Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Neurosci ; 11: 455, 2017.
Article in English | MEDLINE | ID: mdl-28848381

ABSTRACT

Alzheimer's disease is characterized by cognitive decline, neuronal degeneration, and the accumulation of amyloid-beta (Aß). Although, the neurotoxic Aß peptide is widely believed to trigger neuronal dysfunction and degeneration in Alzheimer's disease, the mechanism by which this occurs is poorly defined. Here we describe a novel, Aß-triggered apoptotic pathway in which Aß treatment leads to the upregulation of G-protein activated inwardly rectifying potassium (GIRK/Kir3) channels, causing potassium efflux from neurons and Aß-mediated apoptosis. Although, GIRK channel activity is required for Aß-induced neuronal degeneration, we show that it is not sufficient, with coincident signaling by the p75 neurotrophin receptor (p75NTR) also required for potassium efflux and cell death. Our results identify a novel role for GIRK channels in mediating apoptosis, and provide a previously missing mechanistic link between the excitotoxicity of Aß and its ability to trigger cell death pathways, such as that mediated by p75NTR. We propose that this death-signaling pathway contributes to the dysfunction of neurons in Alzheimer's disease and is responsible for their eventual degeneration.

2.
PLoS One ; 8(3): e60152, 2013.
Article in English | MEDLINE | ID: mdl-23544129

ABSTRACT

The lipid phosphatidylinositol 3,5-bisphosphate (PtdIns(3,5)P 2), synthesised by PIKfyve, regulates a number of intracellular membrane trafficking pathways. Genetic alteration of the PIKfyve complex, leading to even a mild reduction in PtdIns(3,5)P 2, results in marked neurodegeneration via an uncharacterised mechanism. In the present study we have shown that selectively inhibiting PIKfyve activity, using YM-201636, significantly reduces the survival of primary mouse hippocampal neurons in culture. YM-201636 treatment promoted vacuolation of endolysosomal membranes followed by apoptosis-independent cell death. Many vacuoles contained intravacuolar membranes and inclusions reminiscent of autolysosomes. Accordingly, YM-201636 treatment increased the level of the autophagosomal marker protein LC3-II, an effect that was potentiated by inhibition of lysosomal proteases, suggesting that alterations in autophagy could be a contributing factor to neuronal cell death.


Subject(s)
Aminopyridines/pharmacology , Apoptosis/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Neurons/cytology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Autophagy , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Hippocampus/cytology , Immunohistochemistry , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Neuroendocrine Cells/cytology , Neuroendocrine Cells/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Phosphoinositide-3 Kinase Inhibitors , Protein Transport/drug effects , Rats , Vacuoles/drug effects , Vacuoles/metabolism
3.
J Neurosci ; 28(1): 315-24, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18171948

ABSTRACT

The pan neurotrophin receptor p75(NTR) signals programmed cell death both during nervous system development and after neural trauma and disease in the adult. However, the molecular pathways by which death is mediated remain poorly understood. Here, we show that this cell death is initiated by activation of G-protein-coupled inwardly rectifying potassium (GIRK/Kir3) channels and a consequent potassium efflux. Death signals stimulated by neurotrophin-mediated cleavage of p75(NTR) activate GIRK channels through the generation and binding of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2/PIP2] to GIRK channels. Both GIRK channel activity and p75(NTR)-mediated neuronal death are inhibited by sequestration of PtdIns(4,5)P2 and application of GIRK channel inhibitors, whereas pertussis toxin treatment has no effect. Thus, p75(NTR) activates GIRK channels without the need for G(i/o)-proteins. Our results demonstrate a novel mode of activation of GIRK channels, representing an early step in the p75(NTR)-mediated cell death pathway and suggesting a function for these channels during nervous system development.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptors, Nerve Growth Factor/physiology , Animals , Animals, Newborn , Caspases/metabolism , Cell Death/physiology , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Green Fluorescent Proteins/metabolism , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Patch-Clamp Techniques/methods , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Transfection/methods
4.
Mol Cell Neurosci ; 37(2): 346-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055214

ABSTRACT

It has recently been shown that the p75 neurotrophin receptor (p75(NTR)), which is known to mediate neural cell death during development of the nervous system and in a range of adult neurodegenerative conditions, undergoes a regulated process of cell surface receptor cleavage, regulated intramembrane proteolysis (RIP). Here we show that neuronal death signaling occurs only following extracellular metalloprotease cleavage of p75(NTR) and palmitoylation of the resultant C-terminal fragment, causing its translocation to cholesterol-rich domains of the plasma membrane. Furthermore, death signaling is promoted by inhibition of intracellular gamma-secretase cleavage, a process which also occurs within the cholesterol-rich domains. In the presence of TrkA signaling, C-terminal fragment localization in these cholesterol-rich domains is prevented, thereby blocking neuronal death. Thus p75(NTR) activates neuronal death pathways in conditions where the balance of normal RIP is shifted toward extracellular domain cleavage due to increased metalloprotease activity, decreased TrkA activity or compromised gamma-secretase activity, all of which are features of neurodegenerative conditions such as Alzheimer's disease.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Cell Death/genetics , Lipoylation/genetics , Membrane Microdomains/metabolism , Nerve Degeneration/metabolism , Receptors, Nerve Growth Factor/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Extracellular Space/metabolism , Humans , Intracellular Fluid/metabolism , Membrane Microdomains/genetics , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Peptide Hydrolases/metabolism , Protein Structure, Tertiary/genetics , Rats , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...