Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.578
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(28): e2317711121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968101

RÉSUMÉ

Adult neural stem cells (NSCs) reside in the dentate gyrus of the hippocampus, and their capacity to generate neurons and glia plays a role in learning and memory. In addition, neurodegenerative diseases are known to be caused by a loss of neurons and glial cells, resulting in a need to better understand stem cell fate commitment processes. We previously showed that NSC fate commitment toward a neuronal or glial lineage is strongly influenced by extracellular matrix stiffness, a property of elastic materials. However, tissues in vivo are not purely elastic and have varying degrees of viscous character. Relatively little is known about how the viscoelastic properties of the substrate impact NSC fate commitment. Here, we introduce a polyacrylamide-based cell culture platform that incorporates mismatched DNA oligonucleotide-based cross-links as well as covalent cross-links. This platform allows for tunable viscous stress relaxation properties via variation in the number of mismatched base pairs. We find that NSCs exhibit increased astrocytic differentiation as the degree of stress relaxation is increased. Furthermore, culturing NSCs on increasingly stress-relaxing substrates impacts cytoskeletal dynamics by decreasing intracellular actin flow rates and stimulating cyclic activation of the mechanosensitive protein RhoA. Additionally, inhibition of motor-clutch model components such as myosin II and focal adhesion kinase partially or completely reverts cells to lineage distributions observed on elastic substrates. Collectively, our results introduce a unique system for controlling matrix stress relaxation properties and offer insight into how NSCs integrate viscoelastic cues to direct fate commitment.


Sujet(s)
Différenciation cellulaire , Cellules souches neurales , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Cellules souches neurales/physiologie , Animaux , Astrocytes/cytologie , Astrocytes/métabolisme , Astrocytes/physiologie , Souris , Résines acryliques/composition chimique , Protéine G RhoA/métabolisme , Cellules cultivées , Neurones/métabolisme , Neurones/physiologie , Neurones/cytologie , Matrice extracellulaire/métabolisme , Contrainte mécanique
2.
Tissue Eng Regen Med ; 21(6): 809-827, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39004636

RÉSUMÉ

BACKGROUND: In the intricate pathological milieu post-spinal cord injury (SCI), neural stem cells (NSCs) frequently differentiate into astrocytes rather than neurons, significantly limiting nerve repair. Hence, the utilization of biocompatible hydrogel scaffolds in conjunction with exogenous factors to foster the differentiation of NSCs into neurons has the potential for SCI repair. METHODS: In this study, we engineered a 3D-printed porous SilMA hydrogel scaffold (SM) supplemented with pH-/temperature-responsive paclitaxel nanoparticles (PTX-NPs). We analyzed the biocompatibility of a specific concentration of PTX-NPs and its effect on NSC differentiation. We also established an SCI model to explore the ability of composite scaffolds for in vivo nerve repair. RESULTS: The physical adsorption of an optimal PTX-NPs dosage can simultaneously achieve pH/temperature-responsive release and commendable biocompatibility, primarily reflected in cell viability, morphology, and proliferation. An appropriate PTX-NPs concentration can steer NSC differentiation towards neurons over astrocytes, a phenomenon that is also efficacious in simulated injury settings. Immunoblotting analysis confirmed that PTX-NPs-induced NSC differentiation occurred via the MAPK/ERK signaling cascade. The repair of hemisected SCI in rats demonstrated that the composite scaffold augmented neuronal regeneration at the injury site, curtailed astrocyte and fibrotic scar production, and enhanced motor function recovery in rat hind limbs. CONCLUSION: The scaffold's porous architecture serves as a cellular and drug carrier, providing a favorable microenvironment for nerve regeneration. These findings corroborate that this strategy amplifies neuronal expression within the injury milieu, significantly aiding in SCI repair.


Sujet(s)
Différenciation cellulaire , Hydrogels , Nanoparticules , Cellules souches neurales , Neurones , Paclitaxel , Rat Sprague-Dawley , Traumatismes de la moelle épinière , Structures d'échafaudage tissulaires , Traumatismes de la moelle épinière/traitement médicamenteux , Animaux , Paclitaxel/pharmacologie , Structures d'échafaudage tissulaires/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/cytologie , Neurones/métabolisme , Nanoparticules/composition chimique , Hydrogels/composition chimique , Hydrogels/pharmacologie , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Porosité , Rats , Régénération nerveuse/effets des médicaments et des substances chimiques , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/cytologie
3.
Cells ; 13(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891034

RÉSUMÉ

Astrocytes, the most abundant glial cell type in the brain, play crucial roles in maintaining homeostasis within the central nervous system (CNS). Impairment or abnormalities of typical astrocyte functions in the CNS serve as a causative or contributing factor in numerous neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Currently, disease-modeling and drug-screening approaches, primarily focused on human astrocytes, rely on human pluripotent stem cell (hPSC)-derived astrocytes. However, it is important to acknowledge that these hPSC-derived astrocytes exhibit notable differences across studies and when compared to their in vivo counterparts. These differences may potentially compromise translational outcomes if not carefully accounted for. This review aims to explore state-of-the-art in vitro models of human astrocyte development, focusing on the developmental processes, functional maturity, and technical aspects of various hPSC-derived astrocyte differentiation protocols. Additionally, it summarizes their successful application in modeling neurological disorders. The discussion extends to recent advancements in the large-scale production of human astrocytes and their application in developing high-throughput assays conducive to therapeutic drug discovery.


Sujet(s)
Astrocytes , Différenciation cellulaire , Maladies du système nerveux , Cellules souches pluripotentes , Humains , Astrocytes/métabolisme , Astrocytes/cytologie , Maladies du système nerveux/anatomopathologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , , Animaux
4.
Cells ; 13(11)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38891101

RÉSUMÉ

Astrocytes are ubiquitous in the brain and spinal cord and display a complex morphology important for the local interactions with neighboring cells, resulting in the modulation of circuit function. Thus, studies focusing on astrocyte physiology in the healthy and diseased brain generally present analyses of astrocytic structure. The labeling method used to visualize the astrocytic structure defines the morphological level to observe and may vary depending on the anatomical sub-regions. The method choice may significantly affect our understanding of their structural diversity. The main goal of this work was to identify a straightforward and efficient protocol for labeling and reconstructing a detailed astrocytic structure to apply and validate in different brain tissue preparations across laboratories. For that, we explored different tissue processing protocols before GFAP labeling to determine the most effective method for reconstructing astrocytic backbones in the mouse hippocampus. Our results show that the reconstruction of astrocytic structure in vibratome sections labeled by free-floating immunofluorescence protocol provides a more practical method to achieve a higher level of detail and arbor complexity in astrocyte backbone reconstruction. Free-floating immunofluorescence labeling is the most reliable method for obtaining better antibody penetration and more detailed astrocyte structure. Finally, we also show that introducing an antigen retrieval step appears useful for visualizing more complete structural details.


Sujet(s)
Astrocytes , Astrocytes/métabolisme , Astrocytes/cytologie , Animaux , Souris , Hippocampe/cytologie , Protéine gliofibrillaire acide/métabolisme , Souris de lignée C57BL , Mâle , Coloration et marquage/méthodes
5.
Fly (Austin) ; 18(1): 2368336, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38884422

RÉSUMÉ

The Drosophila melanogaster brain is a complex organ with various cell types, orchestrating the development, physiology, and behaviors of the fly. While each cell type in Drosophila brain is known to express a unique gene set, their complete genetic profile is still unknown. Advances in the RNA sequencing techniques at single-cell resolution facilitate identifying novel cell type markers and/or re-examining the specificity of the available ones. In this study, exploiting a single-cell RNA sequencing data of Drosophila optic lobe, we categorized the cells based on their expression pattern for known markers, then the genes with enriched expression in astrocytes were identified. CG11000 was identified as a gene with a comparable expression profile to the Eaat1 gene, an astrocyte marker, in every individual cell inside the Drosophila optic lobe and midbrain, as well as in the entire Drosophila brain throughout its development. Consistent with our bioinformatics data, immunostaining of the brains dissected from transgenic adult flies showed co-expression of CG11000 with Eaat1 in a set of single cells corresponding to the astrocytes in the Drosophila brain. Physiologically, inhibiting CG11000 through RNA interference disrupted the normal development of male D. melanogaster, while having no impact on females. Expression suppression of CG11000 in adult flies led to decreased locomotion activity and also shortened lifespan specifically in astrocytes, indicating the gene's significance in astrocytes. We designated this gene as 'deathstar' due to its crucial role in maintaining the star-like shape of glial cells, astrocytes, throughout their development into adult stage.


Sujet(s)
Astrocytes , Protéines de Drosophila , Drosophila melanogaster , Locomotion , Longévité , Animaux , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Drosophila melanogaster/croissance et développement , Drosophila melanogaster/physiologie , Astrocytes/métabolisme , Astrocytes/cytologie , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Longévité/génétique , Transporteur-1 d'acides aminés excitateurs/métabolisme , Transporteur-1 d'acides aminés excitateurs/génétique , Mâle , Femelle , Encéphale/métabolisme , Encéphale/cytologie , Encéphale/croissance et développement
6.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38927995

RÉSUMÉ

Neural precursor cells (NPCs) that persist in the postnatal/adult subventricular zone (SVZ) express connexins that form hemichannels and gap junctions. Gap junctional communication plays a role in NPC proliferation and differentiation during development, but its relevance on postnatal age remains to be elucidated. In this work we aimed to evaluate the effect of the blockade of gap junctional communication on proliferation and cell fate of NPCs obtained from the SVZ of postnatal rats. NPCs were isolated and expanded in culture as neurospheres. Electron microscopy revealed the existence of gap junctions among neurosphere cells. Treatment of cultures with octanol, a broad-spectrum gap junction blocker, or with Gap27, a specific blocker for gap junctions formed by connexin43, produced a significant decrease in bromodeoxyuridine incorporation. Octanol treatment also exerted a dose-dependent antiproliferative effect on glioblastoma cells. To analyze possible actions on NPC fate, cells were seeded in the absence of mitogens. Treatment with octanol led to an increase in the percentage of astrocytes and oligodendrocyte precursors, whereas the percentage of neurons remained unchanged. Gap27 treatment, in contrast, did not modify the differentiation pattern of SVZ NPCs. Our results indicate that general blockade of gap junctions with octanol induces significant effects on the behavior of postnatal SVZ NPCs, by reducing proliferation and promoting glial differentiation.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Jonctions communicantes , Cellules souches neurales , Névroglie , Octanols , Animaux , Jonctions communicantes/effets des médicaments et des substances chimiques , Jonctions communicantes/métabolisme , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Rats , Octanols/pharmacologie , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Névroglie/cytologie , Cellules cultivées , Ventricules latéraux/cytologie , Ventricules latéraux/métabolisme , Ventricules latéraux/effets des médicaments et des substances chimiques , Connexine 43/métabolisme , Rat Wistar , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/cytologie , Animaux nouveau-nés , Humains
7.
Stem Cell Reports ; 19(7): 946-956, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38876110

RÉSUMÉ

Functionality of the blood-brain barrier (BBB) relies on the interaction between endothelial cells (ECs), pericytes, and astrocytes to regulate molecule transport within the central nervous system. Most experimental models for the BBB rely on freshly isolated primary brain cells. Here, we explored human induced pluripotent stem cells (hiPSCs) as a cellular source for astrocytes in a 3D vessel-on-chip (VoC) model. Self-organized microvascular networks were formed by combining hiPSC-derived ECs, human brain vascular pericytes, and hiPSC-derived astrocytes within a fibrin hydrogel. The hiPSC-ECs and pericytes showed close interactions, but, somewhat unexpectedly, addition of astrocytes disrupted microvascular network formation. However, continuous fluid perfusion or activation of cyclic AMP (cAMP) signaling rescued the vascular organization and decreased vascular permeability. Nevertheless, astrocytes did not affect the expression of proteins related to junction formation, transport, or extracellular matrix, indicating that, despite other claims, hiPSC-derived ECs do not entirely acquire a BBB-like identity in the 3D VoC model.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Cellules endothéliales , Cellules souches pluripotentes induites , Astrocytes/métabolisme , Astrocytes/cytologie , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/cytologie , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Péricytes/cytologie , Péricytes/métabolisme , Différenciation cellulaire , Laboratoires sur puces , Cellules cultivées , Hydrogels , AMP cyclique/métabolisme , Modèles biologiques
8.
PLoS One ; 19(6): e0303901, 2024.
Article de Anglais | MEDLINE | ID: mdl-38917115

RÉSUMÉ

Human induced pluripotent stem cells (hiPSCs) derived into neurons offer a powerful in vitro model to study cellular processes. One method to characterize functional network properties of these cells is using multielectrode arrays (MEAs). MEAs can measure the electrophysiological activity of cellular cultures for extended periods of time without disruption. Here we used WTC11 hiPSCs with a doxycycline-inducible neurogenin 2 (NGN2) transgene differentiated into neurons co-cultured with primary human astrocytes. We achieved a synchrony index ∼0.9 in as little as six-weeks with a mean firing rate of ∼13 Hz. Previous reports show that derived 3D brain organoids can take several months to achieve similar strong network burst synchrony. We also used this co-culture to model aspects of blood-brain barrier breakdown by using human serum. Our fully human co-culture achieved strong network burst synchrony in a fraction of the time of previous reports, making it an excellent first pass, high-throughput method for studying network properties and neurodegenerative diseases.


Sujet(s)
Astrocytes , Différenciation cellulaire , Techniques de coculture , Cellules souches pluripotentes induites , Neurones , Humains , Astrocytes/cytologie , Astrocytes/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Techniques de coculture/méthodes , Neurones/cytologie , Neurones/métabolisme , Cellules cultivées , Protéines de tissu nerveux/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Électrodes , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/cytologie
9.
Glia ; 72(9): 1693-1706, 2024 09.
Article de Anglais | MEDLINE | ID: mdl-38852127

RÉSUMÉ

Astrocytes that reside in superficial (SL) and deep cortical layers have distinct molecular profiles and morphologies, which may underlie specific functions. Here, we demonstrate that the production of SL and deep layer (DL) astrocyte populations from neural progenitor cells in the mouse is temporally regulated. Lineage tracking following in utero and postnatal electroporation with PiggyBac (PB) EGFP and birth dating with EdU and FlashTag, showed that apical progenitors produce astrocytes during late embryogenesis (E16.5) that are biased to the SL, while postnatally labeled (P0) astrocytes are biased to the DL. In contrast, astrocytes born during the predominantly neurogenic window (E14.5) showed a random distribution in the SL and DL. Of interest, E13.5 astrocytes birth dated at E13.5 with EdU showed a lower layer bias, while FT labeling of apical progenitors showed no bias. Finally, examination of the morphologies of "biased" E16.5- and P0-labeled astrocytes demonstrated that E16.5-labeled astrocytes exhibit different morphologies in different layers, while P0-labeled astrocytes do not. Differences based on time of birth are also observed in the molecular profiles of E16.5 versus P0-labeled astrocytes. Altogether, these results suggest that the morphological, molecular, and positional diversity of cortical astrocytes is related to their time of birth from ventricular/subventricular zone progenitors.


Sujet(s)
Astrocytes , Cortex cérébral , Cellules souches neurales , Animaux , Astrocytes/métabolisme , Astrocytes/cytologie , Cortex cérébral/cytologie , Cortex cérébral/métabolisme , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Souris , Souris transgéniques , Femelle , Animaux nouveau-nés , Régulation de l'expression des gènes au cours du développement , Transcriptome , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Ventricules cérébraux/cytologie , Souris de lignée C57BL
10.
J Biol Chem ; 300(7): 107477, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38879014

RÉSUMÉ

Thyroid hormone (TH) is a critical regulator of cellular function and cell fate. The circulating TH level is relatively stable, while tissue TH action fluctuates according to cell type-specific mechanisms. Here, we focused on identifying mechanisms that regulate TH action through the type 2 deiodinase (D2) in glial cells. Dio2 mRNA has an unusually long 3'UTR where we identified multiple putative MSI1 binding sites for Musashi-1 (MSI1), a highly conserved RNA-binding cell cycle regulator. Binding to these sites was confirmed through electrophoretic mobility shift assay. In H4 glioma cells, shRNA-mediated MSI1 knockdown increased endogenous D2 activity, whereas MSI1 overexpression in HEK293T cells decreased D2 expression. This latter effect could be prevented by the deletion of a 3.6 kb region of the 3'UTR of Dio2 mRNA containing MSI1 binding sites. MSI1 immunoreactivity was observed in 2 mouse Dio2-expressing cell types, that is, cortical astrocytes and hypothalamic tanycytes, establishing the anatomical basis for a potential in vivo interaction of Dio2 mRNA and MSl1. Indeed, increased D2 expression was observed in the cortex of mice lacking MSI1 protein. Furthermore, MSI1 knockdown-induced D2 expression slowed down cell proliferation by 56% in primary cultures of mouse cortical astrocytes, establishing the functionality of the MSI1-D2-T3 pathway. In summary, Dio2 mRNA is a target of MSI1 and the MSI1-D2-T3 pathway is a novel regulatory mechanism of astrocyte proliferation with the potential to regulate the pathogenesis of human glioblastoma.


Sujet(s)
Astrocytes , Prolifération cellulaire , Iodide peroxidase , , Protéines de tissu nerveux , Protéines de liaison à l'ARN , Animaux , Iodide peroxidase/métabolisme , Iodide peroxidase/génétique , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Astrocytes/métabolisme , Astrocytes/cytologie , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Cellules HEK293 , Régions 3' non traduites , Souris knockout , Lignée cellulaire tumorale , Hormones thyroïdiennes/métabolisme , Hormones thyroïdiennes/génétique
11.
Sci Signal ; 17(838): eadq5728, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38805584
12.
J Cell Sci ; 137(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38813860

RÉSUMÉ

WW domain-containing transcription regulator 1 (WWTR1, referred to here as TAZ) and Yes-associated protein (YAP, also known as YAP1) are transcriptional co-activators traditionally studied together as a part of the Hippo pathway, and are best known for their roles in stem cell proliferation and differentiation. Despite their similarities, TAZ and YAP can exert divergent cellular effects by differentially interacting with other signaling pathways that regulate stem cell maintenance or differentiation. In this study, we show in mouse neural stem and progenitor cells (NPCs) that TAZ regulates astrocytic differentiation and maturation, and that TAZ mediates some, but not all, of the effects of bone morphogenetic protein (BMP) signaling on astrocytic development. By contrast, both TAZ and YAP mediate the effects on NPC fate of ß1-integrin (ITGB1) and integrin-linked kinase signaling, and these effects are dependent on extracellular matrix cues. These findings demonstrate that TAZ and YAP perform divergent functions in the regulation of astrocyte differentiation, where YAP regulates cell cycle states of astrocytic progenitors and TAZ regulates differentiation and maturation from astrocytic progenitors into astrocytes.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Astrocytes , Différenciation cellulaire , Prolifération cellulaire , Cellules souches neurales , Transduction du signal , Transactivateurs , Transcriptional coactivator with PDZ-binding motif proteins , Protéines de signalisation YAP , Animaux , Astrocytes/métabolisme , Astrocytes/cytologie , Protéines de signalisation YAP/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Souris , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Transactivateurs/métabolisme , Transactivateurs/génétique , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Antigènes CD29/métabolisme , Antigènes CD29/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines morphogénétiques osseuses/métabolisme , Cortex cérébral/cytologie , Cortex cérébral/métabolisme , Protein-Serine-Threonine Kinases
13.
Biomed Mater ; 19(4)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38772389

RÉSUMÉ

The extracellular matrix plays a crucial role in the growth of human neural stem cells (hNSCs) by forming a stem cell niche, bothin vitroandin vivo. The demand for defined synthetic substrates has been increasing recently in stem cell research, reflecting the requirements for precise functions and safety concerns in potential clinical approaches. In this study, we tested the adhesion and expansion of one of the most representative hNSC lines, the ReNcell VM Human Neural Progenitor Cell Line, in a pure-synthesized short peptide-basedin vitroniche using a previously established integrin-binding peptide array. Spontaneous cell differentiation was then induced using two differentin vitroapproaches to further confirm the multipotent features of cells treated with the peptides. Twelve different integrin-binding peptides were capable of supporting hNSC adhesion and expansion at varied proliferation rates. In the ReNcell medium-based differentiation approach, cells detached in almost all peptide-based groups, except integrinα5ß1 binding peptide. In an altered differentiation process induced by retinoic acid containing neural differentiation medium, cell adhesion was retained in all 12 peptide groups. These peptides also appeared to have varied effects on the differentiation potential of hNSCs towards neurons and astrocytes. Our findings provide abundant options for the development ofin vitroneural stem cell niches and will help develop promising tools for disease modeling and future stem cell therapies for neurological diseases.


Sujet(s)
Adhérence cellulaire , Différenciation cellulaire , Prolifération cellulaire , Intégrines , Cellules souches neurales , Peptides , Humains , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Adhérence cellulaire/effets des médicaments et des substances chimiques , Peptides/composition chimique , Peptides/pharmacologie , Intégrines/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Matrice extracellulaire/métabolisme , Neurones/métabolisme , Neurones/cytologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Trétinoïne/pharmacologie , Propriétés de surface , Astrocytes/métabolisme , Astrocytes/cytologie
14.
Mol Brain ; 17(1): 25, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773624

RÉSUMÉ

A growing body of evidence indicates intra- and inter-regional heterogeneity of astrocytes in the brain. However, because of a lack of an efficient method for isolating astrocytes from the spinal cord, little is known about how much spinal cord astrocytes are heterogeneous in adult mice. In this study, we developed a new method for isolating spinal astrocytes from adult mice using a cold-active protease from Bacillus licheniformis with an astrocyte cell surface antigen-2 (ACSA-2) antibody. Using fluorescence-activated cell sorting, isolated spinal ACSA-2+ cells were divided into two distinct populations, ACSA-2high and ACSA-2low. By analyzing the expression of cell-type marker genes, the ACSA-2high and ACSA-2low populations were identified as astrocytes and ependymal cells, respectively. Furthermore, ACSA-2high cells had mRNAs encoding genes that were abundantly expressed in the gray matter (GM) but not white matter astrocytes. By optimizing enzymatic isolation procedures, the yield of GM astrocytes also increased. Therefore, our newly established method enabled the selective and efficient isolation of GM astrocytes from the spinal cord of adult mice and may be useful for bulk- or single-cell RNA-sequencing under physiological and pathological conditions.


Sujet(s)
Astrocytes , Séparation cellulaire , Substance grise , Moelle spinale , Animaux , Astrocytes/métabolisme , Astrocytes/cytologie , Moelle spinale/cytologie , Séparation cellulaire/méthodes , Souris de lignée C57BL , Souris , Mâle , ARN messager/métabolisme , ARN messager/génétique , Vieillissement
15.
Adv Mater ; 36(26): e2312497, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38610101

RÉSUMÉ

This work introduces NeoMag, a system designed to enhance cell mechanics assays in substrate deformation studies. NeoMag uses multidomain magneto-active materials to mechanically actuate the substrate, transmitting reversible mechanical cues to cells. The system boasts full flexibility in alternating loading substrate deformation modes, seamlessly adapting to both upright and inverted microscopes. The multidomain substrates facilitate mechanobiology assays on 2D and 3D cultures. The integration of the system with nanoindenters allows for precise evaluation of cellular mechanical properties under varying substrate deformation modes. The system is used to study the impact of substrate deformation on astrocytes, simulating mechanical conditions akin to traumatic brain injury and ischemic stroke. The results reveal local heterogeneous changes in astrocyte stiffness, influenced by the orientation of subcellular regions relative to substrate strain. These stiffness variations, exceeding 50% in stiffening and softening, and local deformations significantly alter calcium dynamics. Furthermore, sustained deformations induce actin network reorganization and activate Piezo1 channels, leading to an initial increase followed by a long-term inhibition of calcium events. Conversely, fast and dynamic deformations transiently activate Piezo1 channels and disrupt the actin network, causing long-term cell softening. These findings unveil mechanical and functional alterations in astrocytes during substrate deformation, illustrating the multiple opportunities this technology offers.


Sujet(s)
Astrocytes , Astrocytes/métabolisme , Astrocytes/cytologie , Animaux , Calcium/métabolisme , Calcium/composition chimique , Phénomènes biomécaniques , Phénomènes mécaniques , Actines/métabolisme , Canaux ioniques/métabolisme , Souris
16.
J Cell Sci ; 137(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38639242

RÉSUMÉ

WW domain-containing transcription regulator 1 (WWTR1, referred to here as TAZ) and Yes-associated protein (YAP, also known as YAP1) are transcriptional co-activators traditionally studied together as a part of the Hippo pathway, and are best known for their roles in stem cell proliferation and differentiation. Despite their similarities, TAZ and YAP can exert divergent cellular effects by differentially interacting with other signaling pathways that regulate stem cell maintenance or differentiation. In this study, we show in mouse neural stem and progenitor cells (NPCs) that TAZ regulates astrocytic differentiation and maturation, and that TAZ mediates some, but not all, of the effects of bone morphogenetic protein (BMP) signaling on astrocytic development. By contrast, both TAZ and YAP mediate the effects on NPC fate of ß1-integrin (ITGB1) and integrin-linked kinase signaling, and these effects are dependent on extracellular matrix cues. These findings demonstrate that TAZ and YAP perform divergent functions in the regulation of astrocyte differentiation, where YAP regulates cell cycle states of astrocytic progenitors and TAZ regulates differentiation and maturation from astrocytic progenitors into astrocytes.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Astrocytes , Différenciation cellulaire , Cortex cérébral , Protéines de signalisation YAP , Animaux , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Astrocytes/métabolisme , Astrocytes/cytologie , Protéines morphogénétiques osseuses/métabolisme , Différenciation cellulaire/génétique , Prolifération cellulaire , Cortex cérébral/cytologie , Cortex cérébral/métabolisme , Antigènes CD29/métabolisme , Antigènes CD29/génétique , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Protein-Serine-Threonine Kinases , Transduction du signal , Protéines de signalisation YAP/métabolisme
17.
Nanoscale ; 16(19): 9576-9582, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38682293

RÉSUMÉ

Aquaporin-4 (AQP4) facilitates water transport across astrocytic membranes in the brain, forming highly structured nanometric arrays. AQP4 has a central role in regulating cerebrospinal fluid (CSF) circulation and facilitating the clearance of solutes from the extracellular space of the brain. Adrenergic signaling has been shown to modulate the volume of the extracellular space of the brain via AQP4 localized at the end-feet of astrocytes, but the mechanisms by which AQP4 regulates CSF inflow and outflow in the brain remain elusive. Using advanced imaging techniques, including super-resolution microscopy and single-molecule tracking, we investigated the hypothesis that ß-adrenergic receptor activation induces cellular changes that regulate AQP4 array size and mobility, thus influencing water transport in the brain. We report that the ß-adrenergic agonist, isoproterenol hydrochloride, decreases AQP4 array size and enhances its membrane mobility, while hyperosmotic conditions induce the formation of larger, less mobile arrays. These findings reveal that AQP4 arrays are dynamic structures, responsive to adrenergic signals and osmotic changes, highlighting a novel regulatory mechanism of water transport in the brain. Our results provide insights into the molecular control of CSF circulation and extracellular brain space volume, laying the groundwork for understanding the relationship between astrocyte water transport, sleep physiology, and neurodegeneration.


Sujet(s)
Aquaporine-4 , Astrocytes , Isoprénaline , Imagerie de molécules uniques , Aquaporine-4/métabolisme , Astrocytes/métabolisme , Astrocytes/cytologie , Animaux , Isoprénaline/pharmacologie , Souris , Eau/composition chimique , Eau/métabolisme , Cellules cultivées , Récepteurs bêta-adrénergiques/métabolisme , Agonistes bêta-adrénergiques/pharmacologie , Encéphale/métabolisme
18.
Sci Rep ; 14(1): 9846, 2024 04 29.
Article de Anglais | MEDLINE | ID: mdl-38684715

RÉSUMÉ

Astrocytes are glycolytically active cells in the central nervous system playing a crucial role in various brain processes from homeostasis to neurotransmission. Astrocytes possess a complex branched morphology, frequently examined by fluorescent microscopy. However, staining and fixation may impact the properties of astrocytes, thereby affecting the accuracy of the experimental data of astrocytes dynamics and morphology. On the other hand, phase contrast microscopy can be used to study astrocytes morphology without affecting them, but the post-processing of the resulting low-contrast images is challenging. The main result of this work is a novel approach for recognition and morphological analysis of unstained astrocytes based on machine-learning recognition of microscopic images. We conducted a series of experiments involving the cultivation of isolated astrocytes from the rat brain cortex followed by microscopy. Using the proposed approach, we tracked the temporal evolution of the average total length of branches, branching, and area per astrocyte in our experiments. We believe that the proposed approach and the obtained experimental data will be of interest and benefit to the scientific communities in cell biology, biophysics, and machine learning.


Sujet(s)
Astrocytes , Apprentissage machine , Microscopie de contraste de phase , Astrocytes/cytologie , Animaux , Microscopie de contraste de phase/méthodes , Rats , Cellules cultivées , Traitement d'image par ordinateur/méthodes , Cortex cérébral/cytologie
19.
Nature ; 629(8010): 146-153, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38632406

RÉSUMÉ

Astrocytes, the most abundant non-neuronal cell type in the mammalian brain, are crucial circuit components that respond to and modulate neuronal activity through calcium (Ca2+) signalling1-7. Astrocyte Ca2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales-from fast, subcellular activity3,4 to slow, synchronized activity across connected astrocyte networks8-10-to influence many processes5,7,11. However, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon astrocyte imaging while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca2+ activity-propagative activity-differentiates astrocyte network responses to these two main neurotransmitters, and may influence responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over a minutes-long time course, contributing to accumulating evidence that substantial astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales12-14. These findings will enable future studies to investigate the link between specific astrocyte Ca2+ activity and specific functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.


Sujet(s)
Astrocytes , Cortex cérébral , Acide glutamique , Réseau nerveux , Agents neuromédiateurs , Acide gamma-amino-butyrique , Animaux , Femelle , Mâle , Souris , Astrocytes/métabolisme , Astrocytes/cytologie , Calcium/métabolisme , Signalisation calcique , Communication cellulaire , Cortex cérébral/cytologie , Cortex cérébral/métabolisme , Acide gamma-amino-butyrique/métabolisme , Acide glutamique/métabolisme , Souris de lignée C57BL , Réseau nerveux/cytologie , Réseau nerveux/métabolisme , Neurones/métabolisme , Agents neuromédiateurs/métabolisme , Facteurs temps
20.
Bioorg Chem ; 147: 107365, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636436

RÉSUMÉ

Protein prenylation is one example of a broad class of post-translational modifications where proteins are covalently linked to various hydrophobic moieties. To globally identify and monitor levels of all prenylated proteins in a cell simultaneously, our laboratory and others have developed chemical proteomic approaches that rely on the metabolic incorporation of isoprenoid analogues bearing bio-orthogonal functionality followed by enrichment and subsequent quantitative proteomic analysis. Here, several improvements in the synthesis of the alkyne-containing isoprenoid analogue C15AlkOPP are reported to improve synthetic efficiency. Next, metabolic labeling with C15AlkOPP was optimized to obtain useful levels of metabolic incorporation of the probe in several types of primary cells. Those conditions were then used to study the prenylomes of motor neurons (ES-MNs), astrocytes (ES-As), and their embryonic stem cell progenitors (ESCs), which allowed for the identification of 54 prenylated proteins from ESCs, 50 from ES-MNs, and 84 from ES-As, representing all types of prenylation. Bioinformatic analysis revealed specific enriched pathways, including nervous system development, chemokine signaling, Rho GTPase signaling, and adhesion. Hierarchical clustering showed that most enriched pathways in all three cell types are related to GTPase activity and vesicular transport. In contrast, STRING analysis showed significant interactions in two populations that appear to be cell type dependent. The data provided herein demonstrates that robust incorporation of C15AlkOPP can be obtained in ES-MNs and related primary cells purified via magnetic-activated cell sorting allowing the identification and quantification of numerous prenylated proteins. These results suggest that metabolic labeling with C15AlkOPP should be an effective approach for investigating the role of prenylated proteins in primary cells in both normal cells and disease pathologies, including ALS.


Sujet(s)
Alcynes , Astrocytes , Motoneurones , Prénylation des protéines , Astrocytes/métabolisme , Astrocytes/cytologie , Animaux , Alcynes/composition chimique , Alcynes/synthèse chimique , Motoneurones/métabolisme , Motoneurones/cytologie , Terpènes/composition chimique , Terpènes/synthèse chimique , Terpènes/métabolisme , Souris , Structure moléculaire , Cellules cultivées
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE