Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Alzheimers Dis ; 54(2): 679-90, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27567872

RESUMO

Mitochondrial dysfunction and axonal degeneration are early pathological features of Alzheimer's disease (AD)-affected brains. The underlying mechanisms and strategies to rescue it have not been well elucidated. Here, we evaluated axonal mitochondrial transport and function in AD subject-derived mitochondria. We analyzed mitochondrial transport and kinetics in human trans-mitochondrial "cybrid" (cytoplasmic hybrid) neuronal cells whose mitochondria were derived from platelets of patients with sporadic AD and compared these AD cybrid cell lines with cybrid cell lines whose mitochondria were derived from age-matched, cognitively normal subjects. Human AD cybrid cell lines, when induced to differentiate, developed stunted projections. Mitochondrial transport and function within neuronal processes/axons was altered in AD-derived mitochondria. Antioxidants reversed deficits in axonal mitochondrial transport and function. These findings suggest that antioxidants may be able to mitigate the consequences of AD-associated mitochondrial dysfunction. The present study provides evidence of the cause/effect of AD specific mitochondrial defects, which significantly enhances our understanding of the AD pathogenesis and exploring the effective therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/metabolismo , Antioxidantes/metabolismo , Diferenciação Celular/fisiologia , Mitocôndrias/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Transporte Biológico/fisiologia , Linhagem Celular , DNA Mitocondrial/metabolismo , Feminino , Humanos , Masculino , Mitocôndrias/patologia
2.
Hum Mol Genet ; 24(18): 5198-210, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26123488

RESUMO

Accumulation of amyloid-ß (Aß) in synaptic mitochondria is associated with mitochondrial and synaptic injury. The underlying mechanisms and strategies to eliminate Aß and rescue mitochondrial and synaptic defects remain elusive. Presequence protease (PreP), a mitochondrial peptidasome, is a novel mitochondrial Aß degrading enzyme. Here, we demonstrate for the first time that increased expression of active human PreP in cortical neurons attenuates Alzheimer disease's (AD)-like mitochondrial amyloid pathology and synaptic mitochondrial dysfunction, and suppresses mitochondrial oxidative stress. Notably, PreP-overexpressed AD mice show significant reduction in the production of proinflammatory mediators. Accordingly, increased neuronal PreP expression improves learning and memory and synaptic function in vivo AD mice, and alleviates Aß-mediated reduction of long-term potentiation (LTP). Our results provide in vivo evidence that PreP may play an important role in maintaining mitochondrial integrity and function by clearance and degradation of mitochondrial Aß along with the improvement in synaptic and behavioral function in AD mouse model. Thus, enhancing PreP activity/expression may be a new therapeutic avenue for treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Agregação Patológica de Proteínas/metabolismo , Serina Endopeptidases/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Comportamento Animal , Células Cultivadas , Cognição , Modelos Animais de Doenças , Expressão Gênica , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Proteólise , Serina Endopeptidases/genética
3.
J Alzheimers Dis ; 43(2): 451-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25096625

RESUMO

Diabetes is considered to be a risk factor in Alzheimer's disease (AD) pathogenesis. Although recent evidence indicates that diabetes exaggerates pathologic features of AD, the underlying mechanisms are not well understood. To determine whether mitochondrial perturbation is associated with the contribution of diabetes to AD progression, we characterized mouse models of streptozotocin (STZ)-induced type 1 diabetes and transgenic AD mouse models with diabetes. Brains from mice with STZ-induced diabetes revealed a significant increase of cyclophilin D (CypD) expression, reduced respiratory function, and decreased hippocampal long-term potentiation (LTP); these animals had impaired spatial learning and memory. Hyperglycemia exacerbated the upregulation of CypD, mitochondrial defects, synaptic injury, and cognitive dysfunction in the brains of transgenic AD mice overexpressing amyloid-ß as shown by decreased mitochondrial respiratory complex I and IV enzyme activity and greatly decreased mitochondrial respiratory rate. Concomitantly, hippocampal LTP reduction and spatial learning and memory decline, two early pathologic indicators of AD, were enhanced in the brains of diabetic AD mice. Our results suggest that the synergistic interaction between effects of diabetes and AD on mitochondria may be responsible for brain dysfunction that is in common in both diabetes and AD.


Assuntos
Doença de Alzheimer/complicações , Diabetes Mellitus Experimental/complicações , Deficiências da Aprendizagem/etiologia , Transtornos da Memória/etiologia , Mitocôndrias/patologia , Sinapses/patologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Potenciais Pós-Sinápticos Excitadores/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/patologia , Humanos , Técnicas In Vitro , Deficiências da Aprendizagem/patologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/fisiologia , Mutação/genética , Consumo de Oxigênio/fisiologia , Aprendizagem Espacial/efeitos dos fármacos , Aprendizagem Espacial/fisiologia , Fatores de Tempo
4.
Biochim Biophys Acta ; 1842(2): 220-31, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24252614

RESUMO

Mitochondrial dysfunction is an early pathological feature of Alzheimer's disease (AD). The underlying mechanisms and strategies to repair it remain unclear. Here, we demonstrate for the first time the direct consequences and potential mechanisms of mitochondrial functional defects associated with abnormal mitochondrial dynamics in AD. Using cytoplasmic hybrid (cybrid) neurons with incorporated platelet mitochondria from AD and age-matched non-AD human subjects into mitochondrial DNA (mtDNA)-depleted neuronal cells, we observed that AD cybrid cells had significant changes in morphology and function; such changes associate with altered expression and distribution of dynamin-like protein (DLP1) and mitofusin 2 (Mfn2). Treatment with antioxidant protects against AD mitochondria-induced extracellular signal-regulated kinase (ERK) activation and mitochondrial fission-fusion imbalances. Notably, inhibition of ERK activation not only attenuates aberrant mitochondrial morphology and function but also restores the mitochondrial fission and fusion balance. These effects suggest a role of oxidative stress-mediated ERK signal transduction in modulation of mitochondrial fission and fusion events. Further, blockade of the mitochondrial fission protein DLP1 by a genetic manipulation with a dominant negative DLP1 (DLP1(K38A)), its expression with siRNA-DLP1, or inhibition of mitochondrial division with mdivi-1 attenuates mitochondrial functional defects observed in AD cybrid cells. Our results provide new insights into mitochondrial dysfunction resulting from changes in the ERK-fission/fusion (DLP1) machinery and signaling pathway. The protective effect of mdivi-1 and inhibition of ERK signaling on maintenance of normal mitochondrial structure and function holds promise as a potential novel therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Células Híbridas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Antioxidantes/farmacologia , Dinaminas , Feminino , GTP Fosfo-Hidrolases/genética , Humanos , Células Híbridas/patologia , Immunoblotting , Masculino , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Dinâmica Mitocondrial/efeitos dos fármacos , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Mutação , Neurônios/metabolismo , Neurônios/patologia , Probucol/farmacologia , Quinazolinonas/farmacologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
Biochim Biophys Acta ; 1842(12 Pt A): 2517-27, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23507145

RESUMO

The coexistence of neuronal mitochondrial pathology and synaptic dysfunction is an early pathological feature of Alzheimer's disease (AD). Cyclophilin D (CypD), an integral part of mitochondrial permeability transition pore (mPTP), is involved in amyloid beta (Aß)-instigated mitochondrial dysfunction. Blockade of CypD prevents Aß-induced mitochondrial malfunction and the consequent cognitive impairments. Here, we showed the elimination of reactive oxygen species (ROS) by antioxidants probucol or superoxide dismutase (SOD)/catalase blocks Aß-mediated inactivation of protein kinase A (PKA)/cAMP regulatory-element-binding (CREB) signal transduction pathway and loss of synapse, suggesting the detrimental effects of oxidative stress on neuronal PKA/CREB activity. Notably, neurons lacking CypD significantly attenuate Aß-induced ROS. Consequently, CypD-deficient neurons are resistant to Aß-disrupted PKA/CREB signaling by increased PKA activity, phosphorylation of PKA catalytic subunit (PKA C), and CREB. In parallel, lack of CypD protects neurons from Aß-induced loss of synapses and synaptic dysfunction. Furthermore, compared to the mAPP mice, CypD-deficient mAPP mice reveal less inactivation of PKA-CREB activity and increased synaptic density, attenuate abnormalities in dendritic spine maturation, and improve spontaneous synaptic activity. These findings provide new insights into a mechanism in the crosstalk between the CypD-dependent mitochondrial oxidative stress and signaling cascade, leading to synaptic injury, functioning through the PKA/CREB signal transduction pathway.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclofilinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antioxidantes/farmacologia , Catalase/farmacologia , Células Cultivadas , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Immunoblotting , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Técnicas de Patch-Clamp , Probucol/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/farmacologia
6.
PLoS One ; 8(1): e54914, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382999

RESUMO

Normal axonal mitochondrial transport and function is essential for the maintenance of synaptic function. Abnormal mitochondrial motility and mitochondrial dysfunction within axons are critical for amyloid ß (Aß)-induced synaptic stress and the loss of synapses relevant to the pathogenesis of Alzheimer's disease (AD). However, the mechanisms controlling axonal mitochondrial function and transport alterations in AD remain elusive. Here, we report an unexplored role of cyclophilin D (CypD)-dependent mitochondrial permeability transition pore (mPTP) in Aß-impaired axonal mitochondrial trafficking. Depletion of CypD significantly protects axonal mitochondrial motility and dynamics from Aß toxicity as shown by increased axonal mitochondrial density and distribution and improved bidirectional transport of axonal mitochondria. Notably, blockade of mPTP by genetic deletion of CypD suppresses Aß-mediated activation of the p38 mitogen-activated protein kinase signaling pathway, reverses axonal mitochondrial abnormalities, improves synaptic function, and attenuates loss of synapse, suggesting a role of CypD-dependent signaling in Aß-induced alterations in axonal mitochondrial trafficking. The potential mechanisms of the protective effects of lacking CypD on Aß-induced abnormal mitochondrial transport in axon are increased axonal calcium buffer capability, diminished reactive oxygen species (ROS), and suppressing downstream signal transduction P38 activation. These findings provide new insights into CypD-dependent mitochondrial mPTP and signaling on mitochondrial trafficking in axons and synaptic degeneration in an environment enriched for Aß.


Assuntos
Doença de Alzheimer/patologia , Axônios/metabolismo , Axônios/patologia , Ciclofilinas/deficiência , Mitocôndrias/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Axônios/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Deleção de Genes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Fragmentos de Peptídeos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia
7.
Front Biosci (Schol Ed) ; 4(1): 240-50, 2012 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-22202057

RESUMO

RAGE, a receptor for advanced glycation endproducts, is an immunoglobulin-like cell surface receptor that is often described as a pattern recognition receptor due to the structural heterogeneity of its ligand. RAGE is an important cellular cofactor for amyloid beta-peptide (Abeta)-mediated cellular perturbation relevant to the pathogenesis of Alzheimer's disease (AD). The interaction of RAGE with Abeta in neurons, microglia, and vascular cells accelerates and amplifies deleterious effects on neuronal and synaptic function. RAGE-dependent signaling contributes to Abeta-mediated amyloid pathology and cognitive dysfunction observed in the AD mouse model. Blockade of RAGE significantly attenuates neuronal and synaptic injury. In this review, we summarize the role of RAGE in the pathogenesis of AD, specifically in Abeta-induced cellular perturbation.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Receptores Imunológicos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Microglia/metabolismo , Microglia/patologia , Terapia de Alvo Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Receptor para Produtos Finais de Glicação Avançada
8.
J Alzheimers Dis ; 27(1): 75-87, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21750375

RESUMO

Accumulation of amyloid-ß peptide (Aß), the neurotoxic peptide implicated in the pathogenesis of Alzheimer's disease (AD), has been shown in brain mitochondria of AD patients and of AD transgenic mouse models. The presence of Aß in mitochondria leads to free radical generation and neuronal stress. Recently, we identified the presequence protease, PreP, localized in the mitochondrial matrix in mammalian mitochondria as the novel mitochondrial Aß-degrading enzyme. In the present study, we examined PreP activity in the mitochondrial matrix of the human brain's temporal lobe, an area of the brain highly susceptible to Aß accumulation and reactive oxygen species (ROS) production. We found significantly lower hPreP activity in AD brains compared with non-AD age-matched controls. By contrast, in the cerebellum, a brain region typically spared from Aß accumulation, there was no significant difference in hPreP activity when comparing AD samples to non-AD controls. We also found significantly reduced PreP activity in the mitochondrial matrix of AD transgenic mouse brains (Tg mAßPP and Tg mAßPP/ABAD) when compared to non-transgenic aged-matched mice. Furthermore, mitochondrial fractions isolated from AD brains and Tg mAßPP mice had higher levels of 4-hydroxynonenal, an oxidative product, as compared with those from non-AD and nonTg mice. Accordingly, activity of cytochrome c oxidase was significantly reduced in the AD mitochondria. These findings suggest that decreased PreP proteolytic activity, possibly due to enhanced ROS production, contributes to Aß accumulation in mitochondria leading to the mitochondrial toxicity and neuronal death that is exacerbated in AD. Clearance of mitochondrial Aß by PreP may thus be of importance in the pathology of AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/ultraestrutura , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Serina Endopeptidases/metabolismo , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Aldeídos/metabolismo , Peptídeos beta-Amiloides/metabolismo , Análise de Variância , Animais , Encéfalo/patologia , Estudos de Casos e Controles , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mitocôndrias/patologia , Fragmentos de Peptídeos/metabolismo
9.
J Alzheimers Dis ; 20 Suppl 2: S569-78, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20463403

RESUMO

Mitochondrial dysfunction is an early feature of Alzheimer's disease (AD). Abnormalities in mitochondrial properties include impaired energy metabolism, defects in key respiratory enzyme activity/function, accumulation/generation of mitochondrial reactive oxygen species, and formation of membrane permeability transition pore. While the mechanisms underlying mitochondrial dysfunction remain incompletely understood, recent studies provide substantial evidence for the progressive accumulation of mitochondrial Abeta, which directly links to mitochondria-mediated toxicity. In this review, we describe recent studies addressing the following key questions: 1) Does Abeta accumulate in mitochondria of AD brain and AD mouse models? 2) How does Abeta gain access to the mitochondria? 3) If mitochondria are loaded with Abeta, do they develop similar evidence of dysfunction? 4) What are the mechanisms underlying mitochondrial Abeta-induced neuronal toxicity? and 5) What is the impact of interaction of mitochondrial Abeta with its binding partners (cyclophilin D and ABAD) on mitochondrial and neuronal properties/function in an Abeta milieu? The answers to these questions provide new insights into mechanisms of mitochondrial stress related to the pathogenesis of AD and information necessary for developing therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/metabolismo , Doença de Alzheimer/etiologia , Animais , Córtex Cerebral/patologia , Córtex Cerebral/ultraestrutura , Humanos , Doenças Mitocondriais/complicações
10.
FASEB J ; 24(5): 1583-92, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20056715

RESUMO

The immunity-related GTPase Irgm1, also called LRG-47, is known to regulate host resistance to intracellular pathogens through multiple mechanisms that include controlling the survival of T lymphocytes. Here, we address whether Irgm1 also plays a role in the pathogenesis of experimental autoimmune encephalitis (EAE). We find that Irgm1/LRG-47 is a significant factor in the progression of EAE and multiple sclerosis (MS). Expression of Irgm1 was robustly elevated in MS-affected lesions and in the central nervous system (CNS) of myelin basic protein (MBP)-induced EAE mice, especially in cells of lymphoid and mononuclear phagocyte origin. Homozygous Irgm1 null mice were resistant to MBP-induced EAE, and CD4(+) T cells in spleen and CNS of these mice displayed decreased proliferative capacity, increased apoptosis, and up-regulated interferon (IFN)-gamma induction. Therefore, Irgm1-induced survival of autoreactive CD4(+) T cells contributes significantly to the pathogenesis of EAE. Blockade of Irgm1 may be a potential therapeutic strategy for halting multiple sclerosis.


Assuntos
Apoptose/genética , Linfócitos T CD4-Positivos/imunologia , Encefalomielite Autoimune Experimental/genética , Proteínas de Ligação ao GTP/genética , Ativação Linfocitária/genética , Animais , Encefalomielite Autoimune Experimental/imunologia , Humanos , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Medula Espinal
11.
Nat Med ; 14(10): 1097-105, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18806802

RESUMO

Cyclophilin D (CypD, encoded by Ppif) is an integral part of the mitochondrial permeability transition pore, whose opening leads to cell death. Here we show that interaction of CypD with mitochondrial amyloid-beta protein (Abeta) potentiates mitochondrial, neuronal and synaptic stress. The CypD-deficient cortical mitochondria are resistant to Abeta- and Ca(2+)-induced mitochondrial swelling and permeability transition. Additionally, they have an increased calcium buffering capacity and generate fewer mitochondrial reactive oxygen species. Furthermore, the absence of CypD protects neurons from Abeta- and oxidative stress-induced cell death. Notably, CypD deficiency substantially improves learning and memory and synaptic function in an Alzheimer's disease mouse model and alleviates Abeta-mediated reduction of long-term potentiation. Thus, the CypD-mediated mitochondrial permeability transition pore is directly linked to the cellular and synaptic perturbations observed in the pathogenesis of Alzheimer's disease. Blockade of CypD may be a therapeutic strategy in Alzheimer's disease.


Assuntos
Doença de Alzheimer/prevenção & controle , Ciclofilinas/deficiência , Aprendizagem , Memória , Mitocôndrias/metabolismo , Neurônios/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose , Cálcio/metabolismo , Ciclofilinas/antagonistas & inibidores , Ciclofilinas/fisiologia , Modelos Animais de Doenças , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Espécies Reativas de Oxigênio/metabolismo , Sinapses/fisiologia
12.
J Neurosci ; 28(13): 3521-30, 2008 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-18367618

RESUMO

Soluble amyloid-beta (Abeta) peptide is likely to play a key role during early stages of Alzheimer's disease (AD) by perturbing synaptic function and cognitive processes. Receptor for advanced glycation end products (RAGE) has been identified as a receptor involved in Abeta-induced neuronal dysfunction. We investigated the role of neuronal RAGE in Abeta-induced synaptic dysfunction in the entorhinal cortex, an area of the brain important in memory processes that is affected early in AD. We found that soluble oligomeric Abeta peptide (Abeta42) blocked long-term potentiation (LTP), but did not affect long-term depression, paired-pulse facilitation, or basal synaptic transmission. In contrast, Abeta did not inhibit LTP in slices from RAGE-null mutant mice or in slices from wild-type mice treated with anti-RAGE IgG. Similarly, transgenic mice expressing a dominant-negative form of RAGE targeted to neurons showed normal LTP in the presence of Abeta, suggesting that neuronal RAGE functions as a signal transducer for Abeta-mediated LTP impairment. To investigate intracellular pathway transducing RAGE activation by Abeta, we used inhibitors of stress activated kinases. We found that inhibiting p38 mitogen-activated protein kinase (p38 MAPK), but not blocking c-Jun N-terminal kinase activation, was capable of maintaining LTP in Abeta-treated slices. Moreover, Abeta-mediated enhancement of p38 MAPK phosphorylation in cortical neurons was reduced by blocking antibodies to RAGE. Together, our results indicate that Abeta impairs LTP in the entorhinal cortex through neuronal RAGE-mediated activation of p38 MAPK.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Receptores Imunológicos/metabolismo , Sinapses/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Potenciais de Ação/fisiologia , Potenciais de Ação/efeitos da radiação , Animais , Animais Recém-Nascidos , Anticorpos/farmacologia , Células Cultivadas , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Córtex Entorrinal/citologia , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática/métodos , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/efeitos da radiação , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Sinapses/efeitos dos fármacos
13.
J Biol Chem ; 283(9): 5685-91, 2008 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-18167351

RESUMO

Alzheimer patients have increased levels of both the 42 amyloid-beta-peptide (Abeta) and the amyloid binding alcohol dehydrogenase (ABAD), which is an intracellular binding site for Abeta. The overexpression of Abeta and ABAD in transgenic mice has shown that the binding of Abeta to ABAD results in amplified neuronal stress and impairment of learning and memory. From a proteomic analysis of the brains from these animals, we have identified for the first time that the protein endophilin I increases in Alzheimer diseased brain. The increase in endophilin I levels in neurons is linked to an increase in the activation of the stress kinase c-Jun N-terminal kinase with the subsequent death of the neurons. We also demonstrate in living animals that the expression level of endophilin I is an indicator for the interaction of ABAD and Abeta as its expression levels return to normal if this interaction is perturbed. Therefore this identifies endophilin I as a new indicator of the progression of Alzheimer disease.


Assuntos
Aciltransferases/biossíntese , Álcool Desidrogenase/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Aciltransferases/genética , Álcool Desidrogenase/genética , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Animais , Biomarcadores/metabolismo , Encéfalo/patologia , Morte Celular/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Memória , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Ligação Proteica/genética
14.
Expert Rev Neurother ; 7(11): 1517-25, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17997700

RESUMO

Metabolic dysfunction is one of the early features in Alzheimer's disease (AD) affected brain. Amyloid-beta peptide (Abeta), a major peptide deposited in neuritic plaques, has been considered as an important initiating molecule in the pathogenesis of AD. However, the pathogenic role of Abeta remains to be determined. Here, we review current studies showing that progressive accumulation of Abeta occurs within the mitochondria of both transgenic mice overexpressing mutant Abeta peptide precursor protein and autopsied brains from AD patients. Interaction of Abeta with Abeta-binding alcohol dehydrogenase (ABAD), a short-chain alcohol dehydrogenase in the mitochondrial matrix, leads to mitochondrial dysfunction evidenced by increased reactive oxygen species generation, mitochondrial membrane permeability formation and caspase-3-like activity induction, and decreased activities of the Krebs cycle. These effects can be blocked by intracellular transduction of the ABAD decoy peptide. We hypothesize that Abeta-induced and mitochondria-dependent cytotoxic pathways might play an important role in AD pathogenesis and could be a potential therapeutic target.


Assuntos
Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/fisiologia , Proteínas Mitocondriais/efeitos adversos , Proteínas Mitocondriais/fisiologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Animais , Humanos , Proteínas Mitocondriais/toxicidade
15.
J Alzheimers Dis ; 12(2): 177-84, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17917162

RESUMO

As an important molecule in the pathogenesis of Alzheimer's disease (AD), amyloid-beta (Abeta) interferes with multiple aspects of mitochondrial function, including energy metabolism failure, production of reactive oxygen species (ROS) and permeability transition pore formation. Recent studies have demonstrated that Abeta progressively accumulates within mitochondrial matrix, providing a direct link to mitochondrial toxicity. Abeta-binding alcohol dehydrogenase (ABAD) is localized to the mitochondrial matrix and binds to mitochondrial Abeta. Interaction of ABAD with Abeta exaggerates Abeta-mediated mitochondrial and neuronal perturbation, leading to impaired synaptic function, and dysfunctional spatial learning/memory. Thus, blockade of ABAD/Abeta interaction may be a potential therapeutic strategy for AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Doenças Mitocondriais , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Metabolismo Energético , Humanos , Doenças Mitocondriais/epidemiologia , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/fisiopatologia
16.
Mol Cell Neurosci ; 35(2): 377-82, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17490890

RESUMO

Alzheimer's patients have increased levels of both the 42 beta amyloid-beta-peptide (Abeta) and amyloid binding alcohol dehydrogenase (ABAD) which is an intracellular binding site for Abeta. The over-expression of Abeta and ABAD in transgenic mice has shown that the binding of Abeta to ABAD results in exaggerating neuronal stress and impairment of learning and memory. From a proteomic analysis of the brains from these animals we identified that peroxiredoxin II levels increase in Alzheimer's diseased brain. This increase in peroxiredoxin II levels protects neurons against Abeta induced toxicity. We also demonstrate, for the first time in living animals, that the expression level of peroxiredoxin II is an indicator for the interaction of ABAD and Abeta as its expression levels return to normal if this interaction is perturbed. Therefore this indicates the possibility of reversing changes observed in Alzheimer's disease and that the Abeta-ABAD interaction is a suitable drug target.


Assuntos
Álcool Desidrogenase/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas de Choque Térmico/metabolismo , Peroxidases/metabolismo , Regulação para Cima/fisiologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Células Cultivadas , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional/métodos , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peroxirredoxinas , Ligação Proteica , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA