Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microbiol Spectr ; 10(6): e0312522, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36314963

RESUMEN

The cobas 5800 System ("cobas 5800") is a new low- to mid-throughput PCR-based nucleic acid testing system which performs both qualitative and quantitative testing, including viral load (VL) determination. cobas 5800 shares numerous design elements and technical characteristics with the existing cobas 6800/8800 Systems. We compared HBV, HCV, and HIV-1 VL results from cobas 5800 in three different laboratories to those from the same specimens tested on a cobas 6800 system. We also assessed cobas 5800 assay reproducibility by repetitive testing of specimens with VL close to values used as thresholds for patient management or classification. The correlation between VL measurements generated using cobas 5800 versus 6800 was extremely high, with r2 correlation coefficients between 0.990 and 0.999 for the three targets at the different sites. The slope of the Deming regression line ranged from 0.994 (HBV, site 3) to 1.025 (HIV-1, site 1). The standard deviation values ranged from 0.04 to 0.19 log10 IU/mL for HBV, 0.06 to 0.33 log10 IU/mL for HCV, and 0.05 to 0.34 log10 copies/mL for HIV-1. In general, variability was higher at lower VL. Between 98.6% and 100% of results fell within the allowable total difference zone that defines expected variability on the existing 6800/8800 system. This multisite comparison study demonstrates equivalent performance of the new cobas 5800 system compared with cobas 6800. This establishes cobas 5800 as a new option for low- to mid-throughout laboratories seeking to optimize efficiency of their viral molecular testing. IMPORTANCE These are the first published data that demonstrate equivalent performance of the new cobas 5800 system compared with cobas 6800. This fulfills an unmet need for low- to mid-throughout laboratories seeking to optimize efficiency of their viral molecular testing.


Asunto(s)
VIH-1 , Hepatitis C , Humanos , VIH-1/genética , Virus de la Hepatitis B/genética , Carga Viral/métodos , Reproducibilidad de los Resultados , Hepacivirus/genética , Hepatitis C/diagnóstico
2.
Acta Neuropathol ; 129(1): 21-37, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432317

RESUMEN

The key role of APP in the pathogenesis of Alzheimer disease is well established. However, postnatal lethality of double knockout mice has so far precluded the analysis of the physiological functions of APP and the APLPs in the brain. Previously, APP family proteins have been implicated in synaptic adhesion, and analysis of the neuromuscular junction of constitutive APP/APLP2 mutant mice showed deficits in synaptic morphology and neuromuscular transmission. Here, we generated animals with a conditional APP/APLP2 double knockout (cDKO) in excitatory forebrain neurons using NexCre mice. Electrophysiological recordings of adult NexCre cDKOs indicated a strong synaptic phenotype with pronounced deficits in the induction and maintenance of hippocampal LTP and impairments in paired pulse facilitation, indicating a possible presynaptic deficit. These deficits were also reflected in impairments in nesting behavior and hippocampus-dependent learning and memory tasks, including deficits in Morris water maze and radial maze performance. Moreover, while no gross alterations of brain morphology were detectable in NexCre cDKO mice, quantitative analysis of adult hippocampal CA1 neurons revealed prominent reductions in total neurite length, dendritic branching, reduced spine density and reduced spine head volume. Strikingly, the impairment of LTP could be selectively rescued by acute application of exogenous recombinant APPsα, but not APPsß, indicating a crucial role for APPsα to support synaptic plasticity of mature hippocampal synapses on a rapid time scale. Collectively, our analysis reveals an essential role of APP family proteins in excitatory principal neurons for mediating normal dendritic architecture, spine density and morphology, synaptic plasticity and cognition.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/fisiopatología , Plasticidad Neuronal/fisiología , Fragmentos de Péptidos/metabolismo , Sinapsis/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Dendritas/patología , Dendritas/fisiología , Femenino , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Noqueados , Actividad Motora/fisiología , Neuritas/patología , Neuritas/fisiología , Fragmentos de Péptidos/genética , Proteínas Recombinantes/metabolismo , Memoria Espacial/fisiología , Sinapsis/patología
4.
Alzheimers Dement ; 10(2): e27-39, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23850332

RESUMEN

BACKGROUND: Early-onset familial Alzheimer disease (AD) is an autosomal dominant disorder caused by mutations in the amyloid precursor protein, presenilin 1 (PSEN1), or presenilin 2 gene. The objective of this study was to characterize the phenotype in a large family with a PSEN1 F177S mutation by performing detailed clinical assessments, neuroimaging, and neuropathological analysis. METHODS: In two subjects, clinical and neuropsychological assessments, structural magnetic resonance imaging, F-18-2-fluoro-2-deoxy-D-glucose positron emission tomographic imaging, AD biomarkers in cerebrospinal fluid and genetic analysis were available. In three deceased affected subjects, medical records were reviewed. In one subject, a complete neuropathological examination was available. RESULTS: Cognitive impairment and neurological symptoms developed homogeneously around 30 years of age and worsened rapidly. All subjects died about 7 years (range, 6-8 years) after disease onset before 40 years of age. All technical diagnostic information (neuroimaging, cerebrospinal fluid) were typically for AD. Neuropathology showed abundant neuritic plaques and neurofibrillary tangles, typical of severe AD. Antidementia treatment in one subject did not alter the length of survival. CONCLUSIONS: The PSEN1 F177S mutation leads to typical AD starting at age 30 and a homogeneous phenotype with rapid cognitive decline and prominent neurological symptoms. Excessive amyloid beta 42 production in the brain cortex corresponds well with other PSEN1 mutations.


Asunto(s)
Enfermedad de Alzheimer/genética , Salud de la Familia , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Presenilina-1/genética , Adulto , Edad de Inicio , Anciano , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/diagnóstico , Péptidos beta-Amiloides/líquido cefalorraquídeo , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Ondas Encefálicas/genética , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Electroencefalografía , Femenino , Humanos , Masculino , Escala del Estado Mental , Persona de Mediana Edad , Fragmentos de Péptidos/líquido cefalorraquídeo , Fenilalanina/genética , Tomografía de Emisión de Positrones , Radiografía , Serina/genética
5.
Exp Brain Res ; 217(3-4): 441-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22076403

RESUMEN

The amyloid precursor protein (APP) plays a crucial role in the pathogenesis of Alzheimer's disease. Here, we studied whether the lack of APP affects the synaptic properties in the dentate gyrus by measuring granule cell field potentials evoked by perforant path stimulation in anesthetized 9-11-month-old APP-deficient mice in vivo. We found decreased paired-pulse facilitation, indicating altered presynaptic short-term plasticity in the APP-deficient dentate gyrus. In contrast, excitatory synaptic strength and granule cell firing were unchanged in APP knockout mice. Likewise, long-term potentiation (LTP) induced by a theta-burst stimulation protocol was not impaired in the absence of APP. These findings suggest that the deletion of APP may affect presynaptic plasticity of synaptic transmission at the perforant path-granule cell synapse but leaves synaptic efficacy intact and LTP preserved, possibly due to functional redundancy within the APP gene family.


Asunto(s)
Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/fisiología , Giro Dentado/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
BMC Genomics ; 12: 160, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21435241

RESUMEN

BACKGROUND: The ß-amyloid precursor protein (APP) and the related ß-amyloid precursor-like proteins (APLPs) undergo complex proteolytic processing giving rise to several fragments. Whereas it is well established that Aß accumulation is a central trigger for Alzheimer's disease, the physiological role of APP family members and their diverse proteolytic products is still largely unknown. The secreted APPsα ectodomain has been shown to be involved in neuroprotection and synaptic plasticity. The γ-secretase-generated APP intracellular domain (AICD) functions as a transcriptional regulator in heterologous reporter assays although its role for endogenous gene regulation has remained controversial. RESULTS: To gain further insight into the molecular changes associated with knockout phenotypes and to elucidate the physiological functions of APP family members including their proposed role as transcriptional regulators, we performed DNA microarray transcriptome profiling of prefrontal cortex of adult wild-type (WT), APP knockout (APP-/-), APLP2 knockout (APLP2-/-) and APPsα knockin mice (APPα/α) expressing solely the secreted APPsα ectodomain. Biological pathways affected by the lack of APP family members included neurogenesis, transcription, and kinase activity. Comparative analysis of transcriptome changes between mutant and wild-type mice, followed by qPCR validation, identified co-regulated gene sets. Interestingly, these included heat shock proteins and plasticity-related genes that were both down-regulated in knockout cortices. In contrast, we failed to detect significant differences in expression of previously proposed AICD target genes including Bace1, Kai1, Gsk3b, p53, Tip60, and Vglut2. Only Egfr was slightly up-regulated in APLP2-/- mice. Comparison of APP-/- and APPα/α with wild-type mice revealed a high proportion of co-regulated genes indicating an important role of the C-terminus for cellular signaling. Finally, comparison of APLP2-/- on different genetic backgrounds revealed that background-related transcriptome changes may dominate over changes due to the knockout of a single gene. CONCLUSION: Shared transcriptome profiles corroborated closely related physiological functions of APP family members in the adult central nervous system. As expression of proposed AICD target genes was not altered in adult cortex, this may indicate that these genes are not affected by lack of APP under resting conditions or only in a small subset of cells.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Perfilación de la Expresión Génica , Corteza Prefrontal/metabolismo , Animales , Análisis por Conglomerados , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos
7.
Mol Cell Neurosci ; 44(4): 386-93, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20472066

RESUMEN

Impaired proteasomal function is a major hallmark in the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD). Here we investigated the biological properties of the secreted cleavage product of APP (sAPPalpha) in antagonizing stress signalling, dendritic degeneration and neuronal cell death induced by the proteasome inhibitor epoxomicin. Analysis of executioner caspase activation demonstrated that sAPPalpha was able to protect PC12 cells from apoptosis triggered by epoxomicin, as well as by genotoxic stress (UV irradiation). This anti-apoptotic effect of sAPPalpha was associated with inhibition of the stress-triggered c-Jun N-terminal kinase (JNK)-signalling pathway. The anti-apoptotic effect of sAPPalpha could also be confirmed in organotypic slice cultures of Thy1-GFP mouse hippocampi. Confocal time-lapse imaging of CA1 pyramidal neurons revealed that preincubation with sAPPalpha preserves the structural integrity of neurons after epoxomicin treatment. Taken together, our data demonstrate that sAPPalpha is neuroprotective under conditions of proteasomal stress.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Apoptosis , Región CA1 Hipocampal/citología , Espinas Dendríticas/patología , Degeneración Nerviosa , Complejo de la Endopetidasa Proteasomal/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Células Cultivadas , Cromonas/farmacología , Medios de Cultivo Condicionados/química , Técnicas de Cultivo , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Microscopía Confocal , Morfolinas/farmacología , Oligopéptidos/farmacología , Células PC12 , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Proteínas Recombinantes de Fusión , Antígenos Thy-1 , Rayos Ultravioleta
8.
Genesis ; 48(3): 200-6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20140888

RESUMEN

Proteolytical cleavage of the beta-amyloid precursor protein (APP) generates beta-amyloid, which is deposited in the brains of patients suffering from Alzheimer's disease (AD). Despite the well-established key role of APP for AD pathogenesis, the physiological function of APP and its close homologues APLP1 and APLP2 remains poorly understood. Previously, we generated APP(-/-) mice that proved viable, whereas APP(-/-)APLP2(-/-) mice and triple knockouts died shortly after birth, likely due to deficits of neuromuscular synaptic transmission. Here, we generated conditional knockout alleles for both APP and APLP2 in which the promoter and exon1 were flanked by loxP sites. No differences in expression were detectable between wt and floxed alleles, whereas null alleles were obtained upon crossing with Cre-transgenic deleter mice. These mice will now allow for tissue and time-point controlled knockout of both genes.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Marcación de Gen/métodos , Modelos Genéticos , Alelos , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Recién Nacidos , Southern Blotting , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Miembro Anterior/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Fuerza de la Mano , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Tamaño de los Órganos
9.
J Immunol ; 182(12): 7613-24, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494285

RESUMEN

In transgenic animal models, humoral immunity directed against the beta-amyloid peptide (Abeta), which is deposited in the brains of AD patients, can reduce Abeta plaques and restore memory. However, initial clinical trials using active immunization with Abeta1-42 (plus adjuvant) had to be stopped as a subset of patients developed meningoencephalitis, likely due to cytotoxic T cell reactions against Abeta. Previously, we demonstrated that retrovirus-like particles displaying on their surface repetitive arrays of self and foreign Ags can serve as potent immunogens. In this study, we generated retrovirus-like particles that display the 15 N-terminal residues of human Abeta (lacking known T cell epitopes) fused to the transmembrane domain of platelet-derived growth factor receptor (Abeta retroparticles). Western blot analysis, ELISA, and immunogold electron microscopy revealed efficient incorporation of the fusion proteins into the particle membrane. Without the use of adjuvants, single immunization of WT mice with Abeta retroparticles evoked high and long-lived Abeta-specific IgG titers of noninflammatory Th2 isotypes (IgG1 and IgG2b) and led to restimulatable B cell memory. Likewise, immunization of transgenic APP23 model mice induced comparable Ab levels. The CNS of immunized wild-type mice revealed neither infiltrating lymphocytes nor activated microglia, and no peripheral autoreactive T cells were detectable. Importantly, vaccination not only reduced Abeta plaque load to approximately 60% of controls and lowered both insoluble Abeta40 as well as Abeta42 in APP23 brain, but also significantly reduced cerebral soluble Abeta species. In summary, Abeta retroparticle vaccination may thus hold promise as a novel efficient future candidate vaccine for active immunotherapy of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Péptidos beta-Amiloides/ultraestructura , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Encéfalo/ultraestructura , Línea Celular , Sistema Nervioso Central/inmunología , Femenino , Humanos , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Inmunoelectrónica , Solubilidad , Linfocitos T/inmunología , Vacunación , Virión/ultraestructura
10.
Neurobiol Dis ; 33(2): 274-81, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19049874

RESUMEN

The accumulation of amyloid-beta (Abeta) into plaques is a hallmark feature of Alzheimer's disease (AD). While amyloid precursor protein (APP)-related proteins are found in most organisms, only Abeta fragments from human APP have been shown to induce amyloid deposits and progressive neurodegeneration. Therefore, it was suggested that neurotoxic effects are a specific property of human Abeta. Here we show that Abeta fragments derived from the Drosophila orthologue APPL aggregate into intracellular fibrils, amyloid deposits, and cause age-dependent behavioral deficits and neurodegeneration. We also show that APPL can be cleaved by a novel fly beta-secretase-like enzyme. This suggests that Abeta-induced neurotoxicity is a conserved function of APP proteins whereby the lack of conservation in the primary sequence indicates that secondary structural aspects determine their pathogenesis. In addition, we found that the behavioral phenotypes precede extracellular amyloid deposit formation, supporting results that intracellular Abeta plays a key role in AD.


Asunto(s)
Amiloide/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Envejecimiento , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/fisiología , Conducta Animal , Western Blotting , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Expresión Génica , Inmunohistoquímica , Luz , Proteínas de la Membrana/genética , Microscopía Electrónica , Datos de Secuencia Molecular , Degeneración Nerviosa , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/metabolismo , Nexinas de Proteasas , Receptores de Superficie Celular/genética
11.
J Neurosci Res ; 85(12): 2580-90, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17335075

RESUMEN

In neurons, amyloid precursor protein (APP) is localized to the dendritic and axonal compartment. Changes in subcellular localization affect secretase cleavage of APP, altering the generation of Abeta, and presumably also its pathogenic features. It was reported that APP is sorted initially to the axon and transcytosed subsequently to the somatodendritic compartment. This may be carried out by a recessive dendritic sorting signal in the cytoplasmic C-terminus, possibly the tyrosine based basolateral sorting signal (BaSS), and an axonal sorting motif within the extracellular juxtamembraneous domain. We investigated whether the C- or N-terminal domain of APP contains an independent dendritic or axonal sorting signal. We generated different APP deletion mutants, and produced chimeric proteins of APP and a non-related Type I transmembrane protein. Quantitative immunocytochemical analyses of transfected primary neurons showed that similar amounts of all APP mutants, lacking either the N- or C-terminus, were transported to the axonal and dendritic compartment. Investigations of the chimeric proteins showed that neither the N- nor the C-terminus of APP functions as independent sorting signal, whereas another tyrosine based dendritic sorting signal was sufficient to prevent axonal entry of APP. This data shows that, under steady state conditions, Heterologously expressed APP is transported equally to axons and dendrites irrespective of any putative sorting signal in its N- or C-terminus. This shows that APP can enter the axon in absence of the initial axonal sorting motif, indicating the existence of an alternative pathway allowing axonal entry of APP.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Axones/metabolismo , Dendritas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Transporte Axonal/fisiología , Células Cultivadas , Chlorocebus aethiops , Ratones , Proteínas Asociadas a Microtúbulos , Neuronas/citología , Transporte de Proteínas/fisiología , Eliminación de Secuencia/fisiología , Transfección/métodos
12.
Recent Pat CNS Drug Discov ; 1(1): 119-27, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18221198

RESUMEN

It is now almost a century ago that Alois Alzheimer first presented his results in public. Main characteristics of Alzheimer's disease (AD) are massive cerebral accumulation of amyloid, composed of fibrillary aggregates of the Amyloid beta peptide (Abeta) and intracellular accumulation of abnormally phosphorylated tau protein associated with widespread neurodegeneration. The clinical picture is characterized by progressive and irreversible dementia, which is eventually fatal. To date, there is no cure for this severe disease affecting more than of 30 million individuals worldwide. In the last decades, the treatment of Alzheimer patients was mainly focusing on symptomatical strategies. Based on the augmented knowledge about the mechanisms underlying the pathology of AD, particularly the molecular causes and consequences of AD, different therapeutic approaches arose and recently, treatment with Statins, NSAIDs and Abeta vaccines reached the level of clinical trials, showing some indication of efficacy already. According to actual evaluations, these approaches have realistic chances to become established as therapeutic routine in AD within the next 10 years. We will review here some of the most promising novel approaches to cure and prevent rather than to treat the symptoms of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inmunización
13.
Nat Cell Biol ; 7(11): 1118-23, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16227967

RESUMEN

Amyloid beta peptide (Abeta) has a key role in the pathological process of Alzheimer's disease (AD), but the physiological function of Abeta and of the amyloid precursor protein (APP) is unknown. Recently, it was shown that APP processing is sensitive to cholesterol and other lipids. Hydroxymethylglutaryl-CoA reductase (HMGR) and sphingomyelinases (SMases) are the main enzymes that regulate cholesterol biosynthesis and sphingomyelin (SM) levels, respectively. We show that control of cholesterol and SM metabolism involves APP processing. Abeta42 directly activates neutral SMase and downregulates SM levels, whereas Abeta40 reduces cholesterol de novo synthesis by inhibition of HMGR activity. This process strictly depends on gamma-secretase activity. In line with altered Abeta40/42 generation, pathological presenilin mutations result in increased cholesterol and decreased SM levels. Our results demonstrate a biological function for APP processing and also a functional basis for the link that has been observed between lipids and Alzheimer's disease (AD).


Asunto(s)
Péptidos beta-Amiloides/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Colesterol/metabolismo , Metabolismo de los Lípidos , Fragmentos de Péptidos/análisis , Esfingomielinas/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas , Células COS , Células Cultivadas , Chlorocebus aethiops , Endopeptidasas/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Fragmentos de Péptidos/metabolismo , Presenilina-1 , Presenilina-2 , Esfingomielina Fosfodiesterasa/metabolismo
14.
J Neurosci ; 25(11): 2865-73, 2005 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-15772346

RESUMEN

The Drosophila Swiss cheese (sws) mutant is characterized by progressive degeneration of the adult nervous system, glial hyperwrapping, and neuronal apoptosis. The Swiss cheese protein (SWS) shares 39% sequence identity with human neuropathy target esterase (NTE), and a brain-specific deletion of SWS/NTE in mice causes a similar pattern of progressive neuronal degeneration. NTE reacts with organophosphate compounds that cause a paralyzing axonal degeneration in humans and has been shown to degrade endoplasmic reticulum-associated phosphatidylcholine (PtdCho) in cultured mammalian cells. However, its function within the nervous system has remained unknown. Here, we show that both the fly and mouse SWS proteins can rescue the defects that arise in sws mutant flies, whereas a point mutation in the proposed active site cannot restore SWS function. Overexpression of catalytically active SWS caused formation of abnormal intracellular membraneous structures and cell death. Cell-specific expression revealed that not only neurons but also glia depend autonomously on SWS. In wild-type flies, endogenous SWS was detected by immmunohistochemistry in the endoplasmic reticulum (the primary site of PtdCho processing) of neurons and in some glia. sws mutant flies lacked NTE-like esterase activity and had increased levels of PtdCho. Conversely, overexpression of SWS resulted in increased esterase activity and reduced PtdCho. We conclude that SWS is essential for membrane lipid homeostasis and cell survival in both neurons and glia of the adult Drosophila brain and that NTE may play an analogous role in vertebrates.


Asunto(s)
Proteínas de Drosophila/deficiencia , Drosophila/citología , Esterasas/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Neuroglía/fisiología , Neuronas/fisiología , Animales , Animales Modificados Genéticamente , Western Blotting/métodos , Muerte Celular/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica/métodos , Metabolismo de los Lípidos , Ratones , Microscopía Electrónica de Transmisión/métodos , Mutagénesis/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuroglía/ultraestructura , Neuronas/ultraestructura , Fenotipo , Esteroles/metabolismo , Vacuolas/metabolismo
15.
Glia ; 49(1): 59-72, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15390099

RESUMEN

Patients with polyglutamine expansion diseases, like Huntington's disease or several spinocerebellar ataxias, first present with neurological symptoms that can occur in the absence of neurodegeneration. Behavioral symptoms thus appear to be caused by neuronal dysfunction, rather than cell death. Pathogenesis in polyglutamine expansion diseases is largely viewed as a cell-autonomous process in neurons. It is likely, however, that this process is influenced by changes in glial physiology and, at least in the case of DRPLA glial inclusions and glial cell death, seems to be an important part in the pathogenesis. To investigate these aspects in a Drosophila model system, we expressed polyglutamine proteins in the adult nervous system. Glial-specific expression of a polyglutamine (Q)-expanded (n=78) and also a nonexpanded (n=27) truncated version of human ataxin-3 led to the formation of protein aggregates and glial cell death. Behavioral changes were observed prior to cell death. This reveals that glia is susceptible to the toxic action of polyglutamine proteins. Neuronal expression of the same constructs resulted in behavioral changes similar to those resulting from glial expression but did not cause neurodegeneration. Behavioral deficits were selective and affected two analyzed fly behaviors differently. Both glial and neuronal aggregates of Q78 and Q27 appeared early in pathogenesis and, at the electron microscopic resolution, had a fibrillary substructure. This shows that a nonexpanded stretch can cause similar histological and behavioral symptoms as the expanded stretch, however, with a significant delay.


Asunto(s)
Cuerpos de Inclusión/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Sistema Nervioso/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Expansión de Repetición de Trinucleótido/genética , Factores de Edad , Animales , Ataxina-3 , Conducta Animal/fisiología , Muerte Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Núcleo Celular/ultraestructura , Modelos Animales de Enfermedad , Drosophila melanogaster , Femenino , Trastornos Neurológicos de la Marcha/genética , Trastornos Neurológicos de la Marcha/metabolismo , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/ultraestructura , Longevidad/genética , Masculino , Microscopía Electrónica de Transmisión , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/ultraestructura , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/patología , Neuroglía/ultraestructura , Neuronas/patología , Neuronas/ultraestructura , Proteínas Nucleares , Proteínas Represoras
16.
J Neurosci ; 24(16): 3899-906, 2004 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-15102905

RESUMEN

Beta-amyloid peptides that are cleaved from the amyloid precursor protein (APP) play a critical role in Alzheimer's disease (AD) pathophysiology. Here, we show that in Drosophila, the targeted expression of the key genes of AD, APP, the beta-site APP-cleaving enzyme BACE, and the presenilins led to the generation of beta-amyloid plaques and age-dependent neurodegeneration as well as to semilethality, a shortened life span, and defects in wing vein development. Genetic manipulations or pharmacological treatments with secretase inhibitors influenced the activity of the APP-processing proteases and modulated the severity of the phenotypes. This invertebrate model of amyloid plaque pathology demonstrates Abeta-induced neurodegeneration as a basic biological principle and may allow additional genetic analyses of the underlying molecular pathways.


Asunto(s)
Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Drosophila , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Placa Amiloide/patología , Factores de Edad , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Progresión de la Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Endopeptidasas/metabolismo , Marcación de Gen , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Fenotipo , Células Fotorreceptoras de Invertebrados/patología , Placa Amiloide/metabolismo , Presenilinas , Procesamiento Proteico-Postraduccional/fisiología , Retina/metabolismo , Retina/patología , Tasa de Supervivencia , Transgenes
17.
EMBO J ; 21(23): 6367-76, 2002 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-12456644

RESUMEN

The novel Drosophila mutant löchrig (loe) shows progressive neurodegeneration and neuronal cell death, in addition to a low level of cholesterol ester. loe affects a specific isoform of the gamma-subunit of AMP-activated protein kinase (AMPK), a negative regulator of hydroxymethylglutaryl (HMG)-CoA reductase and cholesterol synthesis in vertebrates. Although Drosophila cannot synthesize cholesterol de novo, the regulatory role of fly AMPK on HMG-CoA reductase is conserved. The loe phenotype is modified by the level of HMG-CoA reductase and suppressed by the inhibition of this enzyme by statin, which has been used for the treatment of Alzheimer patients. In addition, the degenerative phenotype of loe is enhanced by a mutation in amyloid precursor protein-like (APPL), the fly homolog of the human amyloid precursor protein involved in Alzheimer's disease. Western analysis revealed that the loe mutation reduces APPL processing, whereas overexpression of Loe increases it. These results describe a novel function of AMPK in neurodegeneration and APPL/APP processing which could be mediated through HMG-CoA reductase and cholesterol ester.


Asunto(s)
Colesterol/metabolismo , Proteínas de Drosophila , Proteínas de la Membrana , Proteínas del Tejido Nervioso/metabolismo , Proteínas Quinasas/genética , Animales , Drosophila/genética , Drosophila/metabolismo , Mutación , Neuronas/metabolismo , Neuronas/patología , Proteínas Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...