Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
Add more filters

Publication year range
1.
Cell ; 158(2): 247-249, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036627

ABSTRACT

The intramembrane protease γ-secretase is a key player in signaling and Alzheimer's disease, but its structural features have remained obscure. A structure reported recently reveals a horseshoe-shaped arrangement of 19 transmembrane helices and an extracellular domain positioned for substrate recognition. This advance bodes well for a finer resolution before long.


Subject(s)
Amyloid Precursor Protein Secretases/chemistry , Models, Molecular , Humans
2.
Mol Cell ; 73(5): 1001-1014.e8, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30527540

ABSTRACT

In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.


Subject(s)
Antiparkinson Agents/pharmacology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Lipid Metabolism/drug effects , Metabolomics/methods , Neurons/drug effects , Parkinson Disease/drug therapy , Stearoyl-CoA Desaturase/antagonists & inhibitors , alpha-Synuclein/toxicity , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Diglycerides/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Lipid Droplets/drug effects , Lipid Droplets/enzymology , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Nerve Degeneration , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neurons/enzymology , Neurons/pathology , Oleic Acid/metabolism , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism , alpha-Synuclein/genetics
3.
Cell ; 147(4): 893-906, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22078885

ABSTRACT

Cells keep their energy balance and avoid oxidative stress by regulating mitochondrial movement, distribution, and clearance. We report here that two Parkinson's disease proteins, the Ser/Thr kinase PINK1 and ubiquitin ligase Parkin, participate in this regulation by arresting mitochondrial movement. PINK1 phosphorylates Miro, a component of the primary motor/adaptor complex that anchors kinesin to the mitochondrial surface. The phosphorylation of Miro activates proteasomal degradation of Miro in a Parkin-dependent manner. Removal of Miro from the mitochondrion also detaches kinesin from its surface. By preventing mitochondrial movement, the PINK1/Parkin pathway may quarantine damaged mitochondria prior to their clearance. PINK1 has been shown to act upstream of Parkin, but the mechanism corresponding to this relationship has not been known. We propose that PINK1 phosphorylation of substrates triggers the subsequent action of Parkin and the proteasome.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , Mice , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Molecular Sequence Data , Parkinson Disease/metabolism , Phosphorylation , Rats , rho GTP-Binding Proteins/chemistry
4.
PLoS Biol ; 20(7): e3001694, 2022 07.
Article in English | MEDLINE | ID: mdl-35862308

ABSTRACT

Strong genetic evidence supports an imbalance between production and clearance of amyloid ß-protein (Aß) in people with Alzheimer disease (AD). Microglia that are potentially involved in alternative mechanisms are actually integral to the amyloid cascade. Fluid biomarkers and brain imaging place accumulation of Aß at the beginning of molecular and clinical changes in the disease. So why have clinical trials of anti-amyloid therapies not provided clear-cut benefits to patients with AD? Can anti-amyloid therapies robustly decrease Aß in the human brain, and if so, could this lowering be too little, too late? These central questions in research on AD are being urgently addressed.


Subject(s)
Alzheimer Disease , Amyloidosis , Cognitive Dysfunction , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Amyloid , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cognitive Dysfunction/therapy , Humans
5.
EMBO Rep ; 24(12): e57145, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37870370

ABSTRACT

α-Synuclein phosphorylation at serine-129 (pS129) is a widely used surrogate marker of pathology in Parkinson's disease and other synucleinopathies. However, we recently demonstrated that phosphorylation of S129 is also a physiological activator of synaptic transmission. In a feed-forward fashion, neuronal activity triggers reversible pS129. Here, we show that Parkinson's disease-linked missense mutations in SNCA impact activity-dependent pS129. Under basal conditions, cytosol-enriched A30P, H50Q, and G51D mutant forms of α-synuclein exhibit reduced pS129 levels in rat primary cortical neurons. A53T pS129 levels are similar to wild-type, and E46K pS129 levels are higher. A30P and E46K mutants show impaired reversibility of pS129 after stimulation. For the engineered profoundly membrane-associated α-synuclein mutant "3K" (E35K + E46K + E61K), de-phosphorylation was virtually absent after blocking stimulation, implying that reversible pS129 is severely compromised. Importantly, pS129 excess resulting from proteasome inhibition is also associated with reduced reversibility by neuronal inhibition, kinase inhibition, or phosphatase activation. Our findings suggest that perturbed pS129 dynamics are probably a shared characteristic of pathology-associated α-synuclein, with possible implications for synucleinopathy treatment and diagnosis.


Subject(s)
Parkinson Disease , Synucleinopathies , Animals , Rats , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Serine/metabolism , Phosphorylation
6.
Cell ; 142(2): 194-6, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20655461

ABSTRACT

A dual goal for treating Alzheimer's disease (AD) is to decrease deposition of neurotoxic amyloid beta-peptide in the brain and to boost repair of damaged neurons. Donmez et al. (2010) now show that SIRT1 may mediate both processes by deacetylating the transcription factor retinoic acid receptor beta, a potential new therapeutic target for AD.

7.
Cell Mol Life Sci ; 81(1): 362, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162859

ABSTRACT

Disease-modifying strategies for Parkinson disease (PD), the most common synucleinopathy, represent a critical unmet medical need. Accumulation of the neuronal protein alpha-synuclein (αS) and abnormal lipid metabolism have each been implicated in PD pathogenesis. Here, we elucidate how retinoid-X-receptor (RXR) nuclear receptor signaling impacts these two aspects of PD pathogenesis. We find that activated RXR differentially regulates fatty acid desaturases, significantly reducing the transcript levels of the largely brain-specific desaturase SCD5 in human cultured neural cells and PD patient-derived neurons. This was associated with reduced perilipin-2 protein levels in patient neurons, reversal of αS-induced increases in lipid droplet (LD) size, and a reduction of triglyceride levels in human cultured cells. With regard to αS proteostasis, our study reveals that RXR agonism stimulates lysosomal clearance of αS. Our data support the involvement of Polo-like kinase 2 activity and αS S129 phosphorylation in mediating this benefit. The lowering of cellular αS levels was associated with reduced cytotoxicity. Compared to RXR activation, the RXR antagonist HX531 had the opposite effects on LD size, SCD, αS turnover, and cytotoxicity, all supporting pathway specificity. Together, our findings show that RXR-activating ligands can modulate fatty acid metabolism and αS turnover to confer benefit in cellular models of PD, including patient neurons. We offer a new paradigm to investigate nuclear receptor ligands as a promising strategy for PD and related synucleinopathies.


Subject(s)
Lipid Metabolism , Lysosomes , Neurons , Retinoid X Receptors , Signal Transduction , alpha-Synuclein , alpha-Synuclein/metabolism , Humans , Lysosomes/metabolism , Neurons/metabolism , Neurons/pathology , Retinoid X Receptors/metabolism , Retinoid X Receptors/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Synucleinopathies/metabolism , Synucleinopathies/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cells, Cultured , Perilipin-2/metabolism , Perilipin-2/genetics , Phosphorylation
8.
Am J Pathol ; 193(5): 520-531, 2023 05.
Article in English | MEDLINE | ID: mdl-36773784

ABSTRACT

Abnormal aggregation of α-synuclein (αS) is thought to initiate neuronal dysfunction and death in Parkinson disease (PD). In addition to higher-molecular-weight, oligomeric, and polymeric forms of αS associated with neurotoxicity and disease, recent findings indicate the occurrence of physiological tetrameric assemblies in healthy neurons in culture and in brain. Herein, the PD-associated neurotoxin paraquat reduced physiological tetramers and led to calpain-truncated monomers and an approximately 70-kDa apparent oligomer different in size from physiological αS multimers. These truncated and oligomeric forms could also be generated by calpain cleavage of pure, recombinant human αS in vitro. Moreover, they were detected in the brains of tetramer-abrogating, E46K-amplified (3K) mice that model PD. These results indicate that paraquat triggers membrane damage and aberrant calpain activity that can induce a pathologic shift of tetramers toward an excess of full-length and truncated monomers, the accumulation of αS oligomers, and insoluble cytoplasmic αS puncta. The findings suggest that an environmental precipitant of PD can alter αS tetramer/monomer equilibrium, as already shown for several genetically caused forms of PD.


Subject(s)
Parkinson Disease , alpha-Synuclein , Mice , Humans , Animals , alpha-Synuclein/toxicity , Calpain , Paraquat/toxicity
9.
Mol Psychiatry ; 28(11): 4877-4888, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37365243

ABSTRACT

We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of ß2-adrenergic receptor (ß2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid ß-protein (oAß). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aß-species. We found that pharmacological activation of ß2-AR, but not ß1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAß-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in ß2-AR knockout mice, suggesting that activating ß2-AR prevents oAß-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of ß-ARs each decreased HDAC2, whereas Aß oligomers increased HDAC2 levels in the hippocampus. Further, oAß-induced inflammatory effects and neurite degeneration were prevented by either ß2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of ß2-AR is a novel potential therapeutic strategy to mitigate oAß-mediated features of AD.


Subject(s)
Amyloid beta-Peptides , Hippocampus , Mice , Animals , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Signal Transduction , Epigenesis, Genetic , Mice, Knockout
10.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34326260

ABSTRACT

Loss-of-function mutations in acid beta-glucosidase 1 (GBA1) are among the strongest genetic risk factors for Lewy body disorders such as Parkinson's disease (PD) and Lewy body dementia (DLB). Altered lipid metabolism in PD patient-derived neurons, carrying either GBA1 or PD αS mutations, can shift the physiological α-synuclein (αS) tetramer-monomer (T:M) equilibrium toward aggregation-prone monomers. A resultant increase in pSer129+ αS monomers provides a likely building block for αS aggregates. 3K αS mice, representing a neuropathological amplification of the E46K PD-causing mutation, have decreased αS T:M ratios and vesicle-rich αS+ aggregates in neurons, accompanied by a striking PD-like motor syndrome. We asked whether enhancing glucocerebrosidase (GCase) expression could benefit αS dyshomeostasis by delivering an adeno-associated virus (AAV)-human wild-type (wt) GBA1 vector into the brains of 3K neonates. Intracerebroventricular AAV-wtGBA1 at postnatal day 1 resulted in prominent forebrain neuronal GCase expression, sustained through 6 mo. GBA1 attenuated behavioral deficits both in working memory and fine motor performance tasks. Furthermore, wtGBA1 increased αS solubility and the T:M ratio in both 3K-GBA mice and control littermates and reduced pS129+ and lipid-rich aggregates in 3K-GBA. We observed GCase distribution in more finely dispersed lysosomes, in which there was increased GCase activity, lysosomal cathepsin D and B maturation, decreased perilipin-stabilized lipid droplets, and a normalized TFEB translocation to the nucleus, all indicative of improved lysosomal function and lipid turnover. Therefore, a prolonged increase of the αS T:M ratio by elevating GCase activity reduced the lipid- and vesicle-rich aggregates and ameliorated PD-like phenotypes in mice, further supporting lipid modulating therapies in PD.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glucosylceramidase/metabolism , alpha-Synuclein/metabolism , Animals , Animals, Newborn , Glucosylceramidase/genetics , Lipid Metabolism , Lipids/chemistry , Maze Learning , Mice , Motor Activity , Recombinant Proteins , alpha-Synuclein/chemistry
11.
Alzheimers Dement ; 20(3): 1573-1585, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38041855

ABSTRACT

INTRODUCTION: A wide array of post-translational modifications of the tau protein occurs in Alzheimer's disease (AD) and they are critical to pathogenesis and biomarker development. Several promising tau markers, pT181, pT217, and pT231, rely on increased phosphorylation within a common molecular motif threonine-proline-proline (TPP). METHODS: We validated new and existing antibodies against pT217, pT231, pT175, and pT181, then combined immunohistochemistry (IHC) and immunoassays (ELISA) to broadly examine the phosphorylation of the tau TPP motif in AD brains. RESULTS: The tau burden, as examined by IHC and ELISA, correlates to Braak stages across all TPP sites. Moreover, we observed regional variability across four TPP motif phosphorylation sites in multiple brains of sporadic AD patients. DISCUSSION: We conclude that there is an elevation of TPP tau phosphorylation in AD brains as disease advances. The regional variability of pTPP tau suggests that examining different phosphorylation sites is essential for a comprehensive assessment of tau pathology.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/pathology , tau Proteins/metabolism , Phosphorylation , Threonine/metabolism , Brain/pathology , Proline/metabolism
12.
J Neurosci ; 42(10): 2116-2130, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35086904

ABSTRACT

α-Synuclein (αS) plays a key role in Parkinson's disease. Although Parkinson's disease is typically "sporadic," inherited αS missense mutations provide crucial insights into molecular mechanisms. Here, we examine two clinical mutants, E46K and G51D, which are both in the conserved N-terminus that mediates transient αS-membrane interactions. However, E46K increases and G51D decreases αS-membrane interactions. Previously, we amplified E46K via the 11-residue repeat motifs, creating "3K" (E35K+E46K+E61K). Here, we engineered these motifs to amplify G51D (V40D+G51D+V66D = "3D") and systematically compared E46K/3K versus G51D/3D. We found that G51D increased cytosolic αS in neural cells and 3D aggravates this. G51D, and 3D even more, reduced αS multimer-to-monomer (αS60:αS14) ratio. Both amplified variants caused cellular stress in rat primary neurons and reduced growth in human neuroblastoma cells. Importantly, both 3K- and 3D-induced stress was ameliorated by pharmacologically inhibiting stearoyl-CoA desaturase or by conditioning the cells in palmitic (16:0) or myristic (14:0) acid. SCD inhibition lowered lipid-droplet accumulation in both 3D- and 3K-expressing cells and benefitted G51D by normalizing multimer:monomer ratio, as reported previously for E46K. Our findings suggest that, despite divergent cytosol/membrane partitioning, both G51D and E46K neurotoxicity can be prevented by decreasing fatty-acid unsaturation as a common therapeutic approach.SIGNIFICANCE STATEMENT α-Synuclein (αS) dyshomeostasis is linked to Parkinson's disease. Here we focus on two contrasting familial-Parkinson's disease αS mutants, E46K and G51D, that alter αS membrane association in opposite directions (E46K increases, G51D decreases it). Taking advantage of αS repeat structure, here we designed αS "3D," an amplified G51D variant (V40D+G51D+V66D). αS 3D further enhanced G51D's cytosolic enrichment. Systematic comparison of G51D/3D with membrane-enriched E46K/its amplified variant 3K revealed that both can elicit stress in human neural cells and primary rodent neurons. This toxicity can be ameliorated by inhibiting stearoyl-CoA desaturase or by saturated fatty acid conditioning. Thus, despite divergent membrane binding, both G51D and E46K αS dyshomeostasis are mitigated by altering fatty acid saturation as a shared target.


Subject(s)
Fatty Acids , Parkinson Disease , alpha-Synuclein , Animals , Cytosol/metabolism , Fatty Acids/metabolism , Homeostasis , Parkinson Disease/metabolism , Rats , Stearoyl-CoA Desaturase/metabolism , alpha-Synuclein/metabolism
13.
Hum Mol Genet ; 30(23): 2332-2346, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34254125

ABSTRACT

α-Synuclein (αS) has been well-documented to play a role in human synucleinopathies such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB). First, the lesions found in PD/DLB brains-Lewy bodies and Lewy neurites-are rich in aggregated αS. Second, genetic evidence links missense mutations and increased αS expression to familial forms of PD/DLB. Third, toxicity and cellular stress can be caused by αS under certain experimental conditions. In contrast, the homologs ß-synuclein (ßS) and γ-synuclein (γS) are not typically found in Lewy bodies/neurites, have not been clearly linked to brain diseases and have been largely non-toxic in experimental settings. In αS, the so-called non-amyloid-ß component of plaques (NAC) domain, constituting amino acids 61-95, has been identified to be critical for aggregation in vitro. This domain is partially absent in ßS and only incompletely conserved in γS, which could explain why both homologs do not cause disease. However, αS in vitro aggregation and cellular toxicity have not been firmly linked experimentally, and it has been proposed that excess αS membrane binding is sufficient to induce neurotoxicity. Indeed, recent characterizations of Lewy bodies have highlighted the accumulation of lipids and membranous organelles, raising the possibility that ßS and γS could also become neurotoxic if they were more prone to membrane/lipid binding. Here, we increased ßS and γS membrane affinity by strategic point mutations and demonstrate that these proteins behave like membrane-associated monomers, are cytotoxic and form round cytoplasmic inclusions that can be prevented by inhibiting stearoyl-CoA desaturase.


Subject(s)
Cell Membrane/metabolism , Inclusion Bodies/metabolism , alpha-Synuclein/metabolism , beta-Synuclein/metabolism , gamma-Synuclein/metabolism , Amino Acid Sequence , Conserved Sequence , Humans , Mutagenesis , Protein Aggregation, Pathological , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Solubility , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , beta-Synuclein/chemistry , beta-Synuclein/genetics , gamma-Synuclein/chemistry , gamma-Synuclein/genetics
14.
Brain ; 145(7): 2528-2540, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35084489

ABSTRACT

Aqueously soluble oligomers of amyloid-ß peptide may be the principal neurotoxic forms of amyloid-ß in Alzheimer's disease, initiating downstream events that include tau hyperphosphorylation, neuritic/synaptic injury, microgliosis and neuron loss. Synthetic oligomeric amyloid-ß has been studied extensively, but little is known about the biochemistry of natural oligomeric amyloid-ß in human brain, even though it is more potent than simple synthetic peptides and comprises truncated and modified amyloid-ß monomers. We hypothesized that monoclonal antibodies specific to neurotoxic oligomeric amyloid-ß could be used to isolate it for further study. Here we report a unique human monoclonal antibody (B24) raised against synthetic oligomeric amyloid-ß that potently prevents Alzheimer's disease brain oligomeric amyloid-ß-induced impairment of hippocampal long-term potentiation. B24 binds natural and synthetic oligomeric amyloid-ß and a subset of amyloid plaques, but only in the presence of Ca2+. The amyloid-ß N terminus is required for B24 binding. Hydroxyapatite chromatography revealed that natural oligomeric amyloid-ß is highly avid for Ca2+. We took advantage of the reversible Ca2+-dependence of B24 binding to perform non-denaturing immunoaffinity isolation of oligomeric amyloid-ß from Alzheimer's disease brain-soluble extracts. Unexpectedly, the immunopurified material contained amyloid fibrils visualized by electron microscopy and amenable to further structural characterization. B24-purified human oligomeric amyloid-ß inhibited mouse hippocampal long-term potentiation. These findings identify a calcium-dependent method for purifying bioactive brain oligomeric amyloid-ß, at least some of which appears fibrillar.


Subject(s)
Alzheimer Disease , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies/metabolism , Brain/metabolism , Calcium/metabolism , Humans , Mice , Plaque, Amyloid/metabolism
16.
Alzheimers Dement ; 19(1): 79-96, 2023 01.
Article in English | MEDLINE | ID: mdl-35278341

ABSTRACT

INTRODUCTION: Identifying CSF-based biomarkers for the ß-amyloidosis that initiates Alzheimer's disease (AD) could provide inexpensive and dynamic tests to distinguish AD from normal aging and predict future cognitive decline. METHODS: We developed immunoassays specifically detecting all C-terminal variants of secreted amyloid ß-protein and identified a novel biomarker, the Aß 37/42 ratio, that outperforms the canonical Aß42/40 ratio as a means to evaluate the γ-secretase activity and brain Aß accumulation. RESULTS: We show that Aß 37/42 can distinguish physiological and pathological status in (1) presenilin-1 mutant vs wild-type cultured cells, (2) AD vs control brain tissue, and (3) AD versus cognitively normal (CN) subjects in CSF, where 37/42 (AUC 0.9622) outperformed 42/40 (AUC 0.8651) in distinguishing CN from AD. DISCUSSION: We conclude that the Aß 37/42 ratio sensitively detects presenilin/γ-secretase dysfunction and better distinguishes CN from AD than Aß42/40 in CSF. Measuring this novel ratio alongside promising phospho-tau analytes may provide highly discriminatory fluid biomarkers for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides , Amyloid Precursor Protein Secretases , tau Proteins , Peptide Fragments , Cognitive Dysfunction/diagnosis , Biomarkers
17.
Alzheimers Dement ; 19(12): 5755-5764, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37438872

ABSTRACT

INTRODUCTION: People with Down syndrome (DS) often develop Alzheimer's disease (AD). Here, we asked whether ultrasensitive plasma immunoassays for a tau N-terminal fragment (NT1-tau) and Aß isoforms predict cognitive impairment. METHODS: Plasma NT1-tau, Aß37 , Aß40 , and Aß42 levels were measured in a longitudinal discovery cohort (N = 85 participants, 220 samples) and a cross-sectional validation cohort (N = 239). We developed linear models and predicted values in the validation cohort. RESULTS: Discovery cohort linear mixed models for NT1-tau, Aß42 , and Aß37:42 were significant for age; there was no main effect of time. In cross-sectional models, NT1-tau increased and Aß42 decreased with age. NT1-tau predicted cognitive and functional scores. The discovery cohort linear model for NT1-tau predicted levels in the validation cohort. DISCUSSION: NT1-tau correlates with age and worse cognition in DS. Further validation of NT1-tau and other plasma biomarkers of AD neuropathology in DS cohorts is important for clinical utility.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Down Syndrome , Humans , tau Proteins , Cross-Sectional Studies , Cognition , Biomarkers , Amyloid beta-Peptides , Peptide Fragments
18.
J Biol Chem ; 296: 100393, 2021.
Article in English | MEDLINE | ID: mdl-33571524

ABSTRACT

γ-Secretase is responsible for the proteolysis of amyloid precursor protein (APP) into amyloid-beta (Aß) peptides, which are centrally implicated in the pathogenesis of Alzheimer's disease (AD). The biochemical mechanism of how processing by γ-secretase is regulated, especially as regards the interaction between enzyme and substrate, remains largely unknown. Here, mutagenesis reveals that the hydrophilic loop-1 (HL-1) of presenilin-1 (PS1) is critical for both γ-secretase step-wise cleavages (processivity) and its allosteric modulation by heterocyclic γ-modulatory compounds. Systematic mutagenesis of HL-1, including all of its familial AD mutations and additional engineered variants, and quantification of the resultant Aß products show that HL-1 is necessary for proper sequential γ-secretase processivity. We identify Y106, L113, and Y115 in HL-1 as key targets for heterocyclic γ-secretase modulators (GSMs) to stimulate processing of pathogenic Aß peptides. Further, we confirm that the GxxxG domain in the APP transmembrane region functions as a critical substrate motif for γ-secretase processivity: a G29A substitution in APP-C99 mimics the beneficial effects of GSMs. Together, these findings provide a molecular basis for the structural regulation of γ-processivity by enzyme and substrate, facilitating the rational design of new GSMs that lower AD-initiating amyloidogenic Aß peptides.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Presenilin-1/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Cells, Cultured , Genetic Predisposition to Disease , Humans , Mutation , Presenilin-1/chemistry , Presenilin-1/genetics , Protein Domains , Proteolysis , Substrate Specificity
19.
Neurobiol Dis ; 172: 105816, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35820646

ABSTRACT

Early cognitive impairment in Alzheimer's disease may result in part from synaptic dysfunction caused by the accumulation oligomeric assemblies of amyloid ß-protein (Aß). Changes in hippocampal function seem critical for cognitive impairment in early Alzheimer's disease (AD). Diffusible oligomers of Aß (oAß) have been shown to block canonical long-term potentiation (LTP) in the CA1 area of hippocampus, but whether there is also a direct effect of oAß on synaptic transmission and plasticity at synapses between mossy fibers (axons) from the dentate gyrus granule cells and CA3 pyramidal neurons (mf-CA3 synapses) is unknown. Studies in APP transgenic mice have suggested an age-dependent impairment of mossy fiber LTP. Here we report that although endogenous AD brain-derived soluble oAß had no effect on mossy-fiber basal transmission, it strongly impaired paired-pulse facilitation in the mossy fiber pathway and presynaptic mossy fiber LTP (mf-LTP). Selective activation of both ß1 and ß2 adrenergic receptors and their downstream cAMP/PKA signaling pathway prevented oAß-mediated inhibition of mf-LTP. Unexpectedly, activation of the cGMP/PKG signaling pathway also prevented oAß-impaired mf-LTP. Our results reveal certain specific pharmacological targets to ameliorate human oAß-mediated impairment at the mf-CA3 synapse.


Subject(s)
Alzheimer Disease , Long-Term Potentiation , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Mice , Mossy Fibers, Hippocampal/physiology , Synapses/metabolism , Synaptic Transmission/physiology
20.
Biochem Biophys Res Commun ; 611: 85-90, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35483223

ABSTRACT

Evidence suggests that plasma levels of tau protein phosphorylated at specific residues such as p-T181, p-T217, and p-T231 can be used as biomarkers for Alzheimer's disease (AD) diagnosis and prognosis. Accurate tools to calibrate immunoassays (calibrators) to precisely detect phosphorylated residues on tau protein will provide important gains in reliability and specificity. This study sought to establish a method to generate those accurate calibrators. We generated a semi-synthetic (chimeric) p-Tau181 calibrator by coupling a recombinant tau fragment (residues 1-174) with a synthetic peptide containing a single phosphorylated residue (p-T181) via thioester bond formation. The generation of a semi-synthetic protein containing both the N-terminal region of tau and the pT181 epitope was demonstrated by mobility shift assays using CBB staining and immunoblotting with N-terminal and pT181-specific antibodies. p-Tau 181 assays performed with the novel calibrator on multiple platforms revealed LLoQs as low as 0.14 pg/ml. Our facile and inexpensive method generates a semi-synthetic tau pT181 calibrator suitable for different immunoassay platforms. The same method can easily be adapted to other AD-relevant phospho-epitopes such as pT217 and pT231.


Subject(s)
Alzheimer Disease , tau Proteins , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Biomarkers/metabolism , Calibration , Humans , Immunoassay , Phosphorylation , Reproducibility of Results , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL