Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters










Publication year range
2.
J Prev Alzheimers Dis ; 7(3): 146-151, 2020.
Article in English | MEDLINE | ID: mdl-32463066

ABSTRACT

The termination of many clinical trials of amyloid-targeting therapies for the treatment of Alzheimer's disease (AD) has had a major impact on the AD clinical research enterprise. However, positive signals in recent studies have reinvigorated support for the amyloid hypothesis and amyloid-targeting strategies. In December 2019, the EU-US Clinical Trials on Alzheimer's Disease (CTAD) Task Force met to share learnings from these studies in order to inform future trials and promote the development of effective AD treatments. Critical factors that have emerged in studies of anti-amyloid monoclonal antibody therapies include developing a better understanding of the specific amyloid species targeted by different antibodies, advancing our insight into the mechanism by which those antibodies may reduce pathology, implementing more comprehensive repertoires of biomarkers into trials, and identifying appropriate doses. Studies suggest that Amyloid-Related Imaging Abnormalities - effusion type (ARIA-E) are a manageable safety concern and that caution should be exercised before terminating studies based on interim analyses. The Task Force concluded that opportunities for developing effective treatments include developing new biomarkers, intervening in early stages of disease, and use of combination therapies.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/administration & dosage , Clinical Trials as Topic , Drug Development/standards , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers , Drug Development/trends , Humans , Research/trends
3.
Dev Biol ; 424(2): 138-146, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284905

ABSTRACT

It is widely accepted that amyloid precursor protein (APP) plays a central role in the pathogenesis of Alzheimer's disease. In addition, APP has been proposed to have functions in numerous biological processes including neuronal proliferation, differentiation, migration, axon guidance, and neurite outgrowth, as well as in synapse formation and function. However, germline knockout of APP yields relatively subtle phenotypes, and brain development appears grossly normal. This is thought to be due in part to functional compensation by APP family members and other type I transmembrane proteins. Here, we have generated a conditional mouse knockout for APP that is controlled temporally using CreER and tamoxifen administration. We show that total cortical expression of APP is reduced following tamoxifen administration during embryonic time points critical for cortical lamination, and that this results in displacement of Reelin-positive cells below the cortical plate with a concurrent elevation in Reelin protein levels. These results support a role for APP in cortical lamination and demonstrate the utility of a conditional knockout approach in which APP can be deleted with temporal control in vivo. This new tool should be useful for many different applications in the study of APP function across the mammalian life span.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/metabolism , Gene Deletion , Mosaicism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Biomarkers/metabolism , Gene Knockdown Techniques , Germ Cells/metabolism , Mice, Knockout , Reelin Protein
4.
Methods Enzymol ; 584: 295-308, 2017.
Article in English | MEDLINE | ID: mdl-28065268

ABSTRACT

Proteolysis within the membrane is catalyzed by a diverse family of proteases immersed within the hydrophobic environment of cellular membranes. These ubiquitous intramembrane-cleaving proteases (I-CLiPs) hydrolyze the transmembrane domains of a large variety of membrane-embedded proteins to facilitate signaling events essential to normal biological functions found in all forms of life. The importance of this unique class of enzyme is highlighted by its central involvement in a variety of human pathologies, including Alzheimer's disease (AD), Parkinson's disease, cancer, and the virulence of a number of viral, bacterial, and fungal pathogens. I-CLiPs therefore represent promising targets for the therapeutic treatment of numerous diseases. The key to understanding the normal biological function of I-CLiPs and capitalizing on their therapeutic potential is through a thorough understanding of the complex catalytic mechanisms that govern this unusual class of enzyme. This is an intrinsically difficult endeavor, given that these enzymes and their substrates reside within lipid membranes, making any in vitro assay technically challenging to design and execute. Here, we describe several in vitro enzymatic assays for the study of the AD-associated γ-secretase protease, which have aided the development of potent γ-secretase-targeting compounds as candidate therapeutics. These assays have also been applied in various forms for the study of other I-CLiPs, providing valuable mechanistic insights into some of the functional similarities and differences between several members of this fascinating family of proteases.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/chemistry , Enzyme Assays/methods , Proteolysis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/biosynthesis , Humans , Membrane Lipids/chemistry , Signal Transduction , Substrate Specificity
5.
Biochem Soc Trans ; 35(Pt 2): 416-20, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371289

ABSTRACT

Overwhelming evidence indicates that the Abeta (amyloid beta-peptide) plays a critical role in the pathogenesis of Alzheimer's disease. Abeta is derived from the APP (amyloid precursor protein) by the action of two aspartyl proteases (beta- and gamma-secretases) that are leading candidates for therapeutic intervention. APP is a member of a multigene family that includes APLP1 (amyloid precursor-like protein 1) and APLP2. Both APLPs are processed in a manner analogous to APP, with all three proteins subject to ectodomain shedding and subsequent cleavage by gamma-secretase. Careful study of the APP family of proteins has already revealed important insights about APP. Here, we will review how knowledge of the similarities and differences between APP and the APLPs may prove useful for the development of novel disease-modifying therapeutics.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/physiology , Brain/physiology , Receptors, Cell Surface/physiology , Animals , Brain/physiopathology , Humans , Protease Nexins , Reference Values
6.
Biochem Soc Trans ; 33(Pt 5): 1087-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16246051

ABSTRACT

Burgeoning evidence suggests that soluble oligomers of Abeta (amyloid beta-protein) are the earliest effectors of synaptic compromise in Alzheimer's disease. Whereas most other investigators have employed synthetic Abeta peptides, we have taken advantage of a beta-amyloid precursor protein-overexpressing cell line (referred to as 7PA2) that secretes sub-nanomolar levels of low-n oligomers of Abeta. These are composed of heterogeneous Abeta peptides that migrate on SDS/PAGE as dimers, trimers and tetramers. When injected into the lateral ventricle of rats in vivo, these soluble oligomers inhibit hippocampal long-term potentiation and alter the memory of a complex learned behaviour. Biochemical manipulation of 7PA2 medium including immunodepletion with Abeta-specific antibodies and fractionation by size-exclusion chromatography allowed us to unambiguously attribute these effects to low-n oligomers. Using this paradigm we have tested compounds directed at three prominent amyloid-based therapeutic targets: inhibition of the secretases responsible for Abeta production, inhibition of Abeta aggregation and immunization against Abeta. In each case, compounds capable of reducing oligomer production or antibodies that avidly bind Abeta oligomers also ameliorate the synaptotoxic effects of these natural, cell-derived oligomers.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Behavior , Humans , Neuronal Plasticity
7.
Biochem Soc Trans ; 30(4): 552-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12196135

ABSTRACT

Despite extensive genetic and animal modelling data that support a central role for the amyloid beta-protein (A beta) in the genesis of Alzheimer's disease, the specific form(s) of A beta which causes injury to neurons in vivo has not been identified. In the present study, we examine the importance of soluble, pre-fibrillar assemblies of A beta as mediators of neurotoxicity. Specifically, we review the role of cell-derived SDS-stable oligomers, their blocking of hippocampal long-term potentiation in vivo and the finding that this blocking can be prevented by prior treatment of oligomer-producing cells with gamma-secretase inhibitors.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Neurotoxins/chemistry , Peptide Fragments/toxicity , Alzheimer Disease/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Cell Death , Humans , Neurons/drug effects , Neurons/pathology , Neurotoxins/toxicity , Peptide Fragments/antagonists & inhibitors
8.
Neuroscience ; 109(1): 1-4, 2002.
Article in English | MEDLINE | ID: mdl-11784695

ABSTRACT

It is generally believed that only L-amino acids have a physiological role in species other than bacteria. Recently, the existence of some D-amino acids, particularly D-aspartate, in various organs of several higher animals has been reported. Here we demonstrate that naturally occurring free D-aspartate is localized subcellularly to the heterochromatin in the nucleoli (but not in either the dendrites or axonal terminals) of magnocellular neurosecretory neurons in the rat hypothalamus, and also of microglia and pericytes in the posterior pituitary. Our results imply that naturally occurring free D-aspartate might have a physiological role in nuclear function in mammals. The findings provide new insight for the biological function of D-stereoisomers of amino acids as well as the organization of the nucleus of at least some eukaryotic cells.


Subject(s)
Cell Nucleus/metabolism , D-Aspartic Acid/metabolism , Hypothalamo-Hypophyseal System/metabolism , Microglia/metabolism , Neurons/metabolism , Animals , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cell Nucleus/ultrastructure , Female , Heterochromatin/metabolism , Heterochromatin/ultrastructure , Immunohistochemistry , Microglia/ultrastructure , Microscopy, Electron , Neurons/ultrastructure , Neurosecretion/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/ultrastructure , Pericytes/metabolism , Pericytes/ultrastructure , Pituitary Gland, Posterior/metabolism , Pituitary Gland, Posterior/ultrastructure , Rats , Rats, Long-Evans , Stereoisomerism , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/ultrastructure
9.
Neuron ; 32(2): 177-80, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11683988

ABSTRACT

Elevated cerebral levels of amyloid beta-protein occur universally in Alzheimer's disease, yet only a few patients show evidence of increased Abeta production. Therefore, defects in proteases that degrade Abeta could underlie some or many cases of familial and sporadic AD. This previously neglected topic has begun receiving serious attention. Understanding how proteolysis regulates Abeta levels in the cerebral cortex has implications for both the pathogenesis and the treatment of this protean disorder.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/enzymology , Endopeptidases/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Animals , Endopeptidases/genetics , Humans , Insulysin/metabolism , Mutation , Neprilysin/metabolism
10.
J Biol Chem ; 276(43): 40288-92, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11544248

ABSTRACT

The beta-amyloid precursor protein (APP) is a ubiquitous receptor-like molecule without a known function. However, the recent recognition that APP and Notch undergo highly similar proteolytic processing has suggested a potential signaling function for APP. After ligand binding, Notch is cleaved by the ADAM-17 metalloprotease followed by an intramembrane cleavage mediated by gamma-secretase. The gamma-secretase cut releases the Notch intracellular domain (NICD), which enters the nucleus and modulates transcription. Because APP is processed similarly by ADAM-17 and gamma-secretase, we reasoned that the APP intracellular domain (AICD) has a role analogous to the NICD. We therefore generated a plasmid encoding the AICD sequence and studied the subcellular localization of the expressed protein (C60). Our results demonstrate that the cytoplasmic domain of APP is a highly labile fragment that is stabilized by forming complexes with Fe65 and can then enter the nucleus in neurons and non-neural cells. These findings strongly support the hypothesis that APP signals in the nucleus in a manner analogous to the function of Notch.


Subject(s)
Active Transport, Cell Nucleus , Amyloid beta-Protein Precursor/metabolism , Drosophila Proteins , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , ADAM Proteins , ADAM17 Protein , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , CREB-Binding Protein , Endopeptidases/metabolism , Half-Life , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Protein Binding , Protein Processing, Post-Translational , Receptors, Notch , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism
11.
Proc Natl Acad Sci U S A ; 98(20): 11039-41, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11572965

ABSTRACT

Elucidation of the proteolytic processing of the amyloid beta-protein precursor (APP) has revealed that one of the two proteases (gamma-secretase) that cleave APP to release amyloid beta-protein (Abeta) is likely to be presenilin. Presenilin also mediates the gamma-secretase-like cleavage of Notch receptors to enable signaling by their cytoplasmic domains. Therefore, APP and Notch may be the first identified substrates of a unique intramembranous aspartyl protease that has presenilin as its active-site component. In view of the evidence for a central role of cerebral build-up of Abeta in the pathogenesis of Alzheimer's disease, this disorder appears to have arisen in the human population as a late-life consequence of the conservation of a critical developmental pathway.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Brain/physiopathology , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , Brain/metabolism , Endopeptidases/metabolism , Humans , Presenilin-1 , Presenilin-2
12.
Proc Natl Acad Sci U S A ; 98(18): 10273-8, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517335

ABSTRACT

Alzheimer's disease is a dementia that involves progressive deposition of amyloid beta-protein (Abeta) in brain regions important for memory and cognition, followed by secondary inflammation that contributes to the neuropathologic process. Immunization with Abeta can reduce cerebral Abeta burden and consequent neuropathologic changes in the brains of mice transgenic for the beta-amyloid precursor protein (APP). We found that transgenic expression of human APP in B6SJL mice, under the prion promoter, results in immune hyporesponsiveness to human Abeta, in terms of both antibody and cellular immune responses. The decreased antibody responses were related not to B cell tolerance but rather to the inability of Abeta-specific T cells to provide help for antibody production. The immune hyporesponsiveness could be overcome if T cell help was provided by coupling an Abeta B cell epitope to BSA. Our results suggest that expression of APP in transgenic mice is associated with an Abeta-specific impaired adaptive immune response that may contribute to the neuropathology. Moreover, humans with life-long elevation of brain and peripheral Abeta (e.g., patients with presenilin mutations or Down syndrome) could have reduced immune responses to Abeta vaccination.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/blood , Animals , Antibody Formation , Gene Expression , Humans , Immune Tolerance , Immunity, Cellular , Immunization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Plaque, Amyloid/immunology
13.
Proc Natl Acad Sci U S A ; 98(16): 9110-5, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11481478

ABSTRACT

alpha-Synuclein (alphaS) is a 140-residue neuronal protein that forms insoluble cytoplasmic aggregates in Parkinson's disease (PD) and several other neurodegenerative disorders. Two missense mutations (A53T and A30P) are linked to rare forms of familial PD. The normal function of alphaS is unknown, and cultured cell systems that model its modification from soluble monomers to aggregated forms have not been reported. Through a systematic centrifugal fractionation of mesencephalic neuronal cell lines and transgenic mouse brains expressing wild-type or A53T human alphaS, we observed unusual, previously unrecognized species of alphaS that migrate well above the 17-kDa monomeric form in denaturing gels. Incubation at 65 degrees C of high-speed cytosols from cells or brains revealed a modified alphaS species migrating at approximately 36 kDa and an extensive higher molecular mass alphaS-reactive smear. Extraction of the cytosols with chloroform/methanol or with a resin (Lipidex 1000) that binds fatty acids resulted in a similar pattern of higher molecular mass alphaS forms. On the basis of this effect of delipidation, we reexamined the primary structure of alphaS and detected a motif at the N and C termini that is homologous to a fatty acid-binding protein signature. In accord, we found that purified human alphaS binds oleic acid, with an apparent K(d) of 12.5 microM. We also observed an enhanced association of A53T alphaS with microsomal membranes in both mesencephalic cells and transgenic mouse brains. We conclude that alphaS has biochemical properties and a structural motif that suggest it is a novel member of the fatty acid-binding protein family and may thus transport fatty acids between the aqueous and membrane phospholipid compartments of the neuronal cytoplasm.


Subject(s)
Carrier Proteins/chemistry , Lipid Metabolism , Neoplasm Proteins , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cytosol/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Lipids/chemistry , Mice , Mice, Transgenic , Molecular Sequence Data , Molecular Weight , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Oleic Acid/metabolism , Protein Binding , Sequence Homology, Amino Acid , Synucleins , alpha-Synuclein
14.
Science ; 293(5528): 263-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11431533

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive accumulation in selected neurons of protein inclusions containing alpha-synuclein and ubiquitin. Rare inherited forms of PD are caused by autosomal dominant mutations in alpha-synuclein or by autosomal recessive mutations in parkin, an E3 ubiquitin ligase. We hypothesized that these two gene products interact functionally, namely, that parkin ubiquitinates alpha-synuclein normally and that this process is altered in autosomal recessive PD. We have now identified a protein complex in normal human brain that includes parkin as the E3 ubiquitin ligase, UbcH7 as its associated E2 ubiquitin conjugating enzyme, and a new 22-kilodalton glycosylated form of alpha-synuclein (alphaSp22) as its substrate. In contrast to normal parkin, mutant parkin associated with autosomal recessive PD failed to bind alphaSp22. In an in vitro ubiquitination assay, alphaSp22 was modified by normal but not mutant parkin into polyubiquitinated, high molecular weight species. Accordingly, alphaSp22 accumulated in a non-ubiquitinated form in parkin-deficient PD brains. We conclude that alphaSp22 is a substrate for parkin's ubiquitin ligase activity in normal human brain and that loss of parkin function causes pathological alphaSp22 accumulation. These findings demonstrate a critical biochemical reaction between the two PD-linked gene products and suggest that this reaction underlies the accumulation of ubiquitinated alpha-synuclein in conventional PD.


Subject(s)
Brain/metabolism , Ligases/metabolism , Nerve Tissue Proteins/metabolism , Parkinson Disease/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitins/metabolism , Brain/enzymology , Brain Stem/enzymology , Brain Stem/metabolism , Cell Line , Detergents , Freezing , Glycosylation , Humans , Lewy Bodies/enzymology , Lewy Bodies/metabolism , Ligases/genetics , Mutation, Missense , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Substrate Specificity , Synucleins , alpha-Synuclein
15.
Biochem J ; 355(Pt 3): 869-77, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11311152

ABSTRACT

In a Flemish kindred, an Ala(692)-->Gly amino acid substitution in the amyloid beta-protein precursor (AbetaPP) causes a form of early-onset Alzheimer's disease (AD) which displays prominent amyloid angiopathy and unusually large senile plaque cores. The mechanistic basis of this Flemish form of AD is unknown. Previous in vitro studies of amyloid beta-protein (Abeta) production in HEK-293 cells transfected with cDNA encoding Flemish AbetaPP have shown that full-length [Abeta(1-40)] and truncated [Abeta(5-40) and Abeta(11-40)] forms of Abeta are produced. In an effort to determine how these peptides might contribute to the pathogenesis of the Flemish disease, comparative biophysical and neurotoxicity studies were performed on wild-type and Flemish Abeta(1-40), Abeta(5-40) and Abeta(11-40). The results revealed that the Flemish amino acid substitution increased the solubility of each form of peptide, decreased the rate of formation of thioflavin-T-positive assemblies, and increased the SDS-stability of peptide oligomers. Although the kinetics of peptide assembly were altered by the Ala(21)-->Gly substitution, all three Flemish variants formed fibrils, as did the wild-type peptides. Importantly, toxicity studies using cultured primary rat cortical cells showed that the Flemish assemblies were as potent a neurotoxin as were the wild-type assemblies. Our results are consistent with a pathogenetic process in which conformational changes in Abeta induced by the Ala(21)-->Gly substitution would facilitate peptide adherence to the vascular endothelium, creating nidi for amyloid growth. Increased peptide solubility and assembly stability would favour formation of larger deposits and inhibit their elimination. In addition, increased concentrations of neurotoxic assemblies would accelerate neuronal injury and death.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Alanine/genetics , Alzheimer Disease/metabolism , Amino Acid Substitution , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Glycine/genetics , Humans , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Rats , Sodium Dodecyl Sulfate/pharmacology , Solubility
16.
Physiol Rev ; 81(2): 741-66, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11274343

ABSTRACT

Rapid progress in deciphering the biological mechanism of Alzheimer's disease (AD) has arisen from the application of molecular and cell biology to this complex disorder of the limbic and association cortices. In turn, new insights into fundamental aspects of protein biology have resulted from research on the disease. This beneficial interplay between basic and applied cell biology is well illustrated by advances in understanding the genotype-to-phenotype relationships of familial Alzheimer's disease. All four genes definitively linked to inherited forms of the disease to date have been shown to increase the production and/or deposition of amyloid beta-protein in the brain. In particular, evidence that the presenilin proteins, mutations in which cause the most aggressive form of inherited AD, lead to altered intramembranous cleavage of the beta-amyloid precursor protein by the protease called gamma-secretase has spurred progress toward novel therapeutics. The finding that presenilin itself may be the long-sought gamma-secretase, coupled with the recent identification of beta-secretase, has provided discrete biochemical targets for drug screening and development. Alternate and novel strategies for inhibiting the early mechanism of the disease are also emerging. The progress reviewed here, coupled with better ability to diagnose the disease early, bode well for the successful development of therapeutic and preventative drugs for this major public health problem.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/physiology , Animals , Apolipoprotein E4 , Apolipoproteins E/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Molecular Sequence Data , Mutation, Missense , Presenilin-1 , Presenilin-2
17.
DNA Cell Biol ; 20(11): 705-11, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11788048

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disease for which there is currently no effective prevention or treatment. The prediction that the number of U.S. patients with AD will triple to approximately 14 million over the next 50 years underscores the urgent need to explore novel therapeutic strategies for AD. The beta-amyloid protein (Abeta) accumulation and accompanying inflammation appear to play key roles in initiating the neuronal degeneration that underlies the signs and symptoms of AD. Interventions geared toward reducing Abeta accumulation and inflammatory responses should delay or prevent the onset of the clinical disease. Recently, several research groups, including ours, have shown that vaccination with Abeta results in a significant lowering of the Abeta burden in the brains of APP transgenic mice and, in some studies, improvement in their cognitive deficits. Our study described a novel approach, namely mucosal (intranasal) Abeta vaccination. Precisely how Abeta vaccination chronically lowers Abeta levels and reduces Abeta-associated pathology remains unclear. Here, we provide an overview of these studies, with particular emphasis on our work with intranasal Abeta vaccination. Examples of other intranasal vaccines and mucosal adjuvants are presented. Taken together, these data have implications for the future development of an intranasal Abeta vaccine for humans.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/administration & dosage , Adjuvants, Immunologic/therapeutic use , Administration, Intranasal , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibody Formation , Brain/metabolism , Disease Models, Animal , Mice , Mice, Transgenic
18.
Science ; 290(5500): 2302-3, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11125142

ABSTRACT

Recent studies suggest that insulin-degrading enzyme (IDE) in neurons and microglia degrades Abeta, the principal component of beta-amyloid and one of the neuropathological hallmarks of Alzheimer's disease (AD). We performed parametric and nonparametric linkage analyses of seven genetic markers on chromosome 10q, six of which map near the IDE gene, in 435 multiplex AD families. These analyses revealed significant evidence of linkage for adjacent markers (D10S1671, D10S583, D10S1710, and D10S566), which was most pronounced in late-onset families. Furthermore, we found evidence for allele-specific association between the putative disease locus and marker D10S583, which has recently been located within 195 kilobases of the IDE gene.


Subject(s)
Alzheimer Disease/genetics , Chromosomes, Human, Pair 10/genetics , Genetic Linkage , Insulysin/genetics , Age of Onset , Aged , Aged, 80 and over , Alleles , Apolipoproteins E/genetics , Chromosome Mapping , Genetic Markers , Humans , Linkage Disequilibrium , Middle Aged
19.
Nat Struct Biol ; 7(12): 1095-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101888

ABSTRACT

We report here that a monomeric de novo designed alpha-helix-turn-alpha-helix peptide, alpha t alpha, when incubated at 37 degrees C in an aqueous buffer at neutral pH, forms nonbranching, protease resistant fibrils that are 6-10 nm in diameter. These fibrils are rich in beta-sheet and bind the amyloidophilic dye Congo red. alpha t alpha fibrils thus display the morphologic, structural, and tinctorial properties of authentic amyloid fibrils. Surprisingly, unlike fibrils formed by peptides such as the amyloid beta-protein or the islet amyloid polypeptide, alpha t alpha fibrils were not toxic to cultured rat primary cortical neurons or PC12 cells. These results suggest that the potential to form fibrils under physiologic conditions is not limited to those proteins associated with amyloidoses and that fibril formation alone is not predictive of cytotoxic activity.


Subject(s)
Helix-Turn-Helix Motifs , Peptides/chemistry , Peptides/metabolism , Plaque, Amyloid/chemistry , Plaque, Amyloid/metabolism , Amino Acid Sequence , Animals , Biopolymers/chemistry , Biopolymers/metabolism , Cell Death/drug effects , Cells, Cultured , Circular Dichroism , Congo Red , Endopeptidase K/metabolism , Formazans , Hydrogen-Ion Concentration , Microscopy, Electron , Models, Biological , Models, Molecular , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , PC12 Cells , Peptides/chemical synthesis , Peptides/toxicity , Plaque, Amyloid/ultrastructure , Protein Conformation , Rats , Static Electricity , Tetrazolium Salts
20.
Neurobiol Dis ; 7(6 Pt B): 673-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11114265

ABSTRACT

To investigate the mechanism of regulation of Ass production by familial Alzheimer's disease (FAD)-linked presenilin 1 (PS1), we used a cell-free system that allows de novo Ass generation to examine whether PS1 participates directly in the gamma-secretase reaction. Optimal Ass generation in vitro was achieved at mildly acidic pH and could be inhibited by the aspartyl protease inhibitor pepstatin A, consistent with the suggestion that gamma-secretase is an aspartyl protease. Dominant negative mutations of the critical transmembrane aspartates in PS1 or full deletion of PS1 did not alter the maturation of APP in the secretory pathway. Instead, PS1 had a direct effect on the inhibition of Ass production by a designed peptidomimetic inhibitor: the inhibition was significantly less effective in cells expressing FAD-causing mutations in either APP or PS1 than in cells expressing the wild-type proteins. Taken together, these findings suggest that PS1 participates physically in a complex with APP during the gamma-secretase cleavage event.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Binding Sites/drug effects , Binding Sites/genetics , CHO Cells , Cell Fractionation , Cell-Free System/metabolism , Cricetinae , Endopeptidases/drug effects , Genes, Dominant , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Macromolecular Substances , Membrane Proteins/genetics , Microsomes/metabolism , Mutation, Missense , Pepstatins/pharmacology , Presenilin-1 , Protein Binding/drug effects , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...