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










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 10(1): 3530, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31387998

RESUMO

Microtubules are a vital component of the cell's cytoskeleton and their organization is crucial for healthy cell functioning. The use of label-free SH imaging of microtubules remains limited, as sensitive detection is required and the true molecular origin and main determinants required to generate SH from microtubules are not fully understood. Using advanced correlative imaging techniques, we identified the determinants of the microtubule-dependent SH signal. Microtubule polarity, number and organization determine SH signal intensity in biological samples. At the molecular level, we show that the GTP-bound tubulin dimer conformation is fundamental for microtubules to generate detectable SH signals. We show that SH imaging can be used to study the effects of microtubule-targeting drugs and proteins and to detect changes in tubulin conformations during neuronal maturation. Our data provide a means to interpret and use SH imaging to monitor changes in the microtubule network in a label-free manner.


Assuntos
Microscopia Intravital/métodos , Microtúbulos/ultraestrutura , Imagem Molecular/métodos , Microscopia de Geração do Segundo Harmônico , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Células Cultivadas , Colchicina/farmacologia , Estudos de Viabilidade , Guanosina Trifosfato/metabolismo , Camundongos , Microscopia Eletrônica , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Neurogênese , Cultura Primária de Células , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/ultraestrutura
2.
Mol Psychiatry ; 18(11): 1225-34, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23399914

RESUMO

Genome-wide association studies (GWAS) have identified a region upstream the BIN1 gene as the most important genetic susceptibility locus in Alzheimer's disease (AD) after APOE. We report that BIN1 transcript levels were increased in AD brains and identified a novel 3 bp insertion allele ∼28 kb upstream of BIN1, which increased (i) transcriptional activity in vitro, (ii) BIN1 expression levels in human brain and (iii) AD risk in three independent case-control cohorts (Meta-analysed Odds ratio of 1.20 (1.14-1.26) (P=3.8 × 10(-11))). Interestingly, decreased expression of the Drosophila BIN1 ortholog Amph suppressed Tau-mediated neurotoxicity in three different assays. Accordingly, Tau and BIN1 colocalized and interacted in human neuroblastoma cells and in mouse brain. Finally, the 3 bp insertion was associated with Tau but not Amyloid loads in AD brains. We propose that BIN1 mediates AD risk by modulating Tau pathology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Predisposição Genética para Doença/genética , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética , Proteínas tau/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Endofenótipos , Expressão Gênica/genética , Humanos , Camundongos , Degeneração Neural/genética , Degeneração Neural/patologia , Proteínas Nucleares/biossíntese , Placa Amiloide/patologia , Polimorfismo de Nucleotídeo Único/genética , Sinaptossomos/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas tau/antagonistas & inibidores
3.
Neurobiol Dis ; 35(2): 193-200, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19379814

RESUMO

Accumulating evidence implicates deregulation of GSK3ss as a converging pathological event in Alzheimer's disease and in neuropsychiatric disorders, including bipolar disorder and schizophrenia. Although these neurological disorders share cognitive dysfunction as a hallmark, the role of GSK3ss in learning and memory remains to be explored in depth. We here report increased phosphorylation of GSK3ss at Serine-9 following cognitive training in two different hippocampus dependent cognitive tasks, i.e. inhibitory avoidance and novel object recognition task. Conversely, transgenic mice expressing the phosphorylation defective mutant GSK3ss[S9A] show impaired memory in these tasks. Furthermore, GSK3ss[S9A] mice displayed impaired hippocampal L-LTP and facilitated LTD. Application of actinomycin, but not anisomycin, mimicked GSK3ss[S9A] induced defects in L-LTP, suggesting that transcriptional activation is affected. This was further supported by decreased expression of the immediate early gene c-Fos, a target gene of CREB. The combined data demonstrate a role for GSK3ss in long term memory formation, by inhibitory phosphorylation at Serine-9. The findings are fundamentally important and relevant in the search for therapeutic strategies in neurological disorders associated with cognitive impairment and deregulated GSK3ss signaling, including AD, bipolar disorder and schizophrenia.


Assuntos
Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Memória/fisiologia , Animais , Aprendizagem da Esquiva/fisiologia , Cognição/fisiologia , Regulação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Potenciais da Membrana/fisiologia , Transtornos Mentais/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Fosforilação , Prática Psicológica , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reconhecimento Psicológico/fisiologia
4.
Neurobiol Aging ; 30(2): 241-56, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17673336

RESUMO

Evidence is accumulating for a role for amyloid peptides in impaired synaptic plasticity and cognition, while the underlying mechanisms remain unclear. We here analyzed the effects of amyloid peptides on NMDA-receptor function in vitro and in vivo. A synthetic amyloid peptide preparation containing monomeric and oligomeric A beta (1-42) peptides was used and demonstrated to bind to synapses expressing NMDA-receptors in cultured hippocampal and cortical neurons. Pre-incubation of primary neuronal cultures with A beta peptides significantly inhibited NMDA-receptor function, albeit not by a direct pharmacological inhibition of NMDA-receptors, since acute application of A beta peptides did not change NMDA-receptor currents in autaptic hippocampal cultures nor in xenopus oocytes expressing recombinant NMDA-receptors. Pre-incubation of primary neuronal cultures with A beta peptides however decreased NR2B-immunoreactive synaptic spines and surface expression of NR2B containing NMDA-receptors. Furthermore, we extended these findings for the first time in vivo, demonstrating decreased concentrations of NMDA-receptor subunit NR2B and PSD-95 as well as activated alpha-CaMKII in postsynaptic density preparations of APP[V717I] transgenic mice. This was associated with impaired NMDA-dependent LTP and decreased NMDA- and AMPA-receptor currents in hippocampal CA1 region in APP[V717I] transgenic mice. In addition, induction of c-Fos following cued and contextual fear conditioning was significantly impaired in the basolateral amygdala and hippocampus of APP[V717I] transgenic mice. Our data demonstrate defects in NMDA-receptor function and learning dependent signaling cascades in vivo in APP[V717I] transgenic mice and point to decreased surface expression of NMDA-receptors as a mechanism involved in early synaptic defects in APP[V717I] transgenic mice in vivo.


Assuntos
Peptídeos beta-Amiloides/administração & dosagem , Precursor de Proteína beta-Amiloide/genética , Neurônios/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Receptores de Superfície Celular/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Nexinas de Proteases , Transdução de Sinais/efeitos dos fármacos
5.
Genes Brain Behav ; 7 Suppl 1: 57-66, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18184370

RESUMO

Phosphorylation is the most common post-translational modification of cellular proteins, essential for most physiological functions. Deregulation of phosphorylation has been invoked in disease mechanisms, and the case of Alzheimer's disease (AD) is no exception: both in the amyloid pathology and in the tauopathy are kinases deeply implicated. The glycogen synthase kinase-3 (GSK-3) isozymes participate in diverse cellular processes and important signalling pathways and have been implicitly linked to diverse medical problems, i.e. from diabetes and cancer to mood disorders and schizophrenia, and in the neurodegeneration of AD. Here, we review specific aspects of GSK-3 isozymes in the framework of recent data that we obtained in novel transgenic mouse models that robustly recapitulate the pathology and mechanistical problems of AD.


Assuntos
Doença de Alzheimer/etiologia , Amiloide/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Tauopatias/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Humanos , Isoenzimas/metabolismo , Lítio/farmacologia , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas tau/metabolismo
6.
Neurobiol Aging ; 29(5): 639-52, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17222948

RESUMO

The function of presenilin1 (PS1) in intra-membrane proteolysis is undisputed, as is its role in neurodegeneration in FAD, in contrast to its exact function in normal conditions. In this study, we analyzed synaptic plasticity and its underlying mechanisms biochemically in brain of mice with a neuron-specific deficiency in PS1 (PS1(n-/-)) and compared them to mice that expressed human mutant PS1[A246E] or wild-type PS1. PS1(n-/-) mice displayed a subtle impairment in Schaffer collateral hippocampal long-term potentiation (LTP) as opposed to normal LTP in wild-type PS1 mice, and a facilitated LTP in mutant PS1[A246E] mice. This finding correlated with, respectively, increased and reduced NMDA receptor responses in PS1[A246E] mice and PS1(n-/-) mice in hippocampal slices. Postsynaptically, levels of NR1/NR2B NMDA-receptor subunits and activated alpha-CaMKII were reduced in PS1(n-/-) mice, while increased in PS1[A246E] mice. In addition, PS1(n-/-) mice, displayed reduced paired pulse facilitation, increased synaptic fatigue and lower number of total and docked synaptic vesicles, implying a presynaptic function for wild-type presenilin1, unaffected by the mutation in PS1[A246E] mice. In contrast to the deficiency in PS1, mutant PS1 activated GSK-3beta by decreasing phosphorylation on Ser-9, which correlated with increased phosphorylation of protein tau at Ser-396-Ser-404 (PHF1/AD2 epitope). The synaptic functions of PS1, exerted on presynaptic vesicles and on postsynaptic NMDA-receptor activity, were concluded to be independent of alterations in GSK-3beta activity and phosphorylation of protein tau.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Presenilina-1/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia , Proteínas tau/metabolismo , Animais , Células Cultivadas , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Neurônios/citologia , Fosforilação , Presenilina-1/genética
7.
Br J Pharmacol ; 150(7): 883-92, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17325649

RESUMO

BACKGROUND AND PURPOSE: A strategy to treat Alzheimer's disease (AD) is to increase the soluble form of amyloid precursor protein (sAPPalpha), a promnesic protein, in the brain. Because strong evidence supports beneficial effects of 5-hydroxytryptamine 5-HT(4) receptor agonists in memory and learning, we investigated the role of 5-HT(4) receptors on APP processing in 8 weeks-old male C57BL/6j mice. EXPERIMENTAL APPROACH: Mice were given, subcutaneously, prucalopride or ML 10302 (s.c.), two highly selective 5-HT(4) receptor agonists and, up to 240 min later, the hippocampus and cortex were analysed by Western blot for sAPPalpha determination. KEY RESULTS: Prucalopride (5 or 10 mg kg(-1)) significantly increased sAPPalpha levels in the hippocampus and cortex, but did not modify the expression level of APP mRNA as detected by quantitative RT-PCR. A selective 5-HT(4) receptor antagonist, GR125487 (1 mg kg(-1), s.c.) inhibited prucalopride induced- increase in sAPPalpha levels. In addition, levels of sAPPalpha were increased by ML10302 only at 20 mg kg(-1) and was limited to the cortex. Also, prucalopride increased sAPPalpha levels in the cortex of a transgenic mouse model of AD, expressing the London mutation of APP. Furthermore, the combined injection of a selective acetylcholinesterase inhibitor, donepezil and prucalopride induced a synergic increase in sAPPalpha levels in the cortex and hippocampus. CONCLUSIONS AND IMPLICATIONS: Our results demonstrate that the 5-HT(4) receptor plays a key role in the non-amyloidogenic pathway of APP metabolism in vivo and give support to the beneficial use of 5-HT(4) agonists for AD treatment.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Agonistas do Receptor 5-HT4 de Serotonina , Aminobenzoatos/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Benzofuranos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Inibidores da Colinesterase/farmacologia , Donepezila , Hipocampo/efeitos dos fármacos , Indanos/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Piperidinas/farmacologia , RNA Mensageiro/metabolismo , Antagonistas do Receptor 5-HT4 de Serotonina , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Sulfonamidas/farmacologia , para-Aminobenzoatos
8.
Rev Neurol (Paris) ; 162(10): 903-7, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17028556

RESUMO

Describing and understanding the pathological processes which devastate the brain of Alzheimer's disease (AD) patients remains a major target for experimental biology. We approached this problem by generating different types of single and double transgenic mice that develop pathological hallmarks of AD. In APP-V717 mice, the progression from intracellular amyloid to diffuse and senile plaques with vascular deposits, is preceded by early defects in cognition and LTP. In Tau-P301L mice, the morbid tauopathy with intracellular filaments, cause mortality before age 1 year. Ageing APP-V717IxTau-P301L double tg mice (14-17 months) have combined AD-like pathology in hippocampus and cortex consisting of amyloid plaques and neurofibrillary tangles. Remarkably, while Tau-P301L mice die before age 1 year, the APP-V717IxTau-P301L double tg mice survive much longer, which correlates with alleviation of tauopathy in hindbrain, despite aggravation in forebrain. This hypothesis is corroborated in Tau-P301LxGSK-3B double transgenic mice, which have also an extended lifespan relative to Tau-P301L mice, that correlates with reduction of brainstem tauopathy. At the same time, Tau-P301LxGSK-3B mice have dramatic forebrain tauopathy, with "tangles in almost all neurons", although without hyper-phosphorylation of Tau. The data corroborate the hypothesis that GSK-3B is the missing link between the amyloid and tau-pathology, and position GSK-3B as prominent player in the pathogenesis in AD.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Quinase 3 da Glicogênio Sintase/genética , Proteínas tau/genética , Envelhecimento/genética , Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Substituição de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Modelos Animais de Doenças , Endopeptidases/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Camundongos Transgênicos
9.
Magn Reson Med ; 53(3): 607-13, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15723413

RESUMO

Transgenic mice overexpressing the London mutant of human amyloid precursor protein (APP[V717I]) in neurons develop amyloid plaques in the brain, thus demonstrating the most prominent neuropathological hallmark of Alzheimer's disease. In vivo 3D T2*-weighted MRI on these mice (24 months of age) revealed hypointense brain inclusions that affected the thalamus almost exclusively. Upon correlating these MRI observations with a panel of different histologic staining techniques, it appeared that only plaques that were positive for both thioflavin-S and iron were visible on the MR images. Numerous thioflavin-S-positive plaques in the cortex that did not display iron staining remained invisible to MRI. The in vivo detection of amyloid plaques in this mouse model, using the intrinsic MRI contrast arising from the iron associated with the plaques, creates an unexpected opportunity for the noninvasive investigation of the longitudinal development of the plaques in the same animal. Thus, this work provides further research opportunities for analyzing younger APP[V717I] mouse models with the knowledge of the final outcome at 24 months of age.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/fisiologia , Ferro/metabolismo , Imageamento por Ressonância Magnética/métodos , Proteínas de Membrana/fisiologia , Placa Amiloide/patologia , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo , Coloração e Rotulagem
10.
Biochem Soc Symp ; (67): 203-10, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11447836

RESUMO

We have reported transgenic mice with neuronal overexpression of the clinical mutant beta-amyloid precursor protein (APP) known as London, which develop an AD-related phenotype [Moechers, Dewachter, Lorent, Reversé, Baekelandt, Nadiu, Tesseur, Spittaels, Van den Haute, Checler, et al. (1999) J. Biol. Chem. 274, 6483-6492]. Characterized early symptoms (3-9 months) include disturbed behaviour, neophobia, aggression, hypersensitivity to kainic acid, hyposensitivity to N-methyl-D-aspartate, defective cognition and memory, and decreased long-term potentiation. Late in life, at 12-15 months, amyloid plaques develop in the brain and correlate with increased levels of beta-amyloid (A beta)40/42 (the 40- and 42-amino-acid forms of A beta). The formation of amyloid plaques is dissociated in time from and not involved in the early phenotype. Hyperphosphorylated protein tau is present but no tangle pathology is observed. In double-transgenic mice, i.e. APP/London x Presenilin 1, the increased production of A beta 42 results in amyloid plaques developing by the age of 6 months. Transgenic mice with overexpression of either human apolipoprotein E4 (ApoE4) or human protein tau in central neurons develop severe axonopathy in the brain and spinal cord. Progressive degeneration of nerves and muscles is demonstrated by motor problems, wasting and premature death. Tau is hyperphosphorylated but there is no formation of filaments or neurofibrillary tangles. The tangle aspect of AD pathology is still missing from all current transgenic amyloid models. Its implementation will require insight into the cellular signalling pathways which regulate the microtubule-stabilizing function by phosphorylation of neuronal tau.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/genética , Animais , Apolipoproteína E4 , Apolipoproteínas E/genética , Modelos Animais de Doenças , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Mutação , Degeneração Neural/etiologia , Degeneração Neural/genética , Presenilina-1 , Proteínas tau/genética
11.
J Biol Chem ; 276(15): 11539-44, 2001 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-11278803

RESUMO

Mutant human presenilin-1 (PS1) causes an Alzheimer's-related phenotype in the brain of transgenic mice in combination with mutant human amyloid precursor protein by means of increased production of amyloid peptides (Dewachter, I., Van Dorpe, J., Smeijers, L., Gilis, M., Kuiperi, C., Laenen, I., Caluwaerts, N., Moechars, D., Checler, F., Vanderstichele, H. & Van Leuven, F. (2000) J. Neurosci. 20, 6452-6458) that aggravate plaques and cerebrovascular amyloid (Van Dorpe, J., Smeijers, L., Dewachter, I., Nuyens, D., Spittaels, K., van den Haute, C., Mercken, M., Moechars, D., Laenen, I., Kuipéri, C., Bruynseels, K., Tesseur, I., Loos, R., Vanderstichele, H., Checler, F., Sciot, R. & Van Leuven, F. (2000) J. Am. Pathol. 157, 1283-1298). This gain of function of mutant PS1 is approached here in three paradigms that relate to glutamate neurotransmission. Mutant but not wild-type human PS1 (i) lowered the excitotoxic threshold for kainic acid in vivo, (ii) facilitated hippocampal long-term potentiation in brain slices, and (iii) increased glutamate-induced intracellular calcium levels in isolated neurons. Prominent higher calcium responses were triggered by thapsigargin and bradykinin, indicating that mutant PS modulates the dynamic release and storage of calcium ions in the endoplasmatic reticulum. In reaction to glutamate, overfilled Ca(2+) stores resulted in higher than normal cytosolic Ca(2+) levels, explaining the facilitated long-term potentiation and enhanced excitotoxicity. The lowered excitotoxic threshold for kainic acid was also observed in mice transgenic for mutant human PS2[N141I] and was prevented by dantrolene, an inhibitor of Ca(2+) release from the endoplasmic reticulum.


Assuntos
Cálcio/metabolismo , Hipocampo/metabolismo , Homeostase/genética , Potenciação de Longa Duração/genética , Proteínas de Membrana/fisiologia , Mutação , Animais , Bradicinina/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Ácido Caínico/farmacologia , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Presenilina-1
12.
Exp Gerontol ; 35(6-7): 831-41, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11053674

RESUMO

In transgenic mice that overexpress mutant Amyloid Precursor Protein [V717I], or APP/London (APP/Lo) (1999a. Early phenotypic changes in transgenic mice that overexpress different mutants of Amyloid Precursor Protein in brain. J. Biol. Chem. 274, 6483-6492; 1999b. Premature death in transgenic mice that overexpress mutant Amyloid precursor protein is preceded by severe neurodegeneration and apoptosis. Neuroscience 91, 819-830) the AD related phenotype of plaque and vascular amyloid pathology is late (12-15 months). This typical and diagnostic pathology is thereby dissociated in time from early symptoms (3-9 months) that include disturbed behavior, neophobia, aggression, glutamate excitotoxicity, defective cognition and decreased LTP. The APP/Lo transgenic mice are therefore a very interesting model to study early as well as late pathology, including the effect of age. In ageing APP*Lo mice, brain soluble and especially "insoluble" amyloid peptides dramatically increased, while normalized levels of secreted APPsalpha and APPsbeta, as well as cell-bound beta-C-stubs, remained remarkably constant, indicating normal alpha- and beta-secretase processing of APP. In double transgenic mice, i.e. APP/LoxPS1, clinical mutant PS1[A246E] but not wild-type human PS1 increased Abeta, and plaques and vascular amyloid developed at age 6-9 months. The PS1 mutant caused increasing Abeta42 production, while ageing did not. Amyloid deposits are thus formed, not by overproduction of Abeta, but by lack of clearance and/or degradation in the brain of ageing APP/Lo transgenic mice. The clearance pathways of the cerebral amyloid peptides are therefore valuable targets for fundamental research and for therapeutic potential. Although hyper-phosphorylated protein tau was evident in swollen neurites around the amyloid plaques, neurofibrillary pathology is not observed and the "tangle" aspect of AD pathology is therefore still missing from all current transgenic "amyloid" models. Also the "ApoE4" risk for late onset AD remains a problem for modeling in transgenic mice. We have generated transgenic mice that overexpress human ApoE4 (2000. Expression of Human Apolipoprotein E4 in neurons causes hyperphosphorylation of Protein tau in the brains of transgenic mice. Am. J. Pathol. 156 (3) 951-964) or human protein tau (1999. Prominent axonopathy in the brain and spinal cord of transgenic mice overexpressing four-repeat human tau protein. Am. J. Pathol. 155, 2153-2165) in their neurons. Both develop a similar although not identical axonopathy, with progressive degeneration of nerves and with muscle wasting resulting in motoric problems. Remarkably, ApoE4 transgenic mice are, like the tau transgenic mice, characterized by progressive hyper-phosphorylation of protein tau also in motor neurons which explains the motoric defects. Further crossing with the APP/Lo transgenic mice is ongoing to yield "multiple" transgenic mouse strains to study new aspects of amyloid and tau pathology.


Assuntos
Envelhecimento , Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/fisiologia , Modelos Animais de Doenças , Proteínas de Membrana/fisiologia , Animais , Apolipoproteína E4 , Apolipoproteínas E/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Presenilina-1 , Proteínas tau/fisiologia
13.
Am J Pathol ; 157(4): 1283-98, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11021833

RESUMO

Deposition of amyloid beta-peptide (Abeta) in cerebral vessel walls (cerebral amyloid angiopathy, CAA) is very frequent in Alzheimer's disease and occurs also as a sporadic disorder. Here, we describe significant CAA in addition to amyloid plaques, in aging APP/Ld transgenic mice overexpressing the London mutant of human amyloid precursor protein (APP) exclusively in neurons. The number of amyloid-bearing vessels increased with age, from approximately 10 to >50 per coronal brain section in APP/Ld transgenic mice, aged 13 to 24 months. Vascular amyloid was preferentially deposited in arterioles and ranged from small focal to large circumferential depositions. Ultrastructural analysis allowed us to identify specific features contributing to weakening of the vessel wall and aneurysm formation, ie, disruption of the external elastic lamina, thinning of the internal elastic lamina, interruption of the smooth muscle layer, and loss of smooth muscle cells. Biochemically, the much lower Abeta42:Abeta40 ratio evident in vascular relative to plaque amyloid, demonstrated that in blood vessel walls Abeta40 was the more abundant amyloid peptide. The exclusive neuronal origin of transgenic APP, the high levels of Abeta in cerebrospinal fluid compared to plasma, and the specific neuroanatomical localization of vascular amyloid strongly suggest specific drainage pathways, rather than local production or blood uptake of Abeta as the primary mechanism underlying CAA. The demonstration in APP/Ld mice of rare vascular amyloid deposits that immunostained only for Abeta42, suggests that, similar to senile plaque formation, Abeta42 may be the first amyloid to be deposited in the vessel walls and that it entraps the more soluble Abeta40. Its ability to diffuse for larger distances along perivascular drainage pathways would also explain the abundance of Abeta40 in vascular amyloid. Consistent with this hypothesis, incorporation of mutant presenilin-1 in APP/Ld mice, which resulted in selectively higher levels of Abeta42, caused an increase in CAA and senile plaques. This mouse model will be useful in further elucidating the pathogenesis of CAA and Alzheimer's disease, and will allow testing of diagnostic and therapeutic strategies.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/patologia , Expressão Gênica , Mutação/fisiologia , Envelhecimento/fisiologia , Amiloide/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Vasos Sanguíneos/ultraestrutura , Angiopatia Amiloide Cerebral/metabolismo , Circulação Cerebrovascular , Humanos , Hipercapnia/fisiopatologia , Imuno-Histoquímica , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Presenilina-1 , Transgenes/fisiologia
14.
J Neurosci ; 20(17): 6452-8, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10964951

RESUMO

Aging of transgenic mice that overexpress the London mutant of amyloid precursor protein (APP/V717I) (Moechars et al., 1999a) was now demonstrated not to affect the normalized levels of alpha- or beta-cleaved secreted APP nor of the beta-C-terminal stubs. This indicated that aging did not markedly disturb either alpha- or beta-secretase cleavage of APP and failed to explain the origin of the massive amounts of amyloid peptides Abeta40 and Abeta42, soluble and precipitated as amyloid plaques in the brain of old APP/V717I transgenic mice. We tested the hypothesis that aging acted on presenilin1 (PS1) to affect gamma-secretase-mediated production of amyloid peptides by comparing aged APP/V717I transgenic mice to double transgenic mice coexpressing human PS1 and APP/V717I. In double transgenic mice with mutant (A246E) but not wild-type human PS1, brain amyloid peptide levels increased and resulted in amyloid plaques when the mice were only 6-9 months old, much earlier than in APP/V717I transgenic mice (12-15 months old). Mutant PS1 increased mainly brain Abeta42 levels, whereas in aged APP/V717I transgenic mice, both Abeta42 and Abeta40 increased. This resulted in a dramatic difference in the Abeta42/Abeta40 ratio of precipitated or plaque-associated amyloid peptides, i.e., 3.11+/-0.22 in double APP/V717I x PS1/A246E transgenic mice compared with 0.43 +/- 0.07 in aged APP/V717I transgenic mice, and demonstrated a clear difference between the effect of aging and the effect of the insertion of a mutant PS1 transgene. In conclusion, we demonstrate that aging did not favor amyloidogenic over nonamyloidogenic processing of APP, nor did it exert a mutant PS1-like effect on gamma-secretase. Therefore, the data are interpreted to suggest that parenchymal and vascular accumulation of amyloid in aging brain resulted from failure to clear the amyloid peptides rather than from increased production.


Assuntos
Envelhecimento , Peptídeos beta-Amiloides/fisiologia , Encéfalo/metabolismo , Proteínas de Membrana/fisiologia , Substituição de Aminoácidos , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Heterozigoto , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Mutação Puntual , Presenilina-1
15.
Neurobiol Dis ; 7(1): 9-22, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10671319

RESUMO

The contribution of mutations in the amyloid precursor protein (APP) gene known as Flemish (APP/A692G) and Dutch (APP/E693Q) to the pathogenesis of Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis of the Dutch type, respectively, was studied in transgenic mice that overexpress the mutant APP in brain. These transgenic mice showed the same early behavioral disturbances and defects and increased premature death as the APP/London (APP V717I), APP/Swedish (K670N, M671L), and other APP transgenic mice described previously. Pathological changes included intense glial reaction, extensive microspongiosis in the white matter, and apoptotic neurons in select areas of the brain, while amyloid deposits were absent, even in mice over 18 months of age. This contrasts with extensive amyloid deposition in APP/London transgenic mice and less pronounced amyloid deposition in APP/Swedish transgenic mice generated identically. It demonstrated, however, that the behavioral deficiencies and the pathological changes in brain resulting from an impaired neuronal function are caused directly by APP or its proteolytic derivative(s). These accelerate or impinge on the normal process of aging and amyloid deposits per se are not essential for this phenotype.


Assuntos
Agressão , Precursor de Proteína beta-Amiloide/genética , Encéfalo/patologia , Mutação Puntual , Doença de Alzheimer/genética , Amiloide/análise , Animais , Encéfalo/ultraestrutura , Núcleo Celular/patologia , Núcleo Celular/ultraestrutura , Angiopatia Amiloide Cerebral/genética , Humanos , Ácido Caínico/toxicidade , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , N-Metilaspartato/toxicidade
16.
Neuroscience ; 94(1): 315-21, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10613521

RESUMO

The role of the receptor-associated protein in controlling the expression of the low-density lipoprotein receptor-related protein was analysed in brain and in cultured neurons of receptor-associated protein - / - mice. In addition, the effect of two important ligands of lipoprotein receptor-related protein in brain, i.e. apolipoprotein E and amyloid precursor protein, was examined by crossing the receptor-associated protein - / - mice with transgenic mice overexpressing these proteins specifically in neurons. The immunohistochemical localization of lipoprotein receptor-related protein and receptor-associated protein in wild-type mouse brain was demonstrated to be congruent over all structures, including the cortex and hippocampus. In primary hippocampal neurons, lipoprotein receptor-related protein was distributed somatodendritically and receptor-associated protein was concentrated perinuclearly. In hippocampal neurons from receptor-associated protein - / - mice, lipoprotein receptor-related protein was redistributed over the cell body at the expense of the dendrites. In the absence of receptor-associated protein, maturation of lipoprotein receptor-related protein is slow, resulting in accumulation of the uncleaved 600,000 mol. wt precursor. Neither the added expression of apolipoprotein E4 nor that of amyloid precursor protein in cultured neurons influenced the maturation of lipoprotein receptor-related protein, in either the presence or absence of receptor-associated protein. This result shows that receptor-associated protein is not needed to allow co-expression of lipoprotein receptor-related protein with these ligands in neurons. Furthermore, the typical ramified neuronal morphology of cultured primary neurons and the histology and architecture of the brain were normal in receptor-associated protein - / - mice and in all of the double transgenic mice. Finally, we demonstrated that the survival of receptor-associated protein - /- hippocampal neurons was normal and unaffected by the genotype of the glial feeder cells, whether they were derived from wild-type mice or from mice deficient in receptor-associated protein or apolipoprotein E. These results show that, despite the dramatic effect on maturation and cellular localization of lipoprotein receptor-related protein, the absence of receptor-associated protein did not result in any notable physiological, functional or morphological effects.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Apolipoproteínas E/genética , Proteínas de Transporte/genética , Glicoproteínas/genética , Hipocampo/química , Neurônios/química , Receptores de LDL/genética , Animais , Apolipoproteína E4 , Química Encefálica/genética , Proteínas de Transporte/análise , Sobrevivência Celular/fisiologia , Células Cultivadas , Expressão Gênica/fisiologia , Genótipo , Glicoproteínas/análise , Hipocampo/citologia , Proteína Associada a Proteínas Relacionadas a Receptor de LDL , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/fisiologia , Receptores de LDL/análise
17.
J Biol Chem ; 274(10): 6483-92, 1999 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-10037741

RESUMO

Transgenic mice overexpressing different forms of amyloid precursor protein (APP), i.e. wild type or clinical mutants, displayed an essentially comparable early phenotype in terms of behavior, differential glutamatergic responses, deficits in maintenance of long term potentiation, and premature death. The cognitive impairment, demonstrated in F1 hybrids of the different APP transgenic lines, was significantly different from nontransgenic littermates as early as 3 months of age. Biochemical analysis of secreted and membrane-bound APP, C-terminal "stubs," and Abeta(40) and Abeta(42) peptides in brain indicated that no single intermediate can be responsible for the complex of phenotypic dysfunctions. As expected, the Abeta(42) levels were most prominent in APP/London transgenic mice and correlated directly with the formation of amyloid plaques in older mice of this line. Plaques were associated with immunoreactivity for hyperphosphorylated tau, eventually signaling some form of tau pathology. In conclusion, the different APP transgenic mouse lines studied display cognitive deficits and phenotypic traits early in life that dissociated in time from the formation of amyloid plaques and will be good models for both early and late neuropathological and clinical aspects of Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Encéfalo/fisiologia , Camundongos Transgênicos/fisiologia , Mutação , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Eletrofisiologia , Expressão Gênica , Hipocampo/fisiologia , Camundongos
18.
Behav Brain Res ; 95(1): 55-64, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9754877

RESUMO

Expression of alpha-secretion mutant APP/RK in mouse brain results in a progressive disorganization of the central nervous system, exemplified by behavioral deficits, premature death and neuropathology. Here we report on the progressive nature of this CNS disorder as indicated by the age dependency of the neophobic reaction in the open-field test. The earlier reported NMDA hypo-sensitivity in the transgenic APP/RK mice is likely to represent a subtle functional disturbance, since no changes in NMDA receptor density or distribution could be detected. None of the typical neuropathological hallmarks of Alzheimer's Disease, i.e. amyloid deposits and neurofibrillary tangles are detected in the brain of these transgenic mice. Nevertheless, the progressive CNS disorder elicited in the transgenic APP/RK mice recapitulates certain features and symptoms of patients with Alzheimer's disease as discussed.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Química Encefálica/genética , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/metabolismo , Mutação/fisiologia , Animais , Autorradiografia , Western Blotting , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/fisiologia , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Coloração pela Prata
19.
EMBO J ; 15(6): 1265-74, 1996 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8635459

RESUMO

A double mutation in the alpha-secretase site in the betaA4 region of mouse amyloid precursor protein (APP) reduced its secretion from COS cells, polarized MDCK cells and rat primary neurons. Expression of this mutant in the brain of mice, using the neuron-specific elements of the mouse Thy-1 gene promoter, resulted in transgenic mice that became progressively hyperactive, displayed seizures and died prematurely. In three different transgenic lines the severity of the phenotype was related directly to the expression levels of the transgene, estimated by both mRNA and protein levels. In addition, homozygous mice derived from each transgenic strain showed more severe symptoms which also occurred earlier in life than in heterozygotes. The observed symptoms were, however, not essentially different in the different lines. Increased aggressiveness, disturbed responses to kainic acid and N-methyl-D-aspartate, neophobia and deficiency in exploratory behavior were demonstrated in these mice. In the brain, the observed neuropathological changes included necrosis, apoptosis and astrogliosis in the hippocampus, cortex and other areas. The data demonstrate that incomplete or incorrect alpha-secretase processing of APP results in severe neurotoxicity and that this effect is expressed in a dominant manner.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encefalopatias Metabólicas/psicologia , Endopeptidases/metabolismo , Mutação , Processamento de Proteína Pós-Traducional , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases , Comportamento Animal , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/mortalidade , Encefalopatias Metabólicas/patologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Hipercinese , Ácido Caínico/farmacologia , Camundongos , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Convulsões
20.
J Biol Chem ; 270(8): 4058-65, 1995 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-7876155

RESUMO

The analysis of potential sorting signals in amyloid precursor protein (APP) by site-directed mutagenesis and the disturbance of metabolic pathways by drugs is used here to define the parameters that determine polarized secretion of APP in Madin-Darby canine kidney cells. Endogenously produced APP751/770 and APP695 produced from transfected constructs are secreted almost exclusively into the basolateral compartment. The sorting mechanism is highly dependent on intracellular pH as demonstrated by its sensitivity to primary amines and inhibitors of the acidifying vacuolar protein ATPase. The role of potential basolateral sorting signals in the cytoplasmic, transmembrane, and beta A4 amyloid region of APP was investigated. Neither deletion of the endocytosis and putative basolateral sorting signal GY.NPTY nor complete deletion of the cytoplasmic domain causes apical secretion of soluble APP. Further deletion of the transmembrane domain and of the beta A4 amyloid region confirmed that the major basolateral sorting determinant resides in the extracellular domain of APP. Increased beta-secretase cleavage of APP after introduction of the "swedish" double mutation causes apical missorting of about 20% of beta-secretase-cleaved APP. The data underline the complexity of processing and sorting APP in polarized cells and suggest a possible problem of protein sorting in Alzheimer's Disease.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Rim/metabolismo , Mutação , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases , Compartimento Celular , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Cães , Endopeptidases/metabolismo , Concentração de Íons de Hidrogênio , Camundongos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...