Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Glia ; 35(1): 72-9, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11424194

RESUMO

We have previously developed and characterized isolated microglia and astrocyte cultures from rapid (<4 h) brain autopsies of Alzheimer's disease (AD) and nondemented elderly control (ND) patients. In the present study, we evaluate the inflammatory repertoire of AD and ND microglia cultured from white matter (corpus callosum) and gray matter (superior frontal gyrus) with respect to three major proinflammatory cytokines, three chemokines, a classical pathway complement component, a scavenger cell growth factor, and a reactive nitrogen intermediate. Significant, dose-dependent increases in the production of pro-interleukin-1beta (pro-IL-1beta), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory peptide-1alpha (MIP-1alpha), IL-8, and macrophage colony-stimulating factor (M-CSF) were observed after exposure to pre-aggregated amyloid beta peptide (1-42) (Abeta1-42). Across constitutive and Abeta-stimulated conditions, secretion of complement component C1q, a reactive nitrogen intermediate, and M-CSF was significantly higher in AD compared with ND microglia. Taken together with previous in situ hybridization findings, these results demonstrate unequivocally that elderly human microglia provide a brain endogenous source for a wide range of inflammatory mediators.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Encéfalo/imunologia , Encefalite/imunologia , Microglia/imunologia , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/farmacologia , Biomarcadores/análise , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Células Cultivadas/metabolismo , Quimiocinas/biossíntese , Complemento C1q/biossíntese , Complemento C1q/efeitos dos fármacos , Corpo Caloso/imunologia , Corpo Caloso/metabolismo , Corpo Caloso/fisiopatologia , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Encefalite/metabolismo , Encefalite/fisiopatologia , Feminino , Lobo Frontal/imunologia , Lobo Frontal/metabolismo , Lobo Frontal/fisiopatologia , Humanos , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Nitritos/metabolismo , Fragmentos de Peptídeos/farmacologia
2.
J Neurosci ; 20(20): 7510-6, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11027208

RESUMO

The expression of alpha(1)-antichymotrypsin (ACT) is significantly enhanced in affected brain regions in Alzheimer's disease. This serine proteinase inhibitor specifically colocalizes with filamentous beta-amyloid deposits and recently has been shown to influence both formation and destabilization of beta-amyloid fibrils. In the brain, ACT is expressed in astrocytes, and interleukin-1 (IL-1), tumor necrosis factor alpha (TNF), oncostatin M (OSM), and IL-6/soluble IL-6 receptor complexes control synthesis of this inhibitor. Here, we characterize a molecular mechanism responsible for both IL-1 and TNF-induced expression of ACT gene in astrocytes. We identify the 5' distal IL-1/TNF-responsive enhancer of the ACT gene located 13 kb upstream of the transcription start site. This 413-bp-long enhancer contains three elements, two of which bind nuclear factor kB (NF-kB) and one that binds activating protein 1 (AP-1). All of these elements contribute to the full responsiveness of the ACT gene to both cytokines, as determined by deletion and mutational analysis. The 5' NF-kB high-affinity binding site and AP-1 element contribute most to the enhancement of gene transcription in response to TNF and IL-1. In addition, we demonstrate that the 5' untranslated region of the ACT mRNA does not contribute to cytokine-mediated activation. Finally, we find that overexpression of the NF-kB inhibitor (IkB) totally inhibits any activation mediated by the newly identified IL-1/TNF enhancer of the ACT gene.


Assuntos
Astrócitos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , alfa 1-Antiquimotripsina/metabolismo , Regiões 5' não Traduzidas/genética , Doença de Alzheimer/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Sítios de Ligação/genética , Células Cultivadas , Elementos Facilitadores Genéticos , Humanos , Interleucina-1/farmacologia , Mutagênese Sítio-Dirigida , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima , alfa 1-Antiquimotripsina/genética
3.
J Neurochem ; 75(5): 2172-7, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11032907

RESUMO

Tissue plasminogen (plgn) activator (tPA) modulates neuronal death in models of stroke, excitotoxicity, and oxidative stress. Amyloid-beta (Abeta) appears central to Alzheimer's disease and is neurotoxic to neurons in vitro. Here, we evaluate tPA effects on Abeta toxicity. We report that tPA alone had no effect on Abeta toxicity. However, in combination with plgn, tPA reduced Abeta toxicity in a robust fashion. Moreover, the combined tPA and plgn treatment markedly inhibited Abeta accumulation. The addition of phenylmethylsulfonyl fluoride, a serine protease inhibitor, to a sample of tPA, plgn, and Abeta resulted in a marked reduction of Abeta degradation. We interpret the actions of tPA and plgn within the context of the ability of plasmin to degrade Abeta.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Plasminogênio/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Fibrinolisina/antagonistas & inibidores , Fibrinolisina/metabolismo , Microscopia Eletrônica , Neurônios/ultraestrutura , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fluoreto de Fenilmetilsulfonil/farmacologia , Plasminogênio/farmacologia , Ratos , Ativador de Plasminogênio Tecidual/farmacologia
4.
Biochemistry ; 39(35): 10831-9, 2000 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-10978169

RESUMO

The progressive aggregation and deposition of amyloid beta-protein (Abeta) in brain regions subserving memory and cognition is an early and invariant feature of Alzheimer's disease, the most common cause of cognitive failure in aged humans. Inhibiting Abeta aggregation is therapeutically attractive because this process is believed to be an exclusively pathological event. Whereas many studies have examined the aggregation of synthetic Abeta peptides under nonphysiological conditions and concentrations, we have detected and characterized the oligomerization of naturally secreted Abeta at nanomolar levels in cultures of APP-expressing CHO cells [Podlisny, M. B., Ostaszewski, B. L., Squazzo, S. L., Koo, E. H., Rydell, R. E., Teplow, D. B., and Selkoe, D. J. (1995) J. Biol. Chem. 270, 9564-9570 (1); Podlisny, M. B., Walsh, D. M., Amarante, P., Ostaszewski, B. L., Stimson, E. R., Maggio, J. E., Teplow, D. B., and Selkoe, D. J. (1998) Biochemistry 37, 3602-3611 (2)]. To determine whether similar species occur in vivo, we probed samples of human cerebrospinal fluid (CSF) and detected SDS-stable dimers of Abeta in some subjects. Incubation of CSF or of CHO conditioned medium at 37 degrees C did not lead to new oligomer formation. This inability to induce oligomers extracellularly as well as the detection of oligomers in cell medium very early during the course of pulse-chase experiments suggested that natural Abeta oligomers might first form intracellularly. We therefore searched for and detected intracellular Abeta oligomers, principally dimers, in primary human neurons and in neuronal and nonneural cell lines. These dimers arose intracellularly rather than being derived from the medium by reuptake. The dimers were particularly detectable in neural cells: the ratio of intracellular to extracellular oligomers was much higher in brain-derived than nonbrain cells. We conclude that the pathogenically critical process of Abeta oligomerization begins intraneuronally.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Líquido Intracelular/metabolismo , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Temperatura Corporal , Células CHO , Sistema Livre de Células/metabolismo , Células Cultivadas , Córtex Cerebral/química , Cricetinae , Meios de Cultivo Condicionados/metabolismo , Dimerização , Espaço Extracelular/metabolismo , Feto , Humanos , Peso Molecular , Neurônios/química , Neurônios/metabolismo , Dodecilsulfato de Sódio/metabolismo , Transfecção
5.
Neurobiol Aging ; 21(3): 383-421, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10858586

RESUMO

Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.


Assuntos
Doença de Alzheimer/patologia , Inflamação/patologia , Encéfalo/patologia , Humanos
6.
J Neurosci ; 20(11): 3937-46, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10818128

RESUMO

Amyloid-beta (Abeta) appears critical to Alzheimer's disease. To clarify possible mechanisms of Abeta action, we have quantified Abeta-induced gene expression in vitro by using Abeta-treated primary cortical neuronal cultures and in vivo by using mice transgenic for the Abeta precursor (AbetaP). Here, we report that aggregated, but not nonaggregated, Abeta increases the level of the mRNAs encoding tissue plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). Moreover, tPA and uPA were also upregulated in aged AbetaP overexpressing mice. Because others have reported that Abeta aggregates can substitute for fibrin aggregates in activating tPA post-translationally, the result of tPA induction by Abeta would be cleavage of plasminogen to the active protease plasmin. To gain insights into the possible actions of plasmin, we evaluated the hypotheses that tPA and plasmin may mediate Abeta in vitro toxicity or, alternatively, that plasmin activation may lead to Abeta degradation. In evaluating these conflicting hypotheses, we found that purified plasmin degrades Abeta with physiologically relevant efficiency, i.e., approximately 1/10th the rate of plasmin on fibrin. Mass spectral analyses show that plasmin cleaves Abeta at multiple sites. Electron microscopy confirms indirect assays suggesting that plasmin degrades Abeta fibrils. Moreover, exogenously added plasmin blocks Abeta neurotoxicity. In summation, we interpret these results as consistent with the possibility that the plasmin pathway is induced by aggregated Abeta, which can lead to Abeta degradation and inhibition of Abeta actions.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Fibrinolisina/fisiologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/efeitos dos fármacos , Animais , Biotransformação , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corantes , Primers do DNA , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Fibrinolisina/efeitos dos fármacos , Fibrinolisina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Ativador de Plasminogênio Tecidual/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
7.
J Neurochem ; 73(6): 2609-12, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10582624

RESUMO

The role of gene expression in neuronal apoptosis may be cell- and apoptotic stimulus-specific. Previously, we and others showed that amyloid beta (Abeta)-induced neuronal apoptosis is accompanied by c-jun induction. Moreover, c-Jun contributes to neuronal death in several apoptosis paradigms involving survival factor withdrawal. To evaluate the role of c-Jun in Abeta toxicity, we compared Abeta-induced apoptosis in neurons from murine fetal littermates that were deficient or wild-type with respect to c-Jun. We report that neurons deficient for c-jun are relatively resistant to Abeta toxicity, suggesting that c-Jun contributes to apoptosis in this model. When changes in gene expression were quantified in neurons treated in parallel, we found that Abeta treatment surprisingly led to an apparent activation of the c-jun promoter in both the c-jun-deficient and wild-type neurons, suggesting that c-Jun is not necessary for activation of the c-jun promoter. Indeed, several genes induced by Abeta in wild-type neurons were also induced in c-jun-deficient neurons, including c-fos, fosB, ngfi-B, and ikappaB. In summary, these results indicate that c-Jun contributes to Abeta-induced neuronal death but that c-Jun is not necessary for c-jun induction.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Apoptose/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Genes Precoces , Genes jun , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-jun/biossíntese , Proteínas Proto-Oncogênicas c-jun/deficiência
8.
Am J Pathol ; 155(3): 853-62, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10487842

RESUMO

We have characterized amyloid beta peptide (Abeta) concentration, Abeta deposition, paired helical filament formation, cerebrovascular amyloid angiopathy, apolipoprotein E (ApoE) allotype, and synaptophysin concentration in entorhinal cortex and superior frontal gyrus of normal elderly control (ND) patients, Alzheimer's disease (AD) patients, and high pathology control (HPC) patients who meet pathological criteria for AD but show no synapse loss or overt antemortem symptoms of dementia. The measures of Abeta deposition, Abeta-immunoreactive plaques with and without cores, thioflavin histofluorescent plaques, and concentrations of insoluble Abeta, failed to distinguish HPC from AD patients and were poor correlates of synaptic change. By contrast, concentrations of soluble Abeta clearly distinguished HPC from AD patients and were a strong inverse correlate of synapse loss. Further investigation revealed that Abeta40, whether in soluble or insoluble form, was a particularly useful measure for classifying ND, HPC, and AD patients compared with Abeta42. Abeta40 is known to be elevated in cerebrovascular amyloid deposits, and Abeta40 (but not Abeta42) levels, cerebrovascular amyloid angiopathy, and ApoE4 allele frequency were all highly correlated with each other. Although paired helical filaments in the form of neurofibrillary tangles or a penumbra of neurites surrounding amyloid cores also distinguished HPC from AD patients, they were less robust predictors of synapse change compared with soluble Abeta, particularly soluble Abeta40. Previous experiments attempting to relate Abeta deposition to the neurodegeneration that underlies AD dementia may have failed because they assayed the classical, visible forms of the molecule, insoluble neuropil plaques, rather than the soluble, unseen forms of the molecule.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Sinapses/patologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Apolipoproteínas E/genética , Encéfalo/metabolismo , Encéfalo/patologia , Angiopatia Amiloide Cerebral/patologia , Diagnóstico Diferencial , Feminino , Frequência do Gene , Humanos , Imuno-Histoquímica , Masculino , Emaranhados Neurofibrilares/patologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/patologia , Valor Preditivo dos Testes
9.
J Neurochem ; 71(2): 506-16, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9681440

RESUMO

Amylin forms large beta-pleated neurotoxic oligomers but shows only 38% sequence similarity to A beta. As patterns of gene expression during neuronal apoptosis appear stimulus and cell type specific, we compared the pattern of amylin-induced gene expression in rat cortical neurons with that shown previously to be induced by A beta in order to evaluate whether these two peptides with different primary but similar secondary structure induce apoptosis similarly. Morphologic and quantitative measures of cell death show widespread apoptotic death after amylin treatment. Amylin treatment results in time- and concentration-dependent inductions of oxidative stress genes, such as cox-2 and IkappaB-alpha. "Apoptotic" genes are also induced in a time- and concentration-dependent manner, including c-jun, junB, c-fos, and fosB, followed temporally by a gene known to be modulated by these transcription factors, i.e., transin. In situ hybridization analyses show that c-fos expression is restricted largely to neurons with condensed chromatin, a hallmark of apoptosis. As these genes are not induced in all models of apoptosis, that amylin-induced neuronal death is genetically similar to that of A beta suggests that these peptides may be neurotoxic through a common mechanism.


Assuntos
Amiloide/toxicidade , Apoptose/efeitos dos fármacos , Neurônios/citologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hibridização In Situ , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/metabolismo , Ratos
10.
J Biol Chem ; 273(7): 4112-8, 1998 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-9461605

RESUMO

alpha1-Antichymotrypsin (ACT) is an acute phase protein expressed in the brain which specifically colocalizes with amyloid-beta during Alzheimer's disease. We analyzed ACT synthesis in cultured human cortical astrocytes in response to various cytokines and growth factors. Oncostatin M (OSM) and interleukin (IL)-1beta were potent stimulators of ACT mRNA expression, whereas tumor necrosis factor-alpha had modest activity, and IL-6 and leukemia inhibitory factor (LIF) were ineffective. The finding that OSM, but not LIF or IL-6, stimulated ACT expression suggests that human astrocytes express a "specific" OSM receptor, but not IL-6 or LIF receptors. However, cotreatment of human astrocytes with soluble IL-6 receptor (sIL-6R).IL-6 complex did result in potent stimulation of ACT expression. When the human ACT gene was cloned, two elements binding STAT1 and STAT3 (signal transducer and activator of transcription) in response to OSM or IL-6.sIL-6R complexes could be identified and characterized. Taken together, these findings indicate that OSM or IL-6.sIL-6 complexes may regulate ACT expression in human astrocytes and thus directly or indirectly contribute to the pathogenesis of Alzheimer's disease.


Assuntos
Astrócitos/efeitos dos fármacos , Quimotripsina/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-6/farmacologia , Peptídeos/farmacologia , Receptores de Interleucina-6/metabolismo , Inibidores de Serina Proteinase/biossíntese , Proteínas de Fase Aguda/biossíntese , Doença de Alzheimer/etiologia , Sequência de Bases , Encéfalo/metabolismo , Clonagem Molecular , Citocinas/farmacologia , Proteínas de Ligação a DNA/análise , Humanos , Dados de Sequência Molecular , Oncostatina M , Regiões Promotoras Genéticas/genética , RNA Mensageiro/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
11.
J Neurosci ; 17(20): 7736-45, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9315895

RESUMO

To gain a molecular understanding of neuronal responses to amyloid-beta peptide (Abeta), we have analyzed the effects of Abeta treatment on neuronal gene expression in vitro by quantitative reverse transcription-PCR and in situ hybridization. Treatment of cultured rat cortical neurons with Abeta1-40 results in a widespread apoptotic neuronal death. Associated with death is an induction of several members of the immediate early gene family. Specifically, we (1) report the time-dependent and robust induction of c-jun, junB, c-fos, and fosB, as well as transin, which is induced by c-Jun/c-Fos heterodimers and encodes an extracellular matrix protease; these gene inductions appear to be selective because other Jun and Fos family members, i.e., junD and fra-1, are induced only marginally; (2) show that the c-jun induction is widespread, whereas c-fos expression is restricted to a subset of neurons, typically those with condensed chromatin, which is a hallmark of apoptosis; (3) correlate gene induction and neuronal death by showing that each has a similar dose-response to Abeta; and (4) demonstrate that both cell death and immediate early gene induction are dependent on Abeta aggregation state. This overall gene expression pattern during this "physiologically inappropriate" apoptotic stimulus is markedly similar to the pattern we previously identified after a "physiologically appropriate" stimulus, i.e., the NGF deprivation-induced death of sympathetic neurons. Hence, the parallels identified here further our understanding of the genetic alterations that may lead neurons to apoptosis in response to markedly different insults.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Apoptose/fisiologia , Córtex Cerebral/citologia , Regulação da Expressão Gênica , Neurônios/fisiologia , Peptídeos beta-Amiloides/intoxicação , Animais , Cromatina/metabolismo , Dosagem de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Precoces , Neurônios/efeitos dos fármacos , Ratos/embriologia , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional
12.
J Neurochem ; 67(5): 1882-96, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8863493

RESUMO

Proteolytic cleavage of beta-amyloid precursor protein (beta APP) by alpha-secretase results in release of one secreted form (sAPP) of APP (sAPP alpha), whereas cleavage by beta-secretase releases a C-terminally truncated sAPP (sAPP beta) plus amyloid beta-peptide (A beta). beta APP mutations linked to some inherited forms of Alzheimer's disease may alter its processing such that levels of sAPP alpha are reduced and levels of sAPP beta increased. sAPP alpha s may play important roles in neuronal plasticity and survival, whereas A beta can be neurotoxic. sAPP alpha was approximately 100-fold more potent than sAPP beta in protecting hippocampal neurons against excitotoxicity, A beta toxicity, and glucose deprivation. Whole-cell patch clamp and calcium imaging analyses showed that sAPP beta was less effective than sAPP alpha in suppressing synaptic activity, activating K+ channels, and attenuating calcium responses to glutamate. Using various truncated sAPP alpha and sAPP beta APP695 products generated by eukaryotic and prokaryotic expression systems, and synthetic sAPP peptides, the activity of sAPP alpha was localized to amino acids 591-612 at the C-terminus. Heparinases greatly reduced the actions of sAPP alpha s, indicating a role for a heparin-binding domain at the C-terminus of sAPP alpha in receptor activation. These findings indicate that alternative processing of beta APP has profound effects on the bioactivity of the resultant sAPP products and suggest that reduced levels of sAPP alpha could contribute to neuronal degeneration in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidases/metabolismo , Heparina/metabolismo , Hipocampo/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/química , Animais , Ácido Aspártico Endopeptidases , Sequência de Bases , Sítios de Ligação , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Clonagem Molecular , Escherichia coli , Feto , Ácido Glutâmico/farmacologia , Glutationa Transferase , Heparina Liase , Humanos , Rim , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neurônios/citologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Polissacarídeo-Liases/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Ratos , Receptores de AMPA/fisiologia , Receptores de Ácido Caínico/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo
13.
J Neurochem ; 67(4): 1595-606, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8858944

RESUMO

H2O2 and free radical-mediated oxidative stresses have been implicated in mediating amyloid beta (1-40) [A beta (1-40)] neurotoxicity to cultured neurons. In this study, we confirm that addition of the H2O2-scavenging enzyme catalase protects neurons in culture against A beta-mediated toxicity; however, it does so by a mechanism that does not involve its ability to scavenge H2O2. A beta-mediated elevation in intracellular H2O2 production is suppressed by addition of a potent H2O2 scavenger without any significant neuroprotection. Three intracellular biochemical markers of H2O2-mediated oxidative stress were unchanged by A beta treatment: (a) glyceraldehyde-3-phosphate dehydrogenase activity, (b) hexose monophosphate shunt activity, and (c) glucose oxidation via the tricarboxylic acid cycle. lonspray mass spectra of A beta in the incubation medium indicated that A beta itself is an unlikely source of reactive oxygen species. In this study we demonstrate that intracellular ATP concentration is compromised during the first 24-h exposure of neurons to A beta. Our results challenge a pivotal role for H2O2 generation in mediating A beta toxicity, and we suggest that impairment of energy homeostasis may be a more significant early factor in the neurodegenerative process.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Córtex Cerebral/metabolismo , Glucose/metabolismo , Peróxido de Hidrogênio/metabolismo , Neurônios/metabolismo , Oxazinas , Estresse Oxidativo , Fragmentos de Peptídeos/farmacologia , Xantenos , Trifosfato de Adenosina/metabolismo , Análise de Variância , Animais , Benzotiazóis , Catalase/antagonistas & inibidores , Sobrevivência Celular , Células Cultivadas , Corantes , Feto , Fluoresceínas , Sequestradores de Radicais Livres , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Glicólise/efeitos dos fármacos , Glioxilatos/farmacologia , L-Lactato Desidrogenase , Neurônios/efeitos dos fármacos , Neurotoxinas/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Tiazóis
15.
Proc Natl Acad Sci U S A ; 92(23): 10738-41, 1995 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-7479875

RESUMO

Neurodegenerative processes in Alzheimer disease (AD) are thought to be driven in part by the deposition of amyloid beta (A beta), a 39- to 43-amino acid peptide product resulting from an alternative cleavage of amyloid precursor protein. Recent descriptions of in vitro neurotoxic effects of A beta support this hypothesis and suggest toxicity might be mediated by A beta-induced neuronal calcium disregulation. In addition, it has been reported that "aging" A beta results in increased toxic potency due to peptide aggregation and formation of a beta-sheet secondary structure. In addition, A beta might also promote neuropathology indirectly by activating immune/inflammatory pathways in affected areas of the brain (e.g., cortex and hippocampus). Here we report that A beta can modulate cytokine secretion [interleukins 6 and 8 (IL-6 and IL-8)] from human astrocytoma cells (U-373 MG). Freshly prepared and aged A beta modestly stimulated IL-6 and IL-8 secretion from U-373 MG cells. However, in the presence of interleukin-1 beta (IL-1 beta), aged, but not fresh, A beta markedly potentiated (3- to 8-fold) cytokine release. In contrast, aged A beta did not potentiate substance P (NK-1)- or histamine (H1)-stimulated cytokine production. Further studies showed that IL-1 beta-induced cytokine release was potentiated by A beta-(25-35), while A beta-(1-16) was inactive. Calcium disregulation may be responsible for the effects of A beta on cytokine production, since the calcium ionophore A23187 similarly potentiated IL-1 beta-induced cytokine secretion and EGTA treatment blocked either A beta or A23187 activity. Thus, chronic neurodegeneration in AD-affected brain regions may be mediated in part by the ability of A beta to exacerbate inflammatory pathways in a conformation-dependent manner.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Astrócitos/metabolismo , Interleucina-1/farmacologia , Interleucinas/metabolismo , Fragmentos de Peptídeos/farmacologia , Doença de Alzheimer/etiologia , Astrócitos/efeitos dos fármacos , Astrocitoma , Cálcio/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Encefalite/etiologia , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Conformação Proteica , Células Tumorais Cultivadas
17.
J Neurochem ; 63(2): 781-4, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8035204

RESUMO

The beta-amyloid precursor protein (beta APP) is the source of the amyloid beta-peptide that accumulates in the brain in Alzheimer's disease. A major processing pathway for beta APP involves an enzymatic cleavage within the amyloid beta-peptide sequence that liberates secreted forms of beta APP (APPss) into the extracellular milieu. We now report that postischemic administration of these APPss intracerebroventricularly protects neurons in the CA1 region of rat hippocampus against ischemic injury. Treatment with APPs695 or APPs751 resulted in increased neuronal survival, and the surviving cells were functional as demonstrated by their ability to synthesize protein. These data provide direct evidence for a neuroprotective action of APPss in vivo.


Assuntos
Precursor de Proteína beta-Amiloide/farmacologia , Encéfalo/patologia , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/prevenção & controle , Neurônios/patologia , Precursor de Proteína beta-Amiloide/administração & dosagem , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Linhagem Celular , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Injeções Intraventriculares , Rim , Masculino , Neurônios/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Ratos , Ratos Wistar , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/farmacologia , Técnicas Estereotáxicas , Transfecção
18.
Mol Pharmacol ; 45(3): 373-9, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8145724

RESUMO

Amyloid beta peptide (A beta), the major protein constituent of senile plaques in patients with Alzheimer's disease, is believed to facilitate the progressive neurodegeneration that occurs in the latter stages of this disease. Early attempts to characterize the structure-activity relationship of A beta toxicity in vitro were compromised by the inability to reproducibly elicit A beta-dependent toxicity across different lots of chemically equivalent peptides. In this study we used CD spectroscopy to demonstrate that A beta secondary structure is an important determinant of A beta toxicity. Solubilized A beta was maximally toxic when the peptide adopted a beta-sheet conformation. Three of the four A beta lots tested had a random coil conformation and were weakly toxic or inactive, whereas the single A beta lot exhibiting toxic activity at low peptide concentrations had significant beta-sheet structure. Incubation of the weakly toxic A beta lots in aqueous stock solutions for several days before use induced a time-dependent conformational transition from random coil to beta-sheet and increased A beta toxicity in three different toxicity assays. Furthermore, the secondary structure of preincubated A beta was dependent upon peptide concentration and pH, so that beta-sheet structures were attenuated when peptide solutions were diluted or buffered at neutral and basic pH. Our data could explain some of the variable toxic activity that has been associated with A beta in the past and provide additional support for the hypothesis that A beta can have a causal role in the molecular neuropathology of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/química , Neurônios/efeitos dos fármacos , Estrutura Secundária de Proteína , Peptídeos beta-Amiloides/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dicroísmo Circular , Concentração de Íons de Hidrogênio , Neurônios/química , Neurônios/citologia , Ratos
19.
Trends Neurosci ; 16(10): 409-14, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7504356

RESUMO

Recent findings link altered processing of beta-amyloid precursor protein (beta APP) to disruption of neuronal Ca2+ homeostasis and an excitotoxic mechanism of cell death in Alzheimer's disease. A major pathway of beta APP metabolism results in the release of secreted forms of beta APP, APPss. These secreted forms are released in response to electrical activity and can modulate neuronal responses to glutamate, suggesting roles in developmental and synaptic plasticity. beta APP is upregulated in response to neural injury and APPss can protect neurons against excitotoxic or ischemic insults by stabilizing the intracellular Ca2+ concentration [Ca2+]i. An alternative beta APP processing pathway liberates intact beta-amyloid peptide, which can form aggregates that disrupt Ca2+ homeostasis and render neurons vulnerable to metabolic or excitotoxic insults. Genetic abnormalities (e.g. certain beta APP mutations or Down syndrome) and age-related changes in brain metabolism (e.g. reduced energy availability or increased oxidative stress) may favor accumulation of [Ca2+]i-destabilizing beta-amyloid peptide and diminish the release of [Ca2+]i-stabilizing, neuroprotective APPss.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cálcio/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/fisiologia , Homeostase , Humanos , Processamento de Proteína Pós-Traducional
20.
Ann N Y Acad Sci ; 695: 158-64, 1993 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-8239276

RESUMO

The suspected involvement of the beta-amyloid precursor protein (beta APP) in the etiology of Alzheimer's disease (AD) has been strengthened by recent genetic evidence, but pursuit of the mechanisms involved will initially require basic cell biology approaches. Several studies have concentrated on toxic activities of beta-amyloid peptide (beta AP) itself, illuminating its contributions to excitotoxicity and calcium-mediated degeneration in general. We now know that generation of beta AP from beta APP also compromises the production of an important set of trophic factors: the secreted forms of beta APP (APPS), which may act--ironically--by conferring protection from calcium-mediated insults. Therefore, conditions which contribute to the formation of beta AP (possibly including ischemia) not only produce an agent which exacerbates calcium-mediated cell death, but also reduce the levels of one of the few factors able to rescue calcium homeostasis. The implications of these postulates and their relationship to the process of aging are discussed.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cálcio/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Homeostase , Humanos , Modelos Biológicos , Mutação , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...