Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Type of study
Publication year range
1.
Lab Invest ; 101(12): 1605-1617, 2021 12.
Article in English | MEDLINE | ID: mdl-34462532

ABSTRACT

Synaptic transfer of tau has long been hypothesized from the human pathology pattern and has been demonstrated in vitro and in vivo, but the precise mechanisms remain unclear. Extracellular vesicles such as exosomes have been suggested as a mechanism, but not all tau is exosomal. The present experiments use a novel flow cytometry assay to quantify depolarization of synaptosomes by KCl after loading with FM2-10, which induces a fluorescence reduction associated with synaptic vesicle release; the degree of reduction in cryopreserved human samples equaled that seen in fresh mouse synaptosomes. Depolarization induced the release of vesicles in the size range of exosomes, along with tetraspanin markers of extracellular vesicles. A number of tau peptides were released, including tau oligomers; released tau was primarily unphosphorylated and C-terminal truncated, with Aß release just above background. When exosomes were immunopurified from release supernatants, a prominent tau band showed a dark smeared appearance of SDS-stable oligomers along with the exosomal marker syntenin-1, and these exosomes induced aggregation in the HEK tau biosensor assay. However, the flow-through did not seed aggregation. Size exclusion chromatography of purified released exosomes shows faint signals from tau in the same fractions that show a CD63 band, an exosomal size signal, and seeding activity. Crude synaptosomes from control, tauopathy, and AD cases demonstrated lower seeding in tauopathy compared to AD that is correlated with the measured Aß42 level. These results show that AD synapses release exosomal tau that is C-terminal-truncated, oligomeric, and with seeding activity that is enhanced by Aß. Taken together with previous findings, these results are consistent with a direct prion-like heterotypic seeding of tau by Aß within synaptic terminals, with subsequent loading of aggregated tau onto exosomes that are released and competent for tau seeding activity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Synapses/metabolism , Synaptosomes/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Extracellular Vesicles/metabolism , Female , Humans , Male , Mice , Middle Aged , Protein Aggregation, Pathological
3.
Mol Brain ; 14(1): 70, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875010

ABSTRACT

AIM: We have previously reported that cambinol (DDL-112), a known inhibitor of neutral sphingomyelinase-2 (nSMase2), suppressed extracellular vesicle (EV)/exosome production in vitro in a cell model and reduced tau seed propagation. The enzyme nSMase2 is involved in the production of exosomes carrying proteopathic seeds and could contribute to cell-to-cell transmission of pathological protein aggregates implicated in neurodegenerative diseases such as Parkinson's disease (PD). Here, we performed in vivo studies to determine if DDL-112 can reduce brain EV/exosome production and proteopathic alpha synuclein (αSyn) spread in a PD mouse model. METHODS: The acute effects of single-dose treatment with DDL-112 on interleukin-1ß-induced extracellular vesicle (EV) release in brain tissue of Thy1-αSyn PD model mice and chronic effects of 5 week DDL-112 treatment on behavioral/motor function and proteinase K-resistant αSyn aggregates in the PD model were determined. RESULTS/DISCUSSION: In the acute study, pre-treatment with DDL-112 reduced EV/exosome biogenesis and in the chronic study, treatment with DDL-112 was associated with a reduction in αSyn aggregates in the substantia nigra and improvement in motor function. Inhibition of nSMase2 thus offers a new approach to therapeutic development for neurodegenerative diseases with the potential to reduce the spread of disease-specific proteopathic proteins.


Subject(s)
Brain/metabolism , Enzyme Inhibitors/pharmacology , Exosomes/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Exosomes/ultrastructure , Mice, Transgenic , Naphthalenes/pharmacology , Protein Aggregates/drug effects , Pyrimidinones/pharmacology , Sirtuins/metabolism , Sphingomyelin Phosphodiesterase/metabolism
4.
ACS Chem Biol ; 15(6): 1671-1684, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32352753

ABSTRACT

We report the discovery of a novel class of compounds that function as dual inhibitors of the enzymes neutral sphingomyelinase-2 (nSMase2) and acetylcholinesterase (AChE). Inhibition of these enzymes provides a unique strategy to suppress the propagation of tau pathology in the treatment of Alzheimer's disease (AD). We describe the key SAR elements that affect relative nSMase2 and/or AChE inhibitor effects and potency, in addition to the identification of two analogs that suppress the release of tau-bearing exosomes in vitro and in vivo. Identification of these novel dual nSMase2/AChE inhibitors represents a new therapeutic approach to AD and has the potential to lead to the development of truly disease-modifying therapeutics.


Subject(s)
Acetylcholinesterase/drug effects , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Structure-Activity Relationship
5.
Sci Rep ; 8(1): 17574, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514854

ABSTRACT

We describe here the results from the testing of a small molecule first-in-class apolipoprotein E4 (ApoE4)-targeted sirtuin1 (SirT1) enhancer, A03, that increases the levels of the neuroprotective enzyme SirT1 while not affecting levels of neurotoxic sirtuin 2 (SirT2) in vitro in ApoE4-transfected cells. A03 was identified by high-throughput screening (HTS) and found to be orally bioavailable and brain penetrant. In vivo, A03 treatment increased SirT1 levels in the hippocampus of 5XFAD-ApoE4 (E4FAD) Alzheimer's disease (AD) model mice and elicited cognitive improvement while inducing no observed toxicity. We were able to resolve the enantiomers of A03 and show using in vitro models that the L-enantiomer was more potent than the corresponding D-enantiomer in increasing SirT1 levels. ApoE4 expression has been shown to decrease the level of the NAD-dependent deacetylase and major longevity determinant SirT1 in brain tissue and serum of AD patients as compared to normal controls. A deficiency in SirT1 level has been recently implicated in increased tau acetylation, a dominant post-translational modification and key pathological event in AD and tauopathies. Therefore, as a novel approach to therapeutic development for AD, we targeted identification of compounds that enhance and normalize brain SirT1 levels.


Subject(s)
Alzheimer Disease/drug therapy , Antiparkinson Agents/pharmacology , Apolipoprotein E4/metabolism , Hippocampus/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sirtuin 1/metabolism , Sirtuin 2/metabolism , Animals , Antiparkinson Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Hippocampus/metabolism , Mice , Mice, Transgenic , Selective Serotonin Reuptake Inhibitors/therapeutic use
6.
FEBS J ; 285(16): 3002-3012, 2018 08.
Article in English | MEDLINE | ID: mdl-29933522

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease which has no effective treatment and is characterized by psychiatric disorders, motor alterations, and dementia, with the cognitive deficits representing a devastating aspect of the disorder. Oxidative stress and elevated levels of lipid peroxidation (LPO) products are found in mouse models and patients with HD, suggesting that strategies to reduce LPO may be beneficial in HD. In contrast with traditional antioxidants, substituting hydrogen with deuterium at bis-allylic sites in polyunsaturated fatty acids (D-PUFA) decreases the rate-limiting initiation step of PUFA autoxidation, a strategy that has shown benefits in other neurodegenerative diseases. Here, we investigated the effect of D-PUFA treatment in a knock-in mouse model of HD (Q140) which presents motor deficits and neuropathology from a few months of age, and progressive cognitive decline. Q140 knock-in mice were fed a diet containing either D- or H-PUFAs for 5 months starting at 1 month of age. D-PUFA treatment significantly decreased F2 -isoprostanes in the striatum by approximately 80% as compared to H-PUFA treatment and improved performance in novel object recognition tests, without significantly changing motor deficits or huntingtin aggregation. Therefore, D-PUFA administration represents a promising new strategy to broadly reduce rates of LPO, and may be useful in improving a subset of the core deficits in HD.


Subject(s)
Cognitive Dysfunction/diet therapy , Deuterium/pharmacology , Huntington Disease/etiology , Linoleic Acid/pharmacology , Lipid Peroxidation/drug effects , Animals , Body Weight/drug effects , Cognitive Dysfunction/metabolism , Deuterium/chemistry , Dietary Supplements , Disease Models, Animal , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/pharmacology , Female , Huntingtin Protein/genetics , Linoleic Acid/chemistry , Male , Mice, Transgenic , Motor Activity/drug effects
7.
Neurobiol Dis ; 117: 170-180, 2018 09.
Article in English | MEDLINE | ID: mdl-29859873

ABSTRACT

In addition to dopaminergic and motor deficits, patients with Parkinson's disease (PD) suffer from non-motor symptoms, including early cognitive and social impairment, that do not respond well to dopaminergic therapy. Cholinergic deficits may contribute to these problems, but cholinesterase inhibitors have limited efficacy. Mice over-expressing α-synuclein, a protein critically associated with PD, show deficits in cognitive and social interaction tests, as well as a decrease in cortical acetylcholine. We have evaluated the effects of chronic administration of nicotine in mice over-expressing wild type human α-synuclein under the Thy1-promoter (Thy1-aSyn mice). Nicotine was administered subcutaneously by osmotic minipump for 6 months from 2 to 8 months of age at 0.4 mg/kg/h and 2.0 mg/kg/h. The higher dose was toxic in the Thy1-aSyn mice, but the low dose was well tolerated and both doses ameliorated cognitive impairment in Y-maze performance after 5 months of treatment. In a separate cohort of Thy1-aSyn mice, nicotine was administered at the lower dose for one month beginning at 5 months of age. This treatment partially eliminated the cognitive deficit in novel object recognition and social impairment. In contrast, chronic nicotine did not improve motor deficits after 2, 4 or 6 months of treatment, nor modified α-synuclein aggregation, tyrosine hydroxylase immunostaining, synaptic and dendritic markers, or microglial activation in Thy1-aSyn mice. These results suggest that cognitive and social impairment in synucleinopathies like PD may result from deficits in cholinergic neurotransmission and may benefit from chronic administration of nicotinic agonists.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Nicotine/administration & dosage , Social Behavior Disorders/drug therapy , Social Behavior Disorders/metabolism , alpha-Synuclein/biosynthesis , Animals , Cognition Disorders/genetics , Drug Administration Schedule , Gene Expression , Humans , Mice , Mice, Transgenic , Nicotinic Agonists/administration & dosage , Social Behavior Disorders/genetics , alpha-Synuclein/genetics
8.
Biochem Biophys Res Commun ; 499(4): 751-757, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29604274

ABSTRACT

Targeting of molecular pathways involved in the cell-to-cell propagation of pathological tau species is a novel approach for development of disease-modifying therapies that could block tau pathology and attenuate cognitive decline in patients with Alzheimer's disease and other tauopathies. We discovered cambinol through a screening effort and show that it is an inhibitor of cell-to-cell tau propagation. Our in vitro data demonstrate that cambinol inhibits neutral sphingomyelinase 2 (nSMase2) enzyme activity in dose response fashion, and suppresses extracellular vesicle (EV) production while reducing tau seed propagation. Our in vivo testing with cambinol shows that it can reduce the nSMase2 activity in the brain after oral administration. Our molecular docking and simulation analysis reveals that cambinol can target the DK-switch in the nSMase2 active site.


Subject(s)
Enzyme Inhibitors/pharmacology , Naphthalenes/pharmacology , Pyrimidinones/pharmacology , Sphingomyelin Phosphodiesterase/chemistry , tau Proteins/metabolism , Animals , Biosensing Techniques , Brain/metabolism , Cell-Free System , Enzyme Inhibitors/chemistry , Extracellular Vesicles/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Models, Molecular , Naphthalenes/chemistry , Permeability , Protein Domains , Pyrimidinones/chemistry , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Tissue Extracts , tau Proteins/antagonists & inhibitors
9.
ACS Chem Neurosci ; 9(3): 462-468, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29161500

ABSTRACT

Humanin (HN), a 24-amino acid bioactive peptide, has been shown to increase cell survival of neurons after exposure to Aß and NMDA-induced toxicity and thus could be beneficial in the treatment of Alzheimer's disease (AD). The neuroprotection by HN is reported to be primarily through its agonist binding properties to the gp130 receptor. However, the peptidic nature of HN presents challenges in its development as a therapeutic for AD. We report here for the first time the elucidation of the binding site of Humanin (HN) peptide to the gp130 receptor extracellular domain through modeling and the synthesis of small molecule mimetics that interact with the HN binding site on the gp130 receptor and provide protection against NMDA-induced neurotoxicity in primary hippocampal neurons. A brain permeable small molecule mimetic was identified through exploratory medicinal chemistry using microfluidic flow chemistry to facilitate the synthesis of new analogues for screening and SAR optimization.


Subject(s)
Amyloid beta-Peptides/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , N-Methylaspartate/toxicity , Neurons/drug effects , Alzheimer Disease/metabolism , Cell Death/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...