sitio-Dirigida , Pliegue de Proteína/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética
6.
J Nat Prod ; 74(1): 38-44, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21141876

RESUMEN

Target-based drug discovery for Alzheimer's disease (AD) centered on modulation of the amyloid ß peptide has met with limited success. Therefore, recent efforts have focused on targeting the microtubule-associated protein tau. Tau pathologically accumulates in more than 15 neurodegenerative diseases and is most closely linked with postsymptomatic progression in AD. We endeavored to identify compounds that decrease tau stability rather than prevent its aggregation. An extract from Myrica cerifera (bayberry/southern wax myrtle) potently reduced both endogenous and overexpressed tau protein levels in cells and murine brain slices. The bayberry flavonoids myricetin and myricitrin were confirmed to contribute to this potency, but a diarylheptanoid, myricanol, was the most effective anti-tau component in the extract, with potency approaching the best targeted lead therapies. (+)-aR,11S-Myricanol, isolated from M. cerifera and reported here for the first time as the naturally occurring aglycone, was significantly more potent than commercially available (±)-myricanol. Myricanol may represent a novel scaffold for drug development efforts targeting tau turnover in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Diarilheptanoides/aislamiento & purificación , Diarilheptanoides/farmacología , Flavonoides/aislamiento & purificación , Flavonoides/farmacología , Myrica/química , Proteínas tau/metabolismo , Animales , Diarilheptanoides/química , Femenino , Flavonoides/química , Células HeLa , Humanos , Masculino , Ratones , Modelos Biológicos , Raíces de Plantas/química , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Proteínas tau/análisis , Proteínas tau/efectos de los fármacos
7.
J Clin Invest ; 117(3): 648-58, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17304350

RESUMEN

A primary pathologic component of Alzheimer's disease (AD) is the formation of neurofibrillary tangles composed of hyperphosphorylated tau (p-tau). Expediting the removal of these p-tau species may be a relevant therapeutic strategy. Here we report that inhibition of Hsp90 led to decreases in p-tau levels independent of heat shock factor 1 (HSF1) activation. A critical mediator of this mechanism was carboxy terminus of Hsp70-interacting protein (CHIP), a tau ubiquitin ligase. Cochaperones were also involved in Hsp90-mediated removal of p-tau, while those of the mature Hsp90 refolding complex prevented this effect. This is the first demonstration to our knowledge that blockade of the refolding pathway promotes p-tau turnover through degradation. We also show that peripheral administration of a novel Hsp90 inhibitor promoted selective decreases in p-tau species in a mouse model of tauopathy, further suggesting a central role for the Hsp90 complex in the pathogenesis of tauopathies. When taken in the context of known high-affinity Hsp90 complexes in affected regions of the AD brain, these data implicate a central role for Hsp90 in the development of AD and other tauopathies and may provide a rationale for the development of novel Hsp90-based therapeutic strategies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Tauopatías/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Barrera Hematoencefálica/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Humanos , Masculino , Ratones , Fosforilación , Pliegue de Proteína , ARN Interferente Pequeño/farmacología , Serina/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 104(26): 11062-7, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17573534

RESUMEN

Proteolytic processing of the amyloid precursor protein (APP) by beta-secretase, beta-site APP cleaving enzyme (BACE1), is the initial step in the production of the amyloid beta (Abeta) peptide, which is involved in the pathogenesis of Alzheimer's disease. The normal cellular function of the prion protein (PrP(C)), the causative agent of the transmissible spongiform encephalopathies such as Creutzfeldt-Jakob disease in humans, remains enigmatic. Because both APP and PrP(C) are subject to proteolytic processing by the same zinc metalloproteases, we tested the involvement of PrP(C) in the proteolytic processing of APP. Cellular overexpression of PrP(C) inhibited the beta-secretase cleavage of APP and reduced Abeta formation. Conversely, depletion of PrP(C) in mouse N2a cells by siRNA led to an increase in Abeta peptides secreted into the medium. In the brains of PrP knockout mice and in the brains from two strains of scrapie-infected mice, Abeta levels were significantly increased. Two mutants of PrP, PG14 and A116V, that are associated with familial human prion diseases failed to inhibit the beta-secretase cleavage of APP. Using constructs of PrP, we show that this regulatory effect of PrP(C) on the beta-secretase cleavage of APP required the localization of PrP(C) to cholesterol-rich lipid rafts and was mediated by the N-terminal polybasic region of PrP(C) via interaction with glycosaminoglycans. In conclusion, this is a mechanism by which the cellular production of the neurotoxic Abeta is regulated by PrP(C) and may have implications for both Alzheimer's and prion diseases.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Priones/fisiología , Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/metabolismo , Animales , Sitios de Unión , Línea Celular , Humanos , Microdominios de Membrana , Ratones , Mutación , Enfermedades por Prión/etiología , Priones/genética , Priones/metabolismo
9.
Neuron ; 47(2): 191-199, 2005 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16039562

RESUMEN

Considerable circumstantial evidence suggests that Abeta42 is the initiating molecule in Alzheimer's disease (AD) pathogenesis. However, the absolute requirement for Abeta42 for amyloid deposition has never been demonstrated in vivo. We have addressed this by developing transgenic models that express Abeta1-40 or Abeta1-42 in the absence of human amyloid beta protein precursor (APP) overexpression. Mice expressing high levels of Abeta1-40 do not develop overt amyloid pathology. In contrast, mice expressing lower levels of Abeta1-42 accumulate insoluble Abeta1-42 and develop compact amyloid plaques, congophilic amyloid angiopathy (CAA), and diffuse Abeta deposits. When mice expressing Abeta1-42 are crossed with mutant APP (Tg2576) mice, there is also a massive increase in amyloid deposition. These data establish that Abeta1-42 is essential for amyloid deposition in the parenchyma and also in vessels.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Vasos Sanguíneos/metabolismo , Encéfalo/metabolismo , Fragmentos de Péptidos/metabolismo , Piamadre/metabolismo , Placa Amiloide/metabolismo , Factores de Edad , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Benzotiazoles , Vasos Sanguíneos/patología , Vasos Sanguíneos/ultraestructura , Northern Blotting/métodos , Western Blotting/métodos , Encéfalo/patología , Encéfalo/ultraestructura , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Hibridación in Situ/métodos , Masculino , Espectrometría de Masas/métodos , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Mutación , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/genética , Piamadre/patología , Piamadre/ultraestructura , Placa Amiloide/patología , Tiazoles/metabolismo
10.
Neuron ; 44(2): 227-38, 2004 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-15473963

RESUMEN

Amyloid beta-peptide (Abeta) accumulation in specific brain regions is a pathological hallmark of Alzheimer's disease (AD). We have previously reported that a well-characterized acyl-coenzyme A: cholesterol acyltransferase (ACAT) inhibitor, CP-113,818, inhibits Abeta production in cell-based experiments. Here, we assessed the efficacy of CP-113,818 in reducing AD-like pathology in the brains of transgenic mice expressing human APP(751) containing the London (V717I) and Swedish (K670M/N671L) mutations. Two months of treatment with CP-113,818 reduced the accumulation of amyloid plaques by 88%-99% and membrane/insoluble Abeta levels by 83%-96%, while also decreasing brain cholesteryl-esters by 86%. Additionally, soluble Abeta(42) was reduced by 34% in brain homogenates. Spatial learning was slightly improved and correlated with decreased Abeta levels. In nontransgenic littermates, CP-113,818 also reduced ectodomain shedding of endogenous APP in the brain. Our results suggest that ACAT inhibition may be effective in the prevention and treatment of AD by inhibiting generation of the Abeta peptide.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Encéfalo/patología , Inhibidores Enzimáticos/uso terapéutico , Piridinas/uso terapéutico , Esterol O-Aciltransferasa/efectos de los fármacos , Glándulas Suprarrenales/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Ésteres del Colesterol/análisis , Ésteres del Colesterol/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/efectos adversos , Femenino , Humanos , Aprendizaje/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Piridinas/efectos adversos , Factores Sexuales , Esterol O-Aciltransferasa/metabolismo
11.
J Neurosci ; 27(29): 7817-26, 2007 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-17634375

RESUMEN

It is well established that the proteolytic processing of the beta-amyloid precursor protein (APP) generates beta-amyloid (Abeta), which plays a central role in the pathogenesis of Alzheimer's disease (AD). In contrast, the physiological role of APP and of its numerous proteolytic fragments and the question of whether a loss of these functions contributes to AD are still unknown. To address this question, we replaced the endogenous APP locus by gene-targeted alleles and generated two lines of knock-in mice that exclusively express APP deletion variants corresponding either to the secreted APP ectodomain (APPs alpha) or to a C-terminal (CT) truncation lacking the YENPTY interaction motif (APPdeltaCT15). Interestingly, the deltaCT15 deletion resulted in reduced turnover of holoAPP, increased cell surface expression, and strongly reduced Abeta levels in brain, likely because of reduced processing in the endocytic pathway. Most importantly, we demonstrate that in both APP knock-in lines the expression of APP N-terminal domains either grossly attenuated or completely rescued the prominent deficits of APP knock-out mice, such as reductions in brain and body weight, grip strength deficits, alterations in circadian locomotor activity, exploratory activity, and the impairment in spatial learning and long-term potentiation. Together, our data suggest that the APP C terminus is dispensable and that APPs alpha is sufficient to mediate the physiological functions of APP assessed by these tests.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/deficiencia , Conducta Animal/fisiología , Encéfalo/patología , Potenciación a Largo Plazo/fisiología , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Péptidos beta-Amiloides/metabolismo , Análisis de Varianza , Animales , Peso Corporal/genética , Encéfalo/metabolismo , Línea Celular , Regulación de la Expresión Génica/genética , Fuerza de la Mano/fisiología , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Actividad Motora/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Eliminación de Secuencia/fisiología , Conducta Espacial/fisiología
12.
FASEB J ; 21(10): 2520-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17403939

RESUMEN

Globoid cell leukodystrophy (GLD), also known as Krabbe disease, is a devastating, degenerative neurological disorder. It is inherited as an autosomal recessive trait caused by loss-of-function mutations in the galactocerebrosidase (GALC) gene. Previously, we have shown that peripheral injection of recombinant GALC, administered every other day, results in a substantial improvement in early clinical phenotype in the twitcher mouse model of GLD. While we did detect active enzyme in the brain following peripheral administration, most of the administered enzyme was localized to the periphery. Given the substantial central nervous system (CNS) involvement in this disease, we were interested in determining whether or not a single-dose administration of the recombinant enzyme directly to the CNS, which could potentially be achieved clinically, would result in any substantial improvement. Following intracerebroventricular (i.c.v.) administration of GALC we noted a significant, 16.5%, reduction in the GALC substrate psychosine, the abnormal accumulation of which is believed to play a pivotal role in the CNS pathology observed in this disease. Moreover, recombinant GALC was found not only in periventricular regions but also at sites distant to the injection such as the cerebral cortex and cerebellum. Most importantly, animals receiving a single i.c.v. dose of the enzyme at postnatal day 20 survived up to 51 days, which compares favorably to the control twitcher animals, which normally only live to postnatal day 40/42. These results indicate that even a single i.c.v. administration of the recombinant enzyme can have significant clinical impact and suggests that other lysosomal storage disorders with significant CNS involvement may similarly benefit.


Asunto(s)
Galactosilceramidasa/uso terapéutico , Leucodistrofia de Células Globoides/tratamiento farmacológico , Animales , Encéfalo/enzimología , Encéfalo/patología , Galactosilceramidasa/administración & dosificación , Galactosilceramidasa/deficiencia , Galactosilceramidasa/genética , Galactosilceramidasa/farmacocinética , Inyecciones Intraventriculares , Cinética , Leucodistrofia de Células Globoides/enzimología , Leucodistrofia de Células Globoides/patología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Especificidad por Sustrato , Distribución Tisular
13.
J Invest Surg ; 21(3): 101-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18569429

RESUMEN

Brain edema secondary to increased blood-brain barrier (BBB) permeability is a lethal complication in fulminant hepatic failure (FHF). Intact tight junctions (TJ) between brain capillary endothelial cells are critical for normal BBB function. However, the role of TJ in FHF has not been explored. We hypothesized that alterations in the composition of TJ proteins would result in increased BBB permeability in FHF. In this study, FHF was induced in C57BL/6J mice by using azoxymethane. BBB permeability was assessed with sodium fluorescein. Expression of TJ proteins was determined by Western blot, and their cellular distribution was examined using immunofluorescent microscopy. Comatose FHF mice had significant cerebral sodium fluorescein extravasation compared with control and precoma FHF mice, indicating increased BBB permeability. Western blot analysis showed a significant decrease in zonula occludens (ZO)-2 expression starting in the precoma stage. Immunofluorescent microscopy showed a significantly altered distribution pattern of ZO-2 in isolated microvessels from precoma FHF mice. These changes were more prominent in comatose FHF animals. Significant alterations in ZO-2 expression and distribution in the tight junctions preceded the increased BBB permeability in FHF mice. These results suggest that ZO-2 may play an important role in the pathogenesis of brain edema in FHF.


Asunto(s)
Barrera Hematoencefálica , Fallo Hepático Agudo/metabolismo , Proteínas de la Membrana/fisiología , Animales , Encéfalo/irrigación sanguínea , Modelos Animales de Enfermedad , Masculino , Proteínas de la Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Proteína de la Zonula Occludens-2
14.
J Neurosci ; 26(27): 7272-80, 2006 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-16822985

RESUMEN

The presence of endogenous stem cell populations in the adult mammalian CNS suggests an innate potential for regeneration and represents a potential resource for neuroregenerative therapy aimed at the treatment of neurodegenerative disorders, such as Parkinson's disease. However, it is first necessary to examine the microenvironmental signals required to activate these innate reparative mechanisms. The small molecule neurotransmitter dopamine has been shown to regulate cell cycle in developing and adult brain, and the D3 receptor is known to play an important role in dopaminergic development. Pharmacological activation of the dopamine D3 receptor has been shown to trigger neurogenesis in the substantia nigra of the adult rat brain. Here, we examined the cell proliferative, neurogenic, and behavioral effects of the dopamine D3 receptor agonist 7-OH-DPAT (7-hydroxy-N,N-di-n-propyl-2-aminotetralin) in a 6-hydroxydopamine model of Parkinson's disease. Consistent with previous findings, we observed a significant induction of cell proliferation in the substantia nigra pars compacta (SN(C)) with a time-dependent adoption of a neuronal dopaminergic phenotype in many of these cells. Indices of nigrostriatal integrity were also affected. Dopaminergic cell counts in the lesioned SN(C) recovered substantially in a time-dependent manner. Similarly, retrograde tracing revealed a restoration of striatal innervation from these cells, with evidence for projections arising from newly generated cells. Finally, we observed a substantial and persistent recovery of locomotor function in these animals. The results of these studies will further our understanding of the environmental signals regulating neurogenesis in the adult brain and could have significant implications for the design of novel treatment strategies for Parkinson's disease.


Asunto(s)
Agonistas de Dopamina/farmacología , Actividad Motora/fisiología , Trastornos Parkinsonianos/tratamiento farmacológico , Receptores de Dopamina D3/agonistas , Tetrahidronaftalenos/farmacología , Animales , División Celular/efectos de los fármacos , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Modelos Animales de Enfermedad , Femenino , Vías Nerviosas , Neuronas/citología , Neuronas/fisiología , Trastornos Parkinsonianos/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D3/fisiología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Sustancia Negra/citología , Sustancia Negra/fisiología
15.
FASEB J ; 20(8): 1269-71, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16636099

RESUMEN

For millennia, ginseng and some of its components have been used to treat a wide variety of medical conditions, including age-related memory impairment. Because of its purported effects and apparently low rate of side effects, ginseng remains one of the top selling natural product remedies in the United States. Given its potential role for improving age-related memory impairments and its common use in China for the treatment of Alzheimer's disease-like symptoms, we analyzed the effects of commercially available preparations of ginseng on the accumulation of the Alzheimer's amyloid beta peptide (Abeta) in a cell-based model system. In this model system, ginseng treatment resulted in a significant reduction in the levels of Abeta in the conditioned medium. We next examined the effects of several compounds isolated from ginseng and found that certain ginsenosides lowered Abeta concentration in a dose-dependent manner with ginsenoside Rg3 having an approximate IC50 of under 25 microM against Abeta42. Furthermore, we found that three of these isolated components, ginsenoside Rg1, Rg3, and RE, resulted in significant reductions in the amount of Abeta detected in the brains of animals after single oral doses of these agents. The results indicate that ginseng itself, or purified ginsenosides, may have similarly useful effects in human disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ginsenósidos/farmacología , Administración Oral , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Cricetinae , Cricetulus , Femenino , Ginsenósidos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Panax/química , Fragmentos de Péptidos/metabolismo , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología
16.
Neurol Clin ; 25(3): 669-82, vi, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17659184

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disease. To rationally develop novel therapeutic and/or preventative agents for AD, an understanding of the etiology and pathogenesis of this complex disease is necessary. This article examines the evidence for the amyloid hypothesis of AD pathogenesis and discusses how it relates to the neurological and neuropathological features of AD, the known genetic risk factors and causative mutations, and the heightened risk associated with advanced age.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Amiloidosis/genética , Amiloidosis/patología , Anciano , Animales , Humanos
18.
FASEB J ; 19(11): 1549-51, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15987783

RESUMEN

Globoid cell leukodystrophy (GLD) or Krabbe disease is a devastating, degenerative neurological disorder caused by mutations in the galactosylceramidase (GALC) gene that severely affect enzyme activity. Currently, treatment options for this disorder are very limited. Enzyme replacement therapy (ERT) has been shown to be effective in lysosomal storage disorders with predominantly peripheral manifestations such as type I Gaucher's and Fabry's disease. Little however is known about the possible benefit of ERT in GLD, which has a substantial central nervous system component. In this study, we examined the effect of peripheral GALC injections in the twitcher mouse model of the disease. Although we were unable to block the precipitous decline that normally occurs just before death, we did observe significant early improvements in motor performance, a substantial attenuation in the initial failure to thrive, and an increase in life span. Immunohistochemical and activity analyses demonstrated GALC uptake in multiple tissues, including the brain. This was associated with a decrease in the abnormal accumulation of the GALC substrate psychosine, which is thought to play a pivotal role in disease pathology. These results indicate that peripheral ERT is likely to be beneficial in GLD.


Asunto(s)
Galactosilceramidasa/uso terapéutico , Leucodistrofia de Células Globoides/tratamiento farmacológico , Animales , Barrera Hematoencefálica , Línea Celular , Modelos Animales de Enfermedad , Insuficiencia de Crecimiento/tratamiento farmacológico , Marcha/efectos de los fármacos , Galactosilceramidasa/análisis , Humanos , Inmunohistoquímica , Leucodistrofia de Células Globoides/enzimología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Psicosina/análisis , Proteínas Recombinantes/uso terapéutico
19.
Neurobiol Aging ; 48: 83-92, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27644077

RESUMEN

Impaired clearance of amyloid-ß peptide (Aß) has been postulated to significantly contribute to the amyloid accumulation typical of Alzheimer's disease. Among the enzymes known to degrade Aß in vivo are endothelin-converting enzyme (ECE)-1, ECE-2, and neprilysin (NEP), and evidence suggests that they regulate independent pools of Aß that may be functionally significant. To better understand the differential regulation of Aß concentration by its physiological degrading enzymes, we characterized the cell and region-specific expression pattern of ECE-1, ECE-2, and NEP by in situ hybridization and immunohistochemistry in brain areas relevant to Alzheimer's disease. In contrast to the broader distribution of ECE-1, ECE-2 and NEP were found enriched in GABAergic neurons. ECE-2 was majorly expressed by somatostatin-expressing interneurons and was active in isolated synaptosomes. NEP messenger RNA was found mainly in parvalbumin-expressing interneurons, with NEP protein localized to perisomatic parvalbuminergic synapses. The identification of somatostatinergic and parvalbuminergic synapses as hubs for Aß degradation is consistent with the possibility that Aß may have a physiological function related to the regulation of inhibitory signaling.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Enzimas Convertidoras de Endotelina/metabolismo , Neuronas GABAérgicas/enzimología , Hipocampo/citología , Hipocampo/enzimología , Neocórtex/citología , Neocórtex/enzimología , Neprilisina/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/fisiología , Animales , Enzimas Convertidoras de Endotelina/genética , Enzimas Convertidoras de Endotelina/fisiología , Expresión Génica , Ratones Transgénicos , Neprilisina/genética , Neprilisina/fisiología , ARN Mensajero/metabolismo , Sinapsis/enzimología
20.
Sci STKE ; 2003(172): RE4, 2003 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-12621149

RESUMEN

Intramembranous proteolysis (IP) is a recently recognized mechanism for transmembrane signal transduction that involves proteolysis of transmembrane proteins within their membrane-spanning domains. Juxtamembranous proteolysis (JP) is similar, but proteolytic cleavage of a transmembrane protein occurs at a site close to, but not within, the transmembrane domain of the target protein. In both IP and JP, a soluble domain of a transmembrane protein is released from its membrane tether. This domain can then transmit a signal either locally or at some distance from the site of cleavage. In certain signaling pathways, JP and IP are linked. JP on one side of the membrane results in secondary IP, which then releases a signaling domain from the membrane. Whereas well-characterized proteases such as caspases, the proteasome, and metalloprotease disintegrins, have been implicated in JP, three families of multipass membrane proteases (MpMPs) have now been shown to carry out IP. Recent studies of events mediated by IP and JP indicate that they regulate key cellular signaling events including pathways involved in sterol regulation, cell fate selection, and growth regulation. Moreover, IP and JP have important roles in certain diseases such as Alzheimer's disease. Because some of the proteases mediating IP and JP can be selectivity inhibited, inhibitors targeting these proteases are likely to alter both physiologic and pathologic events triggered by IP and JP.


Asunto(s)
Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/fisiología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Animales , Apoptosis/fisiología , División Celular/fisiología , Membrana Celular/enzimología , Membrana Celular/patología , Membrana Celular/fisiología , Humanos , Hidrólisis , Modelos Biológicos , Péptido Hidrolasas/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA