Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Phys Rev Lett ; 114(5): 054801, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25699448

RESUMO

The Linac Coherent Light Source has added a self-seeding capability to the soft x-ray range using a grating monochromator system. We report the demonstration of soft x-ray self-seeding with a measured resolving power of 2000-5000, wavelength stability of 10(-4), and an increase in peak brightness by a factor of 2-5 across the photon energy range of 500-1000 eV. By avoiding the need for a monochromator at the experimental station, the self-seeded beam can deliver as much as 50-fold higher brightness to users.

2.
Nat Med ; 4(1): 97-100, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9427614

RESUMO

Genetic causes of Alzheimer's disease (AD) include mutations in the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) genes. The mutant APP(K670N,M671L) transgenic line, Tg2576, shows markedly elevated amyloid beta-protein (A beta) levels at an early age and, by 9-12 months, develops extracellular AD-type A beta deposits in the cortex and hippocampus. Mutant PS1 transgenic mice do not show abnormal pathology, but do display subtly elevated levels of the highly amyloidogenic 42- or 43-amino acid peptide A beta42(43). Here we demonstrate that the doubly transgenic progeny from a cross between line Tg2576 and a mutant PS1M146L transgenic line develop large numbers of fibrillar A beta deposits in cerebral cortex and hippocampus far earlier than their singly transgenic Tg2576 littermates. In the period preceding overt A beta deposition, the doubly transgenic mice show a selective 41% increase in A beta42(43) in their brains. Thus, the development of AD-like pathology is substantially enhanced when a PS1 mutation, which causes a modest increase in A beta42(43), is introduced into Tg2576-derived mice. Remarkably, both doubly and singly transgenic mice showed reduced spontaneous alternation performance in a "Y" maze before substantial A beta deposition was apparent. This suggests that some aspects of the behavioral phenotype in these mice may be related to an event that precedes plaque formation.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Proteínas de Membrana/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/biossíntese , Análise de Variância , Animais , Encéfalo/patologia , Córtex Cerebral/patologia , Cruzamentos Genéticos , Genótipo , Proteína Glial Fibrilar Ácida/análise , Humanos , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Transgênicos , Atividade Motora , Postura , Presenilina-1 , Desempenho Psicomotor , Reflexo , Convulsões
3.
Nat Med ; 2(8): 864-70, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8705854

RESUMO

To determine whether the presenilin 1 (PS1), presenilin 2 (PS2) and amyloid beta-protein precursor (APP) mutations linked to familial Alzheimer's disease (FAD) increase the extracellular concentration of amyloid beta-protein (A beta) ending at A beta 42(43) in vivo, we performed a blinded comparison of plasma A beta levels in carriers of these mutations and controls. A beta 1-42(43) was elevated in plasma from subjects with FAD-linked PS1 (P < 0.0001), PS2N1411 (P = 0.009), APPK670N,M671L (P < 0.0001), and APPV7171 (one subject) mutations. A beta ending at A beta 42(43) was also significantly elevated in fibroblast media from subjects with PS1 (P < 0.0001) or PS2 (P = 0.03) mutations. These findings indicate that the FAD-linked mutations may all cause Alzhelmer's disease by increasing the extracellular concentration of A beta 42(43), thereby fostering cerebral deposition of this highly amyloidogenic peptide.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Proteínas de Membrana/genética , Mutação , Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/sangue , Células Cultivadas , Meios de Cultivo Condicionados , Feminino , Fibroblastos , Humanos , Masculino , Fragmentos de Peptídeos/sangue , Presenilina-1 , Presenilina-2
4.
Science ; 264(5163): 1336-40, 1994 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-8191290

RESUMO

Normal processing of the amyloid beta protein precursor (beta APP) results in secretion of a soluble 4-kilodalton protein essentially identical to the amyloid beta protein (A beta) that forms insoluble fibrillar deposits in Alzheimer's disease. Human neuroblastoma (M17) cells transfected with constructs expressing wild-type beta APP or the beta APP717 mutants linked to familial Alzheimer's disease were compared by (i) isolation of metabolically labeled 4-kilodalton A beta from conditioned medium, digestion with cyanogen bromide, and analysis of the carboxyl-terminal peptides released, or (ii) analysis of the A beta in conditioned medium with sandwich enzyme-linked immunosorbent assays that discriminate A beta 1-40 from the longer A beta 1-42. Both methods demonstrated that the 4-kilodalton A beta released from wild-type beta APP is primarily but not exclusively A beta 1-40. The beta APP717 mutations, which are located three residues carboxyl to A beta 43, consistently caused a 1.5- to 1.9-fold increase in the percentage of longer A beta generated. Long A beta (for example, A beta 1-42) forms insoluble amyloid fibrils more rapidly than A beta 1-40. Thus, the beta APP717 mutants may cause Alzheimer's disease because they secrete increased amounts of long A beta, thereby fostering amyloid deposition.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Mutação , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Meios de Cultivo Condicionados , Ensaio de Imunoadsorção Enzimática , Humanos , Neuroblastoma , Transfecção , Células Tumorais Cultivadas
5.
Science ; 274(5284): 99-102, 1996 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-8810256

RESUMO

Transgenic mice overexpressing the 695-amino acid isoform of human Alzheimer beta-amyloid (Abeta) precursor protein containing a Lys670 --> Asn, Met671 --> Leu mutation had normal learning and memory in spatial reference and alternation tasks at 3 months of age but showed impairment by 9 to 10 months of age. A fivefold increase in Abeta(1-40) and a 14-fold increase in Abeta(1-42/43) accompanied the appearance of these behavioral deficits. Numerous Abeta plaques that stained with Congo red dye were present in cortical and limbic structures of mice with elevated amounts of Abeta. The correlative appearance of behavioral, biochemical, and pathological abnormalities reminiscent of Alzheimer's disease in these transgenic mice suggests new opportunities for exploring the pathophysiology and neurobiology of this disease.


Assuntos
Peptídeos beta-Amiloides/análise , Precursor de Proteína beta-Amiloide/análise , Química Encefálica , Encéfalo/patologia , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Fragmentos de Peptídeos/análise , Envelhecimento , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Deficiências da Aprendizagem/patologia , Aprendizagem em Labirinto , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Desempenho Psicomotor
6.
Science ; 212(4502): 1527-9, 1981 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-7233241

RESUMO

Physical dependence on morphine occurs in a typical fashion during the active state of the mammalian hibernator Citellus lateralis, but does not occur when morphine exposure is confined to the hibernating state. Morphine exposure during hibernation can produce stereotyped behavior, thus demonstrating partial responsiveness of the central nervous system to opioids during this natural state.


Assuntos
Hibernação/efeitos dos fármacos , Morfina/farmacologia , Animais , Barreira Hematoencefálica , Feminino , Humanos , Masculino , Naloxona/farmacologia , Sciuridae , Transtornos Relacionados ao Uso de Substâncias
7.
Science ; 290(5500): 2303-4, 2000 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-11125143

RESUMO

Plasma Abeta42 (amyloid beta42 peptide) is invariably elevated in early-onset familial Alzheimer's disease (AD), and it is also increased in the first-degree relatives of patients with typical late-onset AD (LOAD). To detect LOAD loci that increase Abeta42, we used plasma Abeta42 as a surrogate trait and performed linkage analysis on extended AD pedigrees identified through a LOAD patient with extremely high plasma Abeta. Here, we report linkage to chromosome 10 with a maximal lod score of 3.93 at 81 centimorgans close to D10S1225. Remarkably, linkage to the same region was obtained independently in a genome-wide screen of LOAD sibling pairs. These results provide strong evidence for a novel LOAD locus on chromosome 10 that acts to increase Abeta.


Assuntos
Doença de Alzheimer/sangue , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/sangue , Cromossomos Humanos Par 10/genética , Ligação Genética , Fragmentos de Peptídeos/sangue , Característica Quantitativa Herdável , Adulto , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/genética , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Humanos , Escore Lod , Masculino , Pessoa de Meia-Idade , Linhagem , Fragmentos de Peptídeos/genética , Fenótipo
8.
Science ; 293(5534): 1487-91, 2001 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-11520987

RESUMO

JNPL3 transgenic mice expressing a mutant tau protein, which develop neurofibrillary tangles and progressive motor disturbance, were crossed with Tg2576 transgenic mice expressing mutant beta-amyloid precursor protein (APP), thus modulating the APP-Abeta (beta-amyloid peptide) environment. The resulting double mutant (tau/APP) progeny and the Tg2576 parental strain developed Abeta deposits at the same age; however, relative to JNPL3 mice, the double mutants exhibited neurofibrillary tangle pathology that was substantially enhanced in the limbic system and olfactory cortex. These results indicate that either APP or Abeta influences the formation of neurofibrillary tangles. The interaction between Abeta and tau pathologies in these mice supports the hypothesis that a similar interaction occurs in Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/patologia , Emaranhados Neurofibrilares/patologia , Placa Amiloide/patologia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Sistema Límbico/metabolismo , Sistema Límbico/patologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Degeneração Neural , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Neurônios/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Solubilidade , Medula Espinal/metabolismo , Medula Espinal/patologia , Proteínas tau/genética
9.
Neuron ; 20(3): 603-9, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9539132

RESUMO

Mutations in presenilin 1 (PS1) cosegregate with approximately 25% of early onset familial Alzheimer's disease (FAD) pedigrees. A variety of in vitro and in vivo paradigms have established that one mechanism by which PS1 variants cause AD is by elevating the production of highly amyloidogenic Abeta1-42/43 peptides. PS1 is homologous to sel-12, a C. elegans protein that facilitates signaling mediated by the Notch/lin-12 family of receptors. Wild-type human PS1 complements an egg-laying defect in C. elegans lacking sel-12, while FAD-linked PS1 variants exhibit reduced rescue activity. These data suggested that mutant PS1 may cause disease as a result of reduction in PS1 function. To test the function of FAD-linked PS1 in mammals, we examined the ability of the A246E PS1 variant to complement the embryonic lethality and axial skeletal defects in mice lacking PS1. Finally, to examine the influence of reduced PS1 levels on Abeta production, we quantified Abeta1-42/43 peptide levels in PS1 heterozygous null mice (PS1[+/-] mice). We now report that both human wild-type and A246E PS1 efficiently rescue the phenotypes observed in PS1(-/-) embryos, findings consistent with the view that FAD-linked PS1 mutants retain sufficient normal function during mammalian embryonic development. Moreover, the levels of Abeta1-42/43 and Abeta1-40 peptides between PS1(+/-) and control mice are indistinguishable. Collectively, these data lead us to conclude that mutant PS1 causes AD not by loss of normal PS1 function but by influencing amyloid precursor protein (APP) processing in a manner that elevates Abeta1-42/43 production.


Assuntos
Doença de Alzheimer/genética , Proteínas de Membrana/genética , Receptores de Superfície Celular , Fatores de Transcrição , Doença de Alzheimer/fisiopatologia , Animais , Animais Recém-Nascidos , Osso e Ossos/anormalidades , Osso e Ossos/química , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Variação Genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mesoderma/química , Camundongos , Camundongos Transgênicos , Morfogênese/fisiologia , Mutação , Crista Neural/anormalidades , Crista Neural/química , Gravidez , Presenilina-1 , Receptor Notch1 , Transgenes/fisiologia
10.
Neuron ; 15(5): 1203-18, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7576662

RESUMO

Transgenic FVB/N mice overexpressing human (Hu) or mouse (Mo) Alzheimer amyloid precursor protein (APP695) die early and develop a CNS disorder that includes neophobia and impaired spatial alternation, with diminished glucose utilization and astrogliosis mainly in the cerebrum. Age at onset of neophobia and age at death decrease with increasing levels of brain APP. HuAPP transgenes induce death much earlier than MoAPP transgenes expressed at similar levels. No extracellular amyloid was detected, indicating that some deleterious processes related to APP overexpression are dissociated from formation of amyloid. A similar clinical syndrome occurs spontaneously in approximately 20% of nontransgenic mice when they reach mid- to late-adult life, suggesting that APP overexpression may accelerate a naturally occurring age-related CNS disorder in FVB/N mice.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide/genética , Doenças do Sistema Nervoso Central/genética , Expressão Gênica , Envelhecimento , Animais , Astrócitos/patologia , Sequência de Bases , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Doenças do Sistema Nervoso Central/mortalidade , Doenças do Sistema Nervoso Central/fisiopatologia , Cosmídeos , Feminino , Vetores Genéticos , Glucose/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular
11.
Neuron ; 17(5): 1005-13, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8938131

RESUMO

Mutations in the presenilin 1 (PS1) and presenilin 2 genes cosegregate with the majority of early-onset familial Alzheimer's disease (FAD) pedigrees. We now document that the Abeta1-42(43)/Abeta1-40 ratio in the conditioned media of independent N2a cell lines expressing three FAD-linked PS1 variants is uniformly elevated relative to cells expressing similar levels of wild-type PS1. Similarly, the Abeta1-42(43)/Abeta1-40 ratio is elevated in the brains of young transgenic animals coexpressing a chimeric amyloid precursor protein (APP) and an FAD-linked PS1 variant compared with brains of transgenic mice expressing APP alone or transgenic mice coexpressing wild-type human PS1 and APP. These studies provide compelling support for the view that one mechanism by which these mutant PS1 cause AD is by increasing the extracellular concentration of Abeta peptides terminating at 42(43), species that foster Abeta deposition.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Proteínas de Membrana/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Química Encefálica/genética , Meios de Cultivo Condicionados , Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação/fisiologia , Neuroblastoma , Presenilina-1 , Proteínas Recombinantes de Fusão/fisiologia , Transfecção , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/fisiologia
12.
Nat Neurosci ; 4(9): 887-93, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11528419

RESUMO

Several pathogenic Alzheimer's disease (AD) mutations have been described, all of which cause increased amyloid beta-protein (Abeta) levels. Here we present studies of a pathogenic amyloid precursor protein (APP) mutation, located within the Abeta sequence at codon 693 (E693G), that causes AD in a Swedish family. Carriers of this 'Arctic' mutation showed decreased Abeta42 and Abeta40 levels in plasma. Additionally, low levels of Abeta42 were detected in conditioned media from cells transfected with APPE693G. Fibrillization studies demonstrated no difference in fibrillization rate, but Abeta with the Arctic mutation formed protofibrils at a much higher rate and in larger quantities than wild-type (wt) Abeta. The finding of increased protofibril formation and decreased Abeta plasma levels in the Arctic AD may reflect an alternative pathogenic mechanism for AD involving rapid Abeta protofibril formation leading to accelerated buildup of insoluble Abeta intra- and/or extracellularly.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/fisiologia , Precursor de Proteína beta-Amiloide/genética , Mutação/fisiologia , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/química , Linhagem Celular/metabolismo , Meios de Cultura/metabolismo , Heterozigoto , Humanos , Pessoa de Meia-Idade , Linhagem , Fragmentos de Peptídeos/fisiologia , Suécia
13.
Nat Neurosci ; 2(2): 157-61, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10195200

RESUMO

Peptides derived from proteolytic processing of the beta-amyloid precursor protein (APP), including the amyloid-beta peptide, are important for the pathogenesis of Alzheimer's dementia. We found that transgenic mice overexpressing APP have a profound and selective impairment in endothelium-dependent regulation of the neocortical microcirculation. Such endothelial dysfunction was not found in transgenic mice expressing both APP and superoxide dismutase-1 (SOD1) or in APP transgenics in which SOD was topically applied to the cerebral cortex. These cerebrovascular effects of peptides derived from APP processing may contribute to the alterations in cerebral blood flow and to neuronal dysfunction in Alzheimer's dementia.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Circulação Cerebrovascular/fisiologia , Superóxido Dismutase/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Córtex Cerebral/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Sequestradores de Radicais Livres/farmacologia , Humanos , Camundongos , Camundongos Transgênicos/genética , Superóxido Dismutase/genética , Superóxido Dismutase/farmacologia , Superóxido Dismutase-1 , Vasoconstritores/farmacologia
14.
Biochim Biophys Acta ; 1502(1): 172-87, 2000 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-10899442

RESUMO

Prior to the identification of the various abnormal proteins deposited as fibrillar aggregates in the Alzheimer's disease (AD) brain, there was tremendous controversy over the importance of the various lesions with respect to primacy in the pathology of AD. Nevertheless, based on analogy to systemic amyloidosis, many investigators believed that the amyloid deposits in AD played a causal role and that characterization of these deposits would hold the key to understanding this complex disease. Indeed, in retrospect, it was the initial biochemical purifications of the approximately 4 kDa amyloid beta-peptide (Abeta) from amyloid deposits in the mid 1980s that launched a new era of AD research (Glenner and Wong, Biochem. Biophys. Res. Commun. 122 (1984) 1121-1135; Wong et al., Proc. Natl. Acad Sci. USA 82 (1985) 8729 8732; and Masters et al., Proc. Natl. Acad Sci. USA 82 (1985) 4245-4249). Subsequent studies of the biology of Abeta together with genetic studies of AD have all supported the hypothesis that altered Abeta metabolism leading to aggregation plays a causal role in AD. Although there remains controversy as to whether Abeta deposited as classic amyloid or a smaller, aggregated, form causes AD, the relevance of studying the amyloid deposits has certainly been proven. Despite the significant advances in our understanding of the role of Abeta in AD pathogenesis, many important aspects of Abeta biology remain a mystery. This review will highlight those aspects of Abeta biology that have led to our increased understanding of the pathogenesis of AD as well as areas which warrant additional study.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/análise , Química Encefálica , Encéfalo/metabolismo , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/terapia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Biomarcadores/análise , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Espectrometria de Massas , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutação , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/genética , Presenilina-1 , Presenilina-2 , Isoformas de Proteínas/análise
15.
Neurobiol Aging ; 17(2): 215-22, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8744402

RESUMO

Some forms of familial Alzheimer's disease are caused by mutations in the amyloid beta protein precursor (beta APP), and there is excellent evidence that these mutations foster amyloid deposition by increasing secretion of total amyloid beta protein (A beta) or the highly amyloidogenic A beta 1-42 form. These observations provide a powerful rationale for developing an animal model of AD by generating transgenic mice in which cerebral amyloid deposition is induced by A beta overproduction. To produce substantial A beta in vivo, we generated mice expressing the transgene of signal peptide and 99 residues of carboxyl-terminal fragment (CTF) of beta APP under control of the cytomegalovirus enhancer/chicken beta-actin promoter. The transgenic mRNA was detected in many tissues of these mice, but the levels of transgenic mRNA, CTF, and A beta did not correlate well indicating that tissue-specific posttranslational processing may play an important role in determining the amount of A beta that accumulates in various tissues. A beta was detected biochemically in brain, kidney, and pancreas with the largest amount present in pancreas. In transgenic plasma, there was a marked accumulation of human A beta 1-40 and A beta 1-42(43) to levels over 30-times those observed in normal human plasma. Thus, the transgenic mice produce and secrete considerable A beta. Despite this increase in A beta secretion and the elevated A beta in brain, immunohistochemistry revealed no consistent cerebral A beta deposition. In pancreas, however, intracellular A beta deposits were detected immunohistochemically in acinar cells and interstitial macrophages, some of which showed severe degeneration. In addition, examination of these cells by immunoelectron microscopy revealed many putative amyloid fibrils (7-12 nm) that were stained by anti-A beta antibodies. Overall, our findings indicate that tissue-specific posttranslational processing may play a pivotal role in A beta production and amyloid fibril formation in vivo. By carefully analyzing the changes that occur in the transgenic mice described here as compared to the transgenic line that has recently been shown to form extracellular amyloid plaques in brain, it may be possible to gain considerable insight into the factors that determine the location and amount of A beta that accumulates as amyloid.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Neurofibrilas/metabolismo , Pâncreas/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Citomegalovirus/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Dados de Sequência Molecular , Neurofibrilas/ultraestrutura , Pâncreas/ultraestrutura , Reação em Cadeia da Polimerase , Transgenes
16.
FEBS Lett ; 374(1): 25-8, 1995 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-7589505

RESUMO

PCR primers were designed from the known amino acid (aa) sequence for human red blood cell thioltransferase (hRBC TTase) and the known cDNA sequence for pig liver TTase (82% homologous) and used to amplify thioltransferase from a pool of human brain cDNAs. The PCR product was inserted into the pKK233-2 expression vector. The DNA sequence of the insert agreed with the aa sequence. High level expression of the enzyme was accomplished in E. coli, and Western blot analysis confirmed its identity. Recombinant TTase displayed catalytic properties indistinguishable from natural hRBC TTase.


Assuntos
Oxirredutases/genética , Proteína Dissulfeto Redutase (Glutationa) , Sequência de Aminoácidos , Animais , Sequência de Bases , Catálise , Clonagem Molecular , Eritrócitos/enzimologia , Escherichia coli , Expressão Gênica , Glutarredoxinas , Humanos , Dados de Sequência Molecular , Oxirredutases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Suínos
17.
J Comp Neurol ; 392(4): 428-38, 1998 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-9514508

RESUMO

By using transgenic mice that overexpress human beta-amyloid precursor proteins (APPs) at levels twofold higher than endogenous APPs, following introduction of the human APP gene in a yeast artificial chromosome (YAC), we examined the effects of controlled cortical impact (CCI) brain injury on neuromotor/cognitive dysfunction and the development of Alzheimer's disease (AD)-like neuropathology. Neuropathological analyses included Nissl-staining and immunohistochemistry to detect APPs, beta-amyloid (Abeta), neurofilament proteins, and glial fibrillary acidic protein, whereas Abeta levels were measured in brain homogenates from mice subjected to CCI and control mice by using a sensitive sandwich enzyme-linked immunosorbent assay. Twenty APP-YAC transgenic mice and 17 wild type (WT) littermate controls were anesthetized and subjected to CCI (velocity, 5 m/second; deformation depth, 1 mm). Sham (anesthetized but uninjured) controls (n = 10 APP-YAC; n = 8 WT) also were studied. Motor function was evaluated by using rotarod, inclined-plane, and forelimb/hindlimb flexion tests. The Morris water maze was used to assess memory. Although CCI induced significant motor dysfunction and cognitive deficits, no differences were observed between brain-injured APP-YAC mice and WT mice at 24 hours and 1 week postinjury. By 1 week postinjury, both cortical and hippocampal CA3 neuron loss as well as extensive astrogliosis were observed in all injured animals, suggesting that overexpression of human APPs exhibited no neuroprotective effects. Although AD-like pathology (including amyloid plaques) was not observed in either sham or brain-inj ured animals, a significant decrease in brain concentrations of only Abeta terminating at amino acid 40 (Abeta x-40) was observed following brain injury in APP-YAC mice (P < 0.05 compared with sham control levels). Our data show that the APP-YAC mice do not develop AD-like neuropathology following traumatic brain injury. This may be because this injury does not induce elevated levels of the more amyloidogenic forms of human Abeta (i.e., Abeta x-42/43) in these mice.


Assuntos
Peptídeos beta-Amiloides/genética , Lesões Encefálicas/fisiopatologia , Cognição/fisiologia , Camundongos Transgênicos/fisiologia , Neurônios Motores/fisiologia , Peptídeos beta-Amiloides/análise , Animais , Comportamento Animal , Lesões Encefálicas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Proteína Glial Fibrilar Ácida/análise , Imunoglobulina G/análise , Imunoglobulina G/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/química , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Proteínas de Neurofilamentos/análise , Cloreto de Tolônio
18.
Brain Res Mol Brain Res ; 97(1): 103-13, 2001 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-11744168

RESUMO

The amyloid precursor protein (APP) is cleaved by two enzymes, beta-secretase and gamma-secretase, to generate the pathological amyloid beta (Abeta) peptide. Expression of familial Alzheimer's disease (FAD) mutants of APP in primary neurons causes both intracellular accumulation of the C-terminal beta-secretase cleavage product of APP and increased secretion of Abeta, and eventually results in apoptotic death of the cells. To determine whether either of these two processing products of APP is involved in this apoptotic pathway, we first modeled experimentally the accumulation of the beta-secretase cleavage product in neurons. The C-terminal 100 amino acids (C100) of APP, with and without a signal peptide, was expressed in cells via recombinant herpes simplex virus (HSV) vectors. Both transgene products were targeted to the membrane, and both caused apoptosis in the neurons, implicating the beta-secretase cleavage product of APP in apoptosis caused by FAD APPs. Expression in neurons of a mutant of FAD APP that inhibited beta-secretase cleavage inhibited its ability to cause apoptosis. However, expression in neurons of a mutant of FAD APP that inhibited gamma-secretase cleavage did not inhibit the ability of this mutant to cause apoptosis. These data suggested that the C-terminal beta-secretase cleavage product of APP, but not Abeta, mediates the apoptosis caused by FAD mutants of APP. Consistent with this hypothesis, C31, which is generated from the beta-secretase cleavage product, itself caused neuronal apoptosis. Inhibitors of caspases 3, 6 and 8, but not of caspase 9, inhibited the apoptosis caused by FAD mutants of APP. It may be inferred from these data that beta-secretase cleavage of FAD mutants of APP allows the appropriate caspase access to its site of action to produce C31, which directly causes neuronal apoptosis.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apoptose/fisiologia , Ácido Aspártico Endopeptidases/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Doença de Alzheimer/genética , Substituição de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Animais , Caspases/metabolismo , Meios de Cultivo Condicionados , Endopeptidases/metabolismo , Vetores Genéticos/genética , Humanos , Londres , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/metabolismo , Mutação Puntual , Ratos , Simplexvirus/genética , Suécia , Transgenes
19.
Neuroreport ; 9(14): 3293-9, 1998 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-9831466

RESUMO

Numerous mutations in the presenilin (PS) genes cause early onset familial Alzheimer's disease (FAD). Here we characterize the expression of two naturally occurring alternative PS2 transcripts which lack either exons 3 and 4 (PS2 deltaexon3,4) or exons 3, 4, and 8 (PS2 deltaexon3,4,8). These transcripts do not contain the natural initiation codon within exon 3. The transcripts are efficiently translated as N-terminal truncated proteins. These deleted proteins are still able to regulate formation of endogenous PS fragments, indicating that the C-terminal half of the PS2 protein is sufficient for this phenomenon. Although approximately 50% of the PS1 and both PS2 mutations occur within the N-terminal region lacking in the PS2 deltaexon3,4 and PS2 deltaexon3,4,8 proteins, expression of these truncated proteins does not affect pathological generation of amyloid beta-peptide (Abeta). This suggests that point mutations causing AD are gain of function mutations.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Proteínas de Membrana/genética , Fragmentos de Peptídeos/genética , Splicing de RNA/fisiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/análise , Animais , Células COS , Sondas de DNA , Éxons , Humanos , Rim/citologia , Proteínas de Membrana/análise , Fragmentos de Peptídeos/análise , Presenilina-2 , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/análise , Frações Subcelulares/química
20.
Peptides ; 8(2): 247-50, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-3108865

RESUMO

The effect of impaction-induced spinal trauma on the concentration of immunoreactive TRH (TRH-ir) in the spinal cord was studied. Samples were obtained from tissues proximal to, distal to, and at the site of injury at 30 min, 1 hr, 4 hr, and 6 weeks after impaction. After an initial 38% depletion of TRH-ir at the injury site at 30 min, concentrations were progressively elevated over time at all sites. These elevations reached statistical significance in the proximal and distal segments by 4 hr posttrauma. By 6 weeks, a rostral-caudal gradient of TRH-ir concentration was observed, indicating that axoplasmic transport was restricted. The gradient was characterized by a significant TRH-ir elevation proximal to, and a 60% depletion distal to, the injury. The short-term TRH-ir elevation measured indicates that the ability of exogenously administered TRH to reduce the incidence of posttraumatic functional deficit stems from a restoration of endogenous TRH action. The role of the raphe-spinal tract in the development of traumatic paralysis is considered.


Assuntos
Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Animais , Especificidade de Órgãos , Paraplegia/metabolismo , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA