Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
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
2.
Biochemistry ; 53(24): 3908-21, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24840308

ABSTRACT

Evidence for a central role of amyloid ß-protein (Aß) in the genesis of Alzheimer's disease (AD) has led to advanced human trials of Aß-lowering agents. The "amyloid hypothesis" of AD postulates deleterious effects of small, soluble forms of Aß on synaptic form and function. Because selectively targeting synaptotoxic forms of soluble Aß could be therapeutically advantageous, it is important to understand the full range of soluble Aß derivatives. We previously described a Chinese hamster ovary (CHO) cell line (7PA2 cells) that stably expresses mutant human amyloid precursor protein (APP). Here, we extend this work by purifying an sodium dodecyl sulfate (SDS)-stable, ∼8 kDa Aß species from the 7PA2 medium. Mass spectrometry confirmed its identity as a noncovalently bonded Aß40 homodimer that impaired hippocampal long-term potentiation (LTP) in vivo. We further report the detection of Aß-containing fragments of APP in the 7PA2 medium that extend N-terminal from Asp1 of Aß. These N-terminally extended Aß-containing monomeric fragments are distinct from soluble Aß oligomers formed from Aß1-40/42 monomers and are bioactive synaptotoxins secreted by 7PA2 cells. Importantly, decreasing ß-secretase processing of APP elevated these alternative synaptotoxic APP fragments. We conclude that certain synaptotoxic Aß-containing species can arise from APP processing events N-terminal to the classical ß-secretase cleavage site.


Subject(s)
Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/metabolism , Neuronal Plasticity , Synapses/drug effects , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/isolation & purification , Amyloid beta-Peptides/pharmacology , Amyloid beta-Peptides/toxicity , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Culture Media, Conditioned , Humans , Long-Term Potentiation/drug effects , Male , Mice , Neuronal Plasticity/drug effects , Peptide Fragments , Rats
3.
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
4.
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
5.
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
6.
J Biol Chem ; 285(5): 3417-27, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19923222

ABSTRACT

Immunotherapy targeting of amyloid beta (Abeta) peptide in transgenic mouse models of Alzheimer disease (AD) has been widely demonstrated to resolve amyloid deposition as well as associated neuronal, glial, and inflammatory pathologies. These successes have provided the basis for ongoing clinical trials of immunotherapy for treatment of AD in humans. Acute as well as chronic Abeta-targeted immunotherapy has also been demonstrated to reverse Abeta-related behavioral deficits assessing memory in AD transgenic mouse models. We observe that three antibodies targeting the same linear epitope of Abeta, Abeta(3-7), differ in their ability to reverse contextual fear deficits in Tg2576 mice in an acute testing paradigm. Reversal of contextual fear deficit by the antibodies does not correlate with in vitro recognition of Abeta in a consistent or correlative manner. To better define differences in antigen recognition at the atomic level, we determined crystal structures of Fab fragments in complex with Abeta. The conformation of the Abeta peptide recognized by all three antibodies was highly related and is also remarkably similar to that observed in independently reported Abeta:antibody crystal structures. Sequence and structural differences between the antibodies, particularly in CDR3 of the heavy chain variable region, are proposed to account for differing in vivo properties of the antibodies under study. These findings provide a structural basis for immunotherapeutic strategies targeting Abeta species postulated to underlie cognitive deficits in AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Animals , Behavior, Animal , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray/methods , Disease Models, Animal , Epitopes/chemistry , Heterozygote , Humans , Kinetics , Male , Mice , Molecular Conformation , Recombinant Proteins/chemistry
7.
J Neurosci ; 29(36): 11393-8, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19741145

ABSTRACT

Amyloid beta (Abeta) immunotherapy is emerging as a promising disease-modifying therapy for Alzheimer's disease, although the precise mechanisms whereby anti-Abeta antibodies act against amyloid deposition and cognitive deficits remain elusive. To test the "peripheral sink" theory, which postulates that the effects of anti-Abeta antibodies in the systemic circulation are to promote the Abeta efflux from brain to blood, we studied the clearance of (125)I-Abeta(1-40) microinjected into mouse brains after intraperitoneal administration of an anti-Abeta monoclonal antibody 266. (125)I-Abeta(1-40) was rapidly eliminated from brains with a half-life of approximately 30 min in control mice, whereas 266 significantly retarded the elimination of Abeta, presumably due to formation of Abeta-antibody complex in brains. Administration of 266 to APP transgenic mice increased the levels of monomer Abeta species in an antibody-bound form, without affecting that of total Abeta. We propose a novel mechanism of Abeta immunotherapy by the class of anti-Abeta antibodies that preferentially bind soluble Abeta, i.e., intracerebral, rather than peripheral, sequestration of soluble, monomer form of Abeta, thereby preventing the accumulation of multimeric toxic Abeta species in brains.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Brain/immunology , Immunotherapy, Active/methods , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Brain/metabolism , Humans , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microinjections , Solubility
8.
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
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(3-4): 194-6, 2008.
Article in English | MEDLINE | ID: mdl-18322388

ABSTRACT

BACKGROUND: In a phase 2a clinical trial of AN1792 (Study 201), a potential immunotherapeutic agent for use in Alzheimer's disease (AD), approximately 6% of the treated AD patients (18/300) developed meningoencephalitis (ME). OBJECTIVE: To elucidate potential immune mechanisms of treatment-induced ME. METHODS: Peripheral blood mononuclear cells obtained from patients who received AN1792 were stimulated in vitro either with beta-amyloid (Abeta) or various overlapping peptides of Abeta(1-42), followed by quantification of cytokine-secreting cells by enzyme-linked immunosorbent spot assay. RESULTS: A significant difference in the quality of the T-cell responses between patients in Study 201 and those in earlier studies of AN1792 was noted. T-cell responses specific to the carboxy terminus of Abeta elicited from patients' peripheral blood mononuclear cells in an earlier multiple dose study (Study 102) were Th2 biased, while those from Study 201 were biased toward a proinflammatory Th1 response. Antibody responses in both studies were quantitatively and qualitatively similar (as determined by epitope mapping). The addition of polysorbate 80 to the formulation used in Study 201 is the most likely explanation for the difference in the T-cell response. CONCLUSION: ME following injection of AN1792 may be related to immune response differences driven by a formulation change. To address this, a novel peptide-carrier protein conjugate using an amino-terminal fragment of Abeta (ACC-001) has been developed to avoid potentially harmful T-cell responses, while maintaining a similar antibody response to that of AN1792. Immunotherapeutic trials using this treatment approach in AD patients are in progress.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Vaccines/adverse effects , Amyloid beta-Peptides/adverse effects , Immunotherapy, Active/adverse effects , Meningoencephalitis/chemically induced , Alzheimer Disease/epidemiology , Alzheimer Disease/immunology , Animals , Clinical Trials, Phase II as Topic/methods , Humans , Immunotherapy, Active/methods , Meningoencephalitis/epidemiology , Meningoencephalitis/immunology , Multicenter Studies as Topic/methods , Pilot Projects
11.
Neurodegener Dis ; 5(2): 55-9, 2008.
Article in English | MEDLINE | ID: mdl-18182779

ABSTRACT

BACKGROUND: Alpha-synuclein has been directly linked to Parkinson's disease etiology by mutations in and multiplication of its gene that result in a familial form of Parkinson's disease. Alpha-synuclein has been detected in blood, and was found to be elevated in the blood of those individuals with the alpha-synuclein gene multiplication. OBJECTIVE: A complete analysis of the level of alpha-synuclein in blood has not been performed. In this report, we determine the quantitative distribution of alpha-synuclein in the plasma and different cellular fractions of human blood. The levels of alpha-synuclein in human and mouse blood are compared. METHODS: Alpha-synuclein levels in the different fractions of blood were quantified by a sandwich ELISA with purified recombinant alpha-synuclein as an assay standard. Samples were further characterized by Western immunoblot analysis. RESULTS: More than 99% of the alpha-synuclein resides in the red blood cells (RBCs) with less than 1% of the total detected in the plasma, platelets and peripheral blood mononuclear cells. CONCLUSIONS: More than 99% of the alpha-synuclein in human blood is present in the peripheral blood cells, with the remainder in plasma. Fractionation of peripheral blood cells from human blood and quantification of alpha-synuclein revealed that only a very small amount of the total alpha-synuclein is present in peripheral blood mononuclear cells, and platelets, with the majority of alpha-synuclein in blood being present in RBCs. Considering the abundance and fragility of RBCs, alpha-synuclein levels in these other blood fractions or other bodily fluids such as cerebrospinal fluid may be artificially elevated by contamination with intact or lysed RBCs.


Subject(s)
Erythrocytes/chemistry , alpha-Synuclein/blood , Animals , Humans , Mice , Mice, Knockout , alpha-Synuclein/analysis
12.
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
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.
Curr Opin Immunol ; 16(5): 599-606, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15342006

ABSTRACT

As neuroscientists, we are taught that the brain is immune privileged and thus unlikely to be affected by the peripheral immune system. Accordingly, initial results demonstrating the effectiveness of beta-amyloid (Abeta) immunotherapy in mouse models of Alzheimer's disease (AD) were viewed with considerable surprise and some skepticism. Many groups have since demonstrated efficacy with Abeta immunotherapy in models of AD, using Abeta-based immunogens and anti-Abeta antibodies. Clinical trials involving Abeta immunotherapy for AD are in progress and are providing a wealth of information around the amyloid hypothesis of AD. Abeta immunotherapy is also raising new opportunities and questions about the general role of the immune system in neurodegenerative diseases.


Subject(s)
Alzheimer Disease/therapy , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Immunotherapy , Alzheimer Disease/prevention & control , Alzheimer Vaccines/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Humans
15.
J Chromatogr B Analyt Technol Biomed Life Sci ; 846(1-2): 24-31, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-16939717

ABSTRACT

The measurement of amyloid beta peptides (Abeta) in blood and plasma is expected to be a useful biomarker as potential therapeutics designed to lower Abeta peptide enter clinical trials. Many reports have suggested that Abeta could bind to substances in blood that may influence the recovery of Abeta peptide in plasma, its detection by conventional ELISAs or the actual turnover and half-life of the peptide in blood. In this study we describe a process for analyzing total Abeta in whole blood and plasma using denaturing solid-phase extraction followed by reverse-phase HPLC linked to ELISA. Comparison of total Abeta peptide levels in whole blood and plasma from the same bleed showed that most of the Abeta peptide is captured in the plasma if the samples are first denatured. In contrast, plasma that was assayed without denaturation could show greater than 70% reduction in apparent total Abeta peptide. This suggested that there was a pool of Abeta peptide in non-denatured plasma that is occluded from detection by ELISA, perhaps by binding to plasma proteins.


Subject(s)
Amyloid beta-Peptides/blood , Adult , Chromatography, High Pressure Liquid/methods , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
16.
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
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.
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
19.
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
20.
J Alzheimers Dis ; 10(4): 399-406, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17183151

ABSTRACT

BACKGROUND: Treatment with HMG-CoA reductase inhibitors ("statins") has been variably associated with a reduced risk of Alzheimer's disease (AD) in epidemiologic studies and reduced amyloid-beta (Abeta) deposition in animal models of AD. Putative neuroprotective effects of statins may vary in relation to their ability to penetrate into the central nervous system (CNS). METHODS: We measured levels of cerebrospinal fluid (CSF) AD biomarkers following 14 weeks of treatment with simvastatin (a CNS permeant statin; n=10) at 40 mg/day or pravastatin (a CNS impermeant statin; n=13) at 80 mg/day in hypercholesterolemic subjects without dementia. RESULTS: Simvastatin, but not pravastatin, reduced CSF levels of phospho-tau-181 (p-tau181) in all subjects. There were no differences in CSF levels of total tau, Abeta42, Abeta40, soluble amyloid beta protein precursor (sAbetaPP) alpha or beta, or F2-isoprostanes. CONCLUSIONS: Statins may modulate the phosphorylation of tau in humans and this effect may depend on the CNS availability of the statin. These results suggest another mechanism by which statins may act to reduce the risk of AD.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Neuroprotective Agents/therapeutic use , Pravastatin/therapeutic use , Simvastatin/therapeutic use , Adult , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/cerebrospinal fluid , Brain/metabolism , Cholesterol/blood , Cholesterol, LDL/blood , F2-Isoprostanes/cerebrospinal fluid , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypercholesterolemia/cerebrospinal fluid , Male , Middle Aged , Neuroprotective Agents/pharmacokinetics , Peptide Fragments/cerebrospinal fluid , Pravastatin/pharmacokinetics , Simvastatin/pharmacokinetics , Triglycerides/blood , tau Proteins/cerebrospinal fluid
SELECTION OF CITATIONS
SEARCH DETAIL