Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 383(2): 117-128, 2022 11.
Article in English | MEDLINE | ID: mdl-36116796

ABSTRACT

Using synaptosomes purified from the brains of two transgenic mouse models overexpressing mutated human tau (TgP301S and Tg4510) and brains of patients with sporadic Alzheimer's disease, we showed that aggregated and hyperphosphorylated tau was both present in purified synaptosomes and released in a calcium- and synaptosome-associated protein of 25 kDa (SNAP25)-dependent manner. In all mouse and human synaptosomal preparations, tau release was inhibited by the selective metabotropic glutamate receptor 2/3 (mGluR2/3) agonist LY379268, an effect prevented by the selective mGlu2/3 antagonist LY341495. LY379268 was also able to block pathologic tau propagation between primary neurons in an in vitro microfluidic cellular model. These novel results are transformational for our understanding of the molecular mechanisms mediating tau release and propagation at synaptic terminals in Alzheimer's disease and suggest that these processes could be inhibited therapeutically by the selective activation of presynaptic G protein-coupled receptors. SIGNIFICANCE STATEMENT: Pathological tau release and propagation are key neuropathological events underlying cognitive decline in Alzheimer's disease patients. This paper describes the role of regulated exocytosis, and the soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) protein SNAP25, in mediating tau release from rodent and human synaptosomes. This paper also shows that a selective mGluR2/3 agonist is highly effective in blocking tau release from synaptosomes and tau propagation between neurons, opening the way to the discovery of novel therapeutic approaches to this devastating disease.


Subject(s)
Alzheimer Disease , Receptors, Metabotropic Glutamate , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Animals , Calcium/metabolism , Exocytosis , Humans , Mice , N-Ethylmaleimide-Sensitive Proteins/metabolism , N-Ethylmaleimide-Sensitive Proteins/pharmacology , Receptors, Metabotropic Glutamate/metabolism , SNARE Proteins/metabolism , SNARE Proteins/pharmacology , Synaptosomes/metabolism
2.
Br J Clin Pharmacol ; 87(8): 3177-3189, 2021 08.
Article in English | MEDLINE | ID: mdl-33450079

ABSTRACT

AIMS: The objectives of this study were to investigate the safety, tolerability, pharmacokinetics and pharmacodynamics of single and multiple oral doses of ONO-7684, a novel activated factor XI (FXIa) inhibitor, in healthy subjects. METHODS: This was a first-in-human (FIH), randomised, placebo-controlled, double-blind, single and multiple dose study in healthy subjects under fed and fasted conditions. This study consisted of two parts: single ascending dose (Part A; 1, 5, 20, 80, 150 or 300 mg ONO-7684 or placebo) and multiple ascending doses (Part B; 80, 150 or 250 mg ONO-7684 or placebo daily for 14 days). In both parts, subjects were randomised in a 3:1 ratio to receive ONO-7684 or placebo. RESULTS: ONO-7684 was well tolerated at all dose levels tested following both single and repeated doses, with a low overall incidence of treatment-emergent adverse events. There was no evidence to suggest a bleeding risk. Dose proportionality in exposure was observed for the range of 1-300 mg ONO-7684 in Part A. In Part A, the half-life of ONO-7684 administered in the fasted state ranged from 16.0 to 19.8 hours. In Part B, the half-life of ONO-7684 administered in the fed state ranged from 22.1 to 27.9 hours, supporting once daily oral dosing. ONO-7684 strongly inhibited factor XI coagulation activity (FXI:C) and increased activated partial thromboplastin time (aPTT), with a mean maximum on treatment percentage inhibition versus baseline of 92% and a mean maximum on treatment ratio-to-baseline of 2.78, respectively, at 250 mg ONO-7684 daily. CONCLUSIONS: The data generated in this FIH study demonstrate the promising potential of oral FXIa inhibition and ONO-7684 for indications requiring anticoagulation.


Subject(s)
Administration, Oral , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Half-Life , Humans , Partial Thromboplastin Time
3.
Drug Discov Today ; 24(2): 533-543, 2019 02.
Article in English | MEDLINE | ID: mdl-30395928

ABSTRACT

It is important in drug discovery to demonstrate that activity of novel drugs found by screening on recombinant receptors translates to activity on native human receptors in brain areas affected by disease. In this review, we summarise the development and use of the microtransplantation technique. Native receptors are reconstituted from human brain tissues into oocytes from the frog Xenopus laevis where they can be functionally assessed. Oocytes microtransplanted with hippocampal tissue from an epileptic patient were used to demonstrate that new antiepileptic agents act on receptors in diseased tissue. Furthermore, frozen post-mortem human tissues were used to show that drugs are active on receptors in brain areas associated with a disease; but not in areas associated with side effects.


Subject(s)
Brain/metabolism , Oocytes/metabolism , Receptors, Cell Surface/physiology , Transplantation, Heterologous/methods , Animals , Drug Discovery , Humans
4.
Nat Neurosci ; 21(1): 130-138, 2018 01.
Article in English | MEDLINE | ID: mdl-29203896

ABSTRACT

The postsynaptic proteome of excitatory synapses comprises ~1,000 highly conserved proteins that control the behavioral repertoire, and mutations disrupting their function cause >130 brain diseases. Here, we document the composition of postsynaptic proteomes in human neocortical regions and integrate it with genetic, functional and structural magnetic resonance imaging, positron emission tomography imaging, and behavioral data. Neocortical regions show signatures of expression of individual proteins, protein complexes, biochemical and metabolic pathways. We characterized the compositional signatures in brain regions involved with language, emotion and memory functions. Integrating large-scale GWAS with regional proteome data identifies the same cortical region for smoking behavior as found with fMRI data. The neocortical postsynaptic proteome data resource can be used to link genetics to brain imaging and behavior, and to study the role of postsynaptic proteins in localization of brain functions.


Subject(s)
Neocortex/pathology , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Synaptosomes/metabolism , Animals , Computational Biology , Female , Humans , Image Processing, Computer-Assisted , Male , Membrane Potentials/genetics , Microinjections , Neocortex/diagnostic imaging , Nerve Tissue Proteins/genetics , Oocytes , Oxygen/blood , Patch-Clamp Techniques , Positron-Emission Tomography , Proteomics , Stroke/pathology , Synapses/ultrastructure , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
5.
J Neurochem ; 138(3): 384-96, 2016 08.
Article in English | MEDLINE | ID: mdl-27216696

ABSTRACT

Disruption in the expression and function of synaptic proteins, and ion channels in particular, is critical in the pathophysiology of human neuropsychiatric and neurodegenerative diseases. However, very little is known regarding the functional and pharmacological properties of native synaptic human ion channels, and their potential changes in pathological conditions. Recently, an electrophysiological technique has been enabled for studying the functional and pharmacological properties of ion channels present in crude membrane preparation obtained from post-mortem frozen brains. We here extend these studies by showing that human synaptic ion channels also can be studied in this way. Synaptosomes purified from different regions of rodent and human brain (control and Alzheimer's) were characterized biochemically for enrichment of synaptic proteins, and expression of ion channel subunits. The same synaptosomes were also reconstituted in Xenopus oocytes, in which the functional and pharmacological properties of the native synaptic ion channels were characterized using the voltage clamp technique. We show that we can detect GABA, (RS)-α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and NMDA receptors, and modulate them pharmacologically with selective agonists, antagonists, and allosteric modulators. Furthermore, changes in ion channel expression and function were detected in synaptic membranes from Alzheimer's brains. Our present results demonstrate the possibility to investigate synaptic ion channels from healthy and pathological brains. This method of synaptosomes preparation and injection into oocytes is a significant improvement over the earlier method. It opens the way to directly testing, on native ion channels, the effects of novel drugs aimed at modulating important classes of synaptic targets. Disruption in the expression and function of synaptic ion channels is critical in the pathophysiology of human neurodegenerative diseases. We here show that synaptosomes purified from rodent and human frozen brain (control and Alzheimer disease) can be studied both biochemically and functionally. This method opens the way to directly testing the effects of novel drugs on native ion channels.


Subject(s)
Brain/metabolism , Ion Channels/metabolism , Oocytes/metabolism , Synaptosomes/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Female , Humans , Patch-Clamp Techniques/methods , Rats, Wistar , Receptors, GABA-A/metabolism , Xenopus laevis , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
6.
Biochem Pharmacol ; 86(8): 1063-73, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23830821

ABSTRACT

Chronic nicotine exposure gives rise to neural adaptations that change whole cell physiology and behaviour mainly by interacting with neuronal nicotinic acetylcholine receptors (nAChRs). The major nicotine-induced neuroadaptation is the up-regulation of brain nAChRs by means of cell-delimited post-translational mechanisms. We review what is known of the processes regulating nAChR assembly, degradation and trafficking, and how nicotine-induced modulation of these processes leads to nAChR up-regulation and changes in downstream neuronal plasticity at molecular, cellular and circuit level.


Subject(s)
Protein Transport/physiology , Receptors, Nicotinic/metabolism , Up-Regulation/physiology , Animals , Cell Membrane , Neurons/physiology , Protein Conformation , Receptors, Nicotinic/classification , Receptors, Nicotinic/genetics
7.
J Neurosci ; 33(30): 12316-28, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884938

ABSTRACT

Heteromeric nAChRs are pentameric cation channels, composed of combinations of two or three α and three or two ß subunits, which play key physiological roles in the central and peripheral nervous systems. The prototypical agonist nicotine acts intracellularly to upregulate many nAChR subtypes, a phenomenon that is thought to contribute to the nicotine dependence of cigarette smokers. The α3ß4 subtype has recently been genetically linked to nicotine dependence and lung cancer; however, the mode of action of nicotine on this receptor subtype has been incompletely investigated. Here, using transfected mammalian cells as model system, we characterized the response of the human α3ß4 receptor subtype to nicotine and the mechanism of action of the drug. Nicotine, when present at 1 mm concentration, elicited a ∼5-fold increase of cell surface α3ß4 and showed a more modest upregulatory effect also at concentrations as low as 10 µM. Upregulation was obtained if nicotine was present during, but not after, pentamer assembly and was caused by increased stability and trafficking of receptors assembled in the presence of the drug. Experimental determinations as well as computational studies of subunit stoichiometry showed that nicotine favors assembly of pentamers with (α3)2(ß4)3 stoichiometry; these are less prone than (α3)3(ß4)2 receptors to proteasomal degradation and, because of the presence in the ß subunit of an endoplasmic reticulum export motif, more efficiently transported to the plasma membrane. Our findings uncover a novel mechanism of nicotine-induced α3ß4 nAChR upregulation that may be relevant also for other nAChR subtypes.


Subject(s)
Nicotine/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Receptors, Nicotinic/metabolism , Smoking/physiopathology , Animals , Antibodies/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Male , Models, Chemical , Mutagenesis/physiology , Neuroblastoma , Nicotinic Agonists/pharmacology , Proteasome Endopeptidase Complex/metabolism , Rabbits , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Receptors, Nicotinic/immunology , Smoking/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL