Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Adv ; 8(1): eabl4411, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34985955

ABSTRACT

Many cellular activities, such as cell migration, cell division, phagocytosis, and exo-endocytosis, generate and are regulated by membrane tension gradients. Membrane tension gradients drive membrane flows, but there is controversy over how rapidly plasma membrane flow can relax tension gradients. Here, we show that membrane tension can propagate rapidly or slowly, spanning orders of magnitude in speed, depending on the cell type. In a neuronal terminal specialized for rapid synaptic vesicle turnover, membrane tension equilibrates within seconds. By contrast, membrane tension does not propagate in neuroendocrine adrenal chromaffin cells secreting catecholamines. Stimulation of exocytosis causes a rapid, global decrease in the synaptic terminal membrane tension, which recovers slowly due to endocytosis. Thus, membrane flow and tension equilibration may be adapted to distinct membrane recycling requirements.

2.
Mol Cell Neurosci ; 109: 103562, 2020 12.
Article in English | MEDLINE | ID: mdl-32987141

ABSTRACT

Abnormal dendritic arbor development has been implicated in a number of neurodevelopmental disorders, such as autism and Rett syndrome, and the neuropsychiatric disorder schizophrenia. Postmortem brain samples from subjects with schizophrenia show elevated levels of NOS1AP in the dorsolateral prefrontal cortex, a region of the brain associated with cognitive function. We previously reported that the long isoform of NOS1AP (NOS1AP-L), but not the short isoform (NOS1AP-S), negatively regulates dendrite branching in rat hippocampal neurons. To investigate the role that NOS1AP isoforms play in human dendritic arbor development, we adapted methods to generate human neural progenitor cells and neurons using induced pluripotent stem cell (iPSC) technology. We found that increased protein levels of either NOS1AP-L or NOS1AP-S decrease dendrite branching in human neurons at the developmental time point when primary and secondary branching actively occurs. Next, we tested whether pharmacological agents can decrease the expression of NOS1AP isoforms. Treatment of human iPSC-derived neurons with d-serine, but not clozapine, haloperidol, fluphenazine, or GLYX-13, results in a reduction in endogenous NOS1AP-L, but not NOS1AP-S, protein expression; however, d-serine treatment does not reverse decreases in dendrite number mediated by overexpression of NOS1AP isoforms. In summary, we demonstrate how an in vitro model of human neuronal development can help in understanding the etiology of schizophrenia and can also be used as a platform to screen drugs for patients.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Dendrites/ultrastructure , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Cells, Cultured , Clozapine/pharmacology , Drug Evaluation, Preclinical , Fluphenazine/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/physiology , Haloperidol/pharmacology , Humans , Induced Pluripotent Stem Cells/metabolism , Ion Channels/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Oligopeptides/pharmacology , Patch-Clamp Techniques , Protein Isoforms/physiology , Schizophrenia/etiology , Schizophrenia/genetics , Serine/pharmacology
3.
Methods Mol Biol ; 1860: 263-275, 2019.
Article in English | MEDLINE | ID: mdl-30317511

ABSTRACT

During exocytosis, vesicles fuse with the plasma membrane and release their contents. The fusion pore is the initial, nanometer-sized connection between the plasma membrane and the cargo-laden vesicle. A growing body of evidence points toward the fusion pore being a regulator of exocytosis, but the shortcomings of current experimental techniques to investigate single-fusion pores make it difficult to study factors governing pore behavior. Here we describe an assay that fuses v-SNARE-reconstituted nanodiscs with cells ectopically expressing "flipped" t-SNAREs to monitor dynamics of single fusion pores in a biochemically defined system using electrical recordings. We also describe a fluorescence microscopy-based approach to monitor nanodisc-cell fusion that is much simpler to employ, but cannot resolve single pores.


Subject(s)
Biological Assay/methods , Nanostructures/chemistry , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Membrane/chemistry , Cell Membrane/metabolism , Exocytosis , Genetic Engineering , HeLa Cells , Humans , Membrane Fusion , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Secretory Vesicles/chemistry , Secretory Vesicles/metabolism , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/genetics , Syntaxin 1/chemistry , Syntaxin 1/genetics
4.
Sci Rep ; 6: 27287, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27264104

ABSTRACT

The initial, nanometer-sized connection between the plasma membrane and a hormone- or neurotransmitter-filled vesicle -the fusion pore- can flicker open and closed repeatedly before dilating or resealing irreversibly. Pore dynamics determine release and vesicle recycling kinetics, but pore properties are poorly known because biochemically defined single-pore assays are lacking. We isolated single flickering pores connecting v-SNARE-reconstituted nanodiscs to cells ectopically expressing cognate, "flipped" t-SNAREs. Conductance through single, voltage-clamped fusion pores directly reported sub-millisecond pore dynamics. Pore currents fluctuated, transiently returned to baseline multiple times, and disappeared ~6 s after initial opening, as if the fusion pore fluctuated in size, flickered, and resealed. We found that interactions between v- and t-SNARE transmembrane domains (TMDs) promote, but are not essential for pore nucleation. Surprisingly, TMD modifications designed to disrupt v- and t-SNARE TMD zippering prolonged pore lifetimes dramatically. We propose that the post-fusion geometry of the proteins contribute to pore stability.


Subject(s)
Cell Fusion/methods , Cell Nucleus/metabolism , SNARE Proteins/chemistry , SNARE Proteins/metabolism , Calcium/metabolism , Exocytosis , HeLa Cells , Humans , Membrane Fusion , Neurotransmitter Agents , Protein Binding , Protein Domains , Secretory Vesicles/metabolism
5.
Front Cell Neurosci ; 10: 6, 2016.
Article in English | MEDLINE | ID: mdl-26869880

ABSTRACT

Proper communication between neurons depends upon appropriate patterning of dendrites and correct distribution and structure of spines. Schizophrenia is a neuropsychiatric disorder characterized by alterations in dendrite branching and spine density. Nitric oxide synthase 1 adaptor protein (NOS1AP), a risk gene for schizophrenia, encodes proteins that are upregulated in the dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia. To elucidate the effects of NOS1AP overexpression observed in individuals with schizophrenia, we investigated changes in actin dynamics and spine development when a long (NOS1AP-L) or short (NOS1AP-S) isoform of NOS1AP is overexpressed. Increased NOS1AP-L protein promotes the formation of immature spines when overexpressed in rat cortical neurons from day in vitro (DIV) 14 to DIV 17 and reduces the amplitude of miniature excitatory postsynaptic currents (mEPSCs). In contrast, increased NOS1AP-S protein increases the rate of actin polymerization and the number of immature and mature spines, which may be attributed to a decrease in total Rac1 expression and a reduction in the levels of active cofilin. The increase in the number of mature spines by overexpression of NOS1AP-S is accompanied by an increase in the frequency of mEPSCs. Our findings show that overexpression of NOS1AP-L or NOS1AP-S alters the actin cytoskeleton and synaptic function. However, the mechanisms by which these isoforms induce these changes are distinct. These results are important for understanding how increased expression of NOS1AP isoforms can influence spine development and synaptic function.

SELECTION OF CITATIONS
SEARCH DETAIL
...