Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 40.586
Filtrer
1.
J Neuroinflammation ; 21(1): 169, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961424

RÉSUMÉ

BACKGROUND: Understanding the mechanism behind sepsis-associated encephalopathy (SAE) remains a formidable task. This study endeavors to shed light on the complex cellular and molecular alterations that occur in the brains of a mouse model with SAE, ultimately unraveling the underlying mechanisms of this condition. METHODS: We established a murine model using intraperitoneal injection of lipopolysaccharide (LPS) in wild type and Anxa1-/- mice and collected brain tissues for analysis at 0-hour, 12-hour, 24-hour, and 72-hour post-injection. Utilizing advanced techniques such as single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq, we conducted a comprehensive characterization of the cellular responses and molecular patterns within the brain. RESULTS: Our study uncovered notable temporal differences in the response to LPS challenge between Anxa1-/- (annexin A1 knockout) and wild type mice, specifically at the 12-hour and 24-hour time points following injection. We observed a significant increase in the proportion of Astro-2 and Micro-2 cells in these mice. These cells exhibited a colocalization pattern with the vascular subtype Vas-1, forming a distinct region known as V1A2M2, where Astro-2 and Micro-2 cells surrounded Vas-1. Moreover, through further analysis, we discovered significant upregulation of ligands and receptors such as Timp1-Cd63, Timp1-Itgb1, Timp1-Lrp1, as well as Ccl2-Ackr1 and Cxcl2-Ackr1 within this region. In addition, we observed a notable increase in the expression of Cd14-Itgb1, Cd14-Tlr2, and Cd14-C3ar1 in regions enriched with Micro-2 cells. Additionally, Cxcl10-Sdc4 showed broad upregulation in brain regions containing both Micro-2 and Astro-2 cells. Notably, upon LPS challenge, there was an observed increase in Anxa1 expression in the mouse brain. Furthermore, our study revealed a noteworthy increase in mortality rates following Anxa1 knockdown. However, we did not observe substantial differences in the types, numbers, or distribution of other brain cells between Anxa1-/- and wildtype mice over time. Nevertheless, when comparing the 24-hour post LPS injection time point, we observed a significant decrease in the proportion and distribution of Micro-2 and Astro-2 cells in the vicinity of blood vessels in Anxa1-/- mice. Additionally, we noted reduced expression levels of several ligand-receptor pairs including Cd14-Tlr2, Cd14-C3ar1, Cd14-Itgb1, Cxcl10-Sdc4, Ccl2-Ackr1, and Cxcl2-Ackr1. CONCLUSIONS: By combining snRNA-seq and Stereo-seq techniques, our study successfully identified a distinctive cellular colocalization, referred to as a special pathological niche, comprising Astro-2, Micro-2, and Vas-1 cells. Furthermore, we observed an upregulation of ligand-receptor pairs within this niche. These findings suggest a potential association between this cellular arrangement and the underlying mechanisms contributing to SAE or the increased mortality observed in Anxa1 knockdown mice.


Sujet(s)
Astrocytes , Encéphale , Modèles animaux de maladie humaine , Lipopolysaccharides , Souris knockout , Microglie , Encéphalopathie associée au sepsis , Animaux , Souris , Lipopolysaccharides/toxicité , Encéphalopathie associée au sepsis/anatomopathologie , Encéphalopathie associée au sepsis/génétique , Encéphalopathie associée au sepsis/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Analyse de séquence d'ARN/méthodes , Souris de lignée C57BL , Transcriptome , Mâle
2.
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
3.
Biochemistry (Mosc) ; 89(6): 1045-1060, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38981700

RÉSUMÉ

Astrocytic NMDA receptors (NMDARs) are heterotetramers, whose expression and properties are largely determined by their subunit composition. Astrocytic NMDARs are characterized by a low sensitivity to magnesium ions and low calcium conductivity. Their activation plays an important role in the regulation of various intracellular processes, such as gene expression and mitochondrial function. Astrocytic NMDARs are involved in calcium signaling in astrocytes and can act through the ionotropic and metabotropic pathways. Astrocytic NMDARs participate in the interactions of the neuroglia, thus affecting synaptic plasticity. They are also engaged in the astrocyte-vascular interactions and contribute to the regulation of vascular tone. Astrocytic NMDARs are involved in various pathologies, such as ischemia and hyperammonemia, and their blockade prevents negative changes in astrocytes during these diseases.


Sujet(s)
Astrocytes , Récepteurs du N-méthyl-D-aspartate , Récepteurs du N-méthyl-D-aspartate/métabolisme , Astrocytes/métabolisme , Humains , Animaux , Signalisation calcique , Plasticité neuronale
4.
Cereb Cortex ; 34(7)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38981852

RÉSUMÉ

Previously, we found that dCA1 A1-like polarization of astrocytes contributes a lot to the spatial memory deficit in methamphetamine abstinence mice. However, the underlying mechanism remains unclear, resulting in a lack of promising therapeutic targets. Here, we found that methamphetamine abstinence mice exhibited an increased M1-like microglia and A1-like astrocytes, together with elevated levels of interleukin 1α and tumor necrosis factor α in dCA1. In vitro, the M1-like BV2 microglia cell medium, containing high levels of Interleukin 1α and tumor necrosis factor α, elevated A1-like polarization of astrocytes, which weakened their capacity for glutamate clearance. Locally suppressing dCA1 M1-like microglia activation with minocycline administration attenuated A1-like polarization of astrocytes, ameliorated dCA1 neurotoxicity, and, most importantly, rescued spatial memory in methamphetamine abstinence mice. The effective time window of minocycline treatment on spatial memory is the methamphetamine exposure period, rather than the long-term methamphetamine abstinence.


Sujet(s)
Astrocytes , Troubles de la mémoire , Métamfétamine , Microglie , Minocycline , Mémoire spatiale , Animaux , Métamfétamine/toxicité , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Souris , Troubles de la mémoire/induit chimiquement , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/anatomopathologie , Mémoire spatiale/physiologie , Mémoire spatiale/effets des médicaments et des substances chimiques , Mâle , Minocycline/pharmacologie , Souris de lignée C57BL , Syndrome de sevrage/métabolisme , Syndrome de sevrage/anatomopathologie , Stimulants du système nerveux central/toxicité
5.
Front Immunol ; 15: 1427200, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989284

RÉSUMÉ

Introduction: Glioma, a prevalent and deadly brain tumor, is marked by significant cellular heterogeneity and metabolic alterations. However, the comprehensive cell-of-origin and metabolic landscape in high-grade (Glioblastoma Multiforme, WHO grade IV) and low-grade (Oligoastrocytoma, WHO grade II) gliomas remains elusive. Methods: In this study, we undertook single-cell transcriptome sequencing of these glioma grades to elucidate their cellular and metabolic distinctions. Following the identification of cell types, we compared metabolic pathway activities and gene expressions between high-grade and low-grade gliomas. Results: Notably, astrocytes and oligodendrocyte progenitor cells (OPCs) exhibited the most substantial differences in both metabolic pathways and gene expression, indicative of their distinct origins. The comprehensive analysis identified the most altered metabolic pathways (MCPs) and genes across all cell types, which were further validated against TCGA and CGGA datasets for clinical relevance. Discussion: Crucially, the metabolic enzyme phosphodiesterase 8B (PDE8B) was found to be exclusively expressed and progressively downregulated in astrocytes and OPCs in higher-grade gliomas. This decreased expression identifies PDE8B as a metabolism-related oncogene in IDH-mutant glioma, marking its dual role as both a protective marker for glioma grading and prognosis and as a facilitator in glioma progression.


Sujet(s)
Tumeurs du cerveau , Analyse de profil d'expression de gènes , Gliome , Isocitrate dehydrogenases , Mutation , Analyse sur cellule unique , Humains , Isocitrate dehydrogenases/génétique , Gliome/génétique , Gliome/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Régulation de l'expression des gènes tumoraux , Transcriptome , Astrocytes/métabolisme , Oncogènes , Régulation négative , Précurseurs des oligodendrocytes/métabolisme , Grading des tumeurs , Marqueurs biologiques tumoraux/génétique
6.
Neurology ; 103(3): e209537, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38986050

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Neuroinflammation, particularly early astrocyte reactivity, is a significant driver of Alzheimer disease (AD) pathogenesis. It is unclear how the levels of astrocyte biomarkers change in patients across the AD continuum and which best reflect AD-related change. We performed a systematic review and meta-analysis of 3 blood astrocyte biomarkers (glial fibrillary acidic protein [GFAP], chitinase-3-like protein 1 [YKL-40], and S100B) in patients clinically diagnosed with AD. METHODS: MEDLINE and Web of Science were searched on March 23, 2023, without restrictions on language, time, or study design, for studies reporting blood levels of the astrocyte biomarkers GFAP, YKL-40, or S100B in patients on the AD continuum (including those with mild cognitive impairment [MCI] and dementia) and a cognitively unimpaired (CU) control population. AD diagnosis was based on established diagnostic criteria and/or comprehensive multidisciplinary clinical consensus. Studies reporting indirect biomarker measures (e.g., levels of biomarker autoantibodies) were excluded. Risk of bias assessment was performed using the revised Quality Assessment of Diagnostic Accuracy Studies tool. Pooled effect sizes were determined using the Hedge g method with a random-effects model. The review was prospectively registered on PROSPERO (registration number CRD42023458305). RESULTS: The search identified 1,186 studies; 36 met inclusion criteria (AD continuum n = 3,366, CU n = 4,115). No study was assessed to have a high risk of bias. Compared with CU individuals, patients on the AD continuum had higher GFAP and YKL-40 levels (GFAP effect size 1.15, 95% CI 0.94-1.36, p < 0.0001; YKL-40 effect size 0.38, 95% CI 0.28-0.49, p < 0.0001). Both biomarkers were elevated in more advanced clinical stages of the disease (i.e., in AD dementia compared with MCI due to AD: GFAP effect size 0.48, 95% CI 0.19-0.76, p = 0.0009; YKL-40 effect size 0.34, 95% CI 0.10-0.57, p = 0.0048). No significant differences in blood S100B levels were identified. DISCUSSION: We demonstrated significant elevations in blood GFAP and YKL-40 levels in patients on the AD continuum compared with CU individuals. Furthermore, within the AD clinical spectrum, significant elevation correlated with more advanced disease stage. Our findings suggest that both biomarkers reflect AD-related pathology. Our findings are limited by the lack of cultural and linguistic diversity in the study populations meta-analyzed. Future meta-analyses using a biomarker-defined AD population are warranted.


Sujet(s)
Maladie d'Alzheimer , Astrocytes , Marqueurs biologiques , Protéine-1 similaire à la chitinase-3 , Protéine gliofibrillaire acide , Sous-unité bêta de la protéine liant le calcium S100 , Maladie d'Alzheimer/sang , Maladie d'Alzheimer/diagnostic , Humains , Marqueurs biologiques/sang , Protéine-1 similaire à la chitinase-3/sang , Protéine gliofibrillaire acide/sang , Astrocytes/métabolisme , Sous-unité bêta de la protéine liant le calcium S100/sang , Dysfonctionnement cognitif/sang , Dysfonctionnement cognitif/diagnostic
7.
J Gen Virol ; 105(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38995681

RÉSUMÉ

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with neurological sequelae including haemorrhage, thrombosis and ischaemic necrosis and encephalitis. However, the mechanism by which this occurs is unclear. Neurological disease associated with COVID-19 has been proposed to occur following direct infection of the central nervous system and/or indirectly by local or systemic immune activation. We evaluated the expression of angiotensin-converting enzyme-2 and transmembrane protease, serine 2 (TMPRSS2) in brain tissue from five healthy human donors and observed low-level expression of these proteins in cells morphologically consistent with astrocytes, neurons and choroidal ependymal cells within the frontal cortex and medulla oblongata. Primary human astrocytes, neurons, choroid plexus epithelial cells and pericytes supported productive SARS-CoV-2 infection with ancestral, Alpha, Delta and Omicron variants. Infected cells supported the full viral life cycle, releasing infectious virus particles. In contrast, primary brain microvascular endothelial cells and microglia were refractory to SARS-CoV-2 infection. These data support a model whereby SARS-CoV-2 can infect human brain cells, and the mechanism of viral entry warrants further investigation.


Sujet(s)
Angiotensin-converting enzyme 2 , Astrocytes , COVID-19 , Plexus choroïde , Cellules épithéliales , Neurones , Péricytes , SARS-CoV-2 , Serine endopeptidases , Humains , Péricytes/virologie , SARS-CoV-2/physiologie , Astrocytes/virologie , Plexus choroïde/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Neurones/virologie , COVID-19/virologie , COVID-19/anatomopathologie , Cellules épithéliales/virologie , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Cellules cultivées , Encéphale/virologie , Encéphale/anatomopathologie , Système nerveux central/virologie
8.
Cell Mol Life Sci ; 81(1): 293, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976012

RÉSUMÉ

The function of astrocytes in response to gut microbiota-derived signals has an important role in the pathophysiological processes of central nervous system (CNS) diseases. However, the specific effects of microbiota-derived metabolites on astrocyte activation have not been elucidated yet. Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice as a classical MS model. The alterations of gut microbiota and the levels of short-chain fatty acids (SCFAs) were assessed after EAE induction. We observed that EAE mice exhibit low levels of Allobaculum, Clostridium_IV, Clostridium_XlVb, Lactobacillus genera, and microbial-derived SCFAs metabolites. SCFAs supplementation suppressed astrocyte activation by increasing the level of tryptophan (Trp)-derived AhR ligands that activating the AhR. The beneficial effects of SCFAs supplementation on the clinical scores, histopathological alterations, and the blood brain barrier (BBB)-glymphatic function were abolished by intracisterna magna injection of AAV-GFAP-shAhR. Moreover, SCFAs supplementation suppressed the loss of AQP4 polarity within astrocytes in an AhR-dependent manner. Together, SCFAs potentially suppresses astrocyte activation by amplifying Trp-AhR-AQP4 signaling in EAE mice. Our study demonstrates that SCFAs supplementation may serve as a viable therapy for inflammatory disorders of the CNS.


Sujet(s)
Aquaporine-4 , Astrocytes , Encéphalomyélite auto-immune expérimentale , Acides gras volatils , Souris de lignée C57BL , Récepteurs à hydrocarbure aromatique , Transduction du signal , Tryptophane , Animaux , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Acides gras volatils/pharmacologie , Acides gras volatils/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Souris , Tryptophane/métabolisme , Tryptophane/pharmacologie , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques
9.
Commun Biol ; 7(1): 852, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997325

RÉSUMÉ

Astrocytes play a key role in the regulation of synaptic strength and are thought to orchestrate synaptic plasticity and memory. Yet, how specifically astrocytes and their neuroactive transmitters control learning and memory is currently an open question. Recent experiments have uncovered an astrocyte-mediated feedback loop in CA1 pyramidal neurons which is started by the release of endocannabinoids by active neurons and closed by astrocytic regulation of the D-serine levels at the dendrites. D-serine is a co-agonist for the NMDA receptor regulating the strength and direction of synaptic plasticity. Activity-dependent D-serine release mediated by astrocytes is therefore a candidate for mediating between long-term synaptic depression (LTD) and potentiation (LTP) during learning. Here, we show that the mathematical description of this mechanism leads to a biophysical model of synaptic plasticity consistent with the phenomenological model known as the BCM model. The resulting mathematical framework can explain the learning deficit observed in mice upon disruption of the D-serine regulatory mechanism. It shows that D-serine enhances plasticity during reversal learning, ensuring fast responses to changes in the external environment. The model provides new testable predictions about the learning process, driving our understanding of the functional role of neuron-glia interaction in learning.


Sujet(s)
Astrocytes , Plasticité neuronale , Apprentissage inversé , Animaux , Astrocytes/physiologie , Astrocytes/métabolisme , Plasticité neuronale/physiologie , Souris , Apprentissage inversé/physiologie , Sérine/métabolisme , Modèles neurologiques , Récepteurs du N-méthyl-D-aspartate/métabolisme
10.
Nutrients ; 16(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38999797

RÉSUMÉ

Astrocyte dysfunction and inflammation play a pivotal role in depression. In this study, we evaluated the antidepressant properties of Heracleum moellendorffii root extract (HME), which is traditionally used for inflammation-related diseases, in a mouse model with astrocyte depletion that resembles the prefrontal cortex pathology of depressive patients. Mice were divided into four groups, with 10 mice per group. To induce astrocyte ablation in the mice's prefrontal cortex (PFC), we used astrocytic toxin L-alpha-aminoadipic acid (L-AAA) and administered HME orally at 200 and 500 mg/kg for 22 days. We utilized the tail suspension test (TST) to assess depression-like behaviors and the open field test (OFT) to evaluate anxiety-like activities. Additionally, astrocytic and inflammatory markers in the PFC were evaluated using immunohistochemistry and ELISA. The results showed that infusion of L-AAA significantly decreased the expression of astrocytic glial fibrillary acidic protein (GFAP), which was accompanied by increased depression and anxiety-like behaviors. However, HME significantly reversed these effects by dose-dependently enhancing GFAP expression and modulating inflammatory markers, such as TNF-α, IL-6, and particularly lipocalin-2, a master proinflammatory mediator. These results imply that HME contributes to the alleviation of depression and anxiety-like behaviors by promoting astrocyte recovery and reducing neuroinflammation, especially through lipocalin-2 inhibition.


Sujet(s)
Antidépresseurs , Astrocytes , Comportement animal , Dépression , Modèles animaux de maladie humaine , Lipocaline-2 , Extraits de plantes , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Lipocaline-2/métabolisme , Extraits de plantes/pharmacologie , Dépression/traitement médicamenteux , Souris , Antidépresseurs/pharmacologie , Mâle , Comportement animal/effets des médicaments et des substances chimiques , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Protéine gliofibrillaire acide/métabolisme , Souris de lignée C57BL
11.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000485

RÉSUMÉ

Cytotoxic activity has been reported for the xanthone α-mangostin (AMN) against Glioblastoma multiforme (GBM), an aggressive malignant brain cancer with a poor prognosis. Recognizing that AMN's high degree of hydrophobicity is likely to limit its systemic administration, we formulated AMN using reconstituted high-density lipoprotein (rHDL) nanoparticles. The photophysical characteristics of the formulation, including fluorescence lifetime and steady-state anisotropy, indicated that AMN was successfully incorporated into the rHDL nanoparticles. To our knowledge, this is the first report on the fluorescent characteristics of AMN with an HDL-based drug carrier. Cytotoxicity studies in a 2D culture and 3D spheroid model of LN-229 GBM cells and normal human astrocytes showed an enhanced therapeutic index with the rHDL-AMN formulation compared to the unincorporated AMN and Temozolomide, a standard GBM chemotherapy agent. Furthermore, treatment with the rHDL-AMN facilitated a dose-dependent upregulation of autophagy and reactive oxygen species generation to a greater extent in LN-229 cells compared to astrocytes, indicating the reduced off-target toxicity of this novel formulation. These studies indicate the potential therapeutic benefits to GBM patients via selective targeting using the rHDL-AMN formulation.


Sujet(s)
Glioblastome , Lipoprotéines HDL , Nanoparticules , Sphéroïdes de cellules , Xanthones , Humains , Xanthones/composition chimique , Xanthones/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Lipoprotéines HDL/composition chimique , Lipoprotéines HDL/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique , Espèces réactives de l'oxygène/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Autophagie/effets des médicaments et des substances chimiques
12.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000499

RÉSUMÉ

General anesthetics may accelerate the neuropathological changes related to Alzheimer's disease (AD), of which amyloid beta (Aß)-induced toxicity is one of the main causes. However, the interaction of general anesthetics with different Aß-isoforms remains unclear. In this study, we investigated the effects of sevoflurane (0.4 and 1.2 maximal alveolar concentration (MAC)) on four Aß species-induced changes on dendritic spine density (DSD) in hippocampal brain slices of Thy1-eGFP mice and multiple epidermal growth factor-like domains 10 (MEGF10)-related astrocyte-mediated synaptic engulfment in hippocampal brain slices of C57BL/6 mice. We found that both sevoflurane and Aß downregulated CA1-dendritic spines. Moreover, compared with either sevoflurane or Aß alone, pre-treatment with Aß isoforms followed by sevoflurane application in general further enhanced spine loss. This enhancement was related to MEGF10-related astrocyte-dependent synaptic engulfment, only in AßpE3 + 1.2 MAC sevoflurane and 3NTyrAß + 1.2 MAC sevoflurane condition. In addition, removal of sevoflurane alleviated spine loss in Aß + sevoflurane. In summary, these results suggest that both synapses and astrocytes are sensitive targets for sevoflurane; in the presence of 3NTyrAß, 1.2 MAC sevoflurane alleviated astrocyte-mediated synaptic engulfment and exerted a lasting effect on dendritic spine remodeling.


Sujet(s)
Peptides bêta-amyloïdes , Astrocytes , Région CA1 de l'hippocampe , Épines dendritiques , Souris de lignée C57BL , Sévoflurane , Synapses , Sévoflurane/pharmacologie , Animaux , Épines dendritiques/métabolisme , Épines dendritiques/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Synapses/effets des médicaments et des substances chimiques , Synapses/métabolisme , Souris , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/effets des médicaments et des substances chimiques , Région CA1 de l'hippocampe/cytologie , Mâle , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Anesthésiques par inhalation/pharmacologie
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000512

RÉSUMÉ

Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2-/- female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2-/- female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2-/- brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2-/- hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2-/- mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health.


Sujet(s)
Mitochondries , Mémoire spatiale , Animaux , Souris , Femelle , Mitochondries/métabolisme , Mâle , Troubles de la mémoire/métabolisme , Troubles de la mémoire/génétique , Encéphale/métabolisme , Encéphale/anatomopathologie , Souris knockout , Souris de lignée C57BL , Inflammation/métabolisme , Inflammation/anatomopathologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Microenvironnement cellulaire , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Hippocampe/métabolisme , Hippocampe/anatomopathologie
14.
PLoS Biol ; 22(7): e3002712, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38996200

RÉSUMÉ

Glial cells such as astrocytes can modulate neuronal signaling. Astrocytes can also acquire a reactive phenotype that correlates with cognitive impairments in brain diseases. A study in PLOS Biology shows that prolonged activation of astrocytes can trigger both cognitive impairments and a reactive astrocyte phenotype.


Sujet(s)
Astrocytes , Cognition , Astrocytes/physiologie , Animaux , Humains , Cognition/physiologie , Dysfonctionnement cognitif/physiopathologie , Neurones/physiologie
15.
Medicine (Baltimore) ; 103(28): e38983, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996095

RÉSUMÉ

RATIONALE: Autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy is a rare autoimmune disease of the central nervous system that affects the meninges, brain, spinal cord, and optic nerves. GFAP astrocytopathy can coexist with a variety of antibodies, which is known as overlap syndrome. Anti-NMDAR-positive encephalitis overlap syndrome has been reported; however, encephalitis overlap syndrome with both anti-NMDAR and sulfatide-IgG positivity has not been reported. PATIENT CONCERNS: The patient was a 50-year-old male who was drowsy and had chills and weak limbs for 6 months. His symptoms worsened after admission to our hospital with persistent high fever, dysphoria, gibberish, and disturbance of consciousness. Positive cerebrospinal fluid NMDA, GFAP antibodies, and serum sulfatide antibody IgG were positive. DIAGNOSES: Autoimmune GFAP astrocytopathy with anti-NMDAR and sulfatide-IgG-positive encephalitis overlap syndrome. INTERVENTIONS: In addition to ventilator support and symptomatic supportive treatment, step-down therapy with methylprednisolone (1000 mg/d, halved every 3 days) and pulse therapy with human immunoglobulin (0.4 g/(kg d) for 5 days) were used. OUTCOMES: After 6 days of treatment, the patient condition did not improve, and the family signed up to give up the treatment and left the hospital. CONCLUSIONS: Patients with autoimmune GFAP astrocytopathy may be positive for anti-NMDAR and sulfatide-IgG, and immunotherapy may be effective in patients with severe conditions. LESSONS: Autoimmune GFAP astrocytopathy with nonspecific symptoms is rarely reported and is easy to be missed and misdiagnosed. GFAP astrocytopathy should be considered in patients with fever, headache, disturbance of consciousness, convulsions, and central infections that do not respond to antibacterial and viral agents. Autoimmune encephalopathy-related antibody testing should be performed as soon as possible, early diagnosis should be confirmed, and immunomodulatory therapy should be administered promptly.


Sujet(s)
Protéine gliofibrillaire acide , Sulfoglycosphingolipides , Humains , Mâle , Adulte d'âge moyen , Protéine gliofibrillaire acide/immunologie , Protéine gliofibrillaire acide/sang , Sulfoglycosphingolipides/immunologie , Immunoglobuline G/sang , Immunoglobuline G/liquide cérébrospinal , Autoanticorps/sang , Méthylprednisolone/usage thérapeutique , Encéphalite/diagnostic , Encéphalite/immunologie , Encéphalite à anticorps anti-récepteur N-méthyl-D-aspartate/diagnostic , Encéphalite à anticorps anti-récepteur N-méthyl-D-aspartate/immunologie , Encéphalite à anticorps anti-récepteur N-méthyl-D-aspartate/complications , Astrocytes/immunologie , Astrocytes/anatomopathologie , Maladies auto-immunes du système nerveux/diagnostic , Maladies auto-immunes du système nerveux/immunologie
16.
PLoS Biol ; 22(7): e3002687, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38991663

RÉSUMÉ

Reactive astrocytes are associated with neuroinflammation and cognitive decline in diverse neuropathologies; however, the underlying mechanisms are unclear. We used optogenetic and chemogenetic tools to identify the crucial roles of the hippocampal CA1 astrocytes in cognitive decline. Our results showed that repeated optogenetic stimulation of the hippocampal CA1 astrocytes induced cognitive impairment in mice and decreased synaptic long-term potentiation (LTP), which was accompanied by the appearance of inflammatory astrocytes. Mechanistic studies conducted using knockout animal models and hippocampal neuronal cultures showed that lipocalin-2 (LCN2), derived from reactive astrocytes, mediated neuroinflammation and induced cognitive impairment by decreasing the LTP through the reduction of neuronal NMDA receptors. Sustained chemogenetic stimulation of hippocampal astrocytes provided similar results. Conversely, these phenomena were attenuated by a metabolic inhibitor of astrocytes. Fiber photometry using GCaMP revealed a high level of hippocampal astrocyte activation in the neuroinflammation model. Our findings suggest that reactive astrocytes in the hippocampus are sufficient and required to induce cognitive decline through LCN2 release and synaptic modulation. This abnormal glial-neuron interaction may contribute to the pathogenesis of cognitive disturbances in neuroinflammation-associated brain conditions.


Sujet(s)
Astrocytes , Dysfonctionnement cognitif , Hippocampe , Lipocaline-2 , Potentialisation à long terme , Maladies neuro-inflammatoires , Neurones , Animaux , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/étiologie , Dysfonctionnement cognitif/anatomopathologie , Lipocaline-2/métabolisme , Lipocaline-2/génétique , Souris , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Souris knockout , Mâle , Souris de lignée C57BL , Récepteurs du N-méthyl-D-aspartate/métabolisme , Optogénétique , Région CA1 de l'hippocampe/anatomopathologie , Région CA1 de l'hippocampe/métabolisme , Modèles animaux de maladie humaine
17.
Nat Commun ; 15(1): 5830, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992057

RÉSUMÉ

Impaired ion channels regulating Golgi pH lead to structural alterations in the Golgi apparatus, such as fragmentation, which is found, along with cognitive impairment, in Alzheimer's disease. However, the causal relationship between altered Golgi structure and cognitive impairment remains elusive due to the lack of understanding of ion channels in the Golgi apparatus of brain cells. Here, we identify that a transmembrane protein TMEM87A, renamed Golgi-pH-regulating cation channel (GolpHCat), expressed in astrocytes and neurons that contributes to hippocampus-dependent memory. We find that GolpHCat displays unique voltage-dependent currents, which is potently inhibited by gluconate. Additionally, we gain structural insights into the ion conduction through GolpHCat at the molecular level by determining three high-resolution cryogenic-electron microscopy structures of human GolpHCat. GolpHCat-knockout mice show fragmented Golgi morphology and altered protein glycosylation and functions in the hippocampus, leading to impaired spatial memory. These findings suggest a molecular target for Golgi-related diseases and cognitive impairment.


Sujet(s)
Appareil de Golgi , Hippocampe , Souris knockout , Neurones , Appareil de Golgi/métabolisme , Animaux , Hippocampe/métabolisme , Humains , Souris , Neurones/métabolisme , Concentration en ions d'hydrogène , Astrocytes/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mâle , Souris de lignée C57BL , Cellules HEK293 , Mémoire spatiale/physiologie , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Mémoire/physiologie , Glycosylation , Cryomicroscopie électronique , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/physiopathologie , Dysfonctionnement cognitif/anatomopathologie
18.
Cells ; 13(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38994948

RÉSUMÉ

Excessive inflammatory reactions and oxidative stress are well-recognized molecular findings in autism and these processes can affect or be affected by the epigenetic landscape. Nonetheless, adequate therapeutics are unavailable, as patient-specific brain molecular markers for individualized therapies remain challenging. METHODS: We used iPSC-derived neurons and astrocytes of patients with autism vs. controls (5/group) to examine whether they replicate the postmortem brain expression/epigenetic alterations of autism. Additionally, DNA methylation of 10 postmortem brain samples (5/group) was analyzed for genes affected in PSC-derived cells. RESULTS: We found hyperexpression of TGFB1, TGFB2, IL6 and IFI16 and decreased expression of HAP1, SIRT1, NURR1, RELN, GPX1, EN2, SLC1A2 and SLC1A3 in the astrocytes of patients with autism, along with DNA hypomethylation of TGFB2, IL6, TNFA and EN2 gene promoters and a decrease in HAP1 promoter 5-hydroxymethylation in the astrocytes of patients with autism. In neurons, HAP1 and IL6 expression trended alike. While HAP1 promoter was hypermethylated in neurons, IFI16 and SLC1A3 promoters were hypomethylated and TGFB2 exhibited increased promoter 5-hydroxymethlation. We also found a reduction in neuronal arborization, spine size, growth rate, and migration, but increased astrocyte size and a reduced growth rate in autism. In postmortem brain samples, we found DNA hypomethylation of TGFB2 and IFI16 promoter regions, but DNA hypermethylation of HAP1 and SLC1A2 promoters in autism. CONCLUSION: Autism-associated expression/epigenetic alterations in iPSC-derived cells replicated those reported in the literature, making them appropriate surrogates to study disease pathogenesis or patient-specific therapeutics.


Sujet(s)
Astrocytes , Trouble autistique , Encéphale , Méthylation de l'ADN , Épigenèse génétique , Cellules souches pluripotentes induites , Neurones , Humains , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Trouble autistique/génétique , Trouble autistique/anatomopathologie , Trouble autistique/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Méthylation de l'ADN/génétique , Encéphale/anatomopathologie , Encéphale/métabolisme , Mâle , Femelle , Régions promotrices (génétique)/génétique , Forme de la cellule , Enfant , Régulation de l'expression des gènes , Protéine reeline
19.
Cells ; 13(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38994979

RÉSUMÉ

HIV-associated neurocognitive disorders (HAND) persist under antiretroviral therapy as a complex pathology that has been difficult to study in cellular and animal models. Therefore, we generated an ex vivo human brain slice model of HIV-1 infection from surgically resected adult brain tissue. Brain slice cultures processed for flow cytometry showed >90% viability of dissociated cells within the first three weeks in vitro, with parallel detection of astrocyte, myeloid, and neuronal populations. Neurons within brain slices showed stable dendritic spine density and mature spine morphologies in the first weeks in culture, and they generated detectable activity in multi-electrode arrays. We infected cultured brain slices using patient-matched CD4+ T-cells or monocyte-derived macrophages (MDMs) that were exposed to a GFP-expressing R5-tropic HIV-1 in vitro. Infected slice cultures expressed viral RNA and developed a spreading infection up to 9 days post-infection, which were significantly decreased by antiretrovirals. We also detected infected myeloid cells and astrocytes within slices and observed minimal effect on cellular viability over time. Overall, this human-centered model offers a promising resource to study the cellular mechanisms contributing to HAND (including antiretroviral toxicity, substance use, and aging), infection of resident brain cells, and new neuroprotective therapeutics.


Sujet(s)
Encéphale , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Encéphale/virologie , Encéphale/anatomopathologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Infections à VIH/virologie , Infections à VIH/anatomopathologie , Adulte , Neurones/virologie , Neurones/métabolisme , Macrophages/virologie , Macrophages/métabolisme , Astrocytes/virologie , Lymphocytes T CD4+/virologie , Techniques de culture de tissus
20.
Cells ; 13(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38995011

RÉSUMÉ

Unsuccessful axonal regeneration in transected spinal cord injury (SCI) is mainly attributed to shortage of growth factors, inhibitory glial scar, and low intrinsic regenerating capacity of severely injured neurons. Previously, we constructed an axonal growth permissive pathway in a thoracic hemisected injury by transplantation of Schwann cells overexpressing glial-cell-derived neurotrophic factor (SCs-GDNF) into the lesion gap as well as the caudal cord and proved that this novel permissive bridge promoted the regeneration of descending propriospinal tract (dPST) axons across and beyond the lesion. In the current study, we subjected rats to complete thoracic (T11) spinal cord transections and examined whether these combinatorial treatments can support dPST axons' regeneration beyond the transected injury. The results indicated that GDNF significantly improved graft-host interface by promoting integration between SCs and astrocytes, especially the migration of reactive astrocyte into SCs-GDNF territory. The glial response in the caudal graft area has been significantly attenuated. The astrocytes inside the grafted area were morphologically characterized by elongated and slim process and bipolar orientation accompanied by dramatically reduced expression of glial fibrillary acidic protein. Tremendous dPST axons have been found to regenerate across the lesion and back to the caudal spinal cord which were otherwise difficult to see in control groups. The caudal synaptic connections were formed, and regenerated axons were remyelinated. The hindlimb locomotor function has been improved.


Sujet(s)
Axones , Facteur neurotrophique dérivé des cellules gliales , Régénération nerveuse , Cellules de Schwann , Traumatismes de la moelle épinière , Animaux , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/anatomopathologie , Traumatismes de la moelle épinière/physiopathologie , Cellules de Schwann/métabolisme , Facteur neurotrophique dérivé des cellules gliales/métabolisme , Facteur neurotrophique dérivé des cellules gliales/génétique , Axones/métabolisme , Rats , Rat Sprague-Dawley , Femelle , Astrocytes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...