Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Ann Neurol ; 75(5): 684-99, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24687915

RESUMEN

OBJECTIVE: Proteolytic cleavage of the amyloid precursor protein (APP) generates ß-amyloid (Aß) peptides. Prolonged accumulation of Aß in the brain underlies the pathogenesis of Alzheimer disease (AD) and is regarded as a principal target for development of disease-modifying therapeutics. METHODS: Using Chinese hamster ovary (CHO) APP751SW cells, we identified and characterized effects of 2-([pyridine-2-ylmethyl]-amino)-phenol (2-PMAP) on APP steady-state level and Aß production. Outcomes of 2-PMAP treatment on Aß accumulation and associated memory deficit were studied in APPSW /PS1dE9 AD transgenic model mice. RESULTS: In CHO APP751SW cells, 2-PMAP lowered the steady-state APP level and inhibited Aßx-40 and Aßx-42 production in a dose-response manner with a minimum effective concentration ≤ 0.5µM. The inhibitory effect of 2-PMAP on translational efficiency of APP mRNA into protein was directly confirmed using a 35S-methionine/cysteine metabolic labeling technique, whereas APP mRNA level remained unaltered. Administration of 2-PMAP to APPSW /PS1dE9 mice reduced brain levels of full-length APP and its C-terminal fragments and lowered levels of soluble Aßx-40 and Aßx-42 . Four-month chronic treatment of APPSW /PS1dE9 mice revealed no observable toxicity and improved animals' memory performance. 2-PMAP treatment also caused significant reduction in brain Aß deposition determined by both unbiased quantification of Aß plaque load and biochemical analysis of formic acid-extracted Aßx-40 and Aßx-42 levels and the level of oligomeric Aß. INTERPRETATION: We demonstrate the potential of modulating APP steady-state expression level as a safe and effective approach for reducing Aß deposition in AD transgenic model mice.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Regulación de la Expresión Génica , Placa Amiloide/prevención & control , Precursor de Proteína beta-Amiloide/fisiología , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1/genética
2.
Am J Pathol ; 182(5): 1750-68, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23499462

RESUMEN

Accumulation of ß-amyloid (Aß) in the brain is a key event in Alzheimer disease pathogenesis. Apolipoprotein (Apo) E is a lipid carrier protein secreted by astrocytes, which shows inherent affinity for Aß and has been implicated in the receptor-mediated Aß uptake by neurons. To characterize ApoE involvement in the intraneuronal Aß accumulation and to investigate whether blocking the ApoE/Aß interaction could reduce intraneuronal Aß buildup, we used a noncontact neuronal-astrocytic co-culture system, where synthetic Aß peptides were added into the media without or with cotreatment with Aß12-28P, which is a nontoxic peptide antagonist of ApoE/Aß binding. Compared with neurons cultured alone, intraneuronal Aß content was significantly increased in neurons co-cultured with wild-type but not with ApoE knockout (KO) astrocytes. Neurons co-cultured with astrocytes also showed impaired intraneuronal degradation of Aß, increased level of intraneuronal Aß oligomers, and marked down-regulation of several synaptic proteins. Aß12-28P treatment significantly reduced intraneuronal Aß accumulation, including Aß oligomer level, and inhibited loss of synaptic proteins. Furthermore, we showed significantly reduced intraneuronal Aß accumulation in APPSW/PS1dE9/ApoE KO mice compared with APPSW/PS1dE9/ApoE targeted replacement mice that expressed various human ApoE isoforms. Data from our co-culture and in vivo experiments indicate an essential role of ApoE in the mechanism of intraneuronal Aß accumulation and provide evidence that ApoE/Aß binding antagonists can effectively prevent this process.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Sinapsis/patología , Animales , Astrocitos/patología , Células Cultivadas , Técnicas de Cocultivo , Endocitosis , Espacio Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Humanos , Ratones , Ratones Noqueados , Degeneración Nerviosa/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Fracciones Subcelulares/metabolismo , Sinapsis/metabolismo
3.
Front Aging Neurosci ; 14: 845291, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572125

RESUMEN

The APOE gene is diversified by three alleles ε2, ε3, and ε4 encoding corresponding apolipoprotein (apo) E isoforms. Possession of the ε4 allele is signified by increased risks of age-related cognitive decline, Alzheimer's disease (AD), and the rate of AD dementia progression. ApoE is secreted by astrocytes as high-density lipoprotein-like particles and these are internalized by neurons upon binding to neuron-expressed apoE receptors. ApoE isoforms differentially engage neuronal plasticity through poorly understood mechanisms. We examined here the effects of native apoE lipoproteins produced by immortalized astrocytes homozygous for ε2, ε3, and ε4 alleles on the maturation and the transcriptomic profile of primary hippocampal neurons. Control neurons were grown in the presence of conditioned media from Apoe -/- astrocytes. ApoE2 and apoE3 significantly increase the dendritic arbor branching, the combined neurite length, and the total arbor surface of the hippocampal neurons, while apoE4 fails to produce similar effects and even significantly reduces the combined neurite length compared to the control. ApoE lipoproteins show no systemic effect on dendritic spine density, yet apoE2 and apoE3 increase the mature spines fraction, while apoE4 increases the immature spine fraction. This is associated with opposing effects of apoE2 or apoE3 and apoE4 on the expression of NR1 NMDA receptor subunit and PSD95. There are 1,062 genes differentially expressed across neurons cultured in the presence of apoE lipoproteins compared to the control. KEGG enrichment and gene ontology analyses show apoE2 and apoE3 commonly activate expression of genes involved in neurite branching, and synaptic signaling. In contrast, apoE4 cultured neurons show upregulation of genes related to the glycolipid metabolism, which are involved in dendritic spine turnover, and those which are usually silent in neurons and are related to cell cycle and DNA repair. In conclusion, our work reveals that lipoprotein particles comprised of various apoE isoforms differentially regulate various neuronal arbor characteristics through interaction with neuronal transcriptome. ApoE4 produces a functionally distinct transcriptomic profile, which is associated with attenuated neuronal development. Differential regulation of neuronal transcriptome by apoE isoforms is a newly identified biological mechanism, which has both implication in the development and aging of the CNS.

4.
Acta Neuropathol Commun ; 9(1): 157, 2021 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-34565486

RESUMEN

Prion diseases or prionoses are a group of rapidly progressing and invariably fatal neurodegenerative diseases. The pathogenesis of prionoses is associated with self-replication and connectomal spread of PrPSc, a disease specific conformer of the prion protein. Microglia undergo activation early in the course of prion pathogenesis and exert opposing roles in PrPSc mediated neurodegeneration. While clearance of PrPSc and apoptotic neurons have disease-limiting effect, microglia-driven neuroinflammation bears deleterious consequences to neuronal networks. Apolipoprotein (apo) E is a lipid transporting protein with pleiotropic functions, which include controlling of the phagocytic and inflammatory characteristics of activated microglia in neurodegenerative diseases. Despite the significance of microglia in prion pathogenesis, the role of apoE in prionoses has not been established. We showed here that infection of wild type mice with 22L mouse adapted scrapie strain is associated with significant increase in the total brain apoE protein and mRNA levels and also with a conspicuous cell-type shift in the apoE expression. There is reduced expression of apoE in activated astrocytes and marked upregulation of apoE expression by activated microglia. We also showed apoE ablation exaggerates PrPSc mediated neurodegeneration. Apoe-/- mice have shorter disease incubation period, increased load of spongiform lesion, pronounced neuronal loss, and exaggerated astro and microgliosis. Astrocytes of Apoe-/- mice display salient upregulation of transcriptomic markers defining A1 neurotoxic astrocytes while microglia show upregulation of transcriptomic markers characteristic for microglial neurodegenerative phenotype. There is impaired clearance of PrPSc and dying neurons by microglia in Apoe-/- mice along with increased level of proinflammatory cytokines. Our work indicates that apoE absence renders clearance of PrPSc and dying neurons by microglia inefficient, while the excess of neuronal debris promotes microglial neurodegenerative phenotype aggravating the vicious cycle of neuronal death and neuroinflammation.


Asunto(s)
Apolipoproteínas E/metabolismo , Microglía/patología , Degeneración Nerviosa/patología , Enfermedades por Prión/patología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Fenotipo , Enfermedades por Prión/metabolismo
6.
Mol Neurodegener ; 15(1): 50, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32907613

RESUMEN

BACKGROUND: Disruption of ß-amyloid (Aß) homeostasis is the initial culprit in Alzheimer's disease (AD) pathogenesis. Astrocytes respond to emerging Aß plaques by altering their phenotype and function, yet molecular mechanisms governing astrocytic response and their precise role in countering Aß deposition remain ill-defined. Peroxiredoxin (PRDX) 6 is an enzymatic protein with independent glutathione peroxidase (Gpx) and phospholipase A2 (PLA2) activities involved in repair of oxidatively damaged cell membrane lipids and cellular signaling. In the CNS, PRDX6 is uniquely expressed by astrocytes and its exact function remains unexplored. METHODS: APPswe/PS1dE9 AD transgenic mice were once crossed to mice overexpressing wild-type Prdx6 allele or to Prdx6 knock out mice. Aß pathology and associated neuritic degeneration were assessed in mice aged 10 months. Laser scanning confocal microscopy was used to characterize Aß plaque morphology and activation of plaque-associated astrocytes and microglia. Effect of Prdx6 gene dose on plaque seeding was assessed in mice aged six months. RESULTS: We show that hemizygous knock in of the overexpressing Prdx6 transgene in APPswe/PS1dE9 AD transgenic mice promotes selective enticement of astrocytes to Aß plaques and penetration of plaques by astrocytic processes along with increased number and phagocytic activation of periplaque microglia. This effects suppression of nascent plaque seeding and remodeling of mature plaques consequently curtailing brain Aß load and Aß-associated neuritic degeneration. Conversely, Prdx6 haplodeficiency attenuates astro- and microglia activation around Aß plaques promoting Aß deposition and neuritic degeneration. CONCLUSIONS: We identify here PRDX6 as an important factor regulating response of astrocytes toward Aß plaques. Demonstration that phagocytic activation of periplaque microglia vary directly with astrocytic PRDX6 expression level implies previously unappreciated astrocyte-guided microglia effect in Aß proteostasis. Our showing that upregulation of PRDX6 attenuates Aß pathology may be of therapeutic relevance for AD.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Peroxiredoxina VI/metabolismo , Placa Amiloide/metabolismo , Proteostasis/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/patología
7.
Neurobiol Dis ; 34(2): 267-78, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19385058

RESUMEN

The pathogenesis of prion diseases is related to conformational transformation of cellular prion protein (PrP(C)) into a toxic, infectious, and self-replicating conformer termed PrP(Sc). Following extracerebral inoculation, the replication of PrP(Sc) is confined for months to years to the lymporeticular system (LRS) before the secondary CNS involvement results in occurrence of neurological symptoms. Therefore, replication of PrP(Sc), in the early stage of infection can be targeted by therapeutic approaches, which like passive immunization have limited blood-brain-barrier penetration. In this study, we show that 6D11 anti-PrP monoclonal antibody (Mab) prevents infection on a FDC-P1 myeloid precursor cell line stably infected with 22L mouse adapted scrapie strain. Passive immunization of extracerebrally infected CD-1 mice with Mab 6D11 resulted in effective suppression of PrP(Sc) replication in the LRS. Although, a rebound of PrP(Sc) presence occurred when the Mab 6D11 treatment was stopped, passively immunized mice showed a prolongation of the incubation period by 36.9% (pb0.0001) and a significant decrease in CNS pathology compared to control groups receiving vehicle or murine IgG. Our results indicate that antibody-based therapeutic strategies can be used, even on a short-term basis, to delay or prevent disease in subjects accidentally exposed to prions.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Inmunización Pasiva/métodos , Sistema Linfático/efectos de los fármacos , Células Progenitoras Mieloides/efectos de los fármacos , Proteínas PrPSc/antagonistas & inhibidores , Enfermedades por Prión/tratamiento farmacológico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/metabolismo , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Femenino , Humanos , Sistema Linfático/inmunología , Sistema Linfático/metabolismo , Ratones , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Proteínas PrPSc/biosíntesis , Enfermedades por Prión/inmunología , Enfermedades por Prión/metabolismo , Conformación Proteica/efectos de los fármacos , Scrapie/tratamiento farmacológico , Scrapie/inmunología , Scrapie/metabolismo , Resultado del Tratamiento
8.
Mol Neurobiol ; 56(3): 2073-2091, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29987703

RESUMEN

PrPSc is an infectious and disease-specific conformer of the prion protein, which accumulation in the CNS underlies the pathology of prion diseases. PrPSc replicates by binding to the cellular conformer of the prion protein (PrPC) expressed by host cells and rendering its secondary structure a likeness of itself. PrPC is a plasma membrane anchored protein, which constitutively recirculates between the cell surface and the endocytic compartment. Since PrPSc engages PrPC along this trafficking pathway, its replication process is often referred to as "recycling propagation." Certain monoclonal antibodies (mAbs) directed against prion protein can abrogate the presence of PrPSc from prion-infected cells. However, the precise mechanism(s) underlying their therapeutic propensities remains obscure. Using N2A murine neuroblastoma cell line stably infected with 22L mouse-adapted scrapie strain (N2A/22L), we investigated here the modus operandi of the 6D11 clone, which was raised against the PrPSc conformer and has been shown to permanently clear prion-infected cells from PrPSc presence. We determined that 6D11 mAb engages and sequesters PrPC and PrPSc at the cell surface. PrPC/6D11 and PrPSc/6D11 complexes are then endocytosed from the plasma membrane and are directed to lysosomes, therefore precluding recirculation of nascent PrPSc back to the cell surface. Targeting PrPSc by 6D11 mAb to the lysosomal compartment facilitates its proteolysis and eventually shifts the balance between PrPSc formation and degradation. Ongoing translation of PrPC allows maintaining the steady-state level of prion protein within the cells, which was not depleted under 6D11 mAb treatment. Our findings demonstrate that through disrupting recycling propagation of PrPSc and promoting its degradation, 6D11 mAb restores cellular proteostasis of prion protein.


Asunto(s)
Anticuerpos Monoclonales , Lisosomas/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Proteínas Priónicas/metabolismo , Animales , Línea Celular Tumoral , Ratones , Proteolisis , Proteostasis , Scrapie/metabolismo
9.
Mol Neurodegener ; 12(1): 12, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28143566

RESUMEN

BACKGROUND: APOE genotype is the foremost genetic factor modulating ß-amyloid (Aß) deposition and risk of sporadic Alzheimer's disease (AD). Here we investigated how APOE genotype influences response to anti-Aß immunotherapy. METHODS: APPSW/PS1dE9 (APP) transgenic mice with targeted replacement of the murine Apoe gene for human APOE alleles received 10D5 anti-Aß or TY11-15 isotype control antibodies between the ages of 12 and 15 months. RESULTS: Anti-Aß immunization decreased both the load of fibrillar plaques and the load of Aß immunopositive plaques in mice of all APOE backgrounds. Although the relative reduction in parenchymal Aß plaque load was comparable across all APOE genotypes, APP/ε4 mice showed the greatest reduction in the absolute Aß plaque load values, given their highest baseline. The immunization stimulated phagocytic activation of microglia, which magnitude adjusted for the post-treatment plaque load was the greatest in APP/ε4 mice implying association between the ε4 allele and impaired Aß phagocytosis. Perivascular hemosiderin deposits reflecting ensued microhemorrhages were associated with vascular Aß (VAß) and ubiquitously present in control mice of all APOE genotypes, although in APP/ε3 mice their incidence was the lowest. Anti-Aß immunization significantly reduced VAß burden but increased the number of hemosiderin deposits across all APOE genotypes with the strongest and the weakest effect in APP/ε2 and APP/ε3 mice, respectively. CONCLUSIONS: Our studies indicate that APOE genotype differentially modulates microglia activation and Aß plaque load reduction during anti-Aß immunotherapy. The APOE ε3 allele shows strong protective effect against immunotherapy associated microhemorrhages; while, conversely, the APOE ε2 allele increases risk thereof.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Apolipoproteínas E/genética , Animales , Modelos Animales de Enfermedad , Genotipo , Humanos , Inmunización Pasiva , Inmunohistoquímica , Ratones , Ratones Transgénicos , Placa Amiloide/genética
10.
J Neuropathol Exp Neurol ; 63(5): 418-28, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15198121

RESUMEN

In Alzheimer disease (AD) patients, early memory dysfunction is associated with glucose hypometabolism and neuronal loss in the hippocampus. Double transgenic (Tg) mice co-expressing the M146L presenilin 1 (PS1) and K670N/M671L, the double "Swedish" amyloid precursor protein (APP) mutations, are a model of AD amyloid-beta deposition (Abeta) that exhibits earlier and more profound impairments of working memory and learning than single APP mutant mice. In this study we compared performance on spatial memory tests, regional glucose metabolism, Abeta deposition, and neuronal loss in APP/PS1, PS1, and non-Tg (nTg) mice. At the age of 2 months no significant morphological and metabolic differences were detected between 3 studied genotypes. By 8 months, however, APP/PS1 mice developed selective impairment of spatial memory, which was significantly worse at 22 months and was accompanied by reduced glucose utilization in the hippocampus and a 35.8% dropout of neurons in the CA1 region. PS1 mice exhibited a similar degree of neuronal loss in CA1 but minimal memory deficit and no impairment of glucose utilization compared to nTg mice. Deficits in 22 month APP/PS1 mice were accompanied by a substantially elevated Abeta load, which rose from 2.5% +/- 0.4% at 8 months to 17.4% +/- 4.6%. These findings implicate Abeta or APP in the behavioral and metabolic impairments in APP/PS1 mice and the failure to compensate functionally for PS1-related hippocampal cell loss.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Glucosa/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Animales , Recuento de Células , Muerte Celular/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Hipocampo/patología , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto/fisiología , Proteínas de la Membrana/genética , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Transgénicos , Actividad Motora/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Presenilina-1
11.
J Neuropathol Exp Neurol ; 63(7): 775-84, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15290902

RESUMEN

The diagnosis of prion diseases in humans is challenging due to a lack of specific and sensitive non-invasive tests. Many forms of human prion disease including variant Creutzfeldt-Jakob disease (vCJD), Gerstmann-Sträussler-Scheinker (GSS) syndrome, and 10% of sporadic CJD cases are associated with amyloid deposition. Several positron emission tomography (PET) ligands have recently been developed to directly image beta-amyloid associated with Alzheimer disease. One of them, methoxy-X04, is a fluorescent derivative of Congo red with high binding affinity toward amyloid fibrils and good blood-brain barrier permeability. Using methoxy-X04, we investigated whether amyloid-targeting ligands can be also employed for direct imaging of amyloid deposits associated with some prion diseases. Such a method could potentially become a novel diagnostic approach for these conditions. Studies were performed on MB mice infected with the 87V mouse-adapted scrapie strain. Labeling of PrP amyloid plaques in brains of presymptomatic and symptomatic mice was demonstrated using in vivo transcranial two-photon microscopy after systemic administration of methoxy-X04. During real-time imaging, PrP amyloid deposits could be clearly distinguished 15 min after intravenous administration of methoxy-X04. The ligand showed rapid clearance from brain areas that did not contain amyloid deposits. PrP amyloid deposits could also be detected by direct application of methoxy-X04 on cerebellar sections from GSS patients. These results suggest that methoxy-X04 or similar derivatives could be used as PET imaging agents to improve the diagnosis of human prion diseases associated with amyloid deposition.


Asunto(s)
Alquenos , Péptidos beta-Amiloides/análisis , Derivados del Benceno , Placa Amiloide/patología , Enfermedades por Prión/diagnóstico por imagen , Enfermedades por Prión/patología , Tomografía Computarizada de Emisión/métodos , Alquenos/metabolismo , Alquenos/farmacocinética , Animales , Derivados del Benceno/metabolismo , Derivados del Benceno/farmacocinética , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Tasa de Depuración Metabólica/efectos de los fármacos , Tasa de Depuración Metabólica/fisiología , Ratones , Placa Amiloide/metabolismo , Valor Predictivo de las Pruebas , Enfermedades por Prión/metabolismo , Reproducibilidad de los Resultados , Scrapie/diagnóstico por imagen , Scrapie/metabolismo , Scrapie/patología , Estilbenos
12.
J Neurol Sci ; 196(1-2): 27-32, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11959152

RESUMEN

Hypercortisolemia is thought to be a marker of the stress response following stroke. The aim of this study was to investigate the prevalence and prognostic significance of hypercortisolemia. The circadian variation of cortisol level and the relationship between serum cortisol levels and other stress, inflammatory, and haemostatic markers were also investigated. Seventy consecutive patients with their first ischemic stroke and 24 age- and sex-matched controls were included in the study. Serum cortisol levels (at 6:00 AM, 10:00 AM, 6:00 PM, and 10:00 PM), 24-h urine catecholamine excretion, beta-thromboglobulin levels, and other standard biochemical and haematological parameters were measured on the first day of hospitalisation and in control subjects. Outcome measures used the Barthel Index at Day 30, as well as 30- and 90-day mortality rates. Hypercortisolemia, defined as at least two of the four measurements above the normal range of serum cortisol levels (i.e. >618 nmol/l from the morning samples and >460 nmol/l from the evening samples) was found in 25 (35.7%) of the acute stroke patients and in 3 (12.5%) of the controls (p<0.05). Hypercortisolemia was associated with older age, greater severity of neurological deficit, larger ischemic lesions on CT, and worse prognoses (p<0.05). The study did not find a correlation between serum cortisol levels and other markers of the stress response such as catecholamines excretion and glucose levels. A significant correlation between serum cortisol levels and some markers of the inflammatory response, such as fever, fibrinogen level, white blood cell (WBC) count, and beta-thromboglobulin level, was established in stroke patients. Prognostic significance of hypercortisolemia in acute stroke patients seems to be related to the inflammatory response rather than to the stress response.


Asunto(s)
Hiperfunción de las Glándulas Suprarrenales/sangre , Hidrocortisona/sangre , Estrés Fisiológico/sangre , Accidente Cerebrovascular/sangre , Regulación hacia Arriba/inmunología , Enfermedad Aguda , Hiperfunción de las Glándulas Suprarrenales/inmunología , Hiperfunción de las Glándulas Suprarrenales/fisiopatología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Ritmo Circadiano/inmunología , Femenino , Humanos , Hidrocortisona/inmunología , Inflamación/sangre , Inflamación/inmunología , Inflamación/fisiopatología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Prevalencia , Pronóstico , Factores Sexuales , Estrés Fisiológico/inmunología , Estrés Fisiológico/fisiopatología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/fisiopatología , Tasa de Supervivencia
13.
Przegl Lek ; 59(1): 7-10, 2002.
Artículo en Polaco | MEDLINE | ID: mdl-12108052

RESUMEN

The role of the lipid abnormalities as a risk factor for stroke remains controversial; very likely because only the standard lipid fractions are measured and because different causes of stroke are not considered. LDL phenotype B promotes atherogenesis and is recognized as a risk factor for ischemic heart disease, but its prevalence in ischemic stroke of distinct causes is unknown. Therefore we designed the study to investigate the prevalence of LDL phenotype B in stroke survivors with large vessel disease (LVD) or small vessel disease (SVD) and to establish the relationship between LDL phenotypes and basic lipid fractions in this group of patients. 59 patients (24 patients with LVD and 35 patients with SVD) being at least 3 months after ischemic stroke were included into the study. 30 sex- and age-matched subject served as controls. The concentrations of total cholesterol, HDL- and LDL-cholesterol, as well as triglycerides were measured and the LDL phenotype was determined using the potassium bromide density gradient ultracentrifugation. The LDL phenotype B was more frequent in patients with LVD (67%, p < 0.05) compared to patients with SVD and with controls. LDL phenotype B was also more frequent in patients with SVD (40%) and in all stroke survivors combined (51%), when compared with control group (17%, p < 0.05; chi-square test). Among stroke survivors, controls and studied subjects as a whole, those with LDL phenotype B revealed the lower concentration of HDL-cholesterol (1.19 +/- 0.33 vs. 1.46 +/- 0.42; 1.16 +/- 0.29 vs. 1.53 +/- 0.3 and 1.19 +/- 0.32 vs. 1.49 +/- 0.37, respectively; p < 0.05, Student t-test) when compared with carriers of LDL A phenotype. The LDL phenotype B is more frequent in ischemic stroke survivors compared to controls, and within the group of stroke survivors, LDL B is more prevalent in patients with LVD. The LDL phenotype B is associated with lower concentration of HDL-cholesterol.


Asunto(s)
Isquemia Encefálica/genética , Encéfalo/irrigación sanguínea , LDL-Colesterol/genética , Isquemia Encefálica/sangre , HDL-Colesterol/sangre , HDL-Colesterol/genética , LDL-Colesterol/sangre , Humanos , Fenotipo , Factores de Riesgo
14.
Acta Neuropathol Commun ; 2: 75, 2014 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-24972680

RESUMEN

Accumulation of ß-amyloid (Aß) in the brain is essential to Alzheimer's disease (AD) pathogenesis. Carriers of the apolipoprotein E (APOE) ε4 allele demonstrate greatly increased AD risk and enhanced brain Aß deposition. In contrast, APOE ε2 allele carries show reduced AD risk, later age of disease onset, and lesser Aß accumulation. However, it remains elusive whether the apoE2 isoform exerts truly protective effect against Aß pathology or apoE2 plays deleterious role albeit less pronounced than the apoE4 isoform. Here, we characterized APPSW/PS1dE9/APOE ε2-TR (APP/E2) and APPSW/PS1dE9/APOE ε4-TR (APP/E4) mice, with targeted replacement (TR) of the murine Apoe for human ε2 or ε4 alleles, and used these models to investigate effects of pharmacological inhibition of the apoE/Aß interaction on Aß deposition and neuritic degeneration. APP/E2 and APP/E4 mice replicate differential effect of human apoE isoforms on Aß pathology with APP/E4 mice showing a several-fold greater load of Aß plaques, insoluble brain Aß levels, Aß oligomers, and density of neuritic plaques than APP/E2 mice. Furthermore, APP/E4 mice, but not APP/E2 mice, exhibit memory impairment on object recognition and radial arm maze tests. Between the age of 6 and 10 months APP/E2 and APP/E4 mice received treatment with Aß12-28P, a non-toxic, synthetic peptide homologous to the apoE binding motif within the Aß sequence, which competitively blocks the apoE/Aß interaction. In both lines, the treatment significantly reduced brain Aß accumulation, co-accumulation of apoE within Aß plaques, and neuritic degeneration, and prevented memory deficit in APP/E4 mice. These results indicate that both apoE2 and apoE4 isoforms contribute to Aß deposition and future therapies targeting the apoE/Aß interaction could produce favorable outcome in APOE ε2 and ε4 allele carriers.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E2/metabolismo , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Memoria/fisiología , Ratones , Ratones Transgénicos , Placa Amiloide/etiología , Placa Amiloide/metabolismo
15.
Neurosci Lett ; 538: 26-31, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23370284

RESUMEN

Prionoses are a group of neurodegenerative diseases characterized by misfolding of cellular prion protein (PrP(C)) and accumulation of its diseases specific conformer PrP(Sc) in the brain and neuropathologically, they can be associated with presence or absence of PrP amyloid deposits. Functional molecular chaperones (MCs) that constitute the unfolded protein response include heat shock proteins and glucose-regulated protein families. They protect intracellular milieu against various stress conditions including accumulation of misfolded proteins and oxidative stress, typical of neurodegenerative diseases. Little is known about the role of MCs in pathogenesis of prionoses in mammalian prion model systems. In this study we characterized MCs response pattern in mice infected with various mouse adapted scrapie strains. Rather than uniform upregulation of MCs, we encountered two distinctly different patterns of MCs response distinguishing ME7 and 87V strains from 22L and 139A strains. ME7 and 87V strains are known for the induction of amyloid deposition in infected animals, while in mice infected with 22L and 139A strains amyloid deposits are absent. MCs response pattern similar to that associated with amyloidogenic ME7 and 87V strains was also observed in APPPS1-21 Alzheimer's transgenic mice, which represent an aggressive model of cerebral amyloidosis caused by ß-amyloid deposition. Our results highlight the probability that different mechanisms of MCs regulation exist driven by amyloidogenic and non-amyloidogenic nature of prion strains.


Asunto(s)
Encéfalo/metabolismo , Chaperonas Moleculares/metabolismo , Priones/fisiología , Animales , Encéfalo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Femenino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/patología , Priones/patogenicidad
16.
Oncotarget ; 8(25): 39941-39942, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28537920
18.
Proc Natl Acad Sci U S A ; 103(49): 18787-92, 2006 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17116874

RESUMEN

The amyloid-beta (Abeta) cascade hypothesis of Alzheimer's disease (AD) maintains that accumulation of Abeta peptide constitutes a critical event in the early disease pathogenesis. The direct binding between Abeta and apolipoprotein E (apoE) is an important factor implicated in both Abeta clearance and its deposition in the brain's parenchyma and the walls of meningoencephalic vessels as cerebral amyloid angiopathy. With the aim of testing the effect of blocking the apoE/Abeta interaction in vivo as a potential novel therapeutic target for AD pharmacotherapy, we have developed Abeta12-28P, which is a blood-brain-barrier-permeable nontoxic, and nonfibrillogenic synthetic peptide homologous to the apoE binding site on the full-length Abeta. Abeta12-28P binds with high affinity to apoE, preventing its binding to Abeta, but has no direct effect on Abeta aggregation. Abeta12-28P shows a strong pharmacological effect in vivo. Its systemic administration resulted in a significant reduction of Abeta plaques and cerebral amyloid angiopathy burden and a reduction of the total brain level of Abeta in two AD transgenic mice models. The treatment did not affect the levels of soluble Abeta fraction or Abeta oligomers, indicating that inhibition of the apoE/Abeta interaction in vivo has a net effect of increasing Abeta clearance over deposition and at the same time does not create conditions favoring formation of toxic oligomers. Furthermore, behavioral studies demonstrated that treatment with Abeta12-28P prevents a memory deficit in transgenic animals. These findings provide evidence of another therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Unión Competitiva/fisiología , Femenino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo
19.
Eur J Neurosci ; 23(10): 2635-47, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16817866

RESUMEN

Prion diseases are transmissible and invariably fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K (PK)-resistant conformer, scrapie PrP (PrP(Sc)). Humoral immunity may significantly prolong the incubation period and even prevent disease in murine models of prionoses. However, the mechanism(s) of action of anti-PrP monoclonal antibodies (Mabs) remain(s) obscure. The murine neuroblastoma N2a cell line, infected with the 22L mouse-adapted scrapie strain, was used to screen a large library of Mabs with similar binding affinities to PrP, to identify those antibodies which could clear established infection and/or prevent infection de novo. Three Mabs were found capable of complete and persistent clearing of already-infected N2a cells of PrP(Sc). These antibodies were 6D11 (generated to PK-resistant PrP(Sc) and detecting PrP residues 93-109), and 7H6 and 7A12, which were raised against recombinant PrP and react with neighbouring epitopes of PrP residues 130-140 and 143-155, respectively. Mabs were found to interact with PrP(Sc) formation both on the cell surface and after internalization in the cytosol. Treatment with Mabs was not associated with toxicity nor did it result in decreased expression of PrP(C). Both preincubation of N2a cells with Mabs prior to exposure to 22L inoculum and preincubation of the inoculum with Mabs prior to infecting N2a cells resulted in a significant reduction in PrP(Sc) levels. Information provided in these studies is important for the rational design of humoral immune therapy for prion infection in animals and eventually in humans.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteínas PrPC/efectos de los fármacos , Proteínas PrPSc/efectos de los fármacos , Enfermedades por Prión/inmunología , Enfermedades por Prión/prevención & control , Animales , Afinidad de Anticuerpos , Células Cultivadas , Mapeo Epitopo , Epítopos Inmunodominantes , Ratones , Proteínas PrPC/inmunología , Proteínas PrPSc/inmunología , Proteínas PrPSc/patogenicidad
20.
Dement Geriatr Cogn Disord ; 16(4): 283-6, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14512725

RESUMEN

Inflammatory mechanisms are involved in the pathogenesis of Alzheimer's disease (AD). It is postulated that cytokine synthesis is altered in AD patients compared with nondemented subjects. Glucocorticoids play an important role in cytokine synthesis. We assessed the release of tumor necrosis factor-alpha (TNF-alpha), interleukin-10 (IL-10) and interleukin-12 (IL-12) and its regulation by dexamethasone in AD patients in vitro. Cytokine levels were measured using the ELISA method in unstimulated, LPS-stimulated or whole-blood samples incubated with LPS and dexamethasone from 18 AD patients and 12 controls. The cytokine levels spontaneously produced by blood cells after incubation with LPS or LPS and dexamethasone did not differ significantly between groups. Dexamathasone inhibited TNF-alpha synthesis by LPS-stimulated blood cells more effectively in AD patients than in controls. These results suggest that cytokine synthesis in AD patients could be regulated by glucocorticoids in a different way than in nondemented subjects.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Dexametasona/análogos & derivados , Dexametasona/farmacología , Glucocorticoides/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas In Vitro , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA