Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 28(23): 6030-6, 2008 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-18524908

RESUMO

Analyses of the biologic effects of mutations in the BRI2 (ITM2b) and the amyloid beta precursor protein (APP) genes support the hypothesis that cerebral accumulation of amyloidogenic peptides in familial British and familial Danish dementias and Alzheimer's disease (AD) is associated with neurodegeneration. We have used somatic brain transgenic technology to express the BRI2 and BRI2-Abeta1-40 transgenes in APP mouse models. Expression of BRI2-Abeta1-40 mimics the suppressive effect previously observed using conventional transgenic methods, further validating the somatic brain transgenic methodology. Unexpectedly, we also find that expression of wild-type human BRI2 reduces cerebral Abeta deposition in an AD mouse model. Additional data indicate that the 23 aa peptide, Bri23, released from BRI2 by normal processing, is present in human CSF, inhibits Abeta aggregation in vitro and mediates its anti-amyloidogenic effect in vivo. These studies demonstrate that BRI2 is a novel mediator of Abeta deposition in vivo.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/fisiologia , Encéfalo/metabolismo , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Amiloide/genética , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/administração & dosagem , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Galinhas , Cricetinae , Dependovirus/genética , Feminino , Técnicas de Transferência de Genes , Humanos , Masculino , Glicoproteínas de Membrana , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fenótipo
2.
Nature ; 453(7197): 925-9, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18548070

RESUMO

Selective lowering of Abeta42 levels (the 42-residue isoform of the amyloid-beta peptide) with small-molecule gamma-secretase modulators (GSMs), such as some non-steroidal anti-inflammatory drugs, is a promising therapeutic approach for Alzheimer's disease. To identify the target of these agents we developed biotinylated photoactivatable GSMs. GSM photoprobes did not label the core proteins of the gamma-secretase complex, but instead labelled the beta-amyloid precursor protein (APP), APP carboxy-terminal fragments and amyloid-beta peptide in human neuroglioma H4 cells. Substrate labelling was competed by other GSMs, and labelling of an APP gamma-secretase substrate was more efficient than a Notch substrate. GSM interaction was localized to residues 28-36 of amyloid-beta, a region critical for aggregation. We also demonstrate that compounds known to interact with this region of amyloid-beta act as GSMs, and some GSMs alter the production of cell-derived amyloid-beta oligomers. Furthermore, mutation of the GSM binding site in the APP alters the sensitivity of the substrate to GSMs. These findings indicate that substrate targeting by GSMs mechanistically links two therapeutic actions: alteration in Abeta42 production and inhibition of amyloid-beta aggregation, which may synergistically reduce amyloid-beta deposition in Alzheimer's disease. These data also demonstrate the existence and feasibility of 'substrate targeting' by small-molecule effectors of proteolytic enzymes, which if generally applicable may significantly broaden the current notion of 'druggable' targets.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Animais , Anti-Inflamatórios não Esteroides/química , Sítios de Ligação/efeitos dos fármacos , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Humanos , Camundongos , Ligação Proteica/efeitos dos fármacos , Receptores Notch/genética , Receptores Notch/metabolismo , Especificidade por Substrato/efeitos dos fármacos
3.
Mol Neurodegener ; 2: 11, 2007 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-17559680

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by a decline in cognitive function and accumulation of amyloid-beta peptide (Abeta) in extracellular plaques. Mutations in amyloid precursor protein (APP) and presenilins alter APP metabolism resulting in accumulation of Abeta42, a peptide essential for the formation of amyloid deposits and proposed to initiate the cascade leading to AD. However, the role of Abeta40, the more prevalent Abeta peptide secreted by cells and a major component of cerebral Abeta deposits, is less clear. In this study, virally-mediated gene transfer was used to selectively increase hippocampal levels of human Abeta42 and Abeta40 in adult Wistar rats, allowing examination of the contribution of each to the cognitive deficits and pathology seen in AD. RESULTS: Adeno-associated viral (AAV) vectors encoding BRI-Abeta cDNAs were generated resulting in high-level hippocampal expression and secretion of the specific encoded Abeta peptide. As a comparison the effect of AAV-mediated overexpression of APPsw was also examined. Animals were tested for development of learning and memory deficits (open field, Morris water maze, passive avoidance, novel object recognition) three months after infusion of AAV. A range of impairments was found, with the most pronounced deficits observed in animals co-injected with both AAV-BRI-Abeta40 and AAV-BRI-Abeta42. Brain tissue was analyzed by ELISA and immunohistochemistry to quantify levels of detergent soluble and insoluble Abeta peptides. BRI-Abeta42 and the combination of BRI-Abeta40+42 overexpression resulted in elevated levels of detergent-insoluble Abeta. No significant increase in detergent-insoluble Abeta was seen in the rats expressing APPsw or BRI-Abeta40. No pathological features were noted in any rats, except the AAV-BRI-Abeta42 rats which showed focal, amorphous, Thioflavin-negative Abeta42 deposits. CONCLUSION: The results show that AAV-mediated gene transfer is a valuable tool to model aspects of AD pathology in vivo, and demonstrate that whilst expression of Abeta42 alone is sufficient to initiate Abeta deposition, both Abeta40 and Abeta42 may contribute to cognitive deficits.

4.
FASEB J ; 20(14): 2576-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17068112

RESUMO

A number of hypotheses regarding how anti-Abeta antibodies alter amyloid deposition have been postulated, yet there is no consensus as to how Abeta immunotherapy works. We have examined the in vivo binding properties, pharmacokinetics, brain penetrance, and alterations in Abeta levels after a single peripheral dose of anti-Abeta antibodies to both wild-type (WT) and young non-Abeta depositing APP and BRI-Abeta42 mice. The rapid rise in plasma Abeta observed after antibody (Ab) administration is attributable to prolongation of the half-life of Abeta bound to the Ab. Only a miniscule fraction of Ab enters the brain, and despite dramatic increases in plasma Abeta, we find no evidence that total brain Abeta levels are significantly altered. Surprisingly, cerebral spinal fluid Abeta levels transiently rise, and when Ab:Abeta complex is directly injected into the lateral ventricles of mice, it is rapidly cleared from the brain into the plasma where it remains stable. When viewed in context of daily turnover of Abeta, these data provide a framework to evaluate proposed mechanisms of Abeta attenuation mediated by peripheral administration of an anti-Abeta monoclonal antibody (mAb) effective in passive immunization paradigm. Such quantitative data suggest that the mAbs are either indirectly enhancing clearance of Abeta or targeting a low abundance aggregation intermediate.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais/imunologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Imunização Passiva , Camundongos , Camundongos Transgênicos
5.
J Neuroinflammation ; 3: 17, 2006 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16872492

RESUMO

BACKGROUND: Microglial activation has been proposed to facilitate clearance of amyloid beta protein (Abeta) from the brain following Abeta immunotherapy in amyloid precursor protein (APP) transgenic mice. Interleukin-1 receptor 1 knockout (IL-1 R1-/-) mice are reported to exhibit blunted inflammatory responses to injury. To further define the role of IL-1-mediated inflammatory responses and microglial activation in this paradigm, we examined the efficacy of passive Abeta immunotherapy in Tg2576 mice crossed into the IL-1 R1-/- background. In addition, we examined if loss of IL-1 R1-/- modifies Abeta deposition in the absence of additional manipulations. METHODS: We passively immunized Tg2576 mice crossed into the IL-1 R1-/- background (APP/IL-1 R1-/- mice) with an anti-Abeta1-16 mAb (mAb9, IgG2a) that we previously showed could attenuate Abeta deposition in Tg2576 mice. We also examined whether the IL-1 R1 knockout background modifies Abeta deposition in untreated mice. Biochemical and immunohistochemical Abeta loads and microglial activation was assessed. RESULTS: Passive immunization with anti-Abeta mAb was effective in reducing plaque load in APP/IL-1 R1-/- mice when the immunization was started prior to significant plaque deposition. Similar to previous studies, immunization was not effective in older APP/IL-1 R1-/- mice or IL-1 R1 sufficient wild type Tg2576 mice. Our analysis of Abeta deposition in the untreated APP/IL-1 R1-/- mice did not show differences on biochemical Abeta loads during normal aging of these mice compared to IL-1 R1 sufficient wild type Tg2576 mice. CONCLUSION: We find no evidence that the lack of the IL-1 R1 receptor influences either Abeta deposition or the efficacy of passive immunotherapy. Such results are consistent with other studies in Tg2576 mice that suggest microglial activation may not be required for efficacy in passive immunization approaches.

6.
J Clin Invest ; 116(1): 193-201, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16341263

RESUMO

Accumulation and aggregation of amyloid beta peptide 1-42 (Abeta42) in the brain has been hypothesized as triggering a pathological cascade that causes Alzheimer disease (AD). To determine whether selective targeting of Abeta42 versus Abeta40 or total Abeta is an effective way to prevent or treat AD, we compared the effects of passive immunization with an anti-Abeta42 mAb, an anti-Abeta40 mAb, and multiple Abeta(1-16) mAbs. We established in vivo binding selectivity of the anti-Abeta42 and anti-Abeta40 mAbs using novel TgBRI-Abeta mice. We then conducted a prevention study in which the anti-Abeta mAbs were administered to young Tg2576 mice, which have no significant Abeta deposition, and therapeutic studies in which mAbs were administered to Tg2576 or CRND8 mice with modest levels of preexisting Abeta deposits. Anti-Abeta42, anti-Abeta40, and anti-Abeta(1-16) mAbs attenuated plaque deposition in the prevention study. In contrast, anti-Abeta42 and anti-Abeta40 mAbs were less effective in attenuating Abeta deposition in the therapeutic studies and were not effective in clearing diffuse plaques following direct injection into the cortex. These data suggest that selective targeting of Abeta42 or Abeta40 may be an effective strategy to prevent amyloid deposition, but may have limited benefit in a therapeutic setting.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Amiloide/imunologia , Anticorpos Monoclonais/uso terapêutico , Imunização Passiva , Fragmentos de Peptídeos/imunologia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos
7.
Echocardiography ; 13(1): 57-64, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11442903

RESUMO

In this study, isovolumetric relaxation time (IVRT) flow velocities recorded by pulsed Doppler were induced or augmented in nine patients (aged 60 +/- 20 years) with permanent pacemakers. Six subjects had evidence of IVRT flow (>/=0.2 m/sec) during native rhythm, only three of whom exhibited cavity obliteration. Mean +/- SD left ventricular (LV) ejection fraction and end-diastolic and end-systolic volumes were 60% +/- 5%, 104 +/- 50 mL, and 39 +/- 27 mL, respectively. IVRT flows were induced or augmented (P < 0.003) in all patients during paced ectopic stimulation of the right ventricle and during pacing combined with sublingual nitroglycerin (NTG; 0.4 mg). However, IVRT flows were not significantly increased with NTG alone. IVRT flow duration was prolonged >50% by pacing (P < 0.05). These data suggest that IVRT flows may be present in patients without cavity obliteration and may be accentuated by asynchronous LV relaxation. (ECHOCARDIOGRAPHY, Volume 13, January 1996)

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...