Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Front Neurosci ; 16: 848215, 2022.
Article in English | MEDLINE | ID: mdl-35557606

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disease that affects 50 million people worldwide, with 10 million new cases occurring each year. The emotional and economic impacts of AD on patients and families are devastating. Approved treatments confer modest improvement in symptoms, and recently one treatment obtained accelerated approval from the United States Food and Drug Administration (FDA) and may have modest disease modifying benefit. Research over the past three decades has established a clear causal linkage between AD and elevated brain levels of amyloid ß (Aß) peptide, and substantial evidence now implicates soluble, non-fibrillar Aß oligomers (AßOs) as the molecular assemblies directly responsible for AD-associated memory and cognitive failure and accompanying progressive neurodegeneration. The widely recognized linkage of elevated Aß and AD spawned a comprehensive 20-year therapeutic campaign that focused primarily on two strategies - inhibition of the secretase enzymes responsible for Aß production and clearance of Aß peptide or amyloid plaques with Aß-directed immunotherapeutics. Unfortunately, all clinical trials of secretase inhibitors were unsuccessful. Of the completed phase 3 immunotherapy programs, bapineuzumab (targeting amyloid plaque) and solanezumab (targeting Aß monomers) were negative, and the crenezumab program (targeting Aß monomers and to a small extent oligomers) was stopped for futility. Aducanumab (targeting amyloid plaques), which recently received FDA accelerated approval, had one positive and one negative phase 3 trial. More than 25 negative randomized clinical trials (RCTs) have evaluated Aß-targeting therapeutics, yet none has directly evaluated whether selective blockage of disease-relevant AßOs can stop or reverse AD-associated cognitive decline. Here, we briefly summarize studies that establish the AD therapeutic rationale to target AßOs selectively, and we describe ACU193, the first AßO-selective immunotherapeutic to enter human clinical trials and the first positioned to test the AßO hypothesis of AD.

2.
Sci Rep ; 8(1): 4634, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545579

ABSTRACT

Soluble amyloid ß oligomers (AßOs) are widely recognized neurotoxins that trigger aberrant signaling in specific subsets of neurons, leading to accumulated neuronal damage and memory disorders in Alzheimer's disease (AD). One of the profound downstream consequences of AßO-triggered events is dysregulation of cytosolic calcium concentration ([Ca2+]i), which has been implicated in synaptic failure, cytoskeletal abnormalities, and eventually neuronal death. We have developed an in vitro/in vivo drug screening assay to evaluate putative AßO-blocking candidates by measuring AßO-induced real-time changes in [Ca2+]i. Our screening assay demonstrated that the anti-AßO monoclonal antibody ACU3B3 exhibits potent blocking capability against a broad size range of AßOs. We showed that picomolar concentrations of AßOs were capable of increasing [Ca2+]i in primary neuronal cultures, an effect prevented by ACU3B3. Topical application of 5 nM AßOs onto exposed cortical surfaces also elicited significant calcium elevations in vivo, which was completely abolished by pre-treatment of the brain with 1 ng/mL (6.67 pM) ACU3B3. Our results provide strong support for the utility of this functional screening assay in identifying and confirming the efficacy of AßO-blocking drug candidates such as the human homolog of ACU3B3, which may emerge as the first experimental AD therapeutic to validate the amyloid oligomer hypothesis.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Calcium/metabolism , Image Processing, Computer-Assisted/methods , Aged, 80 and over , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Case-Control Studies , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL
3.
Alzheimers Res Ther ; 6(4): 42, 2014.
Article in English | MEDLINE | ID: mdl-25045405

ABSTRACT

Levels of amyloid-beta monomer and deposited amyloid-beta in the Alzheimer's disease brain are orders of magnitude greater than soluble amyloid-beta oligomer levels. Monomeric amyloid-beta has no known direct toxicity. Insoluble fibrillar amyloid-beta has been proposed to be an in vivo mechanism for removal of soluble amyloid-beta and exhibits relatively low toxicity. In contrast, soluble amyloid-beta oligomers are widely reported to be the most toxic amyloid-beta form, both causing acute synaptotoxicity and inducing neurodegenerative processes. None of the amyloid-beta immunotherapies currently in clinical development selectively target soluble amyloid-beta oligomers, and their lack of efficacy is not unexpected considering their selectivity for monomeric or fibrillar amyloid-beta (or both) rather than soluble amyloid-beta oligomers. Because they exhibit acute, memory-compromising synaptic toxicity and induce chronic neurodegenerative toxicity and because they exist at very low in vivo levels in the Alzheimer's disease brain, soluble amyloid-beta oligomers constitute an optimal immunotherapeutic target that should be pursued more aggressively.

4.
Trends Pharmacol Sci ; 34(5): 261-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23582316

ABSTRACT

Soluble Aß oligomers are now widely recognized as key pathogenic structures in Alzheimer's disease. They inhibit synaptic function, leading to early memory deficits and synaptic degeneration, and they trigger the downstream neuronal signaling responsible for phospho-tau Alzheimer's pathology. The marginal effects observed in recent clinical studies of solanezumab, targeting monomeric Aß, and bapineuzumab, targeting amyloid plaques, prompted expert comments that drug discovery efforts in Alzheimer's disease should focus on soluble forms of Aß rather than fibrillar Aß deposits found in amyloid plaques. Accumulating scientific data suggest that soluble Aß oligomers represent the optimal intervention target within the amyloid manifold. Active drug discovery approaches include antibodies that selectively capture soluble Aß oligomers, selective modifiers of oligomer assembly, and receptor antagonists. The onset of symptomatic clinical benefit is expected to be rapid for such agents, because neuronal memory signaling should normalize on blockage of soluble Aß oligomers. This key feature is not shared by amyloid-lowering therapeutics, and it should translate into streamlined clinical development for oligomer-targeting drugs. Oligomer-targeting drugs should also confer long-term disease modification and slowing of disease progression, because they prevent the downstream signaling responsible for phospho-tau mediated cytoskeletal degeneration.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Humans , Molecular Targeted Therapy/methods
5.
Ann Neurol ; 68(2): 220-30, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20641005

ABSTRACT

OBJECTIVE: Recent evidence suggests that high molecular weight soluble oligomeric Abeta (oAbeta) assemblies (also known as Abeta-derived diffusible ligands, or ADDLs) may represent a primary neurotoxic basis for cognitive failure in Alzheimer disease (AD). To date, most in vivo studies of oAbeta/ADDLs have involved injection of assemblies purified from the cerebrospinal fluid of human subjects with AD or from the conditioned media of Abeta-secreting cells into experimental animals. We sought to study the bioactivities of endogenously formed oAbeta/ADDLs generated in situ from the physiological processing of human amyloid precursor protein (APP) and presenitin1 (PS1) transgenes. METHODS: We produced and histologically characterized single transgenic mice overexpressing APP(E693Q) or APP(E693Q) X PS1DeltaE9 bigenic mice. APP(E693Q) mice were studied in the Morris water maze (MWM) task at 6 and 12 months of age. Following the second MWM evaluation, mice were sacrificed, and brains were assayed for Abetatotal, Abeta40, Abeta42, and oAbeta/ADDLs by enzyme-linked immunosorbent assay (ELISA) and were also histologically examined. Based on results from the oAbeta/ADDL ELISA, we assigned individual APP(E693Q) mice to either an undetectable oAbeta/ADDLs group or a readily detectable oAbeta/ADDLs group. A days to criterion (DTC) analysis was used to determine delays in acquisition of the MWM task. RESULTS: Both single transgenic and bigenic mice developed intraneuronal accumulation of APP/Abeta, although only APP(E693Q) X PS1Delta9 bigenic mice developed amyloid plaques. The APP(E693Q) mice did not develop amyloid plaques at any age studied, up to 30 months. APP(E693Q) mice were tested for spatial learning and memory, and only 12-month-old APP(E693Q) mice with readily detectable oAbeta/ADDLs displayed a significant delay in acquisition of the MWM task when compared to nontransgenic littermates. INTERPRETATION: These data suggest that cerebral oAbeta/ADDL assemblies generated in brain in situ from human APP transgenes may be associated with cognitive impairment. We propose that a DTC analysis may be a sensitive method for assessing the cognitive impact in mice of endogenously generated oligomeric human Abeta assemblies. ANN NEUROL 2010.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Behavior, Animal/drug effects , Peptide Fragments/toxicity , Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/toxicity , Animals , Behavior, Animal/physiology , Brain Chemistry/genetics , Disease Models, Animal , Female , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Processing, Post-Translational/genetics
6.
Neuropharmacology ; 59(4-5): 230-42, 2010.
Article in English | MEDLINE | ID: mdl-20650286

ABSTRACT

Today, it is widely accepted that ADDLs, soluble oligomeric assemblies of the amyloid beta peptide, play a prominent role in triggering the cognitive deficits and neurodegeneration that constitute Alzheimer's disease (AD). Within the past decade, the longstanding emphasis on fibrillar deposits and neuronal death has given way to a new paradigm involving ADDL-triggered aberrant synaptic signaling and consequent memory malfunction and neurodegeneration. As with any paradigm shift in biology, not all molecular details have been elucidated, and not all AD scientists are fully subscribed. Nevertheless, the ADDL paradigm affords a promising framework for ongoing AD research and for development of the first therapeutics endowed with the dual capabilities of immediate symptom reversal and long-term disease modification. In this review we provide a brief account of the discovery of ADDLs, followed by a summary of key results that address questions concerning ADDL structure and assembly, biological activity and therapeutic possibilities.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Signal Transduction/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Humans , Immunologic Factors/physiology , Ligands , Neurons/metabolism , Neurons/pathology , Signal Transduction/drug effects
7.
J Biol Chem ; 284(28): 18742-53, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19406747

ABSTRACT

Accumulation of amyloid beta (Abeta) oligomers in the brain is toxic to synapses and may play an important role in memory loss in Alzheimer disease. However, how these toxins are built up in the brain is not understood. In this study we investigate whether impairments of insulin and insulin-like growth factor-1 (IGF-1) receptors play a role in aggregation of Abeta. Using primary neuronal culture and immortal cell line models, we show that expression of normal insulin or IGF-1 receptors confers cells with abilities to reduce exogenously applied Abeta oligomers (also known as ADDLs) to monomers. In contrast, transfection of malfunctioning human insulin receptor mutants, identified originally from patient with insulin resistance syndrome, or inhibition of insulin and IGF-1 receptors via pharmacological reagents increases ADDL levels by exacerbating their aggregation. In healthy cells, activation of insulin and IGF-1 receptor reduces the extracellular ADDLs applied to cells via seemingly the insulin-degrading enzyme activity. Although insulin triggers ADDL internalization, IGF-1 appears to keep ADDLs on the cell surface. Nevertheless, both insulin and IGF-1 reduce ADDL binding, protect synapses from ADDL synaptotoxic effects, and prevent the ADDL-induced surface insulin receptor loss. Our results suggest that dysfunctions of brain insulin and IGF-1 receptors contribute to Abeta aggregation and subsequent synaptic loss.


Subject(s)
Amyloid beta-Peptides/chemistry , Antigens, CD/chemistry , Insulin/chemistry , Receptor, Insulin/chemistry , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Mice , Models, Biological , Mutation , NIH 3T3 Cells , Neurons/metabolism , Neurotoxins/chemistry , Rats , Synapses/metabolism
8.
J Chem Phys ; 129(21): 214707, 2008 Dec 07.
Article in English | MEDLINE | ID: mdl-19063575

ABSTRACT

Here, we present a novel computational approach for describing the formation of oligomeric assemblies at experimental concentrations and timescales. We propose an extension to the Markovian state model approach, where one includes low concentration oligomeric states analytically. This allows simulation on long timescales (seconds timescale) and at arbitrarily low concentrations (e.g., the micromolar concentrations found in experiments), while still using an all-atom model for protein and solvent. As a proof of concept, we apply this methodology to the oligomerization of an Abeta peptide fragment (Abeta(21-43)). Abeta oligomers are now widely recognized as the primary neurotoxic structures leading to Alzheimer's disease. Our computational methods predict that Abeta trimers form at micromolar concentrations in 10 ms, while tetramers form 1000 times more slowly. Moreover, the simulation results predict specific intermonomer contacts present in the oligomer ensemble as well as putative structures for small molecular weight oligomers. Based on our simulations and statistical models, we propose a novel mutation to stabilize the trimeric form of Abeta in an experimentally verifiable manner.


Subject(s)
Markov Chains , Models, Molecular , Amyloid/chemistry , Hydrophobic and Hydrophilic Interactions , Kinetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Time Factors
9.
FASEB J ; 22(1): 246-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17720802

ABSTRACT

Recent studies have indicated an association between Alzheimer's disease (AD) and central nervous system (CNS) insulin resistance. However, the cellular mechanisms underlying the link between these two pathologies have not been elucidated. Here we show that signal transduction by neuronal insulin receptors (IR) is strikingly sensitive to disruption by soluble Abeta oligomers (also known as ADDLs). ADDLs are known to accumulate in AD brain and have recently been implicated as primary candidates for initiating deterioration of synapse function, composition, and structure. Using mature cultures of hippocampal neurons, a preferred model for studies of synaptic cell biology, we found that ADDLs caused a rapid and substantial loss of neuronal surface IRs specifically on dendrites bound by ADDLs. Removal of dendritic IRs was associated with increased receptor immunoreactivity in the cell body, indicating redistribution of the receptors. The neuronal response to insulin, measured by evoked IR tyrosine autophosphorylation, was greatly inhibited by ADDLs. Inhibition also was seen with added glutamate or potassium-induced depolarization. The effects on IR function were completely blocked by NMDA receptor antagonists, tetrodotoxin, and calcium chelator BAPTA-AM. Downstream from the IR, ADDLs induced a phosphorylation of Akt at serine473, a modification associated with neurodegenerative and insulin resistance diseases. These results identify novel factors that affect neuronal IR signaling and suggest that insulin resistance in AD brain is a response to ADDLs, which disrupt insulin signaling and may cause a brain-specific form of diabetes as part of an overall pathogenic impact on CNS synapses.


Subject(s)
Amyloid beta-Peptides/physiology , Biopolymers/physiology , Neurons/physiology , Receptor, Insulin/physiology , Animals , Cells, Cultured , Dendrites/enzymology , Humans , Insulin/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
10.
J Neurochem ; 100(1): 23-35, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17116235

ABSTRACT

Amyloid beta (Abeta) immunotherapy for Alzheimer's disease has shown initial success in mouse models of Alzheimer's disease and in human patients. However, because of meningoencephalitis in clinical trials of active vaccination, approaches using therapeutic antibodies may be preferred. As a novel antigen to generate monoclonal antibodies, the current study has used Abeta oligomers (amyloid beta-derived diffusible ligands, ADDLs), pathological assemblies known to accumulate in Alzheimer's disease brain. Clones were selected for the ability to discriminate Alzheimer's disease from control brains in extracts and tissue sections. These antibodies recognized Abeta oligomers and fibrils but not the physiologically prevalent Abeta monomer. Discrimination derived from an epitope found in assemblies of Abeta1-28 and ADDLs but not in other sequences, including Abeta1-40. Immunoneutralization experiments showed that toxicity and attachment of ADDLs to synapses in culture could be prevented. ADDL-induced reactive oxygen species (ROS) generation was also inhibited, establishing this response to be oligomer-dependent. Inhibition occurred whether ADDLs were prepared in vitro or obtained from Alzheimer's disease brain. As conformationally sensitive monoclonal antibodies that selectively immunoneutralize binding and function of pathological Abeta assemblies, these antibodies provide tools by which pathological Abeta assemblies from Alzheimer's disease brain might be isolated and evaluated, as well as offering a valuable prototype for new antibodies useful for Alzheimer's disease therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/physiology , Antibody Specificity , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Calmodulin-Binding Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Epitopes , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoblotting/methods , Immunohistochemistry/methods , Mice , Neurons/metabolism , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Protein Binding/drug effects , Rabbits , Reactive Oxygen Species/metabolism , Tetrazolium Salts , Thiazoles
11.
Curr Alzheimer Res ; 4(5): 562-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18220523

ABSTRACT

Amyloid beta-derived diffusible ligands (ADDLs) comprise the neurotoxic subset of soluble Abeta(1-42) oligomers, now widely considered to be the molecular cause of memory malfunction and neurodegeneration in Alzheimer's disease (AD). We have developed a screening cascade which identifies small molecule modulators of ADDL-mediated neurotoxicity. The primary screen involves a fluorescence resonance energy transfer (FRET)-based assay which selects inhibitors of Abeta1-42 oligomer assembly. The identified hits were further characterized by assessing their ability to inhibit the assembly and binding of ADDLs to cultures of primary hippocampal neurons. This approach has led to the identification of a number of small molecules which inhibit ADDL assembly and their subsequent binding to neurons. Here we describe our small molecule discovery efforts to identify ADDL assembly blocker and ADDL binding inhibitors, and to transform validated hits into pre-clinical lead compounds.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antipsychotic Agents/therapeutic use , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Animals , Antipsychotic Agents/chemistry , Drug Design , Humans , Small Molecule Libraries
12.
Curr Top Med Chem ; 6(6): 597-608, 2006.
Article in English | MEDLINE | ID: mdl-16712494

ABSTRACT

The amyloid-beta (Abeta) cascade hypothesis of Alzheimer's disease (AD) has dominated research and subsequent therapeutic drug development for over two decades. Central to this hypothesis is the observation that Abeta is elevated in AD patients and that the disease is ultimately characterized by the central deposition of insoluble senile plaques. More recent evidence, however, suggests that the presence or absence of plaque is insufficient to fully account for the deleterious role of elevated Abeta in AD. Such studies support the basis for an alternate interpretation of the Abeta cascade hypothesis. Namely, that soluble oligomers of Abeta (i.e., ADDLs) accumulate and cause functional deficits prior to overt neuronal cell death or plaque deposition. Accordingly, the following review focuses on research describing the preparation and functional activity of ADDLs in vitro and in vivo. These studies provide the basis for an alternate, ADDL-based, view of the Abeta cascade hypothesis and accounts for the disconnect between plaque burden and cognitive deficits. Possible therapeutic approaches aimed at lowering ADDLs in AD patients are also considered.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Animals , Humans , Ligands
13.
J Neurosci ; 24(45): 10191-200, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537891

ABSTRACT

The cognitive hallmark of early Alzheimer's disease (AD) is an extraordinary inability to form new memories. For many years, this dementia was attributed to nerve-cell death induced by deposits of fibrillar amyloid beta (Abeta). A newer hypothesis has emerged, however, in which early memory loss is considered a synapse failure caused by soluble Abeta oligomers. Such oligomers rapidly block long-term potentiation, a classic experimental paradigm for synaptic plasticity, and they are strikingly elevated in AD brain tissue and transgenic-mouse AD models. The current work characterizes the manner in which Abeta oligomers attack neurons. Antibodies raised against synthetic oligomers applied to AD brain sections were found to give diffuse stain around neuronal cell bodies, suggestive of a dendritic pattern, whereas soluble brain extracts showed robust AD-dependent reactivity in dot immunoblots. Antigens in unfractionated AD extracts attached with specificity to cultured rat hippocampal neurons, binding within dendritic arbors at discrete puncta. Crude fractionation showed ligand size to be between 10 and 100 kDa. Synthetic Abeta oligomers of the same size gave identical punctate binding, which was highly selective for particular neurons. Image analysis by confocal double-label immunofluorescence established that >90% of the punctate oligomer binding sites colocalized with the synaptic marker PSD-95 (postsynaptic density protein 95). Synaptic binding was accompanied by ectopic induction of Arc, a synaptic immediate-early gene, the overexpression of which has been linked to dysfunctional learning. Results suggest the hypothesis that targeting and functional disruption of particular synapses by Abeta oligomers may provide a molecular basis for the specific loss of memory function in early AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Peptide Fragments/pharmacology , Synapses/chemistry , Aged , Aged, 80 and over , Amyloid beta-Peptides/chemistry , Animals , Binding Sites , Cerebral Cortex/chemistry , Cerebrospinal Fluid Proteins/metabolism , Chromatography, High Pressure Liquid , Cytoskeletal Proteins/physiology , Female , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Nerve Tissue Proteins/physiology , Neurons/metabolism , Neurons/ultrastructure , Peptide Fragments/chemistry , Protein Binding , Rats , Synapses/physiology , Tissue Extracts/metabolism , Tissue Extracts/pharmacology
14.
Biochemistry ; 42(44): 12749-60, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14596589

ABSTRACT

Amyloid beta 1-42 (Abeta(1-42)) is a self-associating peptide that becomes neurotoxic upon aggregation. Toxicity originally was attributed to the presence of large, readily formed Abeta fibrils, but a variety of other toxic species are now known. The current study shows that Abeta(1-42) can self-assemble into small, stable globular assemblies free of fibrils and protofibrils. Absence of large molecules was verified by atomic force microscopy (AFM) and nondenaturing gel electrophoresis. Denaturing electrophoresis revealed that the globular assemblies comprised oligomers ranging from trimers to 24mers. Oligomers prepared at 4 degrees C stayed fibril-free for days and remained so when shifted to 37 degrees C, although the spectrum of sizes shifted toward larger oligomers at the higher temperature. The soluble, globular Abeta(1-42) oligomers were toxic to PC12 cells, impairing reduction of MTT and interfering with ERK and Rac signal transduction. Occasionally, oligomers were neither toxic nor recognized by toxicity-neutralizing antibodies, suggesting that oligomers could assume alternative conformations. Tests for oligomerization-blocking activity were carried out by dot-blot immunoassays and showed that neuroprotective extracts of Ginkgo biloba could inhibit oligomer formation at very low doses. The observed neurotoxicity, structure, and stability of synthetic Abeta(1-42) globular assemblies support the hypothesis that Abeta(1-42) oligomers play a role in triggering nerve cell dysfunction and death in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Neurotoxins/chemistry , Neurotoxins/toxicity , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/isolation & purification , Animals , Body Temperature , Cell Survival/drug effects , Diffusion , Epitopes/chemistry , Ginkgo biloba , Humans , Ligands , Neuroprotective Agents/chemistry , Neurotoxins/antagonists & inhibitors , Neurotoxins/isolation & purification , Nitroblue Tetrazolium/chemistry , PC12 Cells , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/isolation & purification , Plant Extracts/chemistry , Protein Conformation , Rats
15.
Proc Natl Acad Sci U S A ; 100(18): 10417-22, 2003 Sep 02.
Article in English | MEDLINE | ID: mdl-12925731

ABSTRACT

A molecular basis for memory failure in Alzheimer's disease (AD) has been recently hypothesized, in which a significant role is attributed to small, soluble oligomers of amyloid beta-peptide (A beta). A beta oligomeric ligands (also known as ADDLs) are known to be potent inhibitors of hippocampal long-term potentiation, which is a paradigm for synaptic plasticity, and have been linked to synapse loss and reversible memory failure in transgenic mouse AD models. If such oligomers were to build up in human brain, their neurological impact could provide the missing link that accounts for the poor correlation between AD dementia and amyloid plaques. This article, using antibodies raised against synthetic A beta oligomers, verifies the predicted accumulation of soluble oligomers in AD frontal cortex. Oligomers in AD reach levels up to 70-fold over control brains. Brain-derived and synthetic oligomers show structural equivalence with respect to mass, isoelectric point, and recognition by conformation-sensitive antibodies. Both oligomers, moreover, exhibit the same striking patterns of attachment to cultured hippocampal neurons, binding on dendrite surfaces in small clusters with ligand-like specificity. Binding assays using solubilized membranes show oligomers to be high-affinity ligands for a small number of nonabundant proteins. Current results confirm the prediction that soluble oligomeric A beta ligands are intrinsic to AD pathology, and validate their use in new approaches to therapeutic AD drugs and vaccines.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Brain Chemistry , Oligopeptides/analysis , Peptide Fragments/analysis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Binding Sites , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Immunoblotting , Ligands , Long-Term Potentiation , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Tissue Extracts/analysis
16.
FASEB J ; 17(1): 118-20, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12424218

ABSTRACT

The prevailing amyloid hypothesis for Alzheimer's disease (AD) holds that amyloid beta-protein (Abeta) causes neuronal degeneration by forming neurotoxic fibrillar structures. Yet, many aspects of AD pathology and symptoms are not well explained by this hypothesis. Here, we present evidence that neurotoxicity of soluble oligomeric Abeta closely corresponds to the selective neurodegeneration so distinctly manifest in AD. Selectivity was first observed in vitro, where only the human central nervous system neuronal cells were susceptible to soluble oligomeric Abeta. Furthermore, in mouse cerebral slice treated with soluble oligomeric Abeta, selective regiospecific toxicity was evident in the hippocampal CA1, a division important for memory, but not in the CA3 subfield. The fibrillar Abeta, however, killed neurons in all regions of the cerebral slice cultures and also in cerebellar slices. Remarkably, even at the highest soluble oligomeric Abeta concentrations, cerebellar neurons were completely spared, consistent with one of the hallmark features of AD pathology. Our observation of the selective neurodegeneration of soluble oligomeric Abeta to neurons involved in cognitive function may provide a new opportunity for the development of an effective AD therapy as well as elucidating the pathological mechanism of AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Nerve Degeneration/chemically induced , Alzheimer Disease/etiology , Amyloid beta-Peptides/chemistry , Animals , Cell Line , Central Nervous System/cytology , Cerebellum/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Mice , Models, Neurological , Nerve Degeneration/pathology , Neurons/drug effects
17.
J Biol Chem ; 278(13): 11612-22, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12499373

ABSTRACT

Extensive research causally links amyloid-beta peptide (A beta) to Alzheimer's disease, although the pathologically relevant A beta conformation remains unclear. A beta spontaneously aggregates into the fibrils that deposit in senile plaques. However, recent in vivo and in vitro reports describe a potent biological activity for oligomeric assemblies of A beta. To consistently prepare in vitro oligomeric and fibrillar forms of A beta 1-42, a detailed knowledge of how solution parameters influence structure is required. This manuscript represents the first study using a single chemically and structurally homogeneous unaggregated starting material to demonstrate that the formation of oligomers, fibrils, and fibrillar aggregates is determined by time, concentration, temperature, pH, ionic strength, and A beta species. We recently reported that oligomers inhibit neuronal viability 10-fold more than fibrils and approximately 40-fold more than unaggregated peptide, with oligomeric A beta 1-42-induced neurotoxicity significant at 10 nm. In addition, we were able to differentiate by structure and neurotoxic activity wild-type A beta1-42 from isoforms containing familial mutations (Dahlgren, K. N., Manelli, A. M., Stine, W. B., Jr., Baker, L. K., Krafft, G. A., and LaDu, M. J. (2002) J. Biol. Chem. 277, 32046-32053). Understanding the biological role of specific A beta conformations may define the link between A beta and Alzheimer's disease, re-focusing therapeutic approaches by identifying the pernicious species of A beta ultimately responsible for the cognitive dysfunction that defines the disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Biopolymers/chemistry , Peptide Fragments/chemistry , Blotting, Western , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Solubility
18.
J Biol Chem ; 277(35): 32046-53, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12058030

ABSTRACT

Genetic evidence predicts a causative role for amyloid-beta (A beta) in Alzheimer's disease. Recent debate has focused on whether fibrils (amyloid) or soluble oligomers of A beta are the active species that contribute to neurodegeneration and dementia. We developed two aggregation protocols for the consistent production of stable oligomeric or fibrillar preparations of A beta-(1-42). Here we report that oligomers inhibit neuronal viability 10-fold more than fibrils and approximately 40-fold more than unaggregated peptide, with oligomeric A beta-(1-42)-induced inhibition significant at 10 nm. Under A beta-(1-42) oligomer- and fibril-forming conditions, A beta-(1-40) remains predominantly as unassembled monomer and had significantly less effect on neuronal viability than preparations of A beta-(1-42). We applied the aggregation protocols developed for wild type A beta-(1-42) to A beta-(1-42) with the Dutch (E22Q) or Arctic (E22G) mutations. Oligomeric preparations of the mutations exhibited extensive protofibril and fibril formation, respectively, but were not consistently different from wild type A beta-(1-42) in terms of inhibition of neuronal viability. However, fibrillar preparations of the mutants appeared larger and induced significantly more inhibition of neuronal viability than wild type A beta-(1-42) fibril preparations. These data demonstrate that protocols developed to produce oligomeric and fibrillar A beta-(1-42) are useful in distinguishing the structural and functional differences between A beta-(1-42) and A beta-(1-40) and genetic mutations of A beta-(1-42).


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/physiology , Cell Survival/physiology , Neurons/cytology , Peptide Fragments/chemistry , Alzheimer Disease , Humans , Microscopy, Atomic Force , Neuroblastoma , Tumor Cells, Cultured
19.
Brain Res ; 924(2): 133-40, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11750898

ABSTRACT

The dementia in Alzheimer disease (AD) is usually attributed to widespread neuronal loss in conjunction with the pathologic hallmarks of intracellular neurofibrillary tangles and extracellular plaques containing amyloid (A beta) in fibrillar form. Recently it has been demonstrated that non-fibrillar assemblies of A beta possess electrophysiologic activity, with the corollary that they may produce dementia by disrupting neuronal signaling prior to cell death. We therefore examined the effects of soluble oligomers of A beta(1-42) on long-term potentiation (LTP) and long-term depression (LTD), two cellular models of memory, in the dentate gyrus of rat hippocampal slices. Compared with vehicle controls, slices pre-incubated 60 min in the presence of A beta-derived diffusible ligands (ADDLs) showed no differences in threshold intensity to evoke a synaptic response, slope of field excitatory post-synaptic potentials (EPSPs), or the input/output function. Tetanus-induced LTP and reversal of LTD were strongly inhibited in ADDLs-treated slices whereas LTD was unaffected. These data suggest that soluble non-fibrillar amyloid may contribute to the pathogenesis of AD both by impairing LTP/memory formation at the cellular level and by creating 'neuroplasticity imbalance' manifested by unopposed LTD in the setting of impaired capacity for neural repair via reversal of LTD or LTP.


Subject(s)
Amyloid beta-Peptides/pharmacology , Dentate Gyrus/physiology , Long-Term Potentiation/drug effects , Neural Inhibition/drug effects , Peptide Fragments/pharmacology , Animals , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Solubility , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...