Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Acta Neuropathol Commun ; 5(1): 7, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28086964

ABSTRACT

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.


Subject(s)
Axons/pathology , Brain/pathology , Immunization, Passive , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Axonal Transport , Axons/metabolism , Brain/metabolism , Disease Models, Animal , Female , Functional Laterality , Genetic Vectors , Humans , Lentivirus , Maze Learning/physiology , Mice, Transgenic , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/psychology , alpha-Synuclein/deficiency , alpha-Synuclein/genetics
2.
J Neurosci ; 34(28): 9441-54, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25009275

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain/immunology , Movement Disorders/immunology , Movement Disorders/therapy , Parkinsonian Disorders/immunology , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , Animals , Brain/drug effects , Disease Models, Animal , Humans , Immunotherapy/methods , Mice , Mice, Transgenic , Tissue Distribution , Treatment Outcome
3.
Alzheimers Dement ; 9(5 Suppl): S105-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583235

ABSTRACT

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.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Blood Vessels/pathology , Brain/pathology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Aquaporin 4/metabolism , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Collagen Type IV/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Intracranial Hemorrhages/etiology , Meninges/pathology , Meninges/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mutation/genetics , Time Factors
4.
Am J Pathol ; 182(3): 940-53, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23313024

ABSTRACT

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.


Subject(s)
Axons/pathology , Lewy Body Disease/pathology , Mutant Proteins/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Antibodies/immunology , Antibody Specificity/immunology , Axons/metabolism , Axons/ultrastructure , Biomarkers/metabolism , Demography , Disease Models, Animal , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Neurites/metabolism , Neurites/pathology , Neurites/ultrastructure , Protein Transport , Synapses/metabolism , Synapses/pathology , Synapses/ultrastructure , Thy-1 Antigens/metabolism , alpha-Synuclein/immunology
5.
J Neurosci ; 32(8): 2696-702, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22357853

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Epitopes/immunology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Antibodies, Neutralizing , Behavioral Symptoms/drug therapy , Behavioral Symptoms/etiology , Behavioral Symptoms/immunology , Biotin/metabolism , Cells, Cultured , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dendritic Spines/drug effects , Disease Models, Animal , Embryo, Mammalian , Epitopes/metabolism , Fear/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/cytology , Humans , Mice , Mice, Transgenic , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/immunology , Neuropeptides/metabolism , Peptide Fragments/immunology , Phosphorylation , Protein Binding/immunology , Protein Structure, Secondary , Protein Transport/drug effects , Rats , Receptors, AMPA/metabolism , Solubility , Vesicular Glutamate Transport Protein 1/immunology , Vesicular Glutamate Transport Protein 1/metabolism
6.
PLoS One ; 6(4): e19338, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559417

ABSTRACT

Dementia with Lewy bodies (DLB) and Parkinson's Disease (PD) are common causes of motor and cognitive deficits and are associated with the abnormal accumulation of alpha-synuclein (α-syn). This study investigated whether passive immunization with a novel monoclonal α-syn antibody (9E4) against the C-terminus (CT) of α-syn was able to cross into the CNS and ameliorate the deficits associated with α-syn accumulation. In this study we demonstrate that 9E4 was effective at reducing behavioral deficits in the water maze, moreover, immunization with 9E4 reduced the accumulation of calpain-cleaved α-syn in axons and synapses and the associated neurodegenerative deficits. In vivo studies demonstrated that 9E4 traffics into the CNS, binds to cells that display α-syn accumulation and promotes α-syn clearance via the lysosomal pathway. These results suggest that passive immunization with monoclonal antibodies against the CT of α-syn may be of therapeutic relevance in patients with PD and DLB.


Subject(s)
Immunization, Passive/methods , Lewy Body Disease/genetics , Lewy Body Disease/therapy , alpha-Synuclein/genetics , Animals , Antibodies, Monoclonal/metabolism , Behavior, Animal , Cell Line, Tumor , Disease Models, Animal , Immunohistochemistry/methods , Lewy Body Disease/immunology , Lysosomes/metabolism , Maze Learning , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Rats
7.
Alzheimers Res Ther ; 2(4): 22, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20682084

ABSTRACT

The amyloid-beta (Aß) hypothesis of Alzheimer's disease (AD) causality is now well into its third decade and is finally entering a phase of rigorous clinical testing in numerous late stage clinical trials. The use of Aß-based animal models of AD has been essential to the discovery and/or preclinical validation of many of these therapeutic approaches. While several neuropathologically based results from preclinical studies have translated nicely into AD patients, the full clinical value of Aß-directed therapies awaits results from trials now in progress.

8.
Learn Mem ; 15(9): 625-32, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18772249

ABSTRACT

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.


Subject(s)
Alzheimer Disease/physiopathology , Memory/physiology , Spatial Behavior/physiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Disease Models, Animal , Humans , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Transgenic
9.
J Neurosci ; 28(27): 6787-93, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18596154

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/immunology , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Amyloid Angiopathy/immunology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/immunology , Immunization, Passive/methods , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Cerebral Amyloid Angiopathy/genetics , Cerebral Arteries/drug effects , Cerebral Arteries/immunology , Cerebral Arteries/metabolism , Cerebral Hemorrhage/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/immunology , Epitopes/immunology , Female , Metabolic Clearance Rate/immunology , Mice , Mice, Transgenic , Treatment Outcome
10.
Neurodegener Dis ; 5(2): 65-71, 2008.
Article in English | MEDLINE | ID: mdl-18182780

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloidosis/immunology , Antibodies/therapeutic use , Cerebral Cortex/immunology , Plaque, Amyloid/immunology , Amyloid beta-Peptides/blood , Amyloidosis/blood , Amyloidosis/therapy , Animals , Antibodies/metabolism , Cerebral Cortex/pathology , Female , Mice , Mice, Transgenic , Plaque, Amyloid/pathology , Protein Binding/immunology , Solubility
11.
J Biol Chem ; 282(36): 26326-34, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17616527

ABSTRACT

The aspartyl protease beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) initiates processing of amyloid precursor protein (APP) into amyloid beta (Abeta) peptide, the major component of Alzheimer disease (AD) plaques. To determine the role that BACE1 plays in the development of Abeta-driven AD-like pathology, we have crossed PDAPP mice, a transgenic mouse model of AD overexpressing human mutated APP, onto mice with either a homozygous or heterozygous BACE1 gene knockout. Analysis of PDAPP/BACE(-/-) mice demonstrated that BACE1 is absolutely required for both Abeta generation and the development of age-associated plaque pathology. Furthermore, synaptic deficits, a neurodegenerative pathology characteristic of AD, were also reversed in the bigenic mice. To determine the extent of BACE1 reduction required to significantly inhibit pathology, PDAPP mice having a heterozygous BACE1 gene knock-out were evaluated for Abeta generation and for the development of pathology. Although the 50% reduction in BACE1 enzyme levels caused only a 12% decrease in Abeta levels in young mice, it nonetheless resulted in a dramatic reduction in Abeta plaques, neuritic burden, and synaptic deficits in older mice. Quantitative analyses indicate that brain Abeta levels in young APP transgenic mice are not the sole determinant for the changes in plaque pathology mediated by reduced BACE1. These observations demonstrate that partial reductions of BACE1 enzyme activity and concomitant Abeta levels lead to dramatic inhibition of Abeta-driven AD-like pathology, making BACE1 an excellent target for therapeutic intervention in AD.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Synaptic Membranes/enzymology , Synaptic Membranes/pathology , Aging/genetics , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Amyloid Precursor Protein Secretases/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/deficiency , Disease Models, Animal , Enzyme Activation/genetics , Humans , Mice , Mice, Knockout , Neurites/enzymology , Neurites/pathology
12.
J Neurosci ; 27(10): 2654-62, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17344403

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Immunization , Maze Learning/physiology , Mutation , Peptide Fragments/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/blood , Behavior, Animal/physiology , Cerebral Cortex/metabolism , Cues , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Peptide Fragments/metabolism , Swimming , Task Performance and Analysis
13.
J Neurosci ; 26(44): 11437-41, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17079673

ABSTRACT

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.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/immunology , Central Nervous System/immunology , Lymphocyte Activation/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/biosynthesis , Alzheimer Disease/metabolism , Alzheimer Vaccines/administration & dosage , Alzheimer Vaccines/immunology , Animals , Cell Count/methods , Central Nervous System/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
14.
J Neuropathol Exp Neurol ; 65(11): 1040-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17086100

ABSTRACT

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.


Subject(s)
Alzheimer Disease/therapy , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/therapeutic use , Brain/pathology , Peptide Fragments/therapeutic use , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Vaccines/immunology , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Cerebral Amyloid Angiopathy/pathology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Microglia/pathology , Microscopy, Confocal , Molecular Sequence Data , Neurofibrillary Tangles/pathology , Neuropil Threads/pathology , Peptide Fragments/immunology , Phagocytosis , Randomized Controlled Trials as Topic
15.
J Alzheimers Dis ; 9(3 Suppl): 133-49, 2006.
Article in English | MEDLINE | ID: mdl-16914852

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Disease Models, Animal , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/immunology , Brain/metabolism , Immunization/methods , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Plaque, Amyloid/pathology , Vaccines/therapeutic use
16.
J Comp Neurol ; 495(1): 70-83, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16432899

ABSTRACT

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.


Subject(s)
Aging/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Proliferation , Dentate Gyrus/physiopathology , Neuronal Plasticity/genetics , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Division/genetics , Cell Survival/genetics , Disease Models, Animal , Female , Genetic Predisposition to Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Neurologic Mutants , Mice, Transgenic , Mutation/genetics , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
17.
J Neurosci ; 25(40): 9096-101, 2005 Oct 05.
Article in English | MEDLINE | ID: mdl-16207868

ABSTRACT

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.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/administration & dosage , Immunotherapy , Nerve Degeneration/therapy , Synapses/drug effects , Age Factors , Amyloid beta-Peptides/immunology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry/methods , Mice , Mice, Transgenic , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Peptides/administration & dosage , Peptides/genetics , Peptides/immunology , Synaptophysin/metabolism
18.
Biochemistry ; 44(42): 13807-19, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16229470

ABSTRACT

Central to the pathology of Alzheimer's disease (AD) is the profuse accumulation of amyloid-beta (Abeta) peptides in the brain of affected individuals, and several amyloid precursor protein (APP) transgenic (Tg) mice models have been created to mimic Abeta deposition. Among these, the PDAPP Tg mice carrying the familial AD APP 717 Val --> Phe mutation have been widely used to test potential AD therapeutic interventions including active and passive anti-Abeta immunizations. The structure and biochemistry of the PDAPP Tg mice Abeta-related peptides were investigated using acid and detergent lysis of brain tissue, ultracentrifugation, FPLC, HPLC, enzymatic and chemical cleavage of peptides, Western blot, immunoprecipitation, and MALDI-TOF and SELDI-TOF mass spectrometry. Our experiments reveal that PDAPP mice produce a variety of C-terminally elongated Abeta peptides in addition to Abeta n-40 and Abeta n-42, as well as N-terminally truncated peptides, suggesting anomalous proteolysis of both APP and Abeta. Important alterations in the overall APP degradation also occur in this model, resulting in a striking comparative lack of CT83 and CT99 fragments, which may be inherent to the strain of mice, a generalized gamma-secretase failure, or the ultimate manifestation of the overwhelming amount of expressed human transgene; these alterations are not observed in other strains of APP Tg mice or in sporadic AD. Understanding at the molecular level the nature of these important animal models will permit a better understanding of therapeutic interventions directed to prevent, delay, or reverse the ravages of sporadic AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Immunoprecipitation , Mass Spectrometry/methods , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/chemistry
19.
J Comp Neurol ; 492(4): 469-76, 2005 Nov 28.
Article in English | MEDLINE | ID: mdl-16228992

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor , Disease Models, Animal , Locus Coeruleus , Neurons/cytology , Neurons/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Humans , Immunohistochemistry , Locus Coeruleus/cytology , Locus Coeruleus/pathology , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurons/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
20.
Neuron ; 46(6): 857-68, 2005 Jun 16.
Article in English | MEDLINE | ID: mdl-15953415

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Immunization/methods , Nerve Tissue Proteins/immunology , Parkinson Disease/immunology , Parkinson Disease/therapy , Animals , Antibodies/metabolism , Antibodies/therapeutic use , Blotting, Western/methods , Cathepsin D/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Diagnostic Imaging/methods , Epitope Mapping/methods , Humans , Immunohistochemistry/methods , Inclusion Bodies/metabolism , Lysosomes/metabolism , Mice , Mice, Transgenic , Models, Immunological , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Presynaptic Terminals/metabolism , Subcellular Fractions/metabolism , Synaptophysin/metabolism , Synucleins , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...