Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Acta Neuropathol ; 129(1): 21-37, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25432317

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/fisiopatologia , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/metabolismo , Sinapses/fisiologia , Precursor de Proteína beta-Amiloide/genética , Animais , Dendritos/patologia , Dendritos/fisiologia , Feminino , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos Knockout , Atividade Motora/fisiologia , Neuritos/patologia , Neuritos/fisiologia , Fragmentos de Peptídeos/genética , Proteínas Recombinantes/metabolismo , Memória Espacial/fisiologia , Sinapses/patologia
2.
Alzheimers Dement ; 10(2): e27-39, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23850332

RESUMO

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.


Assuntos
Doença de Alzheimer/genética , Saúde da Família , Predisposição Genética para Doença/genética , Mutação/genética , Presenilina-1/genética , Adulto , Idade de Início , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/complicações , Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Ondas Encefálicas/genética , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/genética , Eletroencefalografia , Feminino , Humanos , Masculino , Entrevista Psiquiátrica Padronizada , Pessoa de Meia-Idade , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fenilalanina/genética , Tomografia por Emissão de Pósitrons , Radiografia , Serina/genética
3.
Nat Cell Biol ; 7(11): 1118-23, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227967

RESUMO

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).


Assuntos
Peptídeos beta-Amiloides/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Colesterol/metabolismo , Metabolismo dos Lipídeos , Fragmentos de Peptídeos/análise , Esfingomielinas/metabolismo , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases , Células COS , Células Cultivadas , Chlorocebus aethiops , Endopeptidases/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Fragmentos de Peptídeos/metabolismo , Presenilina-1 , Presenilina-2 , Esfingomielina Fosfodiesterase/metabolismo
4.
Exp Brain Res ; 217(3-4): 441-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22076403

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/fisiologia , Giro Denteado/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Microbiol Spectr ; 10(6): e0312522, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36314963

RESUMO

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.


Assuntos
HIV-1 , Hepatite C , Humanos , HIV-1/genética , Vírus da Hepatite B/genética , Carga Viral/métodos , Reprodutibilidade dos Testes , Hepacivirus/genética , Hepatite C/diagnóstico
6.
BMC Genomics ; 12: 160, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21435241

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Perfilação da Expressão Gênica , Córtex Pré-Frontal/metabolismo , Animais , Análise por Conglomerados , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos
7.
J Immunol ; 182(12): 7613-24, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494285

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Encéfalo/ultraestrutura , Linhagem Celular , Sistema Nervoso Central/imunologia , Feminino , Humanos , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica , Solubilidade , Linfócitos T/imunologia , Vacinação , Vírion/ultraestrutura
8.
Mol Cell Neurosci ; 44(4): 386-93, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20472066

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Apoptose , Região CA1 Hipocampal/citologia , Espinhas Dendríticas/patologia , Degeneração Neural , Complexo de Endopeptidases do Proteassoma/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Apoptose/fisiologia , Apoptose/efeitos da radiação , Células Cultivadas , Cromonas/farmacologia , Meios de Cultivo Condicionados/química , Técnicas de Cultura , Inibidores Enzimáticos/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Microscopia Confocal , Morfolinas/farmacologia , Oligopeptídeos/farmacologia , Células PC12 , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Proteínas Recombinantes de Fusão , Antígenos Thy-1 , Raios Ultravioleta
9.
Genesis ; 48(3): 200-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20140888

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Marcação de Genes/métodos , Modelos Genéticos , Alelos , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Southern Blotting , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Membro Anterior/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Força da Mão , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Tamanho do Órgão
11.
Neurobiol Dis ; 33(2): 274-81, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19049874

RESUMO

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.


Assuntos
Amiloide/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Envelhecimento , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/fisiologia , Comportamento Animal , Western Blotting , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Expressão Gênica , Imuno-Histoquímica , Luz , Proteínas de Membrana/genética , Microscopia Eletrônica , Dados de Sequência Molecular , Degeneração Neural , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/metabolismo , Nexinas de Proteases , Receptores de Superfície Celular/genética
12.
J Neurosci ; 25(11): 2865-73, 2005 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-15772346

RESUMO

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.


Assuntos
Proteínas de Drosophila/deficiência , Drosophila/citologia , Esterases/metabolismo , Proteínas do Tecido Nervoso/deficiência , Neuroglia/fisiologia , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Western Blotting/métodos , Morte Celular/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica/métodos , Metabolismo dos Lipídeos , Camundongos , Microscopia Eletrônica de Transmissão/métodos , Mutagênese/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neuroglia/ultraestrutura , Neurônios/ultraestrutura , Fenótipo , Esteróis/metabolismo , Vacúolos/metabolismo
13.
J Neurosci ; 24(16): 3899-906, 2004 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15102905

RESUMO

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.


Assuntos
Doença de Alzheimer/patologia , Modelos Animais de Doenças , Drosophila , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Placa Amiloide/patologia , Fatores Etários , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Progressão da Doença , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Endopeptidases/metabolismo , Marcação de Genes , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Fenótipo , Células Fotorreceptoras de Invertebrados/patologia , Placa Amiloide/metabolismo , Presenilinas , Processamento de Proteína Pós-Traducional/fisiologia , Retina/metabolismo , Retina/patologia , Taxa de Sobrevida , Transgenes
14.
J Neurosci Res ; 85(12): 2580-90, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17335075

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Axônios/metabolismo , Dendritos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Precursor de Proteína beta-Amiloide/genética , Animais , Transporte Axonal/fisiologia , Células Cultivadas , Chlorocebus aethiops , Camundongos , Proteínas Associadas aos Microtúbulos , Neurônios/citologia , Transporte Proteico/fisiologia , Deleção de Sequência/fisiologia , Transfecção/métodos
15.
Recent Pat CNS Drug Discov ; 1(1): 119-27, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18221198

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Anti-Inflamatórios não Esteroides/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Imunização
16.
Glia ; 49(1): 59-72, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15390099

RESUMO

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.


Assuntos
Corpos de Inclusão/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Sistema Nervoso/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Fatores Etários , Animais , Ataxina-3 , Comportamento Animal/fisiologia , Morte Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Núcleo Celular/ultraestrutura , Modelos Animais de Doenças , Drosophila melanogaster , Feminino , Transtornos Neurológicos da Marcha/genética , Transtornos Neurológicos da Marcha/metabolismo , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/ultraestrutura , Longevidade/genética , Masculino , Microscopia Eletrônica de Transmissão , Proteínas do Tecido Nervoso/genética , Sistema Nervoso/ultraestrutura , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neuroglia/patologia , Neuroglia/ultraestrutura , Neurônios/patologia , Neurônios/ultraestrutura , Proteínas Nucleares , Proteínas Repressoras
17.
EMBO J ; 21(23): 6367-76, 2002 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-12456644

RESUMO

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.


Assuntos
Colesterol/metabolismo , Proteínas de Drosophila , Proteínas de Membrana , Proteínas do Tecido Nervoso/metabolismo , Proteínas Quinases/genética , Animais , Drosophila/genética , Drosophila/metabolismo , Mutação , Neurônios/metabolismo , Neurônios/patologia , Proteínas Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA