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.
Glia ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39001577

ABSTRACT

Noradrenaline (norepinephrine) is known to modulate many physiological functions and behaviors. In this study, we tested to what extent astrocytes, a type of glial cell, participate in noradrenergic signaling in mouse primary visual cortex (V1). Astrocytes are essential partners of neurons in the central nervous system. They are central to brain homeostasis, but also dynamically regulate neuronal activity, notably by relaying and regulating neuromodulator signaling. Indeed, astrocytes express receptors for multiple neuromodulators, including noradrenaline, but the extent to which astrocytes are involved in noradrenergic signaling remains unclear. To test whether astrocytes are involved in noradrenergic neuromodulation in mice, we employed both short hairpin RNA mediated knockdown as well as pharmacological manipulation of the major noradrenaline receptor in astrocytes, the α1A-adrenoreceptor. Using acute brain slices, we found that the astrocytic α1A-adrenoreceptor subtype contributes to the generation of large intracellular Ca2+ signals in visual cortex astrocytes, which are generally thought to underlie astrocyte function. To test if reduced α1A-adrenoreceptor signaling in astrocytes affected the function of neuronal circuits in V1, we used both patch-clamp and field potential recordings. These revealed that noradrenergic signaling through the astrocyte α1A-adrenoreceptor is important to not only modulate synaptic activity but also to regulate plasticity in V1, through the potentiation of synaptic responses in circuits involved in visual information processing.

2.
Nanoscale ; 16(12): 6053-6067, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38421016

ABSTRACT

Mesoporous systems are ubiquitous in membrane science and applications due to their high internal surface area and tunable pore size. A new synthesis pathway of hydrolytic ionosilica films with mesopores formed by ionic liquid (IL) templating is proposed and compared with the traditional non-hydrolytic strategy. For both pathways, the multi-scale formation of pores has been studied as a function of IL content, combining the results of thermogravimetric analysis (TGA), nitrogen sorption, and small-angle X-ray scattering (SAXS). The combination of TGA and nitrogen sorption provides access to ionosilica and pore volume fractions, with contributions of meso- and macropores. We then elaborate an original and quantitative geometrical model to analyze the SAXS data based on small spheres (Rs = 1-2 nm) and cylinders (Lcyl = 10-20 nm) with radial polydispersity provided by the nitrogen sorption isotherms. As a result, we found that for a given incorporation of a templating IL, both synthesis pathways produce very similar pore geometries, but the better incorporation efficacy of the new hydrolytic films provides higher mesoporosity. Our combined study provides a coherent view of mesopore geometry, and thereby an optimization pathway of porous ionic membranes in terms of accessible mesoporosity contributing to the specific surface. Possible applications include electrolyte membranes with improved ionic properties, e.g., in fuel cells and batteries, as well as molecular storage.

3.
Development ; 148(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33531432

ABSTRACT

KIF2A is a kinesin motor protein with essential roles in neural progenitor division and axonal pruning during brain development. However, how different KIF2A alternative isoforms function during development of the cerebral cortex is not known. Here, we focus on three Kif2a isoforms expressed in the developing cortex. We show that Kif2a is essential for dendritic arborization in mice and that the functions of all three isoforms are sufficient for this process. Interestingly, only two of the isoforms can sustain radial migration of cortical neurons; a third isoform, lacking a key N-terminal region, is ineffective. By proximity-based interactome mapping for individual isoforms, we identify previously known KIF2A interactors, proteins localized to the mitotic spindle poles and, unexpectedly, also translation factors, ribonucleoproteins and proteins that are targeted to organelles, prominently to the mitochondria. In addition, we show that a KIF2A mutation, which causes brain malformations in humans, has extensive changes to its proximity-based interactome, with depletion of mitochondrial proteins identified in the wild-type KIF2A interactome. Our data raises new insights about the importance of alternative splice variants during brain development.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , Gene Expression Regulation , Kinesins/genetics , Neurons/cytology , Neurons/metabolism , Repressor Proteins/genetics , Alternative Splicing , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Expression Profiling , Kinesins/metabolism , Mice , Mutation , Neurogenesis/genetics , Proteomics/methods , RNA Isoforms , Repressor Proteins/metabolism
4.
Sci Rep ; 9(1): 19697, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873146

ABSTRACT

Excitatory neurons of the mammalian cerebral cortex are organized into six functional layers characterized by unique patterns of connectivity, as well as distinctive physiological and morphological properties. Cortical layers appear after a highly regulated migration process in which cells move from the deeper, proliferative zone toward the superficial layers. Importantly, defects in this radial migration process have been implicated in neurodevelopmental and psychiatric diseases. Here we report that during the final stages of migration, transcription factor Neurogenic Differentiation 2 (Neurod2) contributes to terminal cellular localization within the cortical plate. In mice, in utero knockdown of Neurod2 resulted in reduced numbers of neurons localized to the uppermost region of the developing cortex, also termed the primitive cortical zone. Our ChIP-Seq and RNA-Seq analyses of genes regulated by NEUROD2 in the developing cortex identified a number of key target genes with known roles in Reelin signaling, a critical regulator of neuronal migration. Our focused analysis of regulation of the Reln gene, encoding the extracellular ligand REELIN, uncovered NEUROD2 binding to conserved E-box elements in multiple introns. Furthermore, we demonstrate that knockdown of NEUROD2 in primary cortical neurons resulted in a strong increase in Reln gene expression at the mRNA level, as well as a slight upregulation at the protein level. These data reveal a new role for NEUROD2 during the late stages of neuronal migration, and our analysis of its genomic targets offers new genes with potential roles in cortical lamination.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Serine Endopeptidases/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dendrites/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Neurons/cytology , Neurons/metabolism , Neuropeptides/deficiency , Neuropeptides/genetics , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA-Seq , Reelin Protein
5.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28303257

ABSTRACT

Calcium signaling controls many key processes in neurons, including gene expression, axon guidance, and synaptic plasticity. In contrast to calcium influx through voltage- or neurotransmitter-gated channels, regulatory pathways that control store-operated calcium entry (SOCE) in neurons are poorly understood. Here, we report a transcriptional control of Stim1 (stromal interaction molecule 1) gene, which is a major sensor of endoplasmic reticulum (ER) calcium levels and a regulator of SOCE. By using a genome-wide chromatin immunoprecipitation and sequencing approach in mice, we find that NEUROD2, a neurogenic transcription factor, binds to an intronic element within the Stim1 gene. We show that NEUROD2 limits Stim1 expression in cortical neurons and consequently fine-tunes the SOCE response upon depletion of ER calcium. Our findings reveal a novel mechanism that regulates neuronal calcium homeostasis during cortical development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Animals, Newborn , Cell Cycle Proteins , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Conserved Sequence , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Introns , Mice, Inbred BALB C , Nuclear Proteins , Protein Binding , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL