Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Neurochem ; 128(5): 686-700, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24117625

RESUMO

The WWC1 gene has been genetically associated with human episodic memory performance, and its product KIdney/BRAin protein (KIBRA) has been shown to interact with the atypical protein kinase protein kinase M ζ (PKMζ). Although recently challenged, PKMζ remains a candidate postsynaptic regulator of memory maintenance. Here, we show that PKMζ is subject to rapid proteasomal degradation and that KIBRA is both necessary and sufficient to counteract this process, thus stabilizing the kinase and maintaining its function for a prolonged time. We define the binding sequence on KIBRA, a short amino acid motif near the C-terminus. Both hippocampal knock-down of KIBRA in rats and KIBRA knock-out in mice result in decreased learning and memory performance in spatial memory tasks supporting the notion that KIBRA is a player in episodic memory. Interestingly, decreased memory performance is accompanied by decreased PKMζ protein levels. We speculate that the stabilization of synaptic PKMζ protein levels by KIBRA may be one mechanism by which KIBRA acts in memory maintenance. KIBRA/WWC1 has been genetically associated with human episodic memory. KIBRA has been shown to be post-synaptically localized, but its function remained obscure. Here, we show that KIBRA shields PKMζ, a kinase previously linked to memory maintenance, from proteasomal degradation via direct interaction. KIBRA levels in the rodent hippocampus correlate closely both to spatial memory performance in rodents and to PKMζ levels. Our findings support a role for KIBRA in memory, and unveil a novel function for this protein.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas Correpressoras/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Proteína Quinase C/fisiologia , Sequência de Aminoácidos , Animais , Aprendizagem da Esquiva/fisiologia , Comportamento Animal/fisiologia , Western Blotting , Proteínas de Transporte/metabolismo , Proteínas Correpressoras/metabolismo , Dependovirus/genética , Teste de Complementação Genética , Hipocampo/metabolismo , Hipocampo/fisiologia , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Dados de Sequência Molecular , Fosfoproteínas , Reação em Cadeia da Polimerase , Ligação Proteica , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Técnicas Estereotáxicas
2.
Front Cell Neurosci ; 8: 464, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25653590

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is an incurable fatal motoneuron disease with a lifetime risk of approximately 1:400. It is characterized by progressive weakness, muscle wasting, and death ensuing 3-5 years after diagnosis. Granulocyte-colony stimulating factor (G-CSF) is a drug candidate for ALS, with evidence for efficacy from animal studies and interesting data from pilot clinical trials. To gain insight into the disease mechanisms and mode of action of G-CSF, we performed gene expression profiling on isolated lumbar motoneurons from SOD1(G93A) mice, the most frequently studied animal model for ALS, with and without G-CSF treatment. RESULTS: Motoneurons from SOD1(G93A) mice present a distinct gene expression profile in comparison to controls already at an early disease stage (11 weeks of age), when treatment was initiated. The degree of deregulation increases at a time where motor symptoms are obvious (15 weeks of age). Upon G-CSF treatment, transcriptomic deregulations of SOD1(G93A) motoneurons were notably restored. Discriminant analysis revealed that SOD1 mice treated with G-CSF has a transcriptom close to presymptomatic SOD1 mice or wild type mice. Some interesting genes modulated by G-CSF treatment relate to neuromuscular function such as CCR4-NOT or Prss12. CONCLUSIONS: Our data suggest that G-CSF is able to re-adjust gene expression in symptomatic SOD1(G93A) motoneurons. This provides further arguments for G-CSF as a promising drug candidate for ALS.

3.
J Neurochem ; 119(1): 165-75, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21812782

RESUMO

The stimulation of neurogenesis is an exciting novel therapeutic option for diseases of the central nervous system, ranging from depression to neurodegeneration. One major bottleneck in screening approaches for neurogenesis-inducing compounds is the very demanding in vivo quantification of newborn neurons based on stereological techniques. To effectively develop compounds in this area, novel fast and reliable techniques for quantification of in vivo neurogenesis are needed. In this study, we introduce a flow cytometry-based method for quantifying newly generated neurons in the brain based on the counting of cell nuclei from dissected brain regions. Important steps involve density sedimentation of the cell nuclei, and staining for the proliferation marker bromodeoxy uridine and nuclear cell type markers such as NeuN. We demonstrate the ability of the technique to detect increased neurogenesis in the hippocampus of animals which underwent physical exercise and received fluoxetine treatment.


Assuntos
Encéfalo/fisiologia , Neurogênese/fisiologia , Animais , Antimetabólitos , Encéfalo/efeitos dos fármacos , Bromodesoxiuridina , Contagem de Células , Núcleo Celular/fisiologia , Centrifugação com Gradiente de Concentração , Proteínas de Ligação a DNA , Citometria de Fluxo , Fluoxetina/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Reprodutibilidade dos Testes , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-20552044

RESUMO

The genetic locus encoding KIBRA, a member of the WWC family of proteins, has recently been shown to be associated with human memory performance through genome-wide single nucleotide polymorphism screening. Gene expression analysis and a variety of functional studies have further indicated that such a role is biologically plausible for KIBRA. Here, we review the existing literature, illustrate connections between the different lines of evidence, and derive models based on KIBRA's function(s) in the brain that can be further tested experimentally.

5.
Mol Cell Neurosci ; 41(2): 166-74, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19281847

RESUMO

Alzheimer's disease is characterized by accumulation of toxic beta-amyloid (Abeta) in the brain and neuronal death. Several mutations in presenilin (PS1) and beta-amyloid precursor protein (APP) associate with an increased Abeta(42/40) ratio. Abeta(42), a highly fibrillogenic species, is believed to drive Abeta aggregation. Factors shifting gamma-secretase cleavage of APP to produce Abeta(42) are unclear. We investigate the molecular mechanism underlying altered Abeta(42/40) ratios associated with APP mutations at codon 716 and 717. Using FRET-based fluorescence lifetime imaging to monitor APP-PS1 interactions, we show that I716F and V717I APP mutations increase the proportion of interacting molecules earlier in the secretory pathway, resulting in an increase in Abeta generation. A PS1 conformation assay reveals that, in the presence of mutant APP, PS1 adopts a conformation reminiscent of FAD-associated PS1 mutations, thus influencing APP binding to PS1/gamma-secretase. Mutant APP affects both intracellular location and efficiency of APP-PS1 interactions, thereby changing the Abeta(42/40) ratio.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide , Mutação , Presenilinas/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Transferência Ressonante de Energia de Fluorescência , Humanos , Presenilinas/química , Presenilinas/genética , Conformação Proteica , Transporte Proteico/fisiologia
6.
Brain ; 131(Pt 12): 3335-47, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18835867

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in progressive loss of motoneurons, motor weakness and death within 1-5 years after disease onset. Therapeutic options remain limited despite a substantial number of approaches that have been tested clinically. In particular, various neurotrophic factors have been investigated. Failure in these trials has been largely ascribed to problems of insufficient dosing or inability to cross the blood-brain barrier (BBB). We have recently uncovered the neurotrophic properties of the haematopoietic protein granulocyte-colony stimulating factor (G-CSF). The protein is clinically well tolerated and crosses the intact BBB. This study examined the potential role of G-CSF in motoneuron diseases. We investigated the expression of the G-CSF receptor in motoneurons and studied effects of G-CSF in a motoneuron cell line and in the SOD1(G93A) transgenic mouse model. The neurotrophic growth factor was applied both by continuous subcutaneous delivery and CNS-targeted transgenic overexpression. This study shows that given at the stage of the disease where muscle denervation is already evident, G-CSF leads to significant improvement in motor performance, delays the onset of severe motor impairment and prolongs overall survival of SOD1(G93A)tg mice. The G-CSF receptor is expressed by motoneurons and G-CSF protects cultured motoneuronal cells from apoptosis. In ALS mice, G-CSF increased survival of motoneurons and decreased muscular denervation atrophy. We conclude that G-CSF is a novel neurotrophic factor for motoneurons that is an attractive and feasible drug candidate for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Infusões Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Mutação , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Medula Espinal/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Resultado do Tratamento
7.
Neurosci Lett ; 442(2): 91-5, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18602448

RESUMO

The ability of the low density lipoprotein receptor-related protein (LRP) to form homo-dimers was studied in mouse neuroblastoma and human neuroglioma cells as well as in primary cortical cultures from adult mouse brain. Homo-dimerization of LRP light chain (LC) was shown by several methods including co-immunoprecipitation, fluorescence lifetime imaging microscopy, and bimolecular fluorescence complementation assay. The requirement of intact NPXY motifs of LRP LC for homo-dimerization was ruled out by co-immunoprecipitation assay.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Neurônios/metabolismo , Homologia de Sequência de Aminoácidos , Motivos de Aminoácidos , Animais , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Endocitose , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Processamento de Proteína Pós-Traducional , Transfecção/métodos
8.
PLoS One ; 3(4): e1867, 2008 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-18382657

RESUMO

BACKGROUND: Misfolding, oligomerization, and fibrillization of alpha-synuclein are thought to be central events in the onset and progression of Parkinson's disease (PD) and related disorders. Although fibrillar alpha-synuclein is a major component of Lewy bodies (LBs), recent data implicate prefibrillar, oligomeric intermediates as the toxic species. However, to date, oligomeric species have not been identified in living cells. METHODOLOGY/PRINCIPAL FINDINGS: Here we used bimolecular fluorescence complementation (BiFC) to directly visualize alpha-synuclein oligomerization in living cells, allowing us to study the initial events leading to alpha-synuclein oligomerization, the precursor to aggregate formation. This novel assay provides us with a tool with which to investigate how manipulations affecting alpha-synuclein aggregation affect the process over time. Stabilization of alpha-synuclein oligomers via BiFC results in increased cytotoxicity, which can be rescued by Hsp70 in a process that reduces the formation of alpha-synuclein oligomers. Introduction of PD-associated mutations in alpha-synuclein did not affect oligomer formation but the biochemical properties of the mutant alpha-synuclein oligomers differ from those of wild type alpha-synuclein. CONCLUSIONS/SIGNIFICANCE: This novel application of the BiFC assay to the study of the molecular basis of neurodegenerative disorders enabled the direct visualization of alpha-synuclein oligomeric species in living cells and its modulation by Hsp70, constituting a novel important tool in the search for therapeutics for synucleinopathies.


Assuntos
Mutação , alfa-Sinucleína/química , alfa-Sinucleína/genética , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Proteínas de Choque Térmico HSP70/química , Humanos , Corpos de Lewy/metabolismo , Microscopia de Fluorescência/métodos , Doença de Parkinson/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , alfa-Sinucleína/metabolismo
9.
Biochem Biophys Res Commun ; 370(2): 207-12, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18374657

RESUMO

Cleavage of APP by BACE is the first proteolytic step in the production of Amyloid beta (Abeta, which accumulates in senile plaques in Alzheimer's disease. BACE-cleavage of APP is thought to happen in endosomes. However, there are controversial data whether APP and BACE can already interact on the cell surface dependent on the cholesterol level. To examine whether APP and BACE come into close proximity on the cell surface in living cells, we employed a novel technique by combining time-resolved Förster resonance energy transfer (FRET) measurements with total internal reflection microscopy (TIRET microscopy). Our data indicate that BACE and APP come into close proximity within the cell, but probably not on the cell surface. To analyze the impact of alterations in cholesterol level upon BACE-cleavage, we measured sAPP secretion. Alteration of APP processing and BACE proximity by cholesterol might be explained by alterations in cell membrane fluidity.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Microscopia de Fluorescência/métodos , Receptores de Superfície Celular/metabolismo , Secretases da Proteína Precursora do Amiloide/análise , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/análise , Precursor de Proteína beta-Amiloide/genética , Ácido Aspártico Endopeptidases/análise , Ácido Aspártico Endopeptidases/genética , Linhagem Celular Tumoral , Membrana Celular/química , Colesterol/análise , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , Fluidez de Membrana , Nexinas de Proteases , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/genética
10.
J Neurosci ; 26(39): 9913-22, 2006 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-17005855

RESUMO

The beta-amyloid (Abeta) precursor protein (APP) is cleaved sequentially by beta-site of APP-cleaving enzyme (BACE) and gamma-secretase to release the Abeta peptides that accumulate in plaques in Alzheimer's disease (AD). GGA1, a member of the Golgi-localized gamma-ear-containing ARF-binding (GGA) protein family, interacts with BACE and influences its subcellular distribution. We now report that overexpression of GGA1 in cells increased the APP C-terminal fragment resulting from beta-cleavage but surprisingly reduced Abeta. GGA1 confined APP to the Golgi, in which fluorescence resonance energy transfer analyses suggest that the proteins come into close proximity. GGA1 blunted only APP but not notch intracellular domain release. These results suggest that GGA1 prevented APP beta-cleavage products from becoming substrates for gamma-secretase. Direct binding of GGA1 to BACE was not required for these effects, but the integrity of the GAT (GGA1 and TOM) domain of GGA1 was. GGA1 may act as a specific spatial switch influencing APP trafficking and processing, so that APP-GGA1 interactions may have pathophysiological relevance in AD.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Química Encefálica , Linhagem Celular , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Glicosilação , Complexo de Golgi/metabolismo , Humanos , Rim/citologia , Rim/embriologia , Camundongos , Neuroblastoma/patologia , Fragmentos de Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Relação Estrutura-Atividade , Transfecção
11.
J Biol Chem ; 281(36): 26400-7, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16815845

RESUMO

Presenilin 1 (PS1) in its active heterodimeric form is the catalytic center of the gamma-secretase complex, an enzymatic activity that cleaves amyloid precursor protein (APP) to produce amyloid beta (Abeta). Ubiquilin 1 is a recently described PS1 interacting protein, the overexpression of which increases PS1 holoprotein levels and leads to reduced levels of functionally active PS1 heterodimer. In addition, it has been suggested that splice variants of the UBQLN1 gene are associated with an increased risk of developing Alzheimer disease (AD). However, it is still unclear whether PS1 and ubiquilin 1 interact when expressed at endogenous levels under normal physiological conditions. Here, we employ three novel fluorescence resonance energy transfer-based techniques to investigate the interaction between PS1 and ubiquilin 1 in intact cells. We consistently find that the ubiquilin 1 N terminus is in close proximity to several epitopes on PS1. We show that ubiquilin 1 interacts both with PS1 holoprotein and heterodimer and that the interaction between PS1 and ubiquilin 1 takes place near the cell surface. Furthermore, we show that the PS1-ubiquilin 1 interaction can be detected between endogenous proteins in primary neurons in vitro as well as in brain tissue of healthy controls and Alzheimer disease patients, providing evidence of its physiological relevance.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Microscopia de Fluorescência , Presenilina-1/metabolismo , Conformação Proteica , Proteínas Adaptadoras de Transdução de Sinal , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Proteínas Relacionadas à Autofagia , Encéfalo/citologia , Encéfalo/patologia , Células CHO , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cricetinae , Dimerização , Epitopos , Feminino , Transferência Ressonante de Energia de Fluorescência , Humanos , Masculino , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Pessoa de Meia-Idade , Neurônios/citologia , Neurônios/metabolismo , Presenilina-1/química , Presenilina-1/genética
12.
J Neurosci ; 26(17): 4690-700, 2006 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-16641250

RESUMO

Interactions between dopaminergic and glutamatergic afferents in the striatum are essential for motor learning and the regulation of movement. An important mechanism for these interactions is the ability of dopamine, through D1 receptors, to potentiate NMDA glutamate receptor function. Here we show that, in striatal neurons, D1 receptor activation leads to rapid trafficking of NMDA receptor subunits, with increased NR1 and NR2B subunits in dendrites, enhanced coclustering of these subunits with the postsynaptic density scaffolding molecule postsynaptic density-95, and increased surface expression. The dopamine D1 receptor-mediated NMDA receptor trafficking is blocked by an inhibitor of tyrosine kinases. Blockers of tyrosine phosphatases also induce NMDA subunit trafficking, but this effect is nonselective and alters both NR2A- and NR2B-containing receptors. Furthermore, tyrosine phosphatase inhibition leads to the clustering of tyrosine-phosphorylated NR2B subunit along dendritic shafts. Our findings reveal that D1 receptor activation can potentiate striatal NMDA subunit function by directly promoting the surface insertion of the receptor complexes. This effect is regulated by the reciprocal actions of protein tyrosine phosphatases and tyrosine kinases. Modification of these pathways may be a useful therapeutic target for Parkinson's disease and other basal ganglia disorders in which abnormal function of striatal NMDA receptors contributes to the symptoms of the diseases.


Assuntos
Corpo Estriado/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina/metabolismo , Animais , Células Cultivadas , Fosforilação , Subunidades Proteicas , Transporte Proteico/fisiologia , Ratos , Distribuição Tecidual
13.
Biochemistry ; 45(8): 2618-28, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16489755

RESUMO

SorLA/LR11 is a sorting receptor that regulates the intracellular transport and processing of the amyloid precursor protein (APP) in neurons. SorLA/LR11-mediated binding results in sequestration of APP in the Golgi and in protection from processing into the amyloid-beta peptide (Abeta), the principal component of senile plaques in Alzheimer's disease (AD). To gain insight into the molecular mechanisms governing sorLA and APP interaction, we have dissected the respective protein interacting domains. Using a fluorescence resonance energy transfer (FRET) based assay of protein proximity, we identified binding sites in the extracellular regions of both proteins. Fine mapping by surface plasmon resonance analysis and analytical ultracentrifugation of recombinant APP and sorLA fragments further narrowed down the binding domains to the cluster of complement-type repeats in sorLA that forms a 1:1 stoichiometric complex with the carbohydrate-linked domain of APP. These data shed new light on the molecular determinants of neuronal APP trafficking and processing and on possible targets for intervention with senile plaque formation in patients with AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Neurônios/metabolismo , Transporte Proteico/fisiologia , Receptores de LDL/metabolismo , Precursor de Proteína beta-Amiloide/química , Encéfalo/metabolismo , Células Cultivadas , Humanos , Proteínas Relacionadas a Receptor de LDL , Proteínas de Membrana Transportadoras/química , Microscopia Confocal , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico/genética , Receptores de LDL/química , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Transfecção
14.
J Neurosci ; 26(2): 418-28, 2006 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-16407538

RESUMO

sorLA is a recently identified neuronal receptor for amyloid precursor protein (APP) that is known to interact with APP and affect its intracellular transport and processing. Decreased levels of sorLA in the brain of Alzheimer's disease (AD) patients and elevated levels of amyloid-beta peptide (Abeta) in sorLA-deficient mice point to the importance of the receptor in this neurodegenerative disorder. We analyzed APP cleavage in an APP-shedding assay and found that both sorLA and, surprisingly, a sorLA tail construct inhibited APP cleavage in a beta-site APP-cleaving enzyme (BACE)-dependent manner. In line with this finding, sorLA and the sorLA tail significantly reduced secreted Abeta levels when BACE was overexpressed, suggesting that sorLA influences beta-cleavage. To understand the effect of sorLA on APP cleavage by BACE, we analyzed whether sorLA interacts with APP and/or BACE. Because both full-length sorLA and sorLA C-terminal tail constructs were functionally relevant for APP processing, we analyzed sorLA-APP for a potential cytoplasmatic interaction domain. sorLA and C99 coimmunoprecipitated, pointing toward the existence of a new cytoplasmatic interaction site between sorLA and APP. Moreover, sorLA and BACE also coimmunoprecipitate. Thus, sorLA interacts both with BACE and APP and might therefore directly affect BACE-APP complex formation. To test whether sorLA impacts BACE-APP interactions, we used a fluorescence resonance energy transfer assay to evaluate BACE-APP interactions in cells. We discovered that sorLA significantly reduced BACE-APP interactions in Golgi. We postulate that sorLA acts as a trafficking receptor that prevents BACE-APP interactions and hence BACE cleavage of APP.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidases/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mapeamento de Interação de Proteínas , Receptores de LDL/metabolismo , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/química , Animais , Ácido Aspártico Endopeptidases , Biotinilação , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Citosol , Endocitose , Endopeptidases/química , Complexo de Golgi/metabolismo , Humanos , Rim , Proteínas Relacionadas a Receptor de LDL , Proteínas de Membrana Transportadoras/química , Complexos Multiproteicos , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de LDL/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
15.
Cell ; 122(5): 751-62, 2005 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-16143106

RESUMO

Androgens and estrogens are transported bound to the sex hormone binding globulin (SHBG). SHBG is believed to keep sex steroids inactive and to control the amount of free hormones that enter cells by passive diffusion. Contrary to the free hormone hypothesis, we demonstrate that megalin, an endocytic receptor in reproductive tissues, acts as a pathway for cellular uptake of biologically active androgens and estrogens bound to SHBG. In line with this function, lack of receptor expression in megalin knockout mice results in impaired descent of the testes into the scrotum in males and blockade of vagina opening in females. Both processes are critically dependent on sex-steroid signaling, and similar defects are seen in animals treated with androgen- or estrogen-receptor antagonists. Thus, our findings uncover the existence of endocytic pathways for protein bound androgens and estrogens and their crucial role in development of the reproductive organs.


Assuntos
Sistema Endócrino/fisiologia , Endocitose/fisiologia , Hormônios Esteroides Gonadais/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Androgênios/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Sistema Endócrino/citologia , Estrogênios/metabolismo , Células Eucarióticas/metabolismo , Feminino , Perfilação da Expressão Gênica , Hormônios Esteroides Gonadais/antagonistas & inibidores , Hormônios Esteroides Gonadais/sangue , Humanos , Imuno-Histoquímica , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Knockout , Gravidez , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Globulina de Ligação a Hormônio Sexual/farmacologia , Fatores de Tempo , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/patologia , Sistema Urogenital/embriologia , Sistema Urogenital/patologia
16.
Proc Natl Acad Sci U S A ; 102(38): 13461-6, 2005 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-16174740

RESUMO

sorLA (Sorting protein-related receptor) is a type-1 membrane protein of unknown function that is expressed in neurons. Its homology to sorting receptors that shuttle between the plasma membrane, endosomes, and the Golgi suggests a related function in neuronal trafficking processes. Because expression of sorLA is reduced in the brain of patients with Alzheimer's disease (AD), we tested involvement of this receptor in intracellular transport and processing of the amyloid precursor protein (APP) to the amyloid beta-peptide (Abeta), the principal component of senile plaques. We demonstrate that sorLA interacts with APP in vitro and in living cells and that both proteins colocalize in endosomal and Golgi compartments. Overexpression of sorLA in neurons causes redistribution of APP to the Golgi and decreased processing to Abeta, whereas ablation of sorLA expression in knockout mice results in increased levels of Abeta in the brain similar to the situation in AD patients. Thus, sorLA acts as a sorting receptor that protects APP from processing into Abeta and thereby reduces the burden of amyloidogenic peptide formation. Consequently, reduced receptor expression in the human brain may increase Abeta production and plaque formation and promote spontaneous AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Placa Amiloide/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de LDL/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Proteínas Relacionadas a Receptor de LDL , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Transporte Proteico , Receptores de LDL/genética
17.
J Biol Chem ; 280(18): 17777-85, 2005 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-15749709

RESUMO

BACE is a transmembrane protease with beta-secretase activity that cleaves the amyloid precursor protein (APP). After BACE cleavage, APP becomes a substrate for gamma-secretase, leading to release of amyloid-beta peptide (Abeta), which accumulates in senile plaques in Alzheimer disease. APP and BACE are co-internalized from the cell surface to early endosomes. APP is also known to interact at the cell surface and be internalized by the low density lipoprotein receptor-related protein (LRP), a multifunctional endocytic and signaling receptor. Using a new fluorescence resonance energy transfer (FRET)-based assay of protein proximity, fluorescence lifetime imaging (FLIM), and co-immunoprecipitation we demonstrate that the light chain of LRP interacts with BACE on the cell surface in association with lipid rafts. Surprisingly, the BACE-LRP interaction leads to an increase in LRP C-terminal fragment, release of secreted LRP in the media and subsequent release of the LRP intracellular domain from the membrane. Taken together, these data suggest that there is a close interaction between BACE and LRP on the cell surface, and that LRP is a novel BACE substrate.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases/genética , Células CHO , Linhagem Celular Tumoral , Cricetinae , Endopeptidases , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Especificidade por Substrato/fisiologia
18.
Development ; 132(2): 405-14, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15623804

RESUMO

Megalin is a low-density lipoprotein receptor-related protein (LRP2) expressed in the neuroepithelium and the yolk sac of the early embryo. Absence of megalin expression in knockout mice results in holoprosencephaly, indicating an essential yet unidentified function in forebrain development. We used mice with complete or conditional megalin gene inactivation in the embryo to demonstrate that expression of megalin in the neuroepithelium but not in the yolk sac is crucial for brain development. During early forebrain development, megalin deficiency leads to an increase in bone morphogenic protein (Bmp) 4 expression and signaling in the rostral dorsal neuroepithelium, and a subsequent loss of sonic hedgehog (Shh) expression in the ventral forebrain. As a consequence of absent SHH activity, ventrally derived oligodendroglial and interneuronal cell populations are lost in the forebrain of megalin-/- embryos. Similar defects are seen in models with enhanced signaling through BMPs, central regulators of neural tube patterning. Because megalin mediates endocytic uptake and degradation of BMP4, these findings indicate a role for megalin in neural tube specification, possibly by acting as BMP4 clearance receptor in the neuroepithelium.


Assuntos
Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Telencéfalo/embriologia , Animais , Apoptose , Padronização Corporal , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/metabolismo , Encéfalo/embriologia , Linhagem da Célula , Proliferação de Células , Células Epiteliais/citologia , Epitélio/metabolismo , Proteínas Hedgehog , Imuno-Histoquímica , Hibridização In Situ , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Transgênicos , Crista Neural/citologia , Prosencéfalo/metabolismo , Fatores de Tempo , Transativadores/biossíntese , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...