Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neurosci ; 34(28): 9441-54, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009275

RESUMO

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common neurodegenerative disorders of the aging population, characterized by progressive and abnormal accumulation of α-synuclein (α-syn). Recent studies have shown that C-terminus (CT) truncation and propagation of α-syn play a role in the pathogenesis of PD/DLB. Therefore, we explored the effect of passive immunization against the CT of α-syn in the mThy1-α-syn transgenic (tg) mouse model, which resembles the striato-nigral and motor deficits of PD. Mice were immunized with the new monoclonal antibodies 1H7, 5C1, or 5D12, all directed against the CT of α-syn. CT α-syn antibodies attenuated synaptic and axonal pathology, reduced the accumulation of CT-truncated α-syn (CT-α-syn) in axons, rescued the loss of tyrosine hydroxylase fibers in striatum, and improved motor and memory deficits. Among them, 1H7 and 5C1 were most effective at decreasing levels of CT-α-syn and higher-molecular-weight aggregates. Furthermore, in vitro studies showed that preincubation of recombinant α-syn with 1H7 and 5C1 prevented CT cleavage of α-syn. In a cell-based system, CT antibodies reduced cell-to-cell propagation of full-length α-syn, but not of the CT-α-syn that lacked the 118-126 aa recognition site needed for antibody binding. Furthermore, the results obtained after lentiviral expression of α-syn suggest that antibodies might be blocking the extracellular truncation of α-syn by calpain-1. Together, these results demonstrate that antibodies against the CT of α-syn reduce levels of CT-truncated fragments of the protein and its propagation, thus ameliorating PD-like pathology and improving behavioral and motor functions in a mouse model of this disease.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Encéfalo/imunologia , Transtornos dos Movimentos/imunologia , Transtornos dos Movimentos/terapia , Transtornos Parkinsonianos/imunologia , Transtornos Parkinsonianos/terapia , alfa-Sinucleína/imunologia , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Transgênicos , Distribuição Tecidual , Resultado do Tratamento
2.
Am J Pathol ; 182(3): 940-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23313024

RESUMO

Progressive accumulation of α-synuclein (α-syn) in limbic and striatonigral systems is associated with the neurodegenerative processes in dementia with Lewy bodies (DLB) and Parkinson's disease (PD). The murine Thy-1 (mThy1)-α-syn transgenic (tg) model recapitulates aspects of degenerative processes associated with α-syn accumulation in these disorders. Given that axonal and synaptic pathologies are important features of DLB and PD, we sought to investigate the extent and characteristics of these alterations in mThy1-α-syn tg mice and to determine the contribution of α-syn c-terminally cleaved at amino acid 122 (CT α-syn) to these abnormalities. We generated a novel polyclonal antibody (SYN105) against the c-terminally truncated sequence (amino acids 121 to 123) of α-syn (CT α-syn) and performed immunocytochemical and ultrastructural analyses in mThy1-α-syn tg mice. We found abundant clusters of dystrophic neurites in layers 2 to 3 of the neocortex, the stratum lacunosum, the dentate gyrus, and cornu ammonis 3 of the hippocampus, striatum, thalamus, midbrain, and pons. Dystrophic neurites displayed intense immunoreactivity detected with the SYN105 antibody. Double-labeling studies with antibodies to phosphorylated neurofilaments confirmed the axonal location of full-length and CT α-syn. α-Syn immunoreactive dystrophic neurites contained numerous electrodense laminated structures. These results show that neuritic dystrophy is a prominent pathologic feature of the mThy1-α-syn tg model and suggest that CT α-syn might play an important role in the process of axonal damage in these mice as well as in DLB and PD.


Assuntos
Axônios/patologia , Doença por Corpos de Lewy/patologia , Proteínas Mutantes/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Axônios/metabolismo , Axônios/ultraestrutura , Biomarcadores/metabolismo , Demografia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neuritos/metabolismo , Neuritos/patologia , Neuritos/ultraestrutura , Transporte Proteico , Sinapses/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura , Antígenos Thy-1/metabolismo , alfa-Sinucleína/imunologia
3.
J Neurosci ; 32(8): 2696-702, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22357853

RESUMO

Several anti-amyloid ß (Aß) antibodies are under evaluation for the treatment of Alzheimer's disease (AD). Clinical studies using the N-terminal-directed anti-Aß antibody bapineuzumab have demonstrated reduced brain PET-Pittsburg-B signals, suggesting the reduction of Aß plaques, and reduced levels of total and phosphorylated tau protein in the CSF of treated AD patients. Preclinical studies using 3D6 (the murine form of bapineuzumab) have demonstrated resolution of Aß plaque and vascular burdens, neuritic dystrophy, and preservation of synaptic density in the transgenic APP mouse models. In contrast, few studies have evaluated the direct interaction of this antibody with synaptotoxic soluble Aß species. In the current report, we demonstrated that 3D6 binds to soluble, synaptotoxic assemblies of Aß(1-42) and prevents multiple downstream functional consequences in rat hippocampal neurons including changes in glutamate AMPA receptor trafficking, AD-type tau phosphorylation, and loss of dendritic spines. In vivo, we further demonstrated that 3D6 prevents synaptic loss and acutely reverses the behavioral deficit in the contextual fear conditioning task in transgenic mouse models of AD, two endpoints thought to be linked to synaptotoxic soluble Aß moieties. Importantly C-terminal anti-Aß antibodies were ineffective on these endpoints. These results, taken with prior studies, suggest that N-terminal anti-Aß antibodies effectively interact with both soluble and insoluble forms of Aß and therefore appear particularly well suited for testing the Aß hypothesis of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/imunologia , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Epitopos/imunologia , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Anticorpos Neutralizantes , Sintomas Comportamentais/tratamento farmacológico , Sintomas Comportamentais/etiologia , Sintomas Comportamentais/imunologia , Biotina/metabolismo , Células Cultivadas , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Espinhas Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Embrião de Mamíferos , Epitopos/metabolismo , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/imunologia , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/imunologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/imunologia , Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/imunologia , Fosforilação , Ligação Proteica/imunologia , Estrutura Secundária de Proteína , Transporte Proteico/efeitos dos fármacos , Ratos , Receptores de AMPA/metabolismo , Solubilidade , Proteína Vesicular 1 de Transporte de Glutamato/imunologia , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
4.
Alzheimers Dement ; 9(5 Suppl): S105-15, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23583235

RESUMO

BACKGROUND: Clinical studies of ß-amyloid (Aß) immunotherapy in Alzheimer's disease (AD) patients have demonstrated reduction of central Aß plaque by positron emission tomography (PET) imaging and the appearance of amyloid-related imaging abnormalities (ARIA). To better understand the relationship between ARIA and the pathophysiology of AD, we undertook a series of studies in PDAPP mice evaluating vascular alterations in the context of central Aß pathology and after anti-Aß immunotherapy. METHODS: We analyzed PDAPP mice treated with either 3 mg/kg/week of 3D6, the murine form of bapineuzumab, or isotype control antibodies for periods ranging from 1 to 36 weeks and evaluated the vascular alterations in the context of Aß pathology and after anti-Aß immunotherapy. The number of mice in each treatment group ranged from 26 to 39 and a total of 345 animals were analyzed. RESULTS: The central vasculature displayed morphological abnormalities associated with vascular Aß deposits. Treatment with 3D6 antibody induced clearance of vascular Aß that was spatially and temporally associated with a transient increase in microhemorrhage and in capillary Aß deposition. Microhemorrhage resolved over a time period that was associated with a recovery of vascular morphology and a decrease in capillary Aß accumulation. CONCLUSIONS: These data suggest that vascular leakage events, such as microhemorrhage, may be related to the removal of vascular Aß. With continued treatment, this initial susceptibility period is followed by restoration of vascular morphology and reduced vulnerability to further vascular leakage events. The data collectively suggested a vascular amyloid clearance model of ARIA, which accounts for the currently known risk factors for the incidence of ARIA in clinical studies.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Vasos Sanguíneos/patologia , Encéfalo/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Aquaporina 4/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestrutura , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hemorragias Intracranianas/etiologia , Meninges/patologia , Meninges/ultraestrutura , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Fatores de Tempo
5.
Neuron ; 46(6): 857-68, 2005 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15953415

RESUMO

Abnormal folding of alpha-synuclein (alpha-syn) is thought to lead to neurodegeneration and the characteristic symptoms of Lewy body disease (LBD). Since previous studies suggest that immunization might be a potential therapy for Alzheimer's disease, we hypothesized that immunization with human (h)alpha-syn might have therapeutic effects in LBD. For this purpose, halpha-syn transgenic (tg) mice were vaccinated with halpha-syn. In mice that produced high relative affinity antibodies, there was decreased accumulation of aggregated halpha-syn in neuronal cell bodies and synapses that was associated with reduced neurodegeneration. Furthermore, antibodies produced by immunized mice recognized abnormal halpha-syn associated with the neuronal membrane and promoted the degradation of halpha-syn aggregates, probably via lysosomal pathways. Similar effects were observed with an exogenously applied FITC-tagged halpha-syn antibody. These results suggest that vaccination is effective in reducing neuronal accumulation of halpha-syn aggregates and that further development of this approach might have a potential role in the treatment of LBD.


Assuntos
Modelos Animais de Doenças , Imunização/métodos , Proteínas do Tecido Nervoso/imunologia , Doença de Parkinson/imunologia , Doença de Parkinson/terapia , Animais , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Western Blotting/métodos , Catepsina D/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Diagnóstico por Imagem/métodos , Mapeamento de Epitopos/métodos , Humanos , Imuno-Histoquímica/métodos , Corpos de Inclusão/metabolismo , Lisossomos/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Proteínas do Tecido Nervoso/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Terminações Pré-Sinápticas/metabolismo , Frações Subcelulares/metabolismo , Sinaptofisina/metabolismo , Sinucleínas , alfa-Sinucleína
6.
J Neurosci ; 28(27): 6787-93, 2008 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-18596154

RESUMO

In addition to parenchymal amyloid-beta (Abeta) plaques, Alzheimer's disease (AD) is characterized by Abeta in the cerebral vasculature [cerebral amyloid angiopathy (CAA)] in the majority of patients. Recent studies investigating vascular Abeta (VAbeta) in amyloid precursor protein transgenic mice have suggested that passive immunization with anti-Abeta antibodies may clear parenchymal amyloid but increase VAbeta and the incidence of microhemorrhage. However, the influences of antibody specificity and exposure levels on VAbeta and microhemorrhage rates have not been well established, nor has any clear causal relationship been identified. This report examines the effects of chronic, passive immunization on VAbeta and microhemorrhage in PDAPP mice by comparing antibodies with different Abeta epitopes (3D6, Abeta(1-5); 266, Abeta(16-23)) and performing a 3D6 dose-response study. VAbeta and microhemorrhage were assessed using concomitant Abeta immunohistochemistry and hemosiderin detection. 3D6 prevented or cleared VAbeta in a dose-dependent manner, whereas 266 was without effect. Essentially complete absence of VAbeta was observed at the highest 3D6 dose, whereas altered morphology suggestive of ongoing clearance was seen at lower doses. The incidence of microhemorrhage was increased in the high-dose 3D6 group and limited to focal, perivascular sites. These colocalized with Abeta deposits having altered morphology and apparent clearance in the lower-dose 3D6 group. Our results suggest that passive immunization can reduce VAbeta levels, and modulating antibody dose can significantly mitigate the incidence of microhemorrhage while still preventing or reducing VAbeta. These observations raise the possibility that Abeta immunotherapy can potentially slow or halt the course of CAA development in AD that is implicated in vascular dysfunction.


Assuntos
Peptídeos beta-Amiloides/imunologia , Angiopatia Amiloide Cerebral/tratamento farmacológico , Angiopatia Amiloide Cerebral/imunologia , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/imunologia , Imunização Passiva/métodos , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Angiopatia Amiloide Cerebral/genética , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/imunologia , Artérias Cerebrais/metabolismo , Hemorragia Cerebral/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Epitopos/imunologia , Feminino , Taxa de Depuração Metabólica/imunologia , Camundongos , Camundongos Transgênicos , Resultado do Tratamento
7.
Learn Mem ; 15(9): 625-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18772249

RESUMO

Two experiments were conducted to investigate the possibility of faster forgetting by PDAPP mice (a well-established model of Alzheimer's disease as reported by Games and colleagues in an earlier paper). Experiment 1, using mice aged 13-16 mo, confirmed the presence of a deficit in a spatial reference memory task in the water maze by hemizygous PDAPP mice relative to littermate controls. However, after overtraining to a criterion of equivalent navigational performance, a series of memory retention tests revealed faster forgetting in the PDAPP group. Very limited retraining was sufficient to reinstate good memory in both groups, indicating that their faster forgetting may be due to retrieval failure rather than trace decay. In Experiment 2, 6-mo-old PDAPP and controls were required to learn each of a series of spatial locations to criterion with their memory assessed 10 min after learning each location. No memory deficit was apparent in the PDAPP mice initially, but a deficit built up through the series of locations suggestive of increased sensitivity to interference. Faster forgetting and increased interference may each reflect a difficulty in accessing memory traces. This interpretation of one aspect of the cognitive deficit in human mutant APP mice has parallels to deficits observed in patients with Alzheimer's disease, further supporting the validity of transgenic models of the disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Memória/fisiologia , Comportamento Espacial/fisiologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos
8.
J Neurosci ; 27(10): 2654-62, 2007 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-17344403

RESUMO

The behavioral and biochemical impact of active immunization against human beta-amyloid (Abeta) was assessed using male transgenic (Tg) mice overexpressing a human mutant amyloid precursor protein (heterozygous PDAPP mice) and littermate controls. Administration of aggregated Abeta42 occurred at monthly intervals from 7 months ("prevention") or 11 months ("reversal"), followed by double-blind behavioral training at 16 months on a cued task, then serial spatial learning in a water maze. Using a 2 x 2 design, with Abeta42 adjuvanted with MPL-AF (adjuvant formulation of monophosphoryl lipid A) or MPL-AF alone, PDAPP mice were impaired compared with non-Tg littermates on two separate measures of serial spatial learning. Immunization caused no overall rescue of learning but limited the accumulation of total Abeta and Abeta42 levels in cortex and hippocampus by up to 60%. In immunized PDAPP mice, significant negative correlations were observed between hippocampal and cortical Abeta levels and learning capacity, particularly in the prevention study, and correlations between learning capacity and antibody titer. Moreover, a subset of PDAPP mice with very low Abeta levels (hippocampal Abeta levels of <6000 ng/g or cortical Abeta levels of <1000 ng/g) was indistinguishable from non-Tg controls. Mice in the prevention study were also rescued from cognitive impairment more effectively than those in the reversal study. The combination of variability in antibody response and differential levels of Abeta accumulation across the population of immunized PDAPP mice may be responsible for success in cognitive protection with only a subset of these animals, but the similarity to the findings of certain human vaccination trials is noteworthy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Imunização , Aprendizagem em Labirinto/fisiologia , Mutação , Fragmentos de Peptídeos/imunologia , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/sangue , Comportamento Animal/fisiologia , Córtex Cerebral/metabolismo , Sinais (Psicologia) , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Natação , Análise e Desempenho de Tarefas
9.
Neurodegener Dis ; 5(2): 65-71, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18182780

RESUMO

BACKGROUND: In vivo administration of antibodies against the amyloid-beta (Abeta) peptide has been shown to reduce and reverse the progressive amyloidosis that develops in a variety of mouse models of Alzheimer's disease (AD). This work has been extended to clinical trials where subsequent autopsy cases of AD subjects immunized against Abeta showed similar reductions in parenchymal amyloid plaques, suggesting this approach to reduce neuropathology in man is feasible. OBJECTIVE: Multiple hypotheses have been advanced to explain how anti-Abeta antibodies may lower amyloid burden. In this report, we compare approaches utilizing either plaque-binding or peptide-capturing anti-Abeta antibodies for effectiveness in reducing amyloidosis in a mouse model of AD. METHODS: A plaque-binding monoclonal antibody (3D6) and an Abeta peptide-capturing monoclonal antibody (266) were compared in chronic treatment and prevention paradigms using a transgenic mouse model of AD. The effects of antibody therapy on plaque burden and plasma clearance of Abeta were investigated by quantitative imaging and clearance studies of intravenously injected (125)I-Abeta. RESULTS: The plaque-binding antibody 3D6 was highly effective in either treatment or prevention of amyloidosis. In these studies, the peptide-capture antibody 266 showed no reduction in amyloidosis in either paradigm and showed trends towards increasing amyloidosis. Antibody 266 was also found to greatly prolong (>180-fold) the normally rapid peripheral clearance of Abeta, in contrast to that found with 3D6 (>24-fold). CONCLUSION: Reversing and preventing Alzheimer's type amyloidosis is most effectively accomplished with anti-amyloid antibodies that avidly bind plaque.


Assuntos
Peptídeos beta-Amiloides/imunologia , Amiloidose/imunologia , Anticorpos/uso terapêutico , Córtex Cerebral/imunologia , Placa Amiloide/imunologia , Peptídeos beta-Amiloides/sangue , Amiloidose/sangue , Amiloidose/terapia , Animais , Anticorpos/metabolismo , Córtex Cerebral/patologia , Feminino , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Ligação Proteica/imunologia , Solubilidade
10.
J Neurosci ; 26(44): 11437-41, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079673

RESUMO

Immunotherapy targeting the amyloid beta (Abeta) peptide is a novel therapy under investigation for the treatment of Alzheimer's disease (AD). A clinical trial using Abeta(1-42) (AN1792) as the immunogen was halted as a result of development of meningoencephalitis in a small number of patients. The cytokine TGF-beta1 is a key modulator of immune responses that is increased in the brain in AD. We show here that local overexpression of TGF-beta1 in the brain increases both meningeal and parenchymal T lymphocyte number. Furthermore, TGF-beta1 overexpression in a mouse model for AD [amyloid precursor protein (APP) mice] leads to development of additional T cell infiltrates when mice were immunized at a young but not old age with AN1792. Notably, only mice overproducing both Abeta (APP mice) and TGF-beta1 experienced a rise in T lymphocyte number after immunization. One-third of infiltrating T cells were CD4 positive. We did not observe significant differences in B lymphocyte numbers in any of the genotypes or treatment groups. These results demonstrate that TGF-beta1 overproduction in the brain can promote T cell infiltration, in particular after Abeta(1-42) immunization. Likewise, levels of TGF-beta1 or other immune factors in brains of AD patients may influence the response to Abeta(1-42) immunization.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/imunologia , Sistema Nervoso Central/imunologia , Ativação Linfocitária/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/biossíntese , Doença de Alzheimer/metabolismo , Vacinas contra Alzheimer/administração & dosagem , Vacinas contra Alzheimer/imunologia , Animais , Contagem de Células/métodos , Sistema Nervoso Central/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta1
11.
Acta Neuropathol Commun ; 5(1): 7, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28086964

RESUMO

Neurodegenerative disorders such as Parkinson's Disease (PD), PD dementia (PDD) and Dementia with Lewy bodies (DLB) are characterized by progressive accumulation of α-synuclein (α-syn) in neurons. Recent studies have proposed that neuron-to-neuron propagation of α-syn plays a role in the pathogenesis of these disorders. We have previously shown that antibodies against the C-terminus of α-syn reduce the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy, probably by abrogating the axonal transport and accumulation of α-syn in in vivo models. Here, we assessed the effect of passive immunization against α-syn in a new mouse model of axonal transport and accumulation of α-syn. For these purpose, non-transgenic, α-syn knock-out and mThy1-α-syn tg (line 61) mice received unilateral intra-cerebral injections with a lentiviral (LV)-α-syn vector construct followed by systemic administration of the monoclonal antibody 1H7 (recognizes amino acids 91-99) or control IgG for 3 months. Cerebral α-syn accumulation and axonopathy was assessed by immunohistochemistry and effects on behavior were assessed by Morris water maze. Unilateral LV-α-syn injection resulted in axonal propagation of α-syn in the contra-lateral site with subsequent behavioral deficits and axonal degeneration. Passive immunization with 1H7 antibody reduced the axonal accumulation of α-syn in the contra-lateral side and ameliorated the behavioral deficits. Together this study supports the notion that immunotherapy might improve the deficits in models of synucleinopathy by reducing the axonal propagation and accumulation of α-syn. This represents a potential new mode of action through which α-syn immunization might work.


Assuntos
Axônios/patologia , Encéfalo/patologia , Imunização Passiva , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , alfa-Sinucleína/imunologia , alfa-Sinucleína/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Transporte Axonal , Axônios/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Lateralidade Funcional , Vetores Genéticos , Humanos , Lentivirus , Aprendizagem em Labirinto/fisiologia , Camundongos Transgênicos , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/psicologia , alfa-Sinucleína/deficiência , alfa-Sinucleína/genética
12.
J Neurosci ; 25(40): 9096-101, 2005 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16207868

RESUMO

Alzheimer's disease neuropathology is characterized by key features that include the deposition of the amyloid beta peptide (Abeta) into plaques, the formation of neurofibrillary tangles, and the loss of neurons and synapses in specific brain regions. The loss of synapses, and particularly the associated presynaptic vesicle protein synaptophysin in the hippocampus and association cortices, has been widely reported to be one of the most robust correlates of Alzheimer's disease-associated cognitive decline. The beta-amyloid hypothesis supports the idea that Abeta is the cause of these pathologies. However, the hypothesis is still controversial, in part because the direct role of Abeta in synaptic degeneration awaits confirmation. In this study, we show that Abeta reduction by active or passive Abeta immunization protects against the progressive loss of synaptophysin in the hippocampal molecular layer and frontal neocortex of a transgenic mouse model of Alzheimer's disease. These results, substantiated by quantitative electron microscopic analysis of synaptic densities, strongly support a direct causative role of Abeta in the synaptic degeneration seen in Alzheimer's disease and strengthen the potential of Abeta immunotherapy as a treatment approach for this disease.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/administração & dosagem , Imunoterapia , Degeneração Neural/terapia , Sinapses/efeitos dos fármacos , Fatores Etários , Peptídeos beta-Amiloides/imunologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Peptídeos/administração & dosagem , Peptídeos/genética , Peptídeos/imunologia , Sinaptofisina/metabolismo
13.
J Neuropathol Exp Neurol ; 65(11): 1040-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17086100

RESUMO

Neuropathologic examination of 3 patients with Alzheimer disease in the Elan Pharmaceuticals trial using antibodies specific for different Abeta species showed in one case, 4 months after the immunization, evidence of a stage of active plaque clearance with "moth-eaten" plaques and abundant Abeta phagocytosis by microglia. At 1 to 2 years after immunization, 2 cases showed extensive areas cleared of plaques (69% and 86% of the temporal cortex was plaque-free). Cortex cleared of plaques in all 3 cases had a characteristic constellation of features, including a very low plaque burden, sparse residual dense plaque cores, and phagocytosed Abeta within microglia. There was resolution of tau-containing dystrophic neurites, although other features of tau pathology (tangles and neuropil threads) remained and cerebral amyloid angiopathy persisted. Although most antibodies generated by Abeta42 immunization in humans bind the intact N-terminus, immunohistochemistry with specific antibodies showed clearance of all major species of Abeta (Abeta40, Abeta42, and N-terminus truncated Abeta). Abeta immunotherapy can clear all Abeta species from the cortex. However, if it is to be used for treatment of established Alzheimer disease, then the residual tau pathology and cerebral amyloid angiopathy require further study.


Assuntos
Doença de Alzheimer/terapia , Vacinas contra Alzheimer/uso terapêutico , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/uso terapêutico , Encéfalo/patologia , Fragmentos de Peptídeos/uso terapêutico , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Vacinas contra Alzheimer/imunologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Angiopatia Amiloide Cerebral/patologia , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Microglia/patologia , Microscopia Confocal , Dados de Sequência Molecular , Emaranhados Neurofibrilares/patologia , Filamentos do Neurópilo/patologia , Fragmentos de Peptídeos/imunologia , Fagocitose , Ensaios Clínicos Controlados Aleatórios como Assunto
14.
J Comp Neurol ; 495(1): 70-83, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16432899

RESUMO

Abnormal subgranular zone (SGZ) neurogenesis is proposed to contribute to Alzheimer's disease (AD)-related decreases in hippocampal function. Our goal was to examine hippocampal neurogenesis in the PDAPP mouse, a model of AD with age-dependent accumulation of amyloid-beta(42) (Abeta(42))-containing plaques that is well studied with regard to AD therapies. A secondary goal was to determine whether altered neurogenesis in the PDAPP mouse is associated with abnormal maturation or number of mature cells. A tertiary goal was to provide insight into why hippocampal neurogenesis appears to be increased in AD post-mortem tissue and decreased in most AD mouse models. We report an age-dependent decrease in SGZ proliferation in homozygous PDAPP mice. At 1 year of age, PDAPP mice also had new dentate gyrus granule neurons with abnormal maturation and fewer dying cells relative to control mice. In contrast to decreased SGZ cell birth, PDAPP mice had increased birth of immature neurons in the outer portion of the granule cell layer (oGCL), providing insight into why some studies link AD with increased neurogenesis. However, these ectopic oGCL cells were still rare compared with SGZ proliferating cells, emphasizing that the primary characteristic of PDAPP mice is decreased neurogenesis. The decrease in SGZ neurogenesis was not associated with an age-dependent loss of dentate granule neurons. The altered neurogenesis in the PDAPP mouse may contribute to the age-related cognitive deficits reported in this model of AD and may be a useful adjunct target for assessing the impact of AD therapies.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proliferação de Células , Giro Denteado/fisiopatologia , Plasticidade Neuronal/genética , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Apoptose/genética , Diferenciação Celular/genética , Divisão Celular/genética , Sobrevivência Celular/genética , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Mutação/genética , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia
15.
J Alzheimers Dis ; 9(3 Suppl): 133-49, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16914852

RESUMO

Progress in understanding and treating Alzheimer's disease (AD) has been tremendously bolstered by the era of transgenic models of AD. The identification of disease-causing mutations in proteins such as amyloid-beta precursor protein (betaAPP) and presenilin1 (PS1), together with the discovery of other high risk factors (e.g., Apolipoprotein E4), as well as pathogenic mutations in the tau protein has led to the creation of several transgenic mice, including those expressing bi- and tri-genic constructs. Each model has unique pathologies that provide insights into disease mechanisms and interactive features of neuropathologic cascades. More importantly, therapeutic hypotheses are now testable in a manner unheard of less than 15 years ago. The wealth of new approaches currently in clinical and preclinical evaluations can be directly attributed to the impact of these animals on our ability to model relevant aspects of the disease. As a result, we may see containment or even the elimination of AD in the near future as a direct consequence of these advances.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Imunização/métodos , Camundongos , Camundongos Transgênicos , Degeneração Neural/patologia , Placa Amiloide/patologia , Vacinas/uso terapêutico
16.
J Neurosci ; 22(18): 7873-8, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12223540

RESUMO

Transgenic (Tg) mouse models overexpressing amyloid precursor protein (APP) develop senile plaques similar to those found in Alzheimer's disease in an age-dependent manner. Recent reports demonstrated that immunotherapy is effective at preventing or removing amyloid-beta deposits in the mouse models. To characterize the mechanisms involved in clearance, we used antibodies of either IgG1 (10d5) or IgG2b (3d6) applied directly to the brains of 18-month-old Tg2576 or 20-month-old PDAPP mice. Both 10d5 and 3d6 led to clearance of 50% of diffuse amyloid deposits in both animal models within 3 d. Fc receptor-mediated clearance has been shown to be important in an ex vivo assay showing antibody-mediated clearance of plaques by microglia. We now show, using in vivo multiphoton microscopy, that FITC-labeled F(ab')2 fragments of 3d6 (which lack the Fc region of the antibody) also led to clearance of 45% of the deposits within 3 d, similar to the results obtained with full-length 3d6 antibody. This result suggests that direct disruption of plaques, in addition to Fc-dependent phagocytosis, is involved in the antibody-mediated clearance of amyloid-beta deposits in vivo. Dense-core deposits that were not cleared were reduced in size by approximately 30% with full-length antibodies and F(ab')2 fragments 3 d after a topical treatment. Together, these results indicate that clearance of amyloid deposits in vivo may involve, in addition to Fc-dependent clearance, a non-Fc-mediated disruption of plaque structure.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoterapia , Administração Tópica , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Anticorpos/administração & dosagem , Benzotiazóis , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Corantes Fluorescentes , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Mutação , Fagocitose/imunologia , Placa Amiloide/efeitos dos fármacos , Placa Amiloide/metabolismo , Tiazóis
17.
J Comp Neurol ; 492(4): 469-76, 2005 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-16228992

RESUMO

Alzheimer's disease is characterized by neuronal degeneration in the cerebral cortex and hippocampus and subcortical neuronal degeneration in such nuclei as the locus coeruleus (LC). Transgenic mice overexpressing mutant human amyloid precursor protein V717F, PDAPP mice, develop several Alzheimer's disease-like lesions. The present study sought to determine whether there is also loss of LC noradrenergic neurons or evidence of degenerative changes in these animals. PDAPP hemizygous and wild-type littermate control mice were examined at 23 months of age, at a time when there are numerous amyloid-beta (Abeta) plaques in the neocortex and hippocampus. Tissue sections were stained immunohistochemically with an antibody against tyrosine hydroxylase (TH) to identify LC neurons. Computer imaging procedures were used to count the TH-immunoreactive somata in sections through the rostral-caudal extent of the nucleus. There was no loss of LC neurons in the hemizygous mice. In a second experiment, homozygous PDAPP and wild-type mice were examined, at 2 months and 24 months of age. Again there was no age-related loss of neurons in the homozygous animals. In the portion of the LC where neurons reside that project to the cortex and hippocampus, however, the neurons were decreased in size selectively in the 24-month-old transgenic animals. These data indicate that overt LC cell loss does not occur following abundant overexpression of Abeta peptide. However, the selective size reduction of the LC neuronal population projecting to cortical and hippocampal regions containing Abeta-related neuropathology implies that these cells may be subjected to a retrograde-mediated stress.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide , Modelos Animais de Doenças , Locus Cerúleo , Neurônios/citologia , Neurônios/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Humanos , Imuno-Histoquímica , Locus Cerúleo/citologia , Locus Cerúleo/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Neurônios/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia
18.
J Comp Neurol ; 462(4): 371-81, 2003 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-12811807

RESUMO

Transgenic mice overexpressing mutant human amyloid precursor protein (PDAPP mice) develop several Alzheimer's disease (AD)-like lesions including an age-related accumulation of amyloid-beta (Abeta)-containing neuritic plaques. Although aged, heterozygous PDAPP mice also exhibit synaptic and glial cell changes characteristic of AD pathology, no evidence of widespread neuronal loss has been observed. The present study sought to determine whether homozygous PDAPP mice, which express very high levels of Abeta peptide, exhibit AD-like cholinergic degenerative changes, and whether the changes parallel the deposition of Abeta plaques. Mice were examined at 2 and 4 months and at 1 and 2 years of age. There was an age-related increase in the density of Abeta plaques in the cortex and hippocampus of the PDAPP animals; at 4 months of age there were very few plaques, and at 2 years there was a very high density of plaques. There was an age-related reduction in the density of cholinergic nerve terminals in the cerebral cortex; at 2 months there was a normal density of nerve terminals, but as early as age 4 months there was an approximately 50% reduction. However, at age 2 years there was no difference in the number or size of basal forebrain cholinergic somata compared with 2-month-old PDAPP mice. These data indicated that the homozygous PDAPP mouse exhibits cholinergic nerve terminal degenerative pathology and that the cortical neurodegenerative changes occur before the deposition of Abeta-containing neuritic plaques.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Fibras Colinérgicas/patologia , Placa Amiloide/patologia , Terminações Pré-Sinápticas/patologia , Acetilcolina/metabolismo , Fatores Etários , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Córtex Cerebral/patologia , Colina O-Acetiltransferase/análise , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Hipocampo/patologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Regulação para Cima
19.
Brain Res ; 928(1-2): 126-37, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11844479

RESUMO

Recent studies demonstrated that mice overexpressing the human mutant beta-amyloid precursor protein (hbetaAPP; PDAPP mice) show age-independent and age-related deficits in spatial learning. We used behavioral and electrophysiological techniques to determine in young and aged PDAPP mice whether deficits in spatial learning also involve alterations in sleep-wake states, thermoregulation and motor activity. Consistent with earlier studies, young PDAPP mice exhibited selective age-independent deficits using spatial, but not random and serial strategies in the circular maze. Aged PDAPP mice exhibited deficits using all search strategies. The core body temperature (Tb) in young and aged PDAPP mice was significantly lower than in age-matched non-transgenic (non-Tg) littermates. During the dark period, the motor activity (LMA) was significantly increased in young PDAPP mice, but not in aged PDAPP mice. During the light period, young PDAPP mice showed a reduction in the generation of rapid-eye-movement (REM) sleep. In contrast, aged PDAPP mice exhibited a reduction in the amount of time spent in W and an increase in SWS during the light period. Aged PDAPP mice also showed an increase in the amount of time spent in W and a reduction in REM sleep during the dark period. Our findings support previous reports indicating deficits in spatial learning in young and aged PDAPP mice. These data also suggest that PDAPP mice exhibit age-independent and age-related deficits in neural mechanisms regulating visuospatial learning, the total amount and the circadian distribution of sleep-wake states, thermoregulation and motor activity.


Assuntos
Envelhecimento/genética , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Transtornos Cronobiológicos/genética , Transtornos da Memória/genética , Transtornos do Sono-Vigília/genética , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Regulação da Temperatura Corporal/genética , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Transtornos Cronobiológicos/metabolismo , Transtornos Cronobiológicos/fisiopatologia , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/fisiopatologia
20.
Neurosci Lett ; 330(1): 45-8, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12213631

RESUMO

Mice overexpressing the human mutant beta-amyloid precursor protein (hbetaAPP; PDAPP mice) show deficits in hippocampal-dependent spatial learning and hippocampal short- and long-term plasticity at ages preceding Abeta plaque deposition. We determined whether young PDAPP mice also exhibit alterations in septohippocampal function in vivo, which plays an important role in cognitive function. Electrical stimulation of the medial septum significantly increased neuronal excitability and reduced paired-pulse facilitation in the dentate gyrus. Medial septal-induced facilitation of dentate neuronal excitability was reduced in PDAPP mice. The inhibitory effects of medial septum stimulation on dentate paired-pulse facilitation were also attenuated in PDAPP mice. Young mice overexpressing hbetaAPP exhibit early abnormalities in neural circuits implicated in cognitive function that may play an important role in the more profound deficits observed in aged PDAPP mice.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Giro Denteado/fisiologia , Núcleos Septais/fisiologia , Fatores Etários , Precursor de Proteína beta-Amiloide/genética , Animais , Giro Denteado/metabolismo , Estimulação Elétrica/métodos , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Núcleos Septais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA