Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 7803, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551212

RESUMO

Sotatercept is an activin receptor type IIA-Fc (ActRIIA-Fc) fusion protein that improves cardiopulmonary function in patients with pulmonary arterial hypertension (PAH) by selectively trapping activins and growth differentiation factors. However, the cellular and molecular mechanisms of ActRIIA-Fc action are incompletely understood. Here, we determined through genome-wide expression profiling that inflammatory and immune responses are prominently upregulated in the lungs of a Sugen-hypoxia rat model of severe angio-obliterative PAH, concordant with profiles observed in PAH patients. Therapeutic treatment with ActRIIA-Fc-but not with a vasodilator-strikingly reversed proinflammatory and proliferative gene expression profiles and normalized macrophage infiltration in diseased rodent lungs. Furthermore, ActRIIA-Fc normalized pulmonary macrophage infiltration and corrected cardiopulmonary structure and function in Bmpr2 haploinsufficient mice subjected to hypoxia, a model of heritable PAH. Three high-affinity ligands of ActRIIA-Fc each induced macrophage activation in vitro, and their combined immunoneutralization in PAH rats produced cardiopulmonary benefits comparable to those elicited by ActRIIA-Fc. Our results in complementary experimental and genetic models of PAH reveal therapeutic anti-inflammatory activities of ActRIIA-Fc that, together with its known anti-proliferative effects on vascular cell types, could underlie clinical activity of sotatercept as either monotherapy or add-on to current PAH therapies.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Animais , Modelos Animais de Doenças , Hipertensão Pulmonar Primária Familiar , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/tratamento farmacológico , Inflamação/tratamento farmacológico , Camundongos , Hipertensão Arterial Pulmonar/tratamento farmacológico , Ratos , Proteínas Recombinantes de Fusão
2.
Alzheimers Res Ther ; 12(1): 12, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31931873

RESUMO

BACKGROUND: Pyroglutamate-3 Aß (pGlu-3 Aß) is an N-terminally truncated and post-translationally modified Aß species found in Alzheimer's disease (AD) brain. Its increased peptide aggregation propensity and toxicity make it an attractive emerging treatment strategy for AD. We address the question of how the effector function of an anti-pGlu-3 Aß antibody influences the efficacy of immunotherapy in mouse models with AD-like pathology. METHODS: We compared two different immunoglobulin (Ig) isotypes of the same murine anti-pGlu-3 Aß mAb (07/1 IgG1 and 07/2a IgG2a) and a general N-terminal Aß mAb (3A1 IgG1) for their ability to clear Aß and protect cognition in a therapeutic passive immunotherapy study in aged, plaque-rich APPSWE/PS1ΔE9 transgenic (Tg) mice. We also compared the ability of these antibodies and a CDC-mutant form of 07/2a (07/2a-k), engineered to avoid complement activation, to clear Aß in an ex vivo phagocytosis assay and following treatment in APPSLxhQC double Tg mice, and to activate microglia using longitudinal microPET imaging with TSPO-specific 18F-GE180 tracer following a single bolus antibody injection in young and old Tg mice. RESULTS: We demonstrated significant cognitive improvement, better plaque clearance, and more plaque-associated microglia in the absence of microhemorrhage in aged APPSWE/PS1ΔE9 Tg mice treated with 07/2a, but not 07/1 or 3A1, compared to PBS in our first in vivo study. All mAbs cleared plaques in an ex vivo assay, although 07/2a promoted the highest phagocytic activity. Compared with 07/2a, 07/2a-k showed slightly reduced affinity to Fcγ receptors CD32 and CD64, although the two antibodies had similar binding affinities to pGlu-3 Aß. Treatment of APPSLxhQC mice with 07/2a and 07/2a-k mAbs in our second in vivo study showed significant plaque-lowering with both mAbs. Longitudinal 18F-GE180 microPET imaging revealed different temporal patterns of microglial activation for 3A1, 07/1, and 07/2a mAbs and no difference between 07/2a-k and PBS-treated Tg mice. CONCLUSION: Our results suggest that attenuation of behavioral deficits and clearance of amyloid is associated with strong effector function of the anti-pGlu-3 Aß mAb in a therapeutic treatment paradigm. We present evidence that antibody engineering to reduce CDC-mediated complement binding facilitates phagocytosis of plaques without inducing neuroinflammation in vivo. Hence, the results provide implications for tailoring effector function of humanized antibodies for clinical development.


Assuntos
Doença de Alzheimer , Vacinas contra Alzheimer/farmacologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Neuroglia/efeitos dos fármacos , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Imunização Passiva/métodos , Imunoglobulina G , Camundongos , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Ácido Pirrolidonocarboxílico/metabolismo
3.
Sci Rep ; 9(1): 12118, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31431669

RESUMO

Space travel will expose people to high-energy, heavy particle radiation, and the cognitive deficits induced by this exposure are not well understood. To investigate the short-term effects of space radiation, we irradiated 4-month-old Alzheimer's disease (AD)-like transgenic (Tg) mice and wildtype (WT) littermates with a single, whole-body dose of 10 or 50 cGy 56Fe ions (1 GeV/u) at Brookhaven National Laboratory. At ~1.5 months post irradiation, behavioural testing showed sex-, genotype-, and dose-dependent changes in locomotor activity, contextual fear conditioning, grip strength, and motor learning, mainly in Tg but not WT mice. There was little change in general health, depression, or anxiety. Two months post irradiation, microPET imaging of the stable binding of a translocator protein ligand suggested no radiation-specific change in neuroinflammation, although initial uptake was reduced in female mice independently of cerebral blood flow. Biochemical and immunohistochemical analyses revealed that radiation reduced cerebral amyloid-ß levels and microglia activation in female Tg mice, modestly increased microhemorrhages in 50 cGy irradiated male WT mice, and did not affect synaptic marker levels compared to sham controls. Taken together, we show specific short-term changes in neuropathology and behaviour induced by 56Fe irradiation, possibly having implications for long-term space travel.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Encéfalo/patologia , Encéfalo/efeitos da radiação , Radioisótopos de Ferro/efeitos adversos , Voo Espacial , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Comportamento Animal/efeitos da radiação , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Humanos , Inflamação/patologia , Inflamação/fisiopatologia , Aprendizagem/efeitos da radiação , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Microglia/fisiologia , Microglia/efeitos da radiação , Atividade Motora/efeitos da radiação , Presenilina-1/genética , Presenilina-1/metabolismo , Fatores Sexuais
5.
Mol Neurodegener ; 11(1): 48, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27363697

RESUMO

Immunization against amyloid-ß (Aß) peptides deposited in Alzheimer's disease (AD) has shown considerable therapeutic effect in animal models however, the translation into human Alzheimer's patients is challenging. In recent years, a number of promising Aß immunotherapy trials failed to reach primary study endpoints. Aside from uncertainties in the selection of patients and the start and duration of treatment, these results also suggest that the mechanisms underlying AD are still not fully understood. Thorough characterizations of protein aggregates in AD brain have revealed a conspicuous heterogeneity of Aß peptides enabling the study of the toxic potential of each of the major forms. One such form, amino-terminally truncated and modified pyroglutamate (pGlu)-3 Aß peptide appears to play a seminal role for disease initiation, qualifying it as novel target for immunotherapy approaches.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Vacinas contra Alzheimer , Precursor de Proteína beta-Amiloide , Ácido Pirrolidonocarboxílico , Vacinas contra Alzheimer/imunologia , Precursor de Proteína beta-Amiloide/imunologia , Animais , Humanos , Ácido Pirrolidonocarboxílico/imunologia
6.
Trends Cell Biol ; 26(8): 587-597, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27004698

RESUMO

Microglia are resident macrophages of the central nervous system (CNS), representing 5-10% of total CNS cells. Recent findings reveal that microglia enter the embryonic brain, take up residence before the differentiation of other CNS cell types, and become critical regulators of CNS development. Here, we discuss exciting new work implicating microglia in a range of developmental processes, including regulation of cell number and spatial patterning of CNS cells, myelination, and formation and refinement of neural circuits. Furthermore, we review studies suggesting that these cellular functions result in the modulation of behavior, which has important implications for a variety of neurological disorders.


Assuntos
Microglia/citologia , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Animais , Padronização Corporal , Linhagem da Célula , Humanos , Rede Nervosa/fisiologia , Neurônios/citologia
7.
J Neurosci ; 35(47): 15716-30, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26609163

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia. Neuroinflammation appears to play an important role in AD pathogenesis. Ligands of the 18 kDa translocator protein (TSPO), a marker for activated microglia, have been used as positron emission tomography (PET) tracers to reflect neuroinflammation in humans and mouse models. Here, we used the novel TSPO-targeted PET tracer (18)F-GE180 (flutriciclamide) to investigate differences in neuroinflammation between young and old WT and APP/PS1dE9 transgenic (Tg) mice. In vivo PET scans revealed an overt age-dependent elevation in whole-brain uptake of (18)F-GE180 in both WT and Tg mice, and a significant increase in whole-brain uptake of (18)F-GE180 (peak-uptake and retention) in old Tg mice compared with young Tg mice and all WT mice. Similarly, the (18)F-GE180 binding potential in hippocampus was highest to lowest in old Tg > old WT > young Tg > young WT mice using MRI coregistration. Ex vivo PET and autoradiography analysis further confirmed our in vivo PET results: enhanced uptake and specific binding (SUV75%) of (18)F-GE180 in hippocampus and cortex was highest in old Tg mice followed by old WT, young Tg, and finally young WT mice. (18)F-GE180 specificity was confirmed by an in vivo cold tracer competition study. We also examined (18)F-GE180 metabolites in 4-month-old WT mice and found that, although total radioactivity declined over 2 h, of the remaining radioactivity, ∼90% was due to parent (18)F-GE180. In conclusion, (18)F-GE180 PET scans may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment. SIGNIFICANCE STATEMENT: Microglial activation, a player in Alzheimer's disease (AD) pathogenesis, is thought to reflect neuroinflammation. Using in vivo microPET imaging with a novel TSPO radioligand, (18)F-GE180, we detected significantly enhanced neuroinflammation during normal aging in WT mice and in response to AD-associated pathology in APP/PS1dE9 Tg mice, an AD mouse model. Increased uptake and specific binding of (18)F-GE180 in whole brain and hippocampus were confirmed by ex vivo PET and autoradiography. The binding specificity and stability of (18)F-GE180 was further confirmed by a cold tracer competition study and a metabolite study, respectively. Therefore, (18)F-GE180 PET imaging may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment and may also be useful for other neurodegenerative diseases.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Carbazóis/metabolismo , Radioisótopos de Flúor/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores de GABA/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/diagnóstico por imagem , Animais , Progressão da Doença , Humanos , Inflamação/diagnóstico , Inflamação/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Neurobiol Aging ; 36(12): 3187-3199, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26453001

RESUMO

Pyroglutamate-3 amyloid-beta (pGlu-3 Aß) is an N-terminally truncated Aß isoform likely playing a decisive role in Alzheimer's disease pathogenesis. Here, we describe a prophylactic passive immunization study in APPswe/PS1ΔE9 mice using a novel pGlu-3 Aß immunoglobulin G1 (IgG1) monoclonal antibody, 07/1 (150 and 500 µg, intraperitoneal, weekly) and compare its efficacy with a general Aß IgG1 monoclonal antibody, 3A1 (200 µg, intraperitoneal, weekly) as a positive control. After 28 weeks of treatment, plaque burden was reduced and cognitive performance of 07/1-immunized Tg mice, especially at the higher dose, was normalized to wild-type levels in 2 hippocampal-dependent tests and partially spared compared with phosphate-buffered saline-treated Tg mice. Mice that received 3A1 had reduced plaque burden but showed no cognitive benefit. In contrast with 3A1, treatment with 07/1 did not increase the concentration of Aß in plasma, suggesting different modes of Aß plaque clearance. In conclusion, early selective targeting of pGlu-3 Aß by immunotherapy may be effective in lowering cerebral Aß plaque burden and preventing cognitive decline in the clinical setting. Targeting this pathologically modified form of Aß thereby is unlikely to interfere with potential physiologic function(s) of Aß that have been proposed.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Vacinas contra Alzheimer/administração & dosagem , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais/administração & dosagem , Transtornos Cognitivos/prevenção & controle , Cognição , Imunização Passiva , Placa Amiloide/metabolismo , Ácido Pirrolidonocarboxílico/imunologia , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Encéfalo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/patologia
9.
J Neurosci ; 35(38): 13029-42, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26400934

RESUMO

The complement system is part of the innate immune response responsible for removing pathogens and cellular debris, in addition to helping to refine CNS neuronal connections via microglia-mediated pruning of inappropriate synapses during brain development. However, less is known about the role of complement during normal aging. Here, we studied the role of the central complement component, C3, in synaptic health and aging. We examined behavior as well as electrophysiological, synaptic, and neuronal changes in the brains of C3-deficient male mice (C3 KO) compared with age-, strain-, and gender-matched C57BL/6J (wild-type, WT) control mice at postnatal day 30, 4 months, and 16 months of age. We found the following: (1) region-specific and age-dependent synapse loss in aged WT mice that was not observed in C3 KO mice; (2) age-dependent neuron loss in hippocampal CA3 (but not in CA1) that followed synapse loss in aged WT mice, neither of which were observed in aged C3 KO mice; and (3) significantly enhanced LTP and cognition and less anxiety in aged C3 KO mice compared with aged WT mice. Importantly, CA3 synaptic puncta were similar between WT and C3 KO mice at P30. Together, our results suggest a novel and prominent role for complement protein C3 in mediating aged-related and region-specific changes in synaptic function and plasticity in the aging brain. Significance statement: The complement cascade, part of the innate immune response to remove pathogens, also plays a role in synaptic refinement during brain development by the removal of weak synapses. We investigated whether complement C3, a central component, affects synapse loss during aging. Wild-type (WT) and C3 knock-out (C3 KO) mice were examined at different ages. The mice were similar at 1 month of age. However, with aging, WT mice lost synapses in specific brain regions, especially in hippocampus, an area important for memory, whereas C3 KO mice were protected. Aged C3 KO mice also performed better on learning and memory tests than aged WT mice. Our results suggest that complement C3, or its downstream signaling, is detrimental to synapses during aging.


Assuntos
Envelhecimento/patologia , Complemento C3/deficiência , Hipocampo/patologia , Adaptação Fisiológica/genética , Fatores Etários , Animais , Complemento C3/genética , Condicionamento Psicológico/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Comportamento Exploratório/fisiologia , Medo , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Fosfopiruvato Hidratase/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura , Sinapsinas/metabolismo , Sinaptofisina/metabolismo , Sinaptossomos/metabolismo
10.
Eur J Neurosci ; 41(8): 1025-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25816842

RESUMO

The Shank genes (SHANK1, 2, 3) encode scaffold proteins highly enriched in postsynaptic densities where they regulate synaptic structure in spiny neurons. Mutations in human Shank genes are linked to autism spectrum disorder and schizophrenia. Shank1 mutant mice exhibit intriguing cognitive phenotypes reminiscent of individuals with autism spectrum disorder. However, the molecular mechanisms leading to the human pathophysiological phenotypes and mouse behaviors have not been elucidated. In this study it is shown that Shank1 protein is highly localized in parvalbumin-expressing (PV+) fast-spiking inhibitory interneurons in the hippocampus. Importantly, a lack of Shank1 in hippocampal CA1 PV+ neurons reduced excitatory synaptic inputs and inhibitory synaptic outputs to pyramidal neurons. Furthermore, it is demonstrated that hippocampal CA1 pyramidal neurons in Shank1 mutant mice exhibit a shift in the excitatory and inhibitory balance (E-I balance), a pathophysiological hallmark of autism spectrum disorder. The mutant mice also exhibit lower expression of gephyrin (a scaffold component of inhibitory synapses), supporting the dysregulation of E-I balance in the hippocampus. These results suggest that Shank1 scaffold in PV+ interneurons regulates excitatory synaptic strength and participates in the maintenance of E-I balance in excitatory neurons.


Assuntos
Região CA1 Hipocampal/fisiologia , Neurônios GABAérgicos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células Piramidais/fisiologia , Transmissão Sináptica , Animais , Região CA1 Hipocampal/metabolismo , Proteínas de Transporte/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Potenciais da Membrana , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Inibição Neural , Parvalbuminas/metabolismo , Densidade Pós-Sináptica/metabolismo , Células Piramidais/metabolismo
11.
Am J Pathol ; 183(2): 369-81, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23747948

RESUMO

Amyloid-ß (Aß) peptides, starting with pyroglutamate at the third residue (pyroGlu-3 Aß), are a major species deposited in the brain of Alzheimer disease (AD) patients. Recent studies suggest that this isoform shows higher toxicity and amyloidogenecity when compared to full-length Aß peptides. Here, we report the first comprehensive and comparative IHC evaluation of pyroGlu-3 Aß deposition in humans and animal models. PyroGlu-3 Aß immunoreactivity (IR) is abundant in plaques and cerebral amyloid angiopathy of AD and Down syndrome patients, colocalizing with general Aß IR. PyroGlu-3 Aß is further present in two nontransgenic mammalian models of cerebral amyloidosis, Caribbean vervets, and beagle canines. In addition, pyroGlu-3 Aß deposition was analyzed in 12 different AD-like transgenic mouse models. In contrast to humans, all transgenic models showed general Aß deposition preceding pyroGlu-3 Aß deposition. The findings varied greatly among the mouse models concerning age of onset and cortical brain region. In summary, pyroGlu-3 Aß is a major species of ß-amyloid deposited early in diffuse and focal plaques and cerebral amyloid angiopathy in humans and nonhuman primates, whereas it is deposited later in a subset of focal and vascular amyloid in AD-like transgenic mouse models. Given the proposed decisive role of pyroGlu-3 Aß peptides for the development of human AD pathology, this study provides insights into the usage of animal models in AD studies.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide , Animais , Encéfalo/patologia , Angiopatia Amiloide Cerebral/metabolismo , Chlorocebus aethiops , Modelos Animais de Doenças , Cães , Síndrome de Down/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Placa Amiloide/metabolismo
12.
J Neurosci ; 33(16): 7027-37, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595760

RESUMO

Active amyloid-ß (Aß) immunotherapy is under investigation to prevent or treat early Alzheimer's disease (AD). In 2002, a Phase II clinical trial (AN1792) was halted due to meningoencephalitis in ∼6% of the AD patients, possibly caused by a T-cell-mediated immunological response. Thus, generating a vaccine that safely generates high anti-Aß antibody levels in the elderly is required. In this study, MER5101, a novel conjugate of Aß1-15 peptide (a B-cell epitope fragment) conjugated to an immunogenic carrier protein, diphtheria toxoid (DT), and formulated in a nanoparticular emulsion-based adjuvant, was administered to 10-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (Wt) mice. High anti-Aß antibody levels were observed in both vaccinated APPswe/PS1ΔE9 Tg and Wt mice. Antibody isotypes were mainly IgG1 and IgG2b, suggesting a Th2-biased response. Restimulation of splenocytes with the Aß1-15:DT conjugate resulted in a strong proliferative response, whereas proliferation was absent after restimulation with Aß1-15 or Aß1-40/42 peptides, indicating a cellular immune response against DT while avoiding an Aß-specific T-cell response. Moreover, significant reductions in cerebral Aß plaque burden, accompanied by attenuated microglial activation and increased synaptic density, were observed in MER5101-vaccinated APPswe/PS1ΔE9 Tg mice compared with Tg adjuvant controls. Last, MER5101-immunized APPswe/PS1ΔE9 Tg mice showed improvement of cognitive deficits in both contextual fear conditioning and the Morris water maze. Our novel, highly immunogenic Aß conjugate vaccine, MER5101, shows promise for improving Aß vaccine safety and efficacy and therefore, may be useful for preventing and/or treating early AD.


Assuntos
Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/imunologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/terapia , Toxoide Diftérico/imunologia , Imunização/métodos , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Formação de Anticorpos/imunologia , Células CHO/química , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/imunologia , Condicionamento Clássico/fisiologia , Cricetinae , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Medo , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/imunologia , Gliose/terapia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoprecipitação , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Fragmentos de Peptídeos/imunologia , Presenilina-1/genética , Baço/citologia , Estatísticas não Paramétricas , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transfecção
13.
Glia ; 60(6): 993-1003, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438044

RESUMO

Complement components and their receptors are found within and around amyloid ß (Aß) cerebral plaques in Alzheimer's disease (AD). Microglia defend against pathogens through phagocytosis via complement component C3 and/or engagement of C3 cleavage product iC3b with complement receptor type 3 (CR3, Mac-1). Here, we provide direct evidence that C3 and Mac-1 mediate, in part, phagocytosis and clearance of fibrillar amyloid-ß (fAß) by murine microglia in vitro and in vivo. Microglia took up not only synthetic fAß(42) but also amyloid cores from patients with AD, transporting them to lysosomes in vitro. Fibrillar Aß(42) uptake was significantly attenuated by the deficiency or knockdown of C3 or Mac-1 and scavenger receptor class A ligands. In addition, C3 or Mac-1 knockdown combined with a scavenger receptor ligand, fucoidan, further attenuated fibrillar Aß(42) uptake by N9 microglia. Fluorescent fibrillar Aß(42) microinjected cortically was significantly higher in C3 and Mac-1 knockout mice compared with wild-type mice 5 days after surgery, indicating reduced clearance in vivo. Together, these results demonstrate that C3 and Mac-1 are involved in phagocytosis and clearance of fAß by microglia, providing support for a potential beneficial role for microglia and the complement system in AD pathogenesis. © 2012 Wiley Periodicals, Inc.


Assuntos
Amiloide/metabolismo , Encéfalo/citologia , Complemento C3c/metabolismo , Antígeno de Macrófago 1/metabolismo , Microglia/fisiologia , Fagocitose/fisiologia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Linhagem Celular Transformada , Complemento C3c/deficiência , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Ligantes , Proteínas de Membrana Lisossomal/metabolismo , Lisossomos/metabolismo , Antígeno de Macrófago 1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microinjeções , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Fagocitose/efeitos dos fármacos , Fagocitose/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Depuradores Classe A/metabolismo , Fatores de Tempo , Transfecção/métodos
14.
Neurodegener Dis ; 10(1-4): 265-70, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22343072

RESUMO

BACKGROUND: N-terminally truncated and modified pyroglutamate-3 amyloid-ß protein (pE3-Aß) is present in most, if not all, cerebral plaque and vascular amyloid deposits in human Alzheimer's disease (AD). pE3-Aß deposition is also found in AD-like transgenic (tg) mouse brain, albeit in lesser quantities than general Aß. pE3-Aß resists degradation, is neurotoxic, and may act as a seed for Aß aggregation. OBJECTIVE: We sought to determine if pE3-Aß removal by passive immunization with a highly specific monoclonal antibody (mAb) impacts pathogenesis in a mouse model of Alzheimer's amyloidosis. METHODS: APPswe/PS1ΔE9 tg mice were given weekly intraperitoneal injections of a new anti-pE3-Aß mAb (mAb07/1) or PBS from 5.8 to 13.8 months of age (prevention) or from 23 to 24.7 months of age (therapeutic). Multiple forms of cerebral Aß were quantified pathologically and biochemically. Gliosis and microhemorrhage were examined. RESULTS: Chronic passive immunization with an anti-pE3-Aß mAb significantly reduced total plaque deposition and appeared to lower gliosis in the hippocampus and cerebellum in both the prevention and therapeutic studies. Insoluble Aß levels in hemibrain homogenates were not significantly different between immunized and control mice. Microhemorrhage was not observed with anti-pE3-Aß immunotherapy. CONCLUSIONS: Selective removal of pE3-Aß lowered general Aß plaque deposition suggesting a pro-aggregation or seeding role for pE3-Aß.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Imunização Passiva/métodos , Fragmentos de Peptídeos/imunologia , Ácido Pirrolidonocarboxílico/imunologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Amiloidose/etiologia , Amiloidose/terapia , Animais , Anticorpos/sangue , Encéfalo/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Gliose/etiologia , Gliose/terapia , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Projetos Piloto , Placa Amiloide/etiologia , Placa Amiloide/terapia , Presenilina-1/genética , Estatísticas não Paramétricas , Fatores de Tempo
15.
J Alzheimers Dis ; 28(2): 423-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22045486

RESUMO

S-adenosyl methionine (SAM) contributes to multiple pathways in neuronal homeostasis, several of which are compromised in age-related neurodegeneration and Alzheimer's disease. Dietary supplementation of transgenic mice with SAM maintained acetylcholine levels, cognitive performance, oxidative buffering capacity, and phosphatase activity, and reduced aggression, calcium influx, endogenous PS-1 expression, γ-secretase activity, and levels of amyloid-ß (Aß) and phospho-tau. Herein, we examined whether or not SAM could delay neuropathology in 3xTg-AD mice, which harbor mutant genes for human AßPP, PS-1 and tau. Mice received a standard AIN-76 diet with or without SAM (100 mg/kg diet) for 1 month commencing at 10 months of age or for 3 months commencing at 12.5 months of age; mice were sacrificed and examined for Aß and tau neuropathology at 11 and 15.5 months of age, respectively. SAM supplementation reduced hippocampal intracellular AßPP/Aß and phospho-tau immunoreactivity to a similar extent at both sampling intervals. Supplementation reduced the number of extracellular Aß deposits by 80% (p < 0.01) at 11 months of age after 1 month of treatment but only by 24% (p < 0.34) at 15.5 months of age after 3 months of treatment. As anticipated, neurofibrillary tangles were not observed in mice at these young ages; however, supplementation reduced levels of phospho-tau and caspase-cleaved tau within Sarkosyl-insoluble preparations in mice at 15.5 months of age. These limited analyses indicate that SAM can modulate the time course of AD neuropathology, and support further long-term analyses.


Assuntos
Doença de Alzheimer/dietoterapia , Peptídeos beta-Amiloides/metabolismo , Suplementos Nutricionais , S-Adenosilmetionina/administração & dosagem , Tauopatias/prevenção & controle , Fatores Etários , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Transgênicos , Presenilina-1/genética , Tauopatias/etiologia , Tauopatias/genética , Proteínas tau/genética
16.
PLoS One ; 7(12): e53275, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300905

RESUMO

Galactic Cosmic Radiation consisting of high-energy, high-charged (HZE) particles poses a significant threat to future astronauts in deep space. Aside from cancer, concerns have been raised about late degenerative risks, including effects on the brain. In this study we examined the effects of (56)Fe particle irradiation in an APP/PS1 mouse model of Alzheimer's disease (AD). We demonstrated 6 months after exposure to 10 and 100 cGy (56)Fe radiation at 1 GeV/µ, that APP/PS1 mice show decreased cognitive abilities measured by contextual fear conditioning and novel object recognition tests. Furthermore, in male mice we saw acceleration of Aß plaque pathology using Congo red and 6E10 staining, which was further confirmed by ELISA measures of Aß isoforms. Increases were not due to higher levels of amyloid precursor protein (APP) or increased cleavage as measured by levels of the ß C-terminal fragment of APP. Additionally, we saw no change in microglial activation levels judging by CD68 and Iba-1 immunoreactivities in and around Aß plaques or insulin degrading enzyme, which has been shown to degrade Aß. However, immunohistochemical analysis of ICAM-1 showed evidence of endothelial activation after 100 cGy irradiation in male mice, suggesting possible alterations in Aß trafficking through the blood brain barrier as a possible cause of plaque increase. Overall, our results show for the first time that HZE particle radiation can increase Aß plaque pathology in an APP/PS1 mouse model of AD.


Assuntos
Doença de Alzheimer/etiologia , Encéfalo/efeitos da radiação , Transtornos Cognitivos/etiologia , Radiação Cósmica/efeitos adversos , Placa Amiloide/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...