Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS Genet ; 8(8): e1002918, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952452

RESUMO

Abnormal phosphorylation and toxicity of a microtubule-associated protein tau are involved in the pathogenesis of Alzheimer's disease (AD); however, what pathological conditions trigger tau abnormality in AD is not fully understood. A reduction in the number of mitochondria in the axon has been implicated in AD. In this study, we investigated whether and how loss of axonal mitochondria promotes tau phosphorylation and toxicity in vivo. Using transgenic Drosophila expressing human tau, we found that RNAi-mediated knockdown of milton or Miro, an adaptor protein essential for axonal transport of mitochondria, enhanced human tau-induced neurodegeneration. Tau phosphorylation at an AD-related site Ser262 increased with knockdown of milton or Miro; and partitioning defective-1 (PAR-1), the Drosophila homolog of mammalian microtubule affinity-regulating kinase, mediated this increase of tau phosphorylation. Tau phosphorylation at Ser262 has been reported to promote tau detachment from microtubules, and we found that the levels of microtubule-unbound free tau increased by milton knockdown. Blocking tau phosphorylation at Ser262 site by PAR-1 knockdown or by mutating the Ser262 site to unphosphorylatable alanine suppressed the enhancement of tau-induced neurodegeneration caused by milton knockdown. Furthermore, knockdown of milton or Miro increased the levels of active PAR-1. These results suggest that an increase in tau phosphorylation at Ser262 through PAR-1 contributes to tau-mediated neurodegeneration under a pathological condition in which axonal mitochondria is depleted. Intriguingly, we found that knockdown of milton or Miro alone caused late-onset neurodegeneration in the fly brain, and this neurodegeneration could be suppressed by knockdown of Drosophila tau or PAR-1. Our results suggest that loss of axonal mitochondria may play an important role in tau phosphorylation and toxicity in the pathogenesis of AD.


Assuntos
Doença de Alzheimer , Proteínas de Drosophila , Drosophila , Quinase 3 da Glicogênio Sintase , Proteínas rho de Ligação ao GTP , Proteínas tau , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Axônios/patologia , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fosforilação , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
2.
J Biol Chem ; 287(23): 19715-24, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22511769

RESUMO

Amyloid ß-precursor protein (APP) is primarily cleaved by α- or ß-secretase to generate membrane-bound, C-terminal fragments (CTFs). In turn, CTFs are potentially subject to a second, intramembrane cleavage by γ-secretase, which is active in a lipid raft-like membrane microdomain. Mature APP (N- and O-glycosylated APP), the actual substrate of these secretases, is phosphorylated at the cytoplasmic residue Thr(668) and this phosphorylation changes the overall conformation of the cytoplasmic domain of APP. We found that phosphorylated and nonphosphorylated CTFs exist equally in mouse brain and are kinetically equivalent as substrates for γ-secretase, in vitro. However, in vivo, the level of the phosphorylated APP intracellular domain peptide (pAICD) generated by γ-cleavage of CTFs was very low when compared with the level of nonphosphorylated AICD (nAICD). Phosphorylated CTFs (pCTFs), rather than nonphosphorylated CTFs (nCTFs), were preferentially located outside of detergent-resistant, lipid raft-like membrane microdomains. The APP cytoplasmic domain peptide (APP(648-695)) with Thr(P)(668) did not associate with liposomes composed of membrane lipids from mouse brain to which the nonphosphorylated peptide preferentially bound. In addition, APP lacking the C-terminal 8 amino acids (APP-ΔC8), which are essential for membrane association, decreased Aß generation in N2a cells. These observations suggest that the pCTFs and CTFΔC8 are relatively movable within the membrane, whereas the nCTFs are susceptible to being anchored into the membrane, an interaction made available as a consequence of not being phosphorylated. By this mechanism, nCTFs can be preferentially captured and cleaved by γ-secretase. Preservation of the phosphorylated state of APP-CTFs may be a potential treatment to lower the generation of Aß in Alzheimer disease.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Microdomínios da Membrana/metabolismo , Triptofano/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Química Encefálica/genética , Microdomínios da Membrana/genética , Camundongos , Fosforilação , Estrutura Terciária de Proteína , Triptofano/genética
3.
Hum Mol Genet ; 19(15): 2947-57, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20466736

RESUMO

The amyloid-beta 42 (Abeta42) peptide has been suggested to promote tau phosphorylation and toxicity in Alzheimer's disease (AD) pathogenesis; however, the underlying mechanisms are not fully understood. Using transgenic Drosophila expressing both human Abeta42 and tau, we show here that tau phosphorylation at Ser262 plays a critical role in Abeta42-induced tau toxicity. Co-expression of Abeta42 increased tau phosphorylation at AD-related sites including Ser262, and enhanced tau-induced neurodegeneration. In contrast, formation of either sarkosyl-insoluble tau or paired helical filaments was not induced by Abeta42. Co-expression of Abeta42 and tau carrying the non-phosphorylatable Ser262Ala mutation did not cause neurodegeneration, suggesting that the Ser262 phosphorylation site is required for the pathogenic interaction between Abeta42 and tau. We have recently reported that the DNA damage-activated Checkpoint kinase 2 (Chk2) phosphorylates tau at Ser262 and enhances tau toxicity in a transgenic Drosophila model. We detected that expression of Chk2, as well as a number of genes involved in DNA repair pathways, was increased in the Abeta42 fly brains. The induction of a DNA repair response is protective against Abeta42 toxicity, since blocking the function of the tumor suppressor p53, a key transcription factor for the induction of DNA repair genes, in neurons exacerbated Abeta42-induced neuronal dysfunction. Our results demonstrate that tau phosphorylation at Ser262 is crucial for Abeta42-induced tau toxicity in vivo, and suggest a new model of AD progression in which activation of DNA repair pathways is protective against Abeta42 toxicity but may trigger tau phosphorylation and toxicity in AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/toxicidade , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Fragmentos de Peptídeos/toxicidade , Fosfosserina/metabolismo , Proteínas tau/metabolismo , Animais , Animais Geneticamente Modificados , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Quinase do Ponto de Checagem 2 , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Modelos Animais de Doenças , Olho/efeitos dos fármacos , Olho/patologia , Humanos , Atividade Motora/efeitos dos fármacos , Degeneração Neural/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
4.
Hum Mol Genet ; 19(10): 1930-8, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20159774

RESUMO

Hyperphosphorylation of the microtubule associated protein tau is detected in the brains of individuals with a range of neurodegenerative diseases including Alzheimer's disease (AD). An imbalance in phosphorylation and/or dephosphorylation of tau at disease-related sites has been suggested to initiate the abnormal metabolism and toxicity of tau in disease pathogenesis. However, the mechanisms underlying abnormal phosphorylation of tau in AD are not fully understood. Here, we show that the DNA damage-activated Checkpoint kinase 2 (Chk2) is a novel tau kinase and enhances tau toxicity in a transgenic Drosophila model. Overexpression of Drosophila Chk2 increases tau phosphorylation at Ser262 and enhances tau-induced neurodegeneration in transgenic flies expressing human tau. The non-phosphorylatable Ser262Ala mutation abolishes Chk2-induced enhancement of tau toxicity, suggesting that the Ser262 phosphorylation site is involved in the enhancement of tau toxicity by Chk2. In vitro kinase assays revealed that human Chk2 and a closely related checkpoint kinase 1 (Chk1) directly phosphorylate human tau at Ser262. We also demonstrate that Drosophila Chk2 does not modulate the activity of the fly homolog of microtubule affinity regulating kinase, which has been shown to be a physiological tau Ser262 kinase. Since accumulation of DNA damage has been detected in the brains of AD patients, our results suggest that the DNA damage-activated kinases Chk1 and Chk2 may be involved in tau phosphorylation and toxicity in the pathogenesis of AD.


Assuntos
Dano ao DNA , Degeneração Neural/enzimologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas tau/metabolismo , Animais , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Drosophila melanogaster/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Mutação/genética , Degeneração Neural/patologia , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteínas Recombinantes/metabolismo , Degeneração Retiniana/enzimologia , Degeneração Retiniana/patologia , Proteínas tau/toxicidade
5.
J Biol Chem ; 284(43): 29454-61, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19720829

RESUMO

Activation of c-Jun N-terminal kinase (JNK) signaling in neurons increases stress resistance and extends life span, in part through FOXO-mediated transcription in Drosophila. However, the JNK/FOXO target genes are unknown. Here, we identified Jafrac1, a Drosophila homolog of human Peroxiredoxin II (hPrxII), as a downstream effecter of JNK/FOXO signaling in neurons that enhances stress resistance and extends life span. We found that Jafrac1 was expressed in the adult brain and induced by paraquat, a reactive oxygen species-generating chemical. RNA interference-mediated neuronal knockdown of Jafrac1 enhanced, while neuronal overexpression of Jafrac1 and hPrxII suppressed, paraquat-induced lethality in flies. Neuronal expression of Jafrac1 also significantly reduced ROS levels, restored mitochondrial function, and attenuated JNK activation caused by paraquat. Activation of JNK/FOXO signaling in neurons increased the Jafrac1 expression level under both normal and oxidative stressed conditions. Moreover, neuronal knockdown of Jafrac1 shortened, while overexpression of Jafrac1 and hPrxII extended, the life span in flies. These results support the hypothesis that JNK/FOXO signaling extends life span via amelioration of oxidative damage and mitochondrial dysfunction in neurons.


Assuntos
Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Longevidade/fisiologia , Neurônios/enzimologia , Peroxidases/biossíntese , Transdução de Sinais/fisiologia , Animais , Encéfalo/enzimologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Herbicidas/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Longevidade/efeitos dos fármacos , Mitocôndrias/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Paraquat/farmacologia , Peroxidases/genética , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais/efeitos dos fármacos
6.
J Alzheimers Dis ; 15(4): 523-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19096154

RESUMO

Alzheimer's disease (AD) is the most common form of senile dementia, and a cure is desperately needed. The amyloid-beta42 (Abeta42) has been suggested to play a central role in the pathogenesis of AD. However, the mechanism by which Abeta42 causes AD remains unclear. To understand the pathogenesis and to develop therapeutic avenues, it is crucial to generate animal models of AD in genetically tractable organisms. Drosophila is a well-established model system for which abundant genetic tools are available. Moreover, its well organized brain permits the study of complex behaviors such as learning and memory. We have established transgenic flies that express human Abeta42 in the nervous system. These flies developed age-dependent short-term memory impairment and neurodegeneration. In this review, we will first describe transgenic Abeta42 fly models and discuss the unique features of this system compared to mouse AD models. Secondly, we will discuss the usage of the fly models to evaluate currently proposed therapeutic strategies. Thirdly, we will briefly review the results of a genetic screen for modifiers of Abeta42 toxicity in the fly model. Finally, we will discuss how to dissect the complex mechanisms of Abeta42 toxicity focusing on its aggregation propensity using the fly model system.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Amiloidose/genética , Drosophila/genética , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Amiloidose/patologia , Animais , Doenças Neurodegenerativas/genética
7.
J Biochem ; 137(2): 147-55, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15749829

RESUMO

The processing of beta-amyloid precursor protein (APP) generates the amyloid beta-protein (A beta) and contributes to the development of Alzheimer's disease (AD). Elucidating the regulation of APP processing will, therefore, contribute to the understanding of AD. Many APP-binding proteins, such as FE65, X11s, and JNK-interacting proteins (JIPs), bind the motif 681-GYENPTY-687 within the cytoplasmic domain of APP. Here we found that the human homologue of yeast amino-terminal acetyltransferase ARD1 (hARD1) interacts with a novel motif, 658-HGVVEVD-664, in the cytoplasmic domain of APP695. hARD1 expressed its acetyltransferase activity in association with a human subunit homologous to another yeast amino-acetyltransferase, hNAT1. Co-expression of hARD1 and hNAT1 in cells suppressed A beta40 secretion and the suppression correlated with their enzyme activity. These observations suggest that the association of APP with hARD1 and hNAT1 and/or their N-acetyltransferase activity contributes to the regulation of A beta generation.


Assuntos
Acetiltransferases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Arilamina N-Acetiltransferase/metabolismo , Acetiltransferases/análise , Acetiltransferases/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/análise , Precursor de Proteína beta-Amiloide/química , Arilamina N-Acetiltransferase/genética , Células Cultivadas , Citoplasma/química , Citoplasma/metabolismo , Humanos , Isoenzimas , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Técnicas do Sistema de Duplo-Híbrido
8.
Brain Struct Funct ; 214(2-3): 245-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19967412

RESUMO

Alzheimer's disease (AD) is the most common form of senile dementia. Aggregation of the amyloid-beta42 peptide (Abeta42) and tau proteins are pathological hallmarks in AD brains. Accumulating evidence suggests that Abeta42 plays a central role in the pathogenesis of AD, and tau acts downstream of Abeta42 as a modulator of the disease progression. Tau pathology is also observed in frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) and other related diseases, so called tauopathies. Although most cases are sporadic, genes associated with familial AD and FTDP-17 have been identified, which led to the development of transgenic animal models. Drosophila has been a powerful genetic model system used in many fields of biology, and recently emerges as a model for human neurodegenerative diseases. In this review, we will summarize key features of transgenic Drosophila models of AD and tauopathies and a number of insights into disease mechanisms as well as therapeutic implications gained from these models.


Assuntos
Doença de Alzheimer/genética , Animais Geneticamente Modificados/genética , Drosophila/genética , Tauopatias/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença/genética , Humanos , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Placa Amiloide/genética , Placa Amiloide/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/genética
9.
PLoS One ; 4(12): e8498, 2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-20041126

RESUMO

The cAMP-responsive transcription factor CREB functions in adipose tissue and liver to regulate glycogen and lipid metabolism in mammals. While Drosophila has a homolog of mammalian CREB, dCREB2, its role in energy metabolism is not fully understood. Using tissue-specific expression of a dominant-negative form of CREB (DN-CREB), we have examined the effect of blocking CREB activity in neurons and in the fat body, the primary energy storage depot with functions of adipose tissue and the liver in flies, on energy balance, stress resistance and feeding behavior. We found that disruption of CREB function in neurons reduced glycogen and lipid stores and increased sensitivity to starvation. Expression of DN-CREB in the fat body also reduced glycogen levels, while it did not affect starvation sensitivity, presumably due to increased lipid levels in these flies. Interestingly, blocking CREB activity in the fat body increased food intake. These flies did not show a significant change in overall body size, suggesting that disruption of CREB activity in the fat body caused an obese-like phenotype. Using a transgenic CRE-luciferase reporter, we further demonstrated that disruption of the adipokinetic hormone receptor, which is functionally related to mammalian glucagon and beta-adrenergic signaling, in the fat body reduced CRE-mediated transcription in flies. This study demonstrates that CREB activity in either neuronal or peripheral tissues regulates energy balance in Drosophila, and that the key signaling pathway regulating CREB activity in peripheral tissue is evolutionarily conserved.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Metabolismo Energético , Comportamento Alimentar , Neurônios/metabolismo , Transativadores/metabolismo , Envelhecimento/metabolismo , Animais , Drosophila melanogaster/genética , Corpo Adiposo/metabolismo , Técnicas de Silenciamento de Genes , Glicogênio/metabolismo , Metabolismo dos Lipídeos , Estresse Oxidativo , Receptores de Glucagon/metabolismo , Elementos de Resposta/genética , Inanição , Estresse Fisiológico , Transcrição Gênica
10.
PLoS One ; 4(12): e8310, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20016833

RESUMO

The amyloid-beta 42 (Abeta42) is thought to play a central role in the pathogenesis of Alzheimer's disease (AD). However, the molecular mechanisms by which Abeta42 induces neuronal dysfunction and degeneration remain elusive. Mitochondrial dysfunctions are implicated in AD brains. Whether mitochondrial dysfunctions are merely a consequence of AD pathology, or are early seminal events in AD pathogenesis remains to be determined. Here, we show that Abeta42 induces mitochondrial mislocalization, which contributes to Abeta42-induced neuronal dysfunction in a transgenic Drosophila model. In the Abeta42 fly brain, mitochondria were reduced in axons and dendrites, and accumulated in the somata without severe mitochondrial damage or neurodegeneration. In contrast, organization of microtubule or global axonal transport was not significantly altered at this stage. Abeta42-induced behavioral defects were exacerbated by genetic reductions in mitochondrial transport, and were modulated by cAMP levels and PKA activity. Levels of putative PKA substrate phosphoproteins were reduced in the Abeta42 fly brains. Importantly, perturbations in mitochondrial transport in neurons were sufficient to disrupt PKA signaling and induce late-onset behavioral deficits, suggesting a mechanism whereby mitochondrial mislocalization contributes to Abeta42-induced neuronal dysfunction. These results demonstrate that mislocalization of mitochondria underlies the pathogenic effects of Abeta42 in vivo.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Drosophila melanogaster/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Envelhecimento/patologia , Animais , Axônios/metabolismo , Comportamento Animal , Encéfalo/patologia , Encéfalo/ultraestrutura , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dendritos/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/genética , Humanos , Locomoção , Mitocôndrias/ultraestrutura , Neurônios/enzimologia , Fosfoproteínas/metabolismo , Transporte Proteico , Especificidade por Substrato
11.
PLoS One ; 3(2): e1703, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18301778

RESUMO

Aggregation of the amyloid-beta-42 (Abeta42) peptide in the brain parenchyma is a pathological hallmark of Alzheimer's disease (AD), and the prevention of Abeta aggregation has been proposed as a therapeutic intervention in AD. However, recent reports indicate that Abeta can form several different prefibrillar and fibrillar aggregates and that each aggregate may confer different pathogenic effects, suggesting that manipulation of Abeta42 aggregation may not only quantitatively but also qualitatively modify brain pathology. Here, we compare the pathogenicity of human Abeta42 mutants with differing tendencies to aggregate. We examined the aggregation-prone, EOFAD-related Arctic mutation (Abeta42Arc) and an artificial mutation (Abeta42art) that is known to suppress aggregation and toxicity of Abeta42 in vitro. In the Drosophila brain, Abeta42Arc formed more oligomers and deposits than did wild type Abeta42, while Abeta42art formed fewer oligomers and deposits. The severity of locomotor dysfunction and premature death positively correlated with the aggregation tendencies of Abeta peptides. Surprisingly, however, Abeta42art caused earlier onset of memory defects than Abeta42. More remarkably, each Abeta induced qualitatively different pathologies. Abeta42Arc caused greater neuron loss than did Abeta42, while Abeta42art flies showed the strongest neurite degeneration. This pattern of degeneration coincides with the distribution of Thioflavin S-stained Abeta aggregates: Abeta42Arc formed large deposits in the cell body, Abeta42art accumulated preferentially in the neurites, while Abeta42 accumulated in both locations. Our results demonstrate that manipulation of the aggregation propensity of Abeta42 does not simply change the level of toxicity, but can also result in qualitative shifts in the pathology induced in vivo.


Assuntos
Peptídeos beta-Amiloides/ultraestrutura , Degeneração Neural/etiologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/fisiologia , Animais , Encéfalo , Dimerização , Drosophila , Humanos , Memória , Atividade Motora , Mutação , Degeneração Neural/patologia , Neuritos/patologia , Neurônios/patologia
12.
J Biol Chem ; 283(27): 19066-76, 2008 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-18463098

RESUMO

The amyloid-beta42 (Abeta42) peptide has been suggested to play a causative role in Alzheimer disease (AD). Neprilysin (NEP) is one of the rate-limiting Abeta-degrading enzymes, and its enhancement ameliorates extracellular amyloid pathology, synaptic dysfunction, and memory defects in mouse models of Abeta amyloidosis. In addition to the extracellular Abeta, intraneuronal Abeta42 may contribute to AD pathogenesis. However, the protective effects of neuronal NEP expression on intraneuronal Abeta42 accumulation and neurodegeneration remain elusive. In contrast, sustained NEP activation may be detrimental because NEP can degrade many physiological peptides, but its consequences in the brain are not fully understood. Using transgenic Drosophila expressing human NEP and Abeta42, we demonstrated that NEP efficiently suppressed the formation of intraneuronal Abeta42 deposits and Abeta42-induced neuron loss. However, neuronal NEP overexpression reduced cAMP-responsive element-binding protein-mediated transcription, caused age-dependent axon degeneration, and shortened the life span of the flies. Interestingly, the mRNA levels of endogenous fly NEP genes and phosphoramidon-sensitive NEP activity declined during aging in fly brains, as observed in mammals. Taken together, these data suggest both the protective and detrimental effects of chronically high NEP activity in the brain. Down-regulation of NEP activity in aging brains may be an evolutionarily conserved phenomenon, which could predispose humans to developing late-onset AD.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Axônios/enzimologia , Regulação da Expressão Gênica , Neprilisina/biossíntese , Envelhecimento/genética , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Amiloidose/enzimologia , Amiloidose/genética , Animais , Animais Geneticamente Modificados , Evolução Biológica , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster , Ativação Enzimática/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Neprilisina/genética
13.
PLoS One ; 1: e51, 2006 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17183681

RESUMO

BACKGROUND: Amyloid-beta peptide species ending at positions 40 and 42 (Abeta40, Abeta42) are generated by the proteolytic processing of the Alzheimer's amyloid precursor protein (APP). Abeta peptides accumulate in the brain early in the course of Alzheimer's disease (AD), especially Abeta42. The cytoplasmic domain of APP regulates intracellular trafficking and metabolism of APP and its carboxyl-terminal fragments (CTFalpha, CTFbeta). The role of protein phosphorylation in general, and that of the phosphorylation state of APP at threonine-668 (Thr668) in particular, has been investigated in detail by several laboratories (including our own). Some investigators have recently proposed that the phosphorylation state of Thr668 plays a pivotal role in governing brain Abeta levels, prompting the current study. METHODOLOGY: In order to evaluate whether the phosphorylation state of Thr668 controlled brain Abeta levels, we studied the levels and subcellular distributions of holoAPP, sAPPalpha, sAPPbeta, CTFalpha, CTFbeta, Abeta40 and Abeta42 in brains from "knock-in" mice in which a non-phosphorylatable alanyl residue had been substituted at position 668, replacing the threonyl residue present in the wild-type protein. CONCLUSIONS: The levels and subcellular distributions of holoAPP, sAPPalpha, sAPPbeta, CTFalpha, CTFbeta, Abeta40 and Abeta42 in the brains of Thr668Ala mutant mice were identical to those observed in wild-type mice. These results indicate that, despite speculation to the contrary, the phosphorylation state of APP at Thr668 does not play an obvious role in governing the physiological levels of brain Abeta40 or Abeta42 in vivo.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Animais , Sequência de Bases , Encéfalo/anatomia & histologia , Encéfalo/crescimento & desenvolvimento , Primers do DNA/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Treonina/química
14.
Methods Enzymol ; 393: 302-15, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15817296

RESUMO

The cAMP response Element (CRE)-binding protein (CREB) is involved in many adaptive behaviors, including circadian rhythms. In order to assess CREB activity in vivo, we made transgenic flies carrying a CRE-luciferase reporter and showed that this reporter is CRE and dCREB2 responsive. dCREB2 is the Drosophila homolog of mammalian CREB?CREM. The transgenic luciferase activity cycles with a 24-h periodicity, suggesting that dCREB2 and period are somehow linked. The CRE-luciferase reporter is a useful monitor of circadian activity, and mutations can be found that affect its periodicity, baseline activity, or amplitude. Analysis of such mutations should reveal information about how particular genes affect the molecular machinery of circadian cycling and how different genes affect the activity of dCREB2.


Assuntos
Ritmo Circadiano/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Luciferases/metabolismo , Transativadores/metabolismo , Animais , Neurofibromina 1/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Circadianas Period
15.
Proc Natl Acad Sci U S A ; 102(29): 10261-6, 2005 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-16009936

RESUMO

Gene-specific expansion of polyglutamine-encoding CAG repeats can cause neurodegenerative disorders, including Huntington's disease. It is believed that part of the pathological effect of the expanded protein is due to transcriptional dysregulation. Using Drosophila as a model, we show that cAMP-response element-binding protein (CREB) is involved in expanded polyglutamine-induced toxicity. A mutation in the Drosophila homolog of CREB, dCREB2, enhances lethality due to polyglutamine peptides (polyQ), and an additional copy of dCREB2 partially rescues this lethality. Neuronal expression of expanded polyQ attenuates in vivo CRE-mediated transcription of a reporter gene. As reported previously, overexpression of heat-shock protein 70 (Hsp70) rescues polyglutamine-dependent lethality. However, it does not rescue CREB-mediated transcription. The protective effects of CREB and heat-shock protein 70 against polyQ are additive, suggesting that targeting multiple pathways may be effective for treatment of polyglutamine diseases.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP70/metabolismo , Doenças do Sistema Nervoso/genética , Peptídeos/metabolismo , Transativadores/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Drosophila , Luciferases , Doenças do Sistema Nervoso/metabolismo , Neurônios/metabolismo , Peptídeos/genética , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA