Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Alzheimers Dis ; 43(2): 575-88, 2015.
Article in English | MEDLINE | ID: mdl-25096615

ABSTRACT

Amyloid-ß (Aß) immunotherapy for Alzheimer's disease (AD) has good preclinical support from transgenic mouse models and clinical data suggesting that a long-term treatment effect is possible. Soluble Aß protofibrils have been shown to exhibit neurotoxicity in vitro and in vivo, and constitute an attractive target for immunotherapy. Here, we demonstrate that the humanized antibody BAN2401 and its murine version mAb158 exhibit a strong binding preference for Aß protofibrils over Aß monomers. Further, we confirm the presence of the target by showing that both antibodies efficiently immunoprecipitate soluble Aß aggregates in human AD brain extracts. mAb158 reached the brain and reduced the brain protofibril levels by 42% in an exposure-dependent manner both after long-term and short-term treatment in tg-ArcSwe mice. Notably, a 53% reduction of protofibrils/oligomers in cerebrospinal fluid (CSF) that correlated with reduced brain protofibril levels was observed after long-term treatment, suggesting that CSF protofibrils/oligomers could be used as a potential biomarker. No change in native monomeric Aß42 could be observed in brain TBS extracts after mAb158-treatment in tg-ArcSwe mice. By confirming the specific ability of mAb158 to selectively bind and reduce soluble Aß protofibrils, with minimal binding to Aß monomers, we provide further support in favor of its position as an attractive new candidate for AD immunotherapy. BAN2401 has undergone full phase 1 development, and available data indicate a favorable safety profile in AD patients.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Immunologic Factors/therapeutic use , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Immunologic Factors/pharmacology , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid , Presenilin-1/genetics , Protein Binding/drug effects , Protein Binding/genetics
2.
Neurobiol Dis ; 69: 134-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24851801

ABSTRACT

Several lines of evidence suggest that accumulation of aggregated alpha-synuclein (α-synuclein) in the central nervous system (CNS) is an early pathogenic event in Parkinson's disease and other Lewy body disorders. In recent years, animal studies have indicated immunotherapy with antibodies directed against α-synuclein as a promising novel treatment strategy. Since large α-synuclein oligomers, or protofibrils, have been demonstrated to possess pronounced cytotoxic properties, such species should be particularly attractive as therapeutic targets. In support of this, (Thy-1)-h[A30P] α-synuclein transgenic mice with motor dysfunction symptoms were found to display increased levels of α-synuclein protofibrils in the CNS. An α-synuclein protofibril-selective monoclonal antibody (mAb47) was evaluated in this α-synuclein transgenic mouse model. As measured by ELISA, 14month old mice treated for 14weeks with weekly intraperitoneal injections of mAb47 displayed significantly lower levels of both soluble and membrane-associated protofibrils in the spinal cord. Besides the lower levels of pathogenic α-synuclein demonstrated, a reduction of motor dysfunction in transgenic mice upon peripheral administration of mAb47 was indicated. Thus, immunotherapy with antibodies targeting toxic α-synuclein species holds promise as a future disease-modifying treatment in Parkinson's disease and related disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunization, Passive , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , Animals , Antibodies, Monoclonal/metabolism , Brain/immunology , Brain/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Intraperitoneal , Male , Mice, Transgenic , Motor Activity/physiology , Mutation , Parkinsonian Disorders/immunology , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/pathology , alpha-Synuclein/genetics
3.
Amyloid ; 20(4): 233-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24053224

ABSTRACT

Aggregated α-synuclein is the major component of Lewy bodies, protein inclusions observed in the brain in neurodegenerative disorders such as Parkinson's disease and dementia with Lewy bodies. Experimental evidence indicates that α-synuclein potentially can be transferred between cells and act as a seed to accelerate the aggregation process. Here, we investigated in vitro and in vivo seeding effects of α-synuclein oligomers induced by the reactive aldehyde 4-oxo-2-nonenal (ONE). As measured by a Thioflavin-T based fibrillization assay, there was an earlier onset of aggregation when α-synuclein oligomers were added to monomeric α-synuclein. In contrast, exogenously added α-synuclein oligomers did not induce aggregation in a cell model. However, cells overexpressing α-synuclein that were treated with the oligomers displayed reduced α-synuclein levels, indicating that internalized oligomers either decreased the expression or accelerated the degradation of transfected α-synuclein. Also in vivo there were no clear seeding effects, as intracerebral injections of α-synuclein oligomers into the neocortex of α-synuclein transgenic mice did not induce formation of proteinase K resistant α-synuclein pathology. Taken together, we could observe a seeding effect of the ONE-induced α-synuclein oligomers in a fibrillization assay, but neither in a cell nor in a mouse model.


Subject(s)
alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Aldehydes/chemistry , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Microscopy, Atomic Force , Parkinson Disease/metabolism , Parkinson Disease/pathology
4.
J Neurochem ; 126(1): 131-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23363402

ABSTRACT

Inclusions of intraneuronal alpha-synuclein (α-synuclein) can be detected in brains of patients with Parkinson's disease and dementia with Lewy bodies. The aggregation of α-synuclein is a central feature of the disease pathogenesis. Among the different α-synuclein species, large oligomers/protofibrils have particular neurotoxic properties and should therefore be suitable as both therapeutic and diagnostic targets. Two monoclonal antibodies, mAb38F and mAb38E2, with high affinity and strong selectivity for large α-synuclein oligomers were generated. These antibodies, which do not bind amyloid-beta or tau, recognize Lewy body pathology in brains from patients with Parkinson's disease and dementia with Lewy bodies and detect pathology earlier in α-synuclein transgenic mice than linear epitope antibodies. An oligomer-selective sandwich ELISA, based on mAb38F, was set up to analyze brain extracts of the transgenic mice. The overall levels of α-synuclein oligomers/protofibrils were found to increase with age in these mice, although the levels displayed a large interindividual variation. Upon subcellular fractionation, higher levels of α-synuclein oligomers/protofibrils could be detected in the endoplasmic reticulum around the age when behavioral disturbances develop. In summary, our novel oligomer-selective α-synuclein antibodies recognize relevant pathology and should be important tools to further explore the pathogenic mechanisms in Lewy body disorders. Moreover, they could be potential candidates both for immunotherapy and as reagents in an assay to assess a potential disease biomarker.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain/pathology , Lewy Body Disease/genetics , Lewy Body Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/immunology , Animals , Blotting, Western , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Epitopes , Formates/chemistry , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Mutation/genetics , Mutation/physiology , Subcellular Fractions/metabolism
5.
FEBS J ; 276(4): 995-1006, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19215301

ABSTRACT

Oligomeric assemblies of amyloid-beta (Abeta) are suggested to be central in the pathogenesis of Alzheimer's disease because levels of soluble Abeta correlate much better with the extent of cognitive dysfunctions than do senile plaque counts. Moreover, such Abeta species have been shown to be neurotoxic, to interfere with learned behavior and to inhibit the maintenance of hippocampal long-term potentiation. The tg-ArcSwe model (i.e. transgenic mice with the Arctic and Swedish Alzheimer mutations) expresses elevated levels of Abeta protofibrils in the brain, making tg-ArcSwe a highly suitable model for investigating the pathogenic role of these Abeta assemblies. In the present study, we estimated Abeta protofibril levels in the brain and cerebrospinal fluid of tg-ArcSwe mice, and also assessed their role with respect to cognitive functions. Protofibril levels, specifically measured with a sandwich ELISA, were found to be elevated in young tg-ArcSwe mice compared to several transgenic models lacking the Arctic mutation. In aged tg-ArcSwe mice with considerable plaque deposition, Abeta protofibrils were approximately 50% higher than in younger mice, whereas levels of total Abeta were exponentially increased. Young tg-ArcSwe mice showed deficits in spatial learning, and individual performances in the Morris water maze were correlated inversely with levels of Abeta protofibrils, but not with total Abeta levels. We conclude that Abeta protofibrils accumulate in an age-dependent manner in tg-ArcSwe mice, although to a far lesser extent than total Abeta. Our findings suggest that increased levels of Abeta protofibrils could result in spatial learning impairment.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Animals , Mice , Mice, Transgenic , Mutation , Plaque, Amyloid/pathology
6.
Neurobiol Aging ; 30(8): 1238-44, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18304698

ABSTRACT

The amyloid precursor protein (APP) is the source of beta-amyloid, a pivotal peptide in the pathogenesis of Alzheimer's disease (AD). This study examines the possible effect of APP transgene expression on neuronal size by measuring the volumes of cortical neurons (microm(3)) in transgenic mouse models with familial AD Swedish mutation (APPswe), with or without mutated presenilin1 (PS1dE9), as well as in mice carrying wild-type APP (APPwt). Overexpression of APPswe and APPwt protein, but not of PS1dE9 alone, resulted in a greater percentage of medium-sized neurons and a proportionate decrease in the percentage of small-sized neurons. Our observations indicate that the overexpression of mutant (APPswe) or wild-type APP in transgenic mice is necessary and sufficient for hypertrophy of cortical neurons. This is highly suggestive of a neurotrophic effect and also raises the possibility that the lack of neuronal loss in transgenic mouse models of AD may be attributed to overexpression of APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Size , Cerebral Cortex/cytology , Neurons/cytology , Receptors, Cell Surface/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cerebral Cortex/metabolism , Female , Male , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Protease Nexins , Receptors, Cell Surface/genetics , Sex Characteristics
7.
Neuromolecular Med ; 10(3): 195-207, 2008.
Article in English | MEDLINE | ID: mdl-18543125

ABSTRACT

Amyloid plaques are composed primarily of amyloid-beta (Abeta) peptides derived from proteolytic cleavage of amyloid precursor protein (APP) and are considered to play a pivotal role in Alzheimer's disease (AD) pathogenesis. Presently, AD is diagnosed after the onset of clinical manifestations. With the arrival of novel therapeutic agents for treatment of AD, there is an urgent need for biomarkers to detect early stages of AD. Measurement of plasma Abeta has been suggested as an inexpensive and non-invasive tool to diagnose AD and to monitor Abeta modifying therapies. However, the majority of cross-sectional studies on plasma Abeta levels in humans have not shown differences between individuals with AD compared to controls. Similarly, cross-sectional studies of mouse plasma Abeta have yielded inconsistent trends in different mouse models. However, longitudinal studies appear to be more promising in humans. Recently, efforts to modify plasma Abeta levels using modulators have shown some promise. In this review, we will summarize the present data on plasma Abeta in humans and mouse models of AD. We will discuss the potential of modulators of Abeta levels in plasma, including antibodies and insulin, and the challenges associated with measuring plasma Abeta. Modulators of plasma Abeta may provide an important tool to optimize plasma Abeta levels and may improve the diagnostic potential of this approach.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/blood , Animals , Humans , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism
8.
J Neuropathol Exp Neurol ; 67(1): 30-40, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18091561

ABSTRACT

Recent studies have demonstrated the potential utility of antibodies for the treatment of Alzheimer disease (AD). In transgenic mouse models of AD, peripheral and intracerebral administration of Abeta-specific antibodies reduces amyloid burdens to varied extents. The mechanism may involve clearance of pre-existing amyloid plaques or prevention of new amyloid formation. Here, we have used two transgenic models, the inducible CamKII-ttAxtetAPP/swe/ind (Line 107) and the APPswe/PS1dE9 (Line 85), to test the ability of intracerebral injection of Abeta antibodies to clear amyloid. Because the production of Abeta peptides in the Line 107 model is inducible, whereas production in Line 85 mice is constitutive, we could study the effects of antibody on pre-existing plaques versus continuous plaque formation. In Line 85, injection of antibody resulted in modest but statistically significant reductions in amyloid burden (average, 14%-16%). However, injected antibodies had no effect on amyloid burden in Line 107 under conditions in which the production of Abeta was suppressed, indicating that pre-existing plaques are not rapidly cleared. These results indicate that intracerebral injection of Abeta antibodies produces modest reductions in amyloid deposition in these two models and that the mechanism may involve prevention of amyloid formation rather than clearance of pre-existing plaques.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Immunization, Passive , Immunoglobulin G/therapeutic use , Plaque, Amyloid/drug effects , Amyloid beta-Protein Precursor/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Hum Mol Genet ; 13(15): 1599-610, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15190011

ABSTRACT

Huntington's disease (HD) results from the expansion of a glutamine repeat near the N-terminus of huntingtin (htt). At post-mortem, neurons in the central nervous system of patients have been found to accumulate N-terminal fragments of mutant htt in nuclear and cytoplasmic inclusions. This pathology has been reproduced in transgenic mice expressing the first 171 amino acids of htt with 82 glutamines along with losses of motoric function, hypoactivity and abbreviated life-span. The relative contributions of nuclear versus cytoplasmic mutant htt to the pathogenesis of disease have not been clarified. To examine whether pathogenic processes in the nucleus disproportionately contribute to disease features in vivo, we fused a nuclear localization signal (NLS) derived from atrophin-1 to the N-terminus of an N171-82Q construct. Two lines of mice (lines 8A and 61) that were identified expressed NLS-N171-82Q at comparable levels and developed phenotypes identical to our previously described HD-N171-82Q mice. Western blot and immunohistochemical analyses revealed that NLS-N171-82Q fragments accumulate in nuclear, but not cytoplasmic, compartments. These data suggest that disruption of nuclear processes may account for many of the disease phenotypes displayed in the mouse models generated by expressing mutant N-terminal fragments of htt.


Subject(s)
Cell Nucleus/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptide Fragments/genetics , Age Factors , Animals , Cell Nucleus/pathology , Disease Models, Animal , Genetic Vectors , Humans , Huntingtin Protein , Huntington Disease/metabolism , Immunohistochemistry , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/metabolism , Intranuclear Inclusion Bodies/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL