Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Alzheimers Res Ther ; 14(1): 104, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35897046

ABSTRACT

BACKGROUND: The S209F variant of Abelson Interactor Protein 3 (ABI3) increases risk for Alzheimer's disease (AD), but little is known about its function in relation to AD pathogenesis. METHODS: Here, we use a mouse model that is deficient in Abi3 locus to study how the loss of function of Abi3 impacts two cardinal neuropathological hallmarks of AD-amyloid ß plaques and tau pathology. Our study employs extensive neuropathological and transcriptomic characterization using transgenic mouse models and adeno-associated virus-mediated gene targeting strategies. RESULTS: Analysis of bulk RNAseq data confirmed age-progressive increase in Abi3 levels in rodent models of AD-type amyloidosis and upregulation in AD patients relative to healthy controls. Using RNAscope in situ hybridization, we localized the cellular distribution of Abi3 in mouse and human brains, finding that Abi3 is expressed in both microglial and non-microglial cells. Next, we evaluated Abi3-/- mice and document that both Abi3 and its overlapping gene, Gngt2, are disrupted in these mice. Using multiple transcriptomic datasets, we show that expression of Abi3 and Gngt2 are tightly correlated in rodent models of AD and human brains, suggesting a tight co-expression relationship. RNAseq of the Abi3-Gngt2-/- mice revealed upregulation of Trem2, Plcg2, and Tyrobp, concomitant with induction of an AD-associated neurodegenerative signature, even in the absence of AD-typical neuropathology. In APP mice, loss of Abi3-Gngt2 resulted in a gene dose- and age-dependent reduction in Aß deposition. Additionally, in Abi3-Gngt2-/- mice, expression of a pro-aggregant form of human tau exacerbated tauopathy and astrocytosis. Further, using in vitro culture assays, we show that the AD-associated S209F mutation alters the extent of ABI3 phosphorylation. CONCLUSIONS: These data provide an important experimental framework for understanding the role of Abi3-Gngt2 function and early inflammatory gliosis in AD. Our studies also demonstrate that inflammatory gliosis could have opposing effects on amyloid and tau pathology, highlighting the unpredictability of targeting immune pathways in AD.


Subject(s)
Adaptor Proteins, Signal Transducing , Alzheimer Disease , Amyloidosis , GTP-Binding Protein gamma Subunits , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Brain/metabolism , Disease Models, Animal , Gliosis/metabolism , GTP-Binding Protein gamma Subunits/genetics , Membrane Glycoproteins/metabolism , Mice, Transgenic , Plaque, Amyloid/pathology , Receptors, Immunologic/metabolism , tau Proteins/genetics , tau Proteins/metabolism
2.
Acta Neuropathol ; 141(3): 359-381, 2021 03.
Article in English | MEDLINE | ID: mdl-33496840

ABSTRACT

Accumulation of the tau protein in fibrillar intracellular aggregates is a defining feature of multiple neurodegenerative diseases collectively referred to as tauopathies. Despite intensive study of tau, there is limited information on the formation and clearance dynamics of tau inclusions. Using rAAV vectors to mediate expression of Dendra2-tagged human wild-type, P301L and pro-aggregant P301L/S320F tau proteins, with and without the addition of exogenous tau fibrillar seeds, we evaluated tau inclusion dynamics in organotypic brain slice culture (BSC) models using long-term optical pulse labeling methodology. Our studies reveal that tau inclusions typically form in 12-96 h in tauopathy BSC models. Unexpectedly, we demonstrate appreciable turnover of tau within inclusions with an average half-life of ~ 1 week when inclusions are newly formed. When BSCs with inclusions are aged in culture for extended periods, tau inclusions continue to turnover, but their half-lives increase to ~ 2 weeks and ~ 3 weeks after 1 and 2 months in culture, respectively. Individual tau inclusions can be long-lived structures that can persist for months in these BSC models and for even longer in the human brain. However, our data indicate that tau inclusions, are not 'tombstones', but dynamic structures with appreciable turnover. Understanding the cellular processes mediating this inclusion turnover may lead to new therapeutic strategies that could reverse pathological tau inclusion formation.


Subject(s)
Brain/metabolism , Brain/pathology , Neurons/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice , Neurons/pathology , Organ Culture Techniques , Tauopathies/pathology
3.
Alzheimers Res Ther ; 12(1): 61, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32430033

ABSTRACT

BACKGROUND: γ-Secretase is a multiprotein protease that cleaves amyloid protein precursor (APP) and other type I transmembrane proteins. It has two catalytic subunits, presenilins 1 and 2 (PS1 and 2). In our previous report, we observed subtle differences in PS1- and PS2-mediated cleavages of select substrates and slightly different potencies of PS1 versus PS2 inhibition for select γ-secretase inhibitors (GSIs) on various substrates. In this study, we investigated whether γ-secretase modulators (GSMs) and inverse γ-secretase modulators (iGSMs) modulate γ-secretase processivity using multiple different substrates. We next used HEK 293T cell lines in which PSEN1 or PSEN2 was selectively knocked out to investigate processivity and response to GSMs and iGSMs. METHODS: For cell-free γ-secretase cleavage assay, recombinant substrates were incubated with CHAPSO-solubilized CHO or HEK 293T cell membrane with GSMs or iGSMs in suitable buffer. For cell-based assay, cDNA encoding substrates were transfected into HEK 293T cells. Cells were then treated with GSMs or iGSMs, and conditioned media were collected. Aß and Aß-like peptide production from cell-free and cell-based assay were measured by ELISA and mass spectrometry. RESULT: These studies demonstrated that GSMs are highly selective for effects on APP, whereas iGSMs have a more promiscuous effect on many substrates. Surprisingly, iGSMs actually appear to act as like GSIs on select substrates. The data with PSEN1 or PSEN2 knocked out HEK 293T reveal that PS1 has higher processivity and response to GSMs than PS2, but PS2 has higher response to iGSM. CONCLUSION: Collectively, these data indicate that GSMs are likely to have limited target-based toxicity. In addition, they show that iGSMs may act as substrate-selective GSIs providing a potential new route to identify leads for substrate-selective inhibitors of certain γ-secretase-mediated signaling events. With growing concerns that long-term ß-secretase inhibitor is limited by target-based toxicities, such data supports continued development of GSMs as AD prophylactics.


Subject(s)
Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , HEK293 Cells , Humans , Presenilin-2/genetics , Signal Transduction
4.
J Biol Chem ; 294(29): 11276-11285, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31167792

ABSTRACT

Presenilins 1 and 2 (PS1 and 2) are the catalytic subunits of γ-secretase, a multiprotein protease that cleaves amyloid protein precursor and other type I transmembrane proteins. Previous studies with mouse models or cells have indicated differences in PS1 and PS2 functions. We have recently reported that clinical γ-secretase inhibitors (GSIs), initially developed to manage Alzheimer's disease and now being considered for other therapeutic interventions, are both pharmacologically and functionally distinct. Here, using CRISPR/Cas9-based gene editing, we established human HEK 293T cell lines in which endogenous PS1, PS2, or both have been knocked out. Using these knockout lines to examine differences in PS1- and PS2-mediated cleavage events, we confirmed that PS2 generates more intracellular ß-amyloid than does PS1. Moreover, we observed subtle differences in PS1- and PS2-mediated cleavages of select substrates. In exploring the question of whether differences in activity among clinical GSIs could be attributed to differential inhibition of PS1 or PS2, we noted that select GSIs inhibit PS1 and PS2 activities on specific substrates with slightly different potencies. We also found that endoproteolysis of select PS1 FAD-linked variants in human cells is more efficient than what has been previously reported for mouse cell lines. Overall, these results obtained with HEK293T cells suggest that selective PS1 or PS2 inhibition by a given GSI does not explain the previously observed differences in functional and pharmacological properties among various GSIs.


Subject(s)
Presenilin-1/physiology , Presenilin-2/physiology , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , CRISPR-Cas Systems , Gene Knockdown Techniques , HEK293 Cells , Humans , Hydrolysis , Mice , Presenilin-1/genetics , Presenilin-2/genetics , Substrate Specificity
5.
Alzheimers Res Ther ; 11(1): 16, 2019 02 02.
Article in English | MEDLINE | ID: mdl-30711010

ABSTRACT

BACKGROUND: Recent Genome Wide Association Studies (GWAS) have identified novel rare coding variants in immune genes associated with late onset Alzheimer's disease (LOAD). Amongst these, a polymorphism in phospholipase C-gamma 2 (PLCG2) P522R has been reported to be protective against LOAD. PLC enzymes are key elements in signal transmission networks and are potentially druggable targets. PLCG2 is highly expressed in the hematopoietic system. Hypermorphic mutations in PLCG2 in humans have been reported to cause autoinflammation and immune disorders, suggesting a key role for this enzyme in the regulation of immune cell function. METHODS: We assessed PLCG2 distribution in human and mouse brain tissue via immunohistochemistry and in situ hybridization. We transfected heterologous cell systems (COS7 and HEK293T cells) to determine the effect of the P522R AD-associated variant on enzymatic function using various orthogonal assays, including a radioactive assay, IP-One ELISA, and calcium assays. RESULTS: PLCG2 expression is restricted primarily to microglia and granule cells of the dentate gyrus. Plcg2 mRNA is maintained in plaque-associated microglia in the cerebral tissue of an AD mouse model. Functional analysis of the p.P522R variant demonstrated a small hypermorphic effect of the mutation on enzyme function. CONCLUSIONS: The PLCG2 P522R variant is protective against AD. We show that PLCG2 is expressed in brain microglia, and the p.P522R polymorphism weakly increases enzyme function. These data suggest that activation of PLCγ2 and not inhibition could be therapeutically beneficial in AD. PLCγ2 is therefore a potential target for modulating microglia function in AD, and a small molecule drug that weakly activates PLCγ2 may be one potential therapeutic approach.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Frontal Lobe/metabolism , Frontal Lobe/pathology , Phospholipase C gamma/biosynthesis , Phospholipase C gamma/genetics , Alzheimer Disease/pathology , Animals , Female , Genetic Variation/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic
6.
EMBO Mol Med ; 10(11)2018 11.
Article in English | MEDLINE | ID: mdl-30341064

ABSTRACT

Rare coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) are associated with increased risk for Alzheimer's disease (AD), but how they confer this risk remains uncertain. We assessed binding of TREM2, AD-associated TREM2 variants to various forms of Aß and APOE in multiple assays. TREM2 interacts directly with various forms of Aß, with highest affinity interactions observed between TREM2 and soluble Aß42 oligomers. High-affinity binding of TREM2 to Aß oligomers is characterized by very slow dissociation. Pre-incubation with Aß is shown to block the interaction of APOE In cellular assays, AD-associated variants of TREM2 reduced the amount of Aß42 internalized, and in NFAT assay, the R47H and R62H variants decreased NFAT signaling activity in response to Aß42. These studies demonstrate i) a high-affinity interaction between TREM2 and Aß oligomers that can block interaction with another TREM2 ligand and ii) that AD-associated TREM2 variants bind Aß with equivalent affinity but show loss of function in terms of signaling and Aß internalization.


Subject(s)
Amyloid beta-Peptides/metabolism , Membrane Glycoproteins/metabolism , Protein Multimerization , Receptors, Immunologic/metabolism , Signal Transduction , Alzheimer Disease/metabolism , Apolipoproteins E/metabolism , Endocytosis , HEK293 Cells , Humans , NFATC Transcription Factors/metabolism , Protein Binding , Solubility
7.
EMBO Mol Med ; 9(7): 950-966, 2017 07.
Article in English | MEDLINE | ID: mdl-28539479

ABSTRACT

γ-Secretase inhibitors (GSIs) are being actively repurposed as cancer therapeutics based on the premise that inhibition of NOTCH1 signaling in select cancers is therapeutic. Using novel assays to probe effects of GSIs against a broader panel of substrates, we demonstrate that clinical GSIs are pharmacologically distinct. GSIs show differential profiles of inhibition of the various NOTCH substrates, with some enhancing cleavage of other NOTCH substrates at concentrations where NOTCH1 cleavage is inhibited. Several GSIs are also potent inhibitors of select signal peptide peptidase (SPP/SPPL) family members. Extending these findings to mammosphere inhibition assays in triple-negative breast cancer lines, we establish that these GSIs have different functional effects. We also demonstrate that the processive γ-secretase cleavage pattern established for amyloid precursor protein (APP) occurs in multiple substrates and that potentiation of γ-secretase cleavage is attributable to a direct action of low concentrations of GSIs on γ-secretase. Such data definitively demonstrate that the clinical GSIs are not biological equivalents, and provide an important framework to evaluate results from ongoing and completed human trials with these compounds.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Clinical Trials as Topic , Humans
8.
PLoS One ; 10(12): e0144758, 2015.
Article in English | MEDLINE | ID: mdl-26678856

ABSTRACT

The relative increase in Aß42 peptides from familial Alzheimer disease (FAD) linked APP and PSEN mutations can be related to changes in both ε-cleavage site utilization and subsequent step-wise cleavage. Cleavage at the ε-site releases the amyloid precursor protein (APP) intracellular domain (AICD), and perturbations in the position of ε-cleavage are closely associated with changes in the profile of amyloid ß-protein (Aß) species that are produced and secreted. The mechanisms by which γ-secretase modulators (GSMs) or FAD mutations affect the various γ-secretase cleavages to alter the generation of Aß peptides have not been fully elucidated. Recent studies suggested that GSMs do not modulate ε-cleavage of APP, but the data were derived principally from recombinant truncated epitope tagged APP substrate. Here, using full length APP from transfected cells, we investigated whether GSMs modify the ε-cleavage of APP under more native conditions. Our results confirmed the previous findings that ε-cleavage is insensitive to GSMs. In addition, fenofibrate, an inverse GSM (iGSM), did not alter the position or kinetics of ε-cleavage position in vitro. APH1A and APH1B, a subunit of the γ-secretase complex, also modulated Aß42/Aß40 ratio without any alterations in ε-cleavage, a result in contrast to what has been observed with PS1 and APP FAD mutations. Consequently, GSMs and APH1 appear to modulate γ-secretase activity and Aß42 generation by altering processivity but not ε-cleavage site utilization.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/physiology , Peptide Hydrolases/physiology , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Catalytic Domain , Cricetulus , Endopeptidases , Fenofibrate/pharmacology , HEK293 Cells , Humans , Immunoprecipitation , Membrane Proteins/genetics , Mutation , Peptide Fragments/metabolism , Peptide Hydrolases/genetics , Protein Isoforms/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
J Biol Chem ; 286(46): 39794-803, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21868380

ABSTRACT

A subset of non-steroidal anti-inflammatory drugs modulates the γ cleavage site in the amyloid precursor protein (APP) to selectively reduce production of Aß42. It is unclear precisely how these γ-secretase modulators (GSMs) act to preferentially spare Aß40 production as well as Notch processing and signaling. In an effort to determine the substrate requirements in NSAID/GSM activity, we determined the effects of sulindac sulfide and flurbiprofen on γ-cleavage of artificial constructs containing several γ-secretase substrates. Using FLAG-tagged constructs that expressed extracellularly truncated APP, Notch-1, or CD44, we found that these substrates have different sensitivities to sulindac sulfide. γ-Secretase cleavage of APP was altered by sulindac sulfide, but CD44 and Notch-1 were either insensitive or only minimally altered by this compound. Using chimeric APP constructs, we observed that the transmembrane domain (TMD) of APP played a pivotal role in determining drug sensitivity. Substituting the APP TMD with that of APLP2 retained the sensitivity to γ-cleavage modulation, but replacing TMDs from Notch-1 or ErbB4 rendered the resultant molecules insensitive to drug treatment. Specifically, the GXXXG motif within APP appeared to be critical to GSM activity. Consequently, the modulatory effects on γ-cleavage appears to be substrate-dependent. We hypothesize that the substrate present in the γ-secretase complex influences the conformation of the complex so that the binding site of GSMs is either stabilized or less favorable to influence the cleavage of the respective substrates.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Hyaluronan Receptors/metabolism , Receptor, Notch1/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Protein Structure, Tertiary , Receptor, Notch1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sulindac/analogs & derivatives , Sulindac/pharmacology
10.
J Alzheimers Dis ; 27(2): 307-16, 2011.
Article in English | MEDLINE | ID: mdl-21811021

ABSTRACT

Presenilin-1 (PS1) is a broadly expressed transmembrane protein that is often mutated in familial Alzheimer's disease (AD). In addition to its role in amyloid production, PS1 interacts with several protein partners, including the neural plakophilin-related armadillo protein (NPRAP or δ-catenin). Although studies have suggested that NPRAP affects cell adhesion, other data suggest that it can modulate gene expression. To investigate the transcriptional effects of NPRAP, we over-expressed NPRAP and measured gene expression using a microarray. We found that multiple genes, including BCHE, which has been linked to AD, were regulated by NPRAP. Furthermore, we showed that NPRAP nuclear translocation was required for gene regulation. Our results implicate NPRAP as a brain-specific signaling molecule with distinct roles at the cell junction and the nucleus.


Subject(s)
Catenins/physiology , Cell Nucleus/metabolism , Neurons/metabolism , Presenilin-1/physiology , Cell Line, Tumor , Cell Nucleus/genetics , Humans , Delta Catenin
12.
Cell Signal ; 17(4): 437-45, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15601622

ABSTRACT

Mutations in the presenilin (PS) genes are linked to the development of early-onset Alzheimer's disease by a gain-of-function mechanism that alters proteolytic processing of the amyloid precursor protein (APP). Recent work indicates that Alzheimer's-disease-linked mutations in presenilin1 and presenilin2 attenuate calcium entry and augment calcium release from the endoplasmic reticulum (ER) in different cell types. However, the regulatory mechanisms underlying the altered profile of Ca(2+) signaling are unknown. The present study investigated the influence of two familial Alzheimer's-disease-linked presenilin2 variants (N141I and M239V) and a loss-of-function presenilin2 mutant (D263A) on the activity of the transient receptor potential canonical (TRPC)6 Ca(2+) entry channel. We show that transient coexpression of Alzheimer's-disease-linked presenilin2 mutants and TRPC6 in human embryonic kidney (HEK) 293T cells abolished agonist-induced TRPC6 activation without affecting agonist-induced endogenous Ca(2+) entry. The inhibitory effect of presenilin2 and the Alzheimer's-disease-linked presenilin2 variants was not due to an increase in amyloid beta-peptides in the medium. Despite the strong negative effect of the presenilin2 and Alzheimer's-disease-linked presenilin2 variants on agonist-induced TRPC6 activation, conformational coupling between inositol 1,4,5-trisphosphate receptor type 3 (IP(3)R3) and TRPC6 was unaffected. In cells coexpressing presenilin2 or the FAD-linked presenilin2 variants, Ca(2+) entry through TRPC6 could still be induced by direct activation of TRPC6 with 1-oleoyl-2-acetyl-sn-glycerol (OAG). Furthermore, transient coexpression of a loss-of-function PS2 mutant and TRPC6 in HEK293T cells enhanced angiotensin II (AngII)- and OAG-induced Ca(2+) entry. These results clearly indicate that presenilin2 influences TRPC6-mediated Ca(2+) entry into HEK293 cells.


Subject(s)
Alzheimer Disease/metabolism , Calcium/metabolism , Ion Channels/metabolism , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Cell Line , Endopeptidases/genetics , Humans , Membrane Proteins/genetics , Mutation , Presenilin-2 , TRPC Cation Channels , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...