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











Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36625712

RESUMEN

This work investigates linear and non-linear parametric reduced order models (ROM) capable of replacing computationally expensive high-fidelity simulations of human body models (HBM) through a non-intrusive approach. Conventional crash simulation methods pose a computational barrier that restricts profound analyses such as uncertainty quantification, sensitivity analysis, or optimization studies. The non-intrusive framework couples dimensionality reduction techniques with machine learning-based surrogate models that yield a fast responding data-driven black-box model. A comparative study is made between linear and non-linear dimensionality reduction techniques. Both techniques report speed-ups of a few orders of magnitude with an accurate generalization of the design space. These accelerations make ROMs a valuable tool for engineers.


Asunto(s)
Cuerpo Humano , Aprendizaje Automático , Humanos , Incertidumbre
2.
Acta Psychiatr Scand ; 136(5): 517-525, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28940401

RESUMEN

OBJECTIVE: The posterior superior temporal sulcus (pSTS) plays a critical role in the 'social brain'. Its neurodevelopment and relationship with the social impairment in autism spectrum disorders (ASD) are not well understood. We explored the relationship between social cognition and the neurodevelopment of the pSTS in ASD. METHOD: We included 44 adults with high-functioning ASD and 36 controls. We assessed their performances on the 'Reading the mind in the eyes' test (for 34 of 44 subjects with ASD and 30 of 36 controls), their fixation time on the eyes with eye tracking (for 35 of 44 subjects with ASD and 30 of 36 controls) and the morphology of the caudal branches of the pSTS (length and depth), markers of the neurodevelopment, with structural MRI. RESULTS: The right anterior caudal ramus of the pSTS was significantly longer in patients with ASD compared with controls (52.6 mm vs. 38.3 mm; P = 1.4 × 10-3 ; Cohen's d = 0.76). Its length negatively correlated with fixation time on the eyes (P = 0.03) in the ASD group and with the 'Reading the mind in the eyes' test scores in both groups (P = 0.03). CONCLUSION: Our findings suggest that the neurodevelopment of the pSTS is related to the ASD social impairments.


Asunto(s)
Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/fisiopatología , Percepción Social , Lóbulo Temporal/crecimiento & desarrollo , Adolescente , Adulto , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Lóbulo Temporal/diagnóstico por imagen , Adulto Joven
3.
Sci Rep ; 7: 46543, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28440309

RESUMEN

Adult hippocampal neurogenesis is a remarkable form of brain plasticity through which new neurons are generated throughout life. Despite its important roles in cognition and emotion and its modulation in various preclinical disease models, the functional importance of adult hippocampal neurogenesis in human health has not been revealed because of a lack of tools for monitoring adult neurogenesis in vivo. Therefore, we performed an unbiased proteomics screen to identify novel proteins expressed during neuronal differentiation using a human neural stem cell model, and we identified the proteoglycan Glypican-2 (Gpc2) as a putative secreted marker of immature neurons. Exogenous Gpc2 binds to FGF2 and inhibits FGF2-induced neural progenitor cell proliferation. Gpc2 is enriched in neurogenic regions of the adult brain. Its expression is increased by physiological stimuli that increase hippocampal neurogenesis and decreased in transgenic models in which neurogenesis is selectively ablated. Changes in neurogenesis also result in changes in Gpc2 protein level in cerebrospinal fluid (CSF). Gpc2 is detectable in adult human CSF, and first pilot experiments with a longitudinal cohort indicate a decrease over time. Thus, Gpc2 may serve as a potential marker to monitor adult neurogenesis in both animal and human physiology and disease, warranting future studies.


Asunto(s)
Células Madre Adultas/metabolismo , Glipicanos/líquido cefalorraquídeo , Hipocampo/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Adulto , Células Madre Adultas/citología , Animales , Biomarcadores/líquido cefalorraquídeo , Diferenciación Celular , Proliferación Celular , Hipocampo/citología , Humanos , Masculino , Ratones , Células-Madre Neurales/citología
4.
Schweiz Arch Tierheilkd ; 156(7): 345-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24973323

RESUMEN

A 22-year old mare from Switzerland was admitted to an equine clinic in May 2011. She presented with fever, lethargy, icteric mucous membranes, reduced alertness, an unsteady gait and ataxia. An Anaplasma phagocytophilum infection was confirmed by blood smear and PCR. The mare was treated with oxytetracylin and recovered rapidly, but she still suffered from a slight atactic gait disturbance at 3 weeks post infection.


Asunto(s)
Anaplasma phagocytophilum , Ehrlichiosis , Enfermedades de los Caballos , Animales , Antibacterianos/uso terapéutico , Ehrlichiosis/diagnóstico , Ehrlichiosis/tratamiento farmacológico , Ehrlichiosis/fisiopatología , Ehrlichiosis/veterinaria , Femenino , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/tratamiento farmacológico , Enfermedades de los Caballos/fisiopatología , Caballos , Datos de Secuencia Molecular , Oxitetraciclina/uso terapéutico , Suiza
5.
Neurosci Lett ; 316(3): 145-8, 2001 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-11744223

RESUMEN

There is circumstantial evidence that the reelin signaling pathway may contribute to neurodegeneration in the adult brain and could be linked to Alzheimer's disease (AD). In the present immunohistochemical report we studied the reelin expression profile in double-transgenic mice that express both human mutant beta-amyloid precursor protein (APP) and human mutant presenilin-1. We were able to demonstrate that reelin immunostaining was found together with human APP in the neuritic component of many AD-typical plaques in both hippocampus and neocortex. This observation gives the first evidence for the association of reelin with amyloid deposits.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Proteínas de la Matriz Extracelular/genética , Femenino , Hipocampo/patología , Hipocampo/fisiopatología , Inmunohistoquímica , Interneuronas/metabolismo , Interneuronas/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso , Placa Amiloide/genética , Placa Amiloide/patología , Presenilina-1 , Células Piramidales/metabolismo , Células Piramidales/patología , Proteína Reelina , Serina Endopeptidasas
6.
Neurosci Lett ; 306(1-2): 116-20, 2001 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-11403971

RESUMEN

beta-Amyloid peptides are key molecules that are involved in the pathology of Alzheimer's disease (AD). The source and place of the neurotoxic action of Abeta, however, is still a matter of controversial debates. In the present report, we studied the neuropathological events in a transgenic mouse model expressing human mutant beta-amyloid precursor protein and human mutant presenilin-1 in neurons. Western blot and immunohistochemical analysis revealed that intracellular Abeta staining preceded plaque deposition, which started in the hippocampal formation. At later stages, many neuritic Abeta positive plaques were found in all cortical, hippocampal and many other brain areas. Interestingly, intraneuronal Abeta staining was no longer detected in the brain of aged double-transgenic mice, which correlates with the typical neuropathology in the brain of chronic AD patients.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Gliosis/genética , Gliosis/patología , Gliosis/fisiopatología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos/genética , Ratones Transgénicos/metabolismo , Mutación/genética , Neuronas/patología , Placa Amiloide/genética , Placa Amiloide/patología , Presenilina-1
7.
Neurobiol Dis ; 8(2): 331-42, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11300728

RESUMEN

Many cases of early-onset inherited Alzheimer's disease (AD) are caused by mutations in the presenilin-1 (PS1) gene. Expression of PS1 mutations in cell culture systems and in primary neurons from transgenic mice increases their vulnerability to cell death. Interestingly, enhanced vulnerability to cell death has also been demonstrated for peripheral lymphocytes from AD patients. We now report that lymphocytes from PS1 mutant transgenic mice show a similar hypersensitivity to cell death as do peripheral cells from AD patients and several cell culture systems expressing PS1 mutations. The cell death-enhancing action of mutant PS1 was associated with increased production of reactive oxygen species and altered calcium regulation, but not with changes of mitochondrial cytochrome c. Our study further emphasizes the pathogenic role of mutant PS1 and may provide the fundamental basis for new efforts to close the gap between studies using neuronal cell lines transfected with mutant PS1, neurons from transgenic animals, and peripheral cells from AD patients.


Asunto(s)
Enfermedad de Alzheimer/genética , Apoptosis/genética , Química Encefálica/genética , Linfocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Transgénicos/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Calcio/metabolismo , Células Cultivadas/metabolismo , Células Cultivadas/patología , Grupo Citocromo c/genética , Grupo Citocromo c/metabolismo , Citometría de Flujo , Linfocitos/patología , Ratones , Ratones Transgénicos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación/fisiología , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/genética , Presenilina-1
8.
Brain Pathol ; 11(1): 1-11, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11145195

RESUMEN

During the last years it has become evident that the beta-amyloid (Abeta) component of senile plaques may be the key molecule in the pathology of Alzheimer's disease (AD). The source and place of the neurotoxic action of Abeta, however, is still a matter of controversy. The precursor of the beta-amyloid peptide is the predominantly neuronal beta-amyloid precursor protein. We, and others, hypothesize that intraneuronal misregulation of APP leads to an accumulation of Abeta peptides in intracellular compartments. This accumulation impairs APP trafficking, which starts a cascade of pathological changes and causes the pyramidal neurons to degenerate. Enhanced Abeta secretion as a function of stressed neurons and remnants of degenerated neurons provide seeds for extracellular Abeta aggregates, which induce secondary degenerative events involving neighboring cells such as neurons, astroglia and macrophages/microglia. Beta-amyloid precursor protein has a pivotal role in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Animales , Transporte Axonal , Cobre , Humanos , Plasticidad Neuronal , Transmisión Sináptica
9.
Biochem Biophys Res Commun ; 275(3): 910-5, 2000 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-10973821

RESUMEN

Presenilins 1 and 2 are two homologous proteins which, when mutated, appear responsible for most of the early-onset familial forms of Alzheimer's disease. Among various functional aspects, presenilins appear to behave as chaperoning partners of a series of proteins including the beta-amyloid precursor protein. Recently, presenilins were shown to interact with Rab11, a GTPase involved in intracellular transport. This suggested that Rab11-presenilin interaction could influence the routing of betaAPP and thereby modulate its maturation. In this context, we examined whether overexpression of Rab11 or its constitutively active mutant Rab11Q70L could affect betaAPP maturation in human HEK293 cells. We show here that the overexpression of both Rab11-related proteins does not modify the recovery of secreted sAPPalpha or Abeta in HEK293 cells expressing wild-type betaAPP or betaAPP harboring the Swedish double mutation. These data indicate that Rab11 does not influence betaAPP processing in HEK293 cells. However, it does not preclude the possibility for Rab11 to modulate other presenilin-mediated functions in human cells.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Mutación/genética , Proteínas de Unión al GTP rab/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Línea Celular , Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Presenilina-1 , Presenilina-2 , Unión Proteica , Procesamiento Proteico-Postraduccional , Transfección , Proteínas de Unión al GTP rab/genética
10.
Neurosci Lett ; 292(2): 87-90, 2000 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-10998555

RESUMEN

Alzheimer's disease-related mutations in the presenilin-1 gene (PS1) are leading to an elevated production of neurotoxic beta-amyloid 1-42 and may additionally enhance oxidative stress. Here, we provide in vivo evidence indicating that brains of transgenic mice expressing different human Alzheimer-linked PS1 mutations exhibit a reduced activity of two antioxidant enzymes. For this purpose, mice transgenic for human PS1 and for single and multiple PS1 mutations were generated. Mice with multiple PS1 mutations showed a significantly decreased activity of the antioxidant enzymes Cu/Zn superoxide dismutase and glutathione reductase already at an age of 3-4 months. As expected, this effect was less pronounced for the mice with a single PS1 mutation. By contrast, animals bearing normal human PS1 showed significantly elevated enzyme activities relative to non-transgenic littermate controls.


Asunto(s)
Antioxidantes/metabolismo , Encéfalo/enzimología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Glutatión Reductasa/metabolismo , Humanos , Peroxidación de Lípido/fisiología , Ratones , Ratones Transgénicos , Mutagénesis/fisiología , Degeneración Nerviosa/metabolismo , Presenilina-1 , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Transgenes/fisiología
11.
Neuropathol Appl Neurobiol ; 26(2): 117-23, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10840274

RESUMEN

Presenilin 1 has been shown to be mutated in a high proportion of cases of familial Alzheimer's disease. Immunoreactive epitopes of the protein have been found mainly in neurones devoid of neurofibrillary tangles - an observation that has led to the conclusion that presenilin 1 could have a protective role. In this study, the relationship between deposits of Abeta peptide (both the 40 and 42 isoforms), tau positive neurofibrillary tangles and presenilin 1-positive neuronal profiles were analysed in three cases of presenilin 1 mutation, four cases of sporadic Alzheimer's disease and five controls. Immunohistochemistry was performed in a sample from the supramarginal gyrus. The proportion of volume occupied by the Abeta1-40 and Abeta1-42 deposits (amyloid load) was evaluated by a point-counting technique. Tau-positive neurofibrillary tangles, and presenilin 1-positive neuronal profiles were directly counted. The location of the lesions in the thickness of the cortex was recorded. The density of PS1-positive neuronal profiles in Alzheimer's disease cases was lower than in the controls. The deficit was significant only in the upper layers of the cortex. The density of presenilin 1 neuronal profiles was negatively correlated with Abeta1-40 and Abeta1-42 loads, and with the density of tau-positive neurofibrillary tangles. Multivariate analysis showed that the Abeta1-42 load was the best determinant of the decrease in presenilin 1-positive neuronal profiles. Presenilin 1-positive neurones appear to be lost rather than protected in the course of Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Corteza Cerebral/patología , Proteínas de la Membrana/análisis , Ovillos Neurofibrilares/patología , Proteínas tau/análisis , Adulto , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/inmunología , Amiloidosis/patología , Anticuerpos , Corteza Cerebral/química , Femenino , Humanos , Masculino , Proteínas de la Membrana/inmunología , Persona de Mediana Edad , Ovillos Neurofibrilares/química , Neuronas/química , Neuronas/patología , Presenilina-1 , Proteínas tau/inmunología
12.
Prog Neurobiol ; 60(4): 363-84, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10670705

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia in the elderly population. Dementia is associated with massive accumulation of fibrillary aggregates in various cortical and subcortical regions of the brain. These aggregates appear intracellularly as neurofibrillary tangles, extracellularly as amyloid plaques and perivascular amyloid in cerebral blood vessels. The causative factors in AD etiology implicate both, genetic and environmental factors. The large majority of early-onset familial Alzheimer's disease (FAD) cases are linked to mutations in the genes coding for presenilin 1 (PS1) and presenilin 2 (PS2). The corresponding proteins are 467 (PS1) and 448 (PS2) amino-acids long, respectively. Both are membrane proteins with multiple transmembrane regions. Presenilins show a high degree of conservation between species and a presenilin homologue with definite conservation of the hydrophobic structure has been identified even in the plant Arabidopsis thaliana. More than 50 missense mutations in PS1 and two missense mutations in PS2 were identified which are causative for FAD. PS mutations lead to the same functional consequence as mutations on amyloid precursor protein (APP), altering the processing of APP towards the release of the more amyloidogenic form 1-42 of Abeta (Abeta42). In this regard, the physical interaction between APP and presenilins in the endoplasmic reticulum has been demonstrated and might play a key role in Abeta42 production. It was hypothesized that PS1 might directly cleave APP. However, extracellular amyloidogenesis and Abeta production might not be the sole factor involved in AD pathology and several lines of evidence support a role of apoptosis in the massive neuronal loss observed. Presenilins were shown to modify the apoptotic response in several cellular systems including primary neuronal cultures. Some evidence is accumulating which points towards the beta-catenin signaling pathways to be causally involved in presenilin mediated cell death. Increased degradation of beta-catenin has been shown in brain of AD patients with PS1 mutations and reduced beta-catenin signaling increased neuronal vulnerability to apoptosis in cell culture models. The study of presenilin physiological functions and the pathological mechanisms underlying their role in pathogenesis clearly advanced our understanding of cellular mechanisms underlying the neuronal cell death and will contribute to the identification of novel drug targets for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/genética , Animales , Femenino , Humanos , Embarazo , Presenilina-1 , Presenilina-2
13.
J Biol Chem ; 275(3): 1525-8, 2000 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-10636839

RESUMEN

Presenilin-1 (PS1) is required for the release of the intracellular domain of Notch from the plasma membrane as well as for the cleavage of the amyloid precursor protein (APP) at the gamma-secretase cleavage site. It remains to be demonstrated whether PS1 acts by facilitating the activity of the protease concerned or is the protease itself. PS1 could have a gamma-secretase activity by itself or could traffic APP and Notch to the appropriate cellular compartment for processing. Human APP 695 and PS1 were coexpressed in Sf9 insect cells, in which endogenous gamma-secretase activity is not detected. In baculovirus-infected Sf9 cells, PS1 undergoes endoproteolysis and interacts with APP. However, PS1 does not cleave APP in Sf9 cells. In CHO cells, endocytosis of APP is required for Abeta secretion. Deletion of the cytoplasmic sequence of APP (APPDeltaC) inhibits both APP endocytosis and Abeta production. When APPDeltaC and PS1 are coexpressed in CHO cells, Abeta is secreted without endocytosis of APP. Taken together, these results conclusively show that, although PS1 does not cleave APP in Sf9 cells, PS1 allows the secretion of Abeta without endocytosis of APP by CHO cells.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/fisiología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas , Baculoviridae/metabolismo , Western Blotting , Células CHO , Línea Celular , Cricetinae , Endocitosis , Humanos , Presenilina-1 , Proteínas Recombinantes/metabolismo , Transfección
14.
J Neurochem ; 72(6): 2479-87, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10349858

RESUMEN

To study amyloid beta-protein (A beta) production and aggregation in vivo, we created two transgenic (Tg) mouse lines expressing the C-terminal 100 amino acids of human amyloid precursor protein (APP): Tg C100.V717F and Tg C100.WT. Western blot analysis showed that human APP-C100 and A beta were produced in brain and some peripheral tissues and A beta was produced in serum. Using antibodies specific for the A beta C terminus we found that Tg C100.V717F produced a 1.6-fold increase in A beta42/A beta40 compared with Tg C100.WT. Approximately 30% of total brain A beta (approximately 122 ng/g of wet tissue) was water-soluble. The remaining 70% of A beta partitioned into the particulate fraction and was completely sodium dodecyl sulfate-soluble. In contrast, human Alzheimer's disease brain has predominantly sodium dodecyl sulfate-insoluble A beta. Immunohistochemistry with an A beta(5-8) antibody showed that A beta or A beta-containing fragments accumulated intracellularly in the hippocampus of aged Tg C100.V717F mice. The soluble A beta levels in Tg brain are similar to those in normal human brain, and this may explain the lack of microscopic amyloid deposits in the Tg mice. However, this mouse model provides a system to study the intracellular processing and accumulation of A beta or A beta-containing fragments and to screen for compounds directed at the gamma-secretase activity.


Asunto(s)
Envejecimiento/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo , Precursor de Proteína beta-Amiloide/aislamiento & purificación , Animales , Detergentes , Hipocampo/crecimiento & desarrollo , Humanos , Cinética , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/aislamiento & purificación , Solubilidad
15.
Hum Mol Genet ; 8(7): 1263-9, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10369872

RESUMEN

Presenilin 1 (PS1) mutations account for the majority of early-onset dominant cases of familial Alzheimer's disease. Presenilins (PSs) are located in many intra-cellular compartments such as the endoplasmic reticulum, Golgi apparatus, nuclear region and vesicular structures. These proteins include from seven to nine putative transmembrane domains, with the N- and C-terminal ends and a large hydrophilic loop orientated towards the cytoplasm. We report an interaction between the human PS1 or PS2 hydrophilic loop and Rab11, a small GTPase belonging to the Ras-related superfamily. Interaction domains were mapped to codons 374-400 for PS1 and to codons 106-179 for Rab11, a region including the fourth GTP-binding domain. Considering the implication of Rab proteins in vesicular transport pathways, the PS-Rab11 inter-action suggests that PSs might be involved in amyloid precursor protein vesicular routing.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP rab , Secuencia de Aminoácidos , Animales , Sitios de Unión , Transporte Biológico/fisiología , Células COS , Humanos , Datos de Secuencia Molecular , Presenilina-1 , Conformación Proteica , Saccharomyces cerevisiae/metabolismo
16.
Neurobiol Dis ; 6(1): 43-55, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10078972

RESUMEN

Mutations in presenilin 1 and presenilin 2 (PS1 and PS2, respectively) genes cause the large majority of familial forms of early-onset Alzheimer's disease. The physical interaction between presenilins and APP has been recently described using coimmunoprecipitation. With a similar technique, we confirmed this interaction and have mapped the interaction domains on both PS2 and APP. Using several carboxy-terminal truncated forms of PS2, we demonstrated that the hydrophilic amino terminus of PS2 (residues 1 to 87, PS2NT) was sufficient for interaction with APP. Interestingly, only a construct with a leader peptide for secretion (SecPS2NT) and not its cytosolic counterpart was shown to interact with APP. For APP, we could demonstrate interaction of PS2 with the last 100 but not the last 45 amino acids of APP, including therefore the A beta region. Accordingly, SecPS2NT is capable of binding to A beta-immunoreactive species in conditioned medium. In addition, a second region in the extracellular domain of APP also interacted with PS2. Comparable results with PS1 indicate that the two presenilins share similar determinants of binding to APP. Confirming these results, SecPS2NT is able to inhibit PS1/APP interaction. Such a competition makes it unlikely that the PS/APP interaction results from nonspecific aggregation of PS in transfected cells. The physical interaction of presenilins with a region encompassing the A beta sequence of APP could be causally related to the misprocessing of APP and the production of A beta1-42.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/química , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Células COS , Mapeo Cromosómico , ADN Complementario , Expresión Génica/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutagénesis/fisiología , Presenilina-1 , Presenilina-2 , Estructura Terciaria de Proteína , Transfección
17.
Proc Natl Acad Sci U S A ; 95(23): 13703-8, 1998 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-9811864

RESUMEN

The importance of the amyloid precursor protein (APP) in the pathogenesis of Alzheimer's disease (AD) became apparent through the identification of distinct mutations in the APP gene, causing early onset familial AD with the accumulation of a 4-kDa peptide fragment (betaA4) in amyloid plaques and vascular deposits. However, the physiological role of APP is still unclear. In this work, Drosophila melanogaster is used as a model system to analyze the function of APP by expressing wild-type and various mutant forms of human APP in fly tissue culture cells as well as in transgenic fly lines. After expression of full-length APP forms, secretion of APP but not of betaA4 was observed in both systems. By using SPA4CT, a short APP form in which the signal peptide was fused directly to the betaA4 region, transmembrane domain, and cytoplasmic tail, we observed betaA4 release in flies and fly-tissue culture cells. Consequently, we showed a gamma-secretase activity in flies. Interestingly, transgenic flies expressing full-length forms of APP have a blistered-wing phenotype. As the wing is composed of interacting dorsal and ventral epithelial cell layers, this phenotype suggests that human APP expression interferes with cell adhesion/signaling pathways in Drosophila, independently of betaA4 generation.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Drosophila melanogaster/genética , Endopeptidasas/genética , Regulación de la Expresión Génica , Genes de Insecto , Secretasas de la Proteína Precursora del Amiloide , Animales , Animales Modificados Genéticamente , Ácido Aspártico Endopeptidasas , Adhesión Celular/genética , Humanos , Fenotipo
18.
J Biol Chem ; 273(44): 28931-6, 1998 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-9786896

RESUMEN

Recombinant adenoviruses were used for the expression of human amyloid precursor protein (APP) of Alzheimer's disease in primary cultures of rat cortical neurons and astrocytes. The catabolic pathways of human APP were studied 3 to 4 days after infection, when the equilibrium of APP production was reached. Although the expression of human wild type APP (WtAPP) by rat neurons induced the production of both extracellular and intraneuronal amyloid peptide (Abeta), Abeta was not detected in the culture medium of rat astrocytes producing human WtAPP. Because a low beta-secretase activity was previously reported in rodent astrocytes, we wondered whether modifications of the APP amino acid sequence at the beta-secretase clipping site would modify the astrocytic production of Abeta. Interestingly, rat astrocytes produced high amounts of Abeta after expression of human APP carrying a double amino acid substitution responsible for Alzheimer's disease in a large Swedish family (SwAPP). In both rat cortical neurons and astrocytes, the beta-secretase cleavage of the human SwAPP occurred very early in the secretion process in a cellular compartment in which a different sorting of SwAPP and WtAPP seems unlikely. These results suggest that human WtAPP and SwAPP could be processed by different beta-secretase activities.


Asunto(s)
Adenoviridae/genética , Precursor de Proteína beta-Amiloide/metabolismo , Astrocitos/enzimología , Corteza Cerebral/enzimología , Endopeptidasas/metabolismo , Neuronas/enzimología , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas , Corteza Cerebral/citología , Humanos , Ratas , Recombinación Genética , beta-Galactosidasa/genética
19.
Neuroscience ; 87(2): 325-36, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9740395

RESUMEN

Mutations in the gene for presenilin 1 are causative for the majority of cases of early onset familial Alzheimer's disease. Yet, the physiological function of presenilin 1 and the pathological mechanisms of the mutations leading to Alzheimer's disease are still unknown. To analyse potential pathological effects of presenilin 1 over-expression, we have generated transgenic rats which express high levels of human presenilin 1 protein in the brain. The over-expression of presenilin 1 leads to saturation of its normal processing and to the appearance of full-length protein in the transgenic rat brain. The transgenic protein is expressed throughout the brain and is predominantly found in neuronal cells. Cultured primary cortical neurons derived from these transgenic rats are significantly more sensitive than non-transgenic controls to apoptosis induced by standard culture conditions and to apoptosis induced by trophic factor withdrawal. Furthermore, the observed apoptosis is directly correlated with the expression of the transgenic protein. The results further emphasize the role of presenilin 1 in apoptotic cell death in native neuronal cultures.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apoptosis/fisiología , Proteínas de la Membrana/análisis , Neuronas/fisiología , Animales , Animales Modificados Genéticamente , Northern Blotting , Western Blotting , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Presenilina-1 , Ratas , Ratas Endogámicas F344
20.
J Neurochem ; 71(2): 723-31, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9681463

RESUMEN

Acetylcholinesterase (AChE) expression is markedly affected in Alzheimer's disease (AD). AChE activity is lower in most regions of the AD brain, but it is increased within and around amyloid plaques. We have previously shown that AChE expression in P19 cells is increased by the amyloid beta protein (A beta). The aim of this study was to investigate AChE expression using a transgenic mouse model of A beta overproduction. The beta-actin promoter was used to drive expression of a transgene encoding the 100-amino acid C-terminal fragment of the human amyloid precursor protein (APP CT100). Analysis of extracts from transgenic mice revealed that the human sequences of full-length human APP CT100 and A beta were overexpressed in the brain. Levels of salt-extractable AChE isoforms were increased in the brains of APP CT100 mice. There was also an increase in amphiphilic monomeric form (G1A) of AChE in the APP CT100 mice, whereas other isoforms were not changed. An increase in the proportion of G1A AChE was also detected in samples of frontal cortex from AD patients. Analysis of AChE by lectin binding revealed differences in the glycosylation pattern in APP CT100 mice similar to those observed in frontal cortex samples from AD. The results are consistent with the possibility that changes in AChE isoform levels and glycosylation patterns in the AD brain may be a direct consequence of altered APP metabolism.


Asunto(s)
Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Ratones Transgénicos/metabolismo , Fragmentos de Péptidos/genética , Anciano , Anciano de 80 o más Años , Animales , Butirilcolinesterasa/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Colina O-Acetiltransferasa/metabolismo , Síndrome de Down/metabolismo , Expresión Génica/fisiología , Glicosilación , Humanos , Isoenzimas/metabolismo , Ratones , Persona de Mediana Edad , Neuronas/enzimología , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA