Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.839
Filtrer
1.
Neurosci Lett ; 841: 137959, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39218293

RÉSUMÉ

Understanding the sequence of cellular responses and their contributions to pathomorphogical changes in spinal white matter injuries is a prerequisite for developing efficient therapeutic strategies for spinal cord injury (SCI) as well as neurodegenerative and inflammatory diseases of the spinal cord such as amyotrophic lateral sclerosis and multiple sclerosis. We have developed several types of surgical procedures suitable for acute one-time and chronic recurrent in vivo multiphoton microscopy of spinal white matter [1]. Sophisticated surgical procedures were combined with transgenic mouse technology to image spinal tissue labeled with up to four fluorescent proteins (FPs) in axons, astrocytes, microglia, and blood vessels. To clearly separate the simultaneously excited FPs, spectral unmixing including iterative procedures was performed after imaging the diversely labeled spinal white matter with a custom-made 4-channel two-photon laser-scanning microscope. In our longitudinal multicellular studies of injured spinal white matter, we imaged axonal dynamics and invasion of microglia and astrocytes for a time course of over 200 days after SCI. Our methods offer ideal platforms for investigating acute and chronic cellular dynamics, cell-cell interactions, and metabolite fluctuations in health and disease as well as pharmacological manipulations in vivo.


Sujet(s)
Axones , Souris transgéniques , Traumatismes de la moelle épinière , Substance blanche , Animaux , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Substance blanche/imagerie diagnostique , Traumatismes de la moelle épinière/anatomopathologie , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/imagerie diagnostique , Axones/anatomopathologie , Axones/métabolisme , Névroglie/métabolisme , Névroglie/anatomopathologie , Souris , Microscopie de fluorescence multiphotonique/méthodes , Moelle spinale/anatomopathologie , Moelle spinale/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Astrocytes/métabolisme , Astrocytes/anatomopathologie
2.
Cancer Biol Ther ; 25(1): 2398285, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39238191

RÉSUMÉ

Breast cancer stands as the most prevalent cancer diagnosed worldwide, often leading to brain metastasis, a challenging complication characterized by high mortality rates and a grim prognosis. Understanding the intricate mechanisms governing breast cancer brain metastasis (BCBM) remains an ongoing challenge. The unique microenvironment in the brain fosters an ideal setting for the colonization of breast cancer cells. The tumor microenvironment (TME) in brain metastases plays a pivotal role in the initiation and progression of BCBM, shaping the landscape for targeted therapeutic interventions. Current research primarily concentrates on unraveling the complexities of the TME in BCBM, with a particular emphasis on neuroglia and immune cells, such as microglia, monocyte-derived macrophages (MDMs), astrocytes and T cells. This comprehensive review delves deeply into these elements within the TME of BCBM, shedding light on their interplay, mechanisms, and potential as therapeutic targets to combat BCBM.


Sujet(s)
Tumeurs du cerveau , Tumeurs du sein , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Femelle , Névroglie/anatomopathologie , Névroglie/immunologie , Animaux
3.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39273535

RÉSUMÉ

Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.


Sujet(s)
Sclérose en plaques , Névroglie , Maladies neuro-inflammatoires , Humains , Sclérose en plaques/métabolisme , Sclérose en plaques/anatomopathologie , Névroglie/métabolisme , Névroglie/anatomopathologie , Animaux , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Microglie/métabolisme , Microglie/anatomopathologie , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Encéphale/métabolisme , Encéphale/anatomopathologie
4.
Nat Commun ; 15(1): 7880, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251599

RÉSUMÉ

Progressive supranuclear palsy (PSP), a rare Parkinsonian disorder, is characterized by problems with movement, balance, and cognition. PSP differs from Alzheimer's disease (AD) and other diseases, displaying abnormal microtubule-associated protein tau by both neuronal and glial cell pathologies. Genetic contributors may mediate these differences; however, the genetics of PSP remain underexplored. Here we conduct the largest genome-wide association study (GWAS) of PSP which includes 2779 cases (2595 neuropathologically-confirmed) and 5584 controls and identify six independent PSP susceptibility loci with genome-wide significant (P < 5 × 10-8) associations, including five known (MAPT, MOBP, STX6, RUNX2, SLCO1A2) and one novel locus (C4A). Integration with cell type-specific epigenomic annotations reveal an oligodendrocytic signature that might distinguish PSP from AD and Parkinson's disease in subsequent studies. Candidate PSP risk gene prioritization using expression quantitative trait loci (eQTLs) identifies oligodendrocyte-specific effects on gene expression in half of the genome-wide significant loci, and an association with C4A expression in brain tissue, which may be driven by increased C4A copy number. Finally, histological studies demonstrate tau aggregates in oligodendrocytes that colocalize with C4 (complement) deposition. Integrating GWAS with functional studies, epigenomic and eQTL analyses, we identify potential causal roles for variation in MOBP, STX6, RUNX2, SLCO1A2, and C4A in PSP pathogenesis.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Locus de caractère quantitatif , Paralysie supranucléaire progressive , Protéines tau , Humains , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/métabolisme , Sujet âgé , Mâle , Femelle , Protéines tau/génétique , Protéines tau/métabolisme , Transcriptome , Polymorphisme de nucléotide simple , Névroglie/métabolisme , Névroglie/anatomopathologie , Sujet âgé de 80 ans ou plus , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Adulte d'âge moyen , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Études cas-témoins , Protéines de la myéline
5.
Int J Mol Sci ; 25(17)2024 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-39273659

RÉSUMÉ

GM1 gangliosidosis is a lysosomal storage disorder characterized by the accumulation of GM1 ganglioside, leading to severe neurodegeneration and early mortality. The disease primarily affects the central nervous system, causing progressive neurodegeneration, including widespread neuronal loss and gliosis. To gain a deeper understanding of the neuropathology associated with GM1 gangliosidosis, we employed single-nucleus RNA sequencing to analyze brain tissues from both GM1 gangliosidosis model mice and control mice. No significant changes in cell proportions were detected between the two groups of animals. Differential expression analysis revealed cell type-specific changes in gene expression in neuronal and glial cells. Functional analysis highlighted the neurodegenerative processes, oxidative phosphorylation, and neuroactive ligand-receptor interactions as the significantly affected pathways. The contribution of the impairment of neurotransmitter system disruption and neuronal circuitry disruption was more important than neuroinflammatory responses to GM1 pathology. In 16-week-old GM1 gangliosidosis mice, no microglial or astrocyte activation or increased expression of innate immunity genes was detected. This suggested that nerve degeneration did not induce the inflammatory response but rather promoted glial cell clearance. Our findings provide a crucial foundation for understanding the cellular and molecular mechanisms of GM1 gangliosidosis, potentially guiding future therapeutic strategies.


Sujet(s)
Modèles animaux de maladie humaine , Gangliosidose à GM1 , Animaux , Gangliosidose à GM1/génétique , Gangliosidose à GM1/métabolisme , Gangliosidose à GM1/anatomopathologie , Souris , Transcriptome , Névroglie/métabolisme , Névroglie/anatomopathologie , Analyse de profil d'expression de gènes , Neurones/métabolisme , Neurones/anatomopathologie , Système nerveux central/métabolisme , Système nerveux central/anatomopathologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Ganglioside GM1/métabolisme , Analyse sur cellule unique , Souris de lignée C57BL
6.
Handb Clin Neurol ; 204: 263-271, 2024.
Article de Anglais | MEDLINE | ID: mdl-39322383

RÉSUMÉ

Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is an adult-onset, inherited white matter disorder encompassing two previously identified clinicopathologically similar entities: pigmentary orthochromatic leukodystrophy (POLD) and hereditary diffuse leukoencephalopathy with spheroids (HDLS). In this chapter, we discuss how advances in our genetic understanding of the condition have further delineated three distinct clinical entities within ALSP, namely CSF1R-related ALSP, AARS2-related leukoencephalopathy (AARS2-L), and AARS (HDLS-S). We provide descriptions of the clinical, radiologic, pathologic, and pathophysiologic findings in each entity, detailing their similarities and differences, and discuss current and future treatment options where available.


Sujet(s)
Leucoencéphalopathies , Névroglie , Humains , Leucoencéphalopathies/génétique , Leucoencéphalopathies/anatomopathologie , Névroglie/anatomopathologie , Adulte , Axones/anatomopathologie
7.
Nat Commun ; 15(1): 8158, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39289367

RÉSUMÉ

The neurovascular unit (NVU) is a complex multicellular structure that helps maintain cerebral homeostasis and blood-brain barrier (BBB) integrity. While extensive evidence links NVU alterations to cerebrovascular diseases and neurodegeneration, the underlying molecular mechanisms remain unclear. Here, we use zebrafish embryos carrying a mutation in Scavenger Receptor B2, a highly conserved endolysosomal protein expressed predominantly in Radial Glia Cells (RGCs), to investigate the interplay among different NVU components. Through live imaging and genetic manipulations, we demonstrate that compromised acidification of the endolysosomal compartment in mutant RGCs leads to impaired Notch3 signaling, thereby inducing excessive neurogenesis and reduced glial differentiation. We further demonstrate that alterations to the neuron/glia balance result in impaired VEGF and Wnt signaling, leading to severe vascular defects, hemorrhages, and a leaky BBB. Altogether, our findings provide insights into NVU formation and function and offer avenues for investigating diseases involving white matter defects and vascular abnormalities.


Sujet(s)
Barrière hémato-encéphalique , Lysosomes , Neurogenèse , Protéines de poisson-zèbre , Danio zébré , Animaux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Lysosomes/métabolisme , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Cellules épendymogliales/métabolisme , Cellules épendymogliales/anatomopathologie , Endosomes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Névroglie/métabolisme , Névroglie/anatomopathologie , Différenciation cellulaire , Cellules souches/métabolisme , Voie de signalisation Wnt , Mutation , Néovascularisation physiologique , Animal génétiquement modifié , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/vascularisation , Transduction du signal ,
8.
J Neuroinflammation ; 21(1): 242, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39334365

RÉSUMÉ

Cryptococcus neoformans (Cn) is an opportunistic encapsulated fungal pathogen that causes life-threatening meningoencephalitis in immunosuppressed individuals. Since IL-6 is important for blood-brain barrier support and its deficiency has been shown to facilitate Cn brain invasion, we investigated the impact of IL-6 on systemic Cn infection in vivo, focusing on central nervous system (CNS) colonization and glial responses, specifically microglia and astrocytes. IL-6 knock-out (IL-6-/-) mice showed faster mortality than C57BL/6 (Wild-type) and IL-6-/- supplemented with recombinant IL-6 (rIL-6; 40 pg/g/day) mice. Despite showing early lung inflammation but no major histological differences in pulmonary cryptococcosis progression among the experimental groups, IL-6-/- mice had significantly higher blood and brain tissue fungal burden at 7-days post infection. Exposure of cryptococci to rIL-6 in vitro increased capsule growth. In addition, IL-6-/- brains were characterized by an increased dystrophic microglia number during Cn infection, which are associated with neurodegeneration and senescence. In contrast, the brains of IL-6-producing or -supplemented mice displayed high numbers of activated and phagocytic microglia, which are related to a stronger anti-cryptococcal response or tissue repair. Likewise, culture of rIL-6 with microglia-like cells promoted high fungal phagocytosis and killing, whereas IL-6 silencing in microglia decreased fungal phagocytosis. Lastly, astrogliosis was high and moderate in infected brains removed from Wild-type and IL-6-/- supplemented with rIL-6 animals, respectively, while minimal astrogliosis was observed in IL-6-/- tissue, highlighting the potential of astrocytes in containing and combating cryptococcal infection. Our findings suggest a critical role for IL-6 in Cn CNS dissemination, neurocryptococcosis development, and host defense.


Sujet(s)
Cryptococcose , Cryptococcus neoformans , Interleukine-6 , Souris de lignée C57BL , Souris knockout , Névroglie , Animaux , Souris , Interleukine-6/métabolisme , Névroglie/anatomopathologie , Névroglie/métabolisme , Névroglie/microbiologie , Cryptococcose/anatomopathologie , Cryptococcose/immunologie , Cryptococcose/microbiologie , Encéphale/anatomopathologie , Encéphale/métabolisme
9.
Bull Exp Biol Med ; 177(4): 528-533, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39266920

RÉSUMÉ

CNS diseases associated with compromised blood supply and/or vascular integrity are one of the leading causes of mortality and disability in adults worldwide and are also among 10 most common causes of death in children. Angiogenesis is an essential element of regeneration processes upon nervous tissue damage and can play a crucial role in neuroprotection. Here we review the features of cerebral vascular regeneration after ischemic stroke, including the complex interactions between endothelial cells and other brain cell types (neural stem cells, astrocytes, microglia, and oligodendrocytes). The mechanisms of reciprocal influence of angiogenesis and neurogenesis, the role of astrocytes in the formation of the blood-brain barrier, and roles of microglia and oligodendrocytes in vascular regeneration are discussed. Understanding the mechanisms of angiogenesis regulation in CNS is of critical importance for the development of new treatments of neurovascular pathologies.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Accident vasculaire cérébral ischémique , Néovascularisation physiologique , Cellules souches neurales , Neurogenèse , Humains , Accident vasculaire cérébral ischémique/physiopathologie , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Barrière hémato-encéphalique/physiopathologie , Néovascularisation physiologique/physiologie , Neurogenèse/physiologie , Animaux , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Astrocytes/physiologie , Cellules souches neurales/métabolisme , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Oligodendroglie/physiologie , Microglie/anatomopathologie , Microglie/métabolisme , Microglie/physiologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Névroglie/métabolisme , Névroglie/anatomopathologie , Encéphalopathie ischémique/physiopathologie , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/anatomopathologie , Système nerveux central/vascularisation , Système nerveux central/métabolisme , Système nerveux central/anatomopathologie , Encéphale/vascularisation , Encéphale/anatomopathologie , Encéphale/métabolisme , Encéphale/physiopathologie ,
10.
Cells ; 13(18)2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39329738

RÉSUMÉ

Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.


Sujet(s)
Ferroptose , Inflammation , Molécule-1 d'adhérence intercellulaire , Maladie de Parkinson , Ferroptose/génétique , Molécule-1 d'adhérence intercellulaire/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Humains , Animaux , Inflammation/anatomopathologie , Inflammation/métabolisme , Névroglie/métabolisme , Névroglie/anatomopathologie , Fer/métabolisme
11.
Nat Commun ; 15(1): 7857, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251578

RÉSUMÉ

Glioblastoma remains one of the deadliest brain malignancies. First-line therapy consists of maximal surgical tumor resection, accompanied by chemotherapy and radiotherapy. Malignant cells escape surgical resection by migrating into the surrounding healthy brain tissue, where they give rise to the recurrent tumor. Based on gene expression, tumor cores can be subtyped into mesenchymal, proneural, and classical tumors, each being associated with differences in genetic alterations and cellular composition. In contrast, the adjacent brain parenchyma where infiltrating malignant cells escape surgical resection is less characterized in patients. Using spatial transcriptomics (n = 11), we show that malignant cells within proneural or mesenchymal tumor cores display spatially organized differences in gene expression, although such differences decrease within the infiltrated brain tissue. Malignant cells residing in infiltrated brain tissue have increased expression of genes related to neurodevelopmental pathways and glial cell differentiation. Our findings provide an updated view of the spatial landscape of glioblastomas and further our understanding of the malignant cells that infiltrate the healthy brain, providing new avenues for the targeted therapy of these cells after surgical resection.


Sujet(s)
Tumeurs du cerveau , Encéphale , Régulation de l'expression des gènes tumoraux , Glioblastome , Récepteurs Notch , Transduction du signal , Humains , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Encéphale/métabolisme , Encéphale/anatomopathologie , Transcriptome , Synapses/métabolisme , Mâle , Femelle , Lignée cellulaire tumorale , Névroglie/métabolisme , Névroglie/anatomopathologie , Différenciation cellulaire/génétique
12.
Acta Neuropathol Commun ; 12(1): 139, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39217398

RÉSUMÉ

CSF1R-related disorder (CSF1R-RD) is a neurodegenerative condition that predominantly affects white matter due to genetic alterations in the CSF1R gene, which is expressed by microglia. We studied an elderly man with a hereditary, progressive dementing disorder of unclear etiology. Standard genetic testing for leukodystrophy and other neurodegenerative conditions was negative. Brain autopsy revealed classic features of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), including confluent white matter degeneration with axonal spheroids and pigmented glial cells in the affected white matter, consistent with CSF1R-RD. Subsequent long-read sequencing identified a novel deletion in CSF1R that was not detectable with short-read exome sequencing. To gain insight into potential mechanisms underlying white matter degeneration in CSF1R-RD, we studied multiple brain regions exhibiting varying degrees of white matter pathology. We found decreased CSF1R transcript and protein across brain regions, including intact white matter. Single nuclear RNA sequencing (snRNAseq) identified two disease-associated microglial cell states: lipid-laden microglia (expressing GPNMB, ATG7, LGALS1, LGALS3) and inflammatory microglia (expressing IL2RA, ATP2C1, FCGBP, VSIR, SESN3), along with a small population of CD44+ peripheral monocyte-derived macrophages exhibiting migratory and phagocytic signatures. GPNMB+ lipid-laden microglia with ameboid morphology represented the end-stage disease microglia state. Disease-associated oligodendrocytes exhibited cell stress signatures and dysregulated apoptosis-related genes. Disease-associated oligodendrocyte precursor cells (OPCs) displayed a failure in their differentiation into mature myelin-forming oligodendrocytes, as evidenced by upregulated LRP1, PDGFRA, SOX5, NFIA, and downregulated NKX2-2, NKX6.2, SOX4, SOX8, TCF7L2, YY1, ZNF488. Overall, our findings highlight microglia-oligodendroglia crosstalk in demyelination, with CSF1R dysfunction promoting phagocytic and inflammatory microglia states, an arrest in OPC differentiation, and oligodendrocyte depletion.


Sujet(s)
Névroglie , Récepteur de facteur de croissance granulocyte-macrophage , Humains , Mâle , Récepteur de facteur de croissance granulocyte-macrophage/génétique , Récepteur de facteur de croissance granulocyte-macrophage/métabolisme , Névroglie/anatomopathologie , Névroglie/métabolisme , Leucoencéphalopathies/génétique , Leucoencéphalopathies/anatomopathologie , Leucoencéphalopathies/métabolisme , Sujet âgé , Microglie/anatomopathologie , Microglie/métabolisme , Analyse de profil d'expression de gènes , Transcriptome , Substance blanche/anatomopathologie , Substance blanche/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Récepteur du facteur de stimulation des colonies de macrophages
14.
Life Sci ; 354: 122953, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39122110

RÉSUMÉ

Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.


Sujet(s)
Névroglie , Humains , Névroglie/métabolisme , Névroglie/anatomopathologie , Animaux , Thérapie génétique/méthodes , Transplantation de cellules souches/méthodes
15.
Cell Rep Med ; 5(8): 101680, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39121861

RÉSUMÉ

The role of central nervous system (CNS) glia in sustaining self-autonomous inflammation and driving clinical progression in multiple sclerosis (MS) is gaining scientific interest. We applied a single transcription factor (SOX10)-based protocol to accelerate oligodendrocyte differentiation from human induced pluripotent stem cell (hiPSC)-derived neural precursor cells, generating self-organizing forebrain organoids. These organoids include neurons, astrocytes, oligodendroglia, and hiPSC-derived microglia to achieve immunocompetence. Over 8 weeks, organoids reproducibly generated mature CNS cell types, exhibiting single-cell transcriptional profiles similar to the adult human brain. Exposed to inflamed cerebrospinal fluid (CSF) from patients with MS, organoids properly mimic macroglia-microglia neurodegenerative phenotypes and intercellular communication seen in chronic active MS. Oligodendrocyte vulnerability emerged by day 6 post-MS-CSF exposure, with nearly 50% reduction. Temporally resolved organoid data support and expand on the role of soluble CSF mediators in sustaining downstream events leading to oligodendrocyte death and inflammatory neurodegeneration. Such findings support the implementation of this organoid model for drug screening to halt inflammatory neurodegeneration.


Sujet(s)
Encéphale , Différenciation cellulaire , Cellules souches pluripotentes induites , Sclérose en plaques , Névroglie , Organoïdes , Phénotype , Humains , Sclérose en plaques/anatomopathologie , Sclérose en plaques/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/anatomopathologie , Organoïdes/anatomopathologie , Organoïdes/métabolisme , Névroglie/métabolisme , Névroglie/anatomopathologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Cellules souches neurales/métabolisme , Cellules souches neurales/anatomopathologie , Microglie/métabolisme , Microglie/anatomopathologie
16.
J Neuroinflammation ; 21(1): 213, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39217340

RÉSUMÉ

BACKGROUND: Neuropathic pain (NP), which results from injury or lesion of the somatosensory nervous system, is intimately associated with glial cells. The roles of microglia and astrocytes in NP have been broadly described, while studies on oligodendrocytes have largely focused on axonal myelination. The mechanisms of oligodendrocytes and their interactions with other glial cells in NP development remain uncertain. METHODS: To explore the function of the interaction of the three glial cells and their interactions on myelin development in NP, we evaluated changes in NP and myelin morphology after a chronic constriction injury (CCI) model in mice, and used single-cell sequencing to reveal the subpopulations characteristics of oligodendrocytes, microglia, and astrocytes in the spinal cord tissues, as well as their relationship with myelin lesions; the proliferation and differentiation trajectories of oligodendrocyte subpopulations were also revealed using pseudotime cell trajectory and RNA velocity analysis. In addition, we identified chemokine ligand-receptor pairs between glial cells by cellular communication and verified them using immunofluorescence. RESULTS: Our study showed that NP peaked on day 7 after CCI in mice, a time at which myelin lesions were present in both the spinal cord and sciatic nerve. Oligodendrocytes, microglia, and astrocytes subpopulations in spinal cord tissue were heterogeneous after CCI and all were involved in suppressing the process of immune defense and myelin production. In addition, the differentiation trajectory of oligodendrocytes involved a unidirectional lattice process of OPC-1-Oligo-9, which was arrested at the Oligo-2 stage under the influence of microglia and astrocytes. And the CADM1-CADM1, NRP1-VEGFA interactions between glial cells are enhanced after CCI and they had a key role in myelin lesions and demyelination. CONCLUSIONS: Our study reveals the close relationship between the differentiation block of oligodendrocytes after CCI and their interaction with microglia and astrocytes-mediated myelin lesions and NP. CADM1/CADM1 and NRP-1/VEGFA may serve as potential therapeutic targets for use in the treatment of NP.


Sujet(s)
Souris de lignée C57BL , Gaine de myéline , Névralgie , Névroglie , Moelle spinale , Animaux , Souris , Moelle spinale/anatomopathologie , Moelle spinale/métabolisme , Gaine de myéline/anatomopathologie , Gaine de myéline/métabolisme , Névralgie/anatomopathologie , Névralgie/métabolisme , Névroglie/anatomopathologie , Névroglie/métabolisme , Mâle , Analyse sur cellule unique
17.
J Virol ; 98(8): e0056024, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39087762

RÉSUMÉ

Powassan virus (POWV) is an emergent tick-borne flavivirus that causes fatal encephalitis in the elderly and long-term neurologic sequelae in survivors. How age contributes to severe POWV encephalitis remains an enigma, and no animal models have assessed age-dependent POWV neuropathology. Inoculating C57BL/6 mice with a POWV strain (LI9) currently circulating in Ixodes ticks resulted in age-dependent POWV lethality 10-20 dpi. POWV infection of 50-week-old mice was 82% fatal with lethality sequentially reduced by age to 7.1% in 10-week-old mice. POWV LI9 was neuroinvasive in mice of all ages, causing acute spongiform CNS pathology and reactive gliosis 5-15 dpi that persisted in survivors 30 dpi. High CNS viral loads were found in all mice 10 dpi. However, by 15 dpi, viral loads decreased by 2-4 logs in 10- to 40-week-old mice, while remaining at high levels in 50-week-old mice. Age-dependent differences in CNS viral loads 15 dpi occurred concomitantly with striking changes in CNS cytokine responses. In the CNS of 50-week-old mice, POWV induced Th1-type cytokines (IFNγ, IL-2, IL-12, IL-4, TNFα, IL-6), suggesting a neurodegenerative pro-inflammatory M1 microglial program. By contrast, in 10-week-old mice, POWV-induced Th2-type cytokines (IL-10, TGFß, IL-4) were consistent with a neuroprotective M2 microglial phenotype. These findings correlate age-dependent CNS cytokine responses and viral loads with POWV lethality and suggest potential neuroinflammatory therapeutic targets. Our results establish the age-dependent lethality of POWV in a murine model that mirrors human POWV severity and long-term CNS pathology in the elderly. IMPORTANCE: Powassan virus is an emerging tick-borne flavivirus causing lethal encephalitis in aged individuals. We reveal an age-dependent POWV murine model that mirrors human POWV encephalitis and long-term CNS damage in the elderly. We found that POWV is neuroinvasive and directs reactive gliosis in all age mice, but at acute stages selectively induces pro-inflammatory Th1 cytokine responses in 50-week-old mice and neuroprotective Th2 cytokine responses in 10-week-old mice. Our findings associate CNS viral loads and divergent cytokine responses with age-dependent POWV lethality and survival outcomes. Responses of young mice suggest potential therapeutic targets and approaches for preventing severe POWV encephalitis that may be broadly applicable to other neurodegenerative diseases. Our age-dependent murine POWV model permits analysis of vaccines that prevent POWV lethality, and therapeutics that resolve severe POWV encephalitis.


Sujet(s)
Cytokines , Modèles animaux de maladie humaine , Virus de l'encéphalite à tiques (sous-groupe) , Encéphalites à tiques , Souris de lignée C57BL , Névroglie , Charge virale , Animaux , Souris , Virus de l'encéphalite à tiques (sous-groupe)/immunologie , Encéphalites à tiques/immunologie , Encéphalites à tiques/virologie , Encéphalites à tiques/mortalité , Encéphalites à tiques/anatomopathologie , Cytokines/métabolisme , Cytokines/immunologie , Névroglie/virologie , Névroglie/immunologie , Névroglie/anatomopathologie , Femelle , Facteurs âges , Ixodes/virologie , Ixodes/immunologie , Système nerveux central/virologie , Système nerveux central/immunologie , Système nerveux central/anatomopathologie , Encéphale/virologie , Encéphale/anatomopathologie , Encéphale/immunologie
18.
Cell Rep ; 43(8): 114586, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39137113

RÉSUMÉ

Our understanding of human fetal cerebellum development during the late second trimester, a critical period for the generation of astrocytes, oligodendrocytes, and unipolar brush cells (UBCs), remains limited. Here, we performed single-cell RNA sequencing (scRNA-seq) in human fetal cerebellum samples from gestational weeks (GWs) 18-25. We find that proliferating UBC progenitors distribute in the subventricular zone of the rhombic lip (RLSVZ) near white matter (WM), forming a layer structure. We also delineate two trajectories from astrogenic radial glia (ARGs) to Bergmann glial progenitors (BGPs) and recognize oligodendrogenic radial glia (ORGs) as one source of primitive oligodendrocyte progenitor cells (PriOPCs). Additionally, our scRNA-seq analysis of the trisomy 21 fetal cerebellum at this stage reveals abnormal upregulated genes in pathways such as the cell adhesion pathway and focal adhesion pathway, which potentially promote neuronal differentiation. Overall, our research provides valuable insights into normal and abnormal development of the human fetal cerebellum.


Sujet(s)
Cervelet , Syndrome de Down , Foetus , Deuxième trimestre de grossesse , Humains , Cervelet/embryologie , Cervelet/malformations , Cervelet/métabolisme , Syndrome de Down/génétique , Syndrome de Down/anatomopathologie , Grossesse , Femelle , Différenciation cellulaire , Oligodendroglie/métabolisme , Oligodendroglie/cytologie , Névroglie/métabolisme , Névroglie/anatomopathologie , Régulation de l'expression des gènes au cours du développement
19.
Lupus ; 33(10): 1116-1129, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39148457

RÉSUMÉ

BACKGROUND: Neuropsychiatric systemic lupus erythematosus (NPSLE) is a poorly understood and heterogeneous manifestation of SLE. Common major NPSLE syndromes include strokes, seizures, myelitis, and aseptic meningitis. Easily obtainable biomarkers are needed to assist in early diagnosis and improve outcomes for NPSLE. A frequent end-result of major syndromes is neuronal or glial injury. Blood-based neurofilament light (NfL) and glial fibrillary acidic protein (GFAP) have been utilized as markers for monitoring disease activity and/or severity in other neurodegenerative and neuroinflammatory diseases; however, they have not been evaluated in active major NPSLE. METHODS: This was a case-control study. We enrolled patients aged 12-60 years with active major NPSLE, SLE without active major NPSLE, and healthy controls. Active NPSLE was defined as being <6 months from last new or worsening neuropsychiatric symptom. Demographics, clinical data, and serum or plasma biosamples were collected. RESULTS: Thirteen patients with active major NPSLE, 13 age/sex/kidney function matched SLE controls without active major NPSLE, and 13 age/sex matched healthy controls (mean ages 26.8, 27.3, 26.6 years) were included. 92% of each group were female. Major syndromes included stroke (5), autonomic disorder (3), demyelinating disease (2), aseptic meningitis (2), sensorimotor polyneuropathy (2), cranial neuropathy (1), seizures (1), and myelopathy (2). Mean (standard deviation) blood NfL and GFAP were 3.6 pg/ml (2.0) and 50.4 pg/ml (15.0), respectively, for the healthy controls. Compared to healthy controls, SLE without active major NPSLE had mean blood NfL and GFAP levels 1.3 pg/ml (p = .42) and 1.2 pg/ml higher (p = .53), respectively. Blood NfL was on average 17.9 pg/ml higher (95% CI: 9.2, 34.5; p < .001) and blood GFAP was on average 3.2 pg/ml higher (95% CI: 1.9, 5.5; p < .001) for cases of active major NPSLE compared to SLE without active major NPSLE. In a subset of 6 patients sampled at multiple time points, blood NfL and GFAP decreased after immunotherapy. CONCLUSIONS: Blood NfL and GFAP levels are elevated in persons with SLE with active major NPSLE compared to disease matched controls and may lower after immunotherapy initiation. Larger and longitudinal studies are needed to ascertain their utility in a clinical setting.


Sujet(s)
Marqueurs biologiques , Protéine gliofibrillaire acide , Vascularite lupique du système nerveux central , Protéines neurofilamenteuses , Humains , Femelle , Marqueurs biologiques/sang , Études cas-témoins , Adulte , Mâle , Vascularite lupique du système nerveux central/sang , Protéine gliofibrillaire acide/sang , Protéines neurofilamenteuses/sang , Jeune adulte , Adolescent , Adulte d'âge moyen , Enfant , Névroglie/anatomopathologie , Névroglie/métabolisme , Neurones/anatomopathologie
20.
Neurobiol Dis ; 200: 106631, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39111701

RÉSUMÉ

Friedreich ataxia is a hereditary neurodegenerative disorder resulting from reduced levels of the protein frataxin due to an expanded GAA repeat in the FXN gene. This deficiency causes progressive degeneration of specific neuronal populations in the cerebellum and the consequent loss of movement coordination and equilibrium, which are some of the main symptoms observed in affected individuals. Like in other neurodegenerative diseases, previous studies suggest that glial cells could be involved in the neurodegenerative process and disease progression in patients with Friedreich ataxia. In this work, we followed and characterized the progression of changes in the cerebellar cortex in the latest version of Friedreich ataxia humanized mouse model, YG8-800 (Fxnnull:YG8s(GAA)>800), which carries a human FXN transgene containing >800 GAA repeats. Comparative analyses of behavioral, histopathological, and biochemical parameters were conducted between the control strain Y47R and YG8-800 mice at different time points. Our findings revealed that YG8-800 mice exhibit an ataxic phenotype characterized by poor motor coordination, decreased body weight, cerebellar atrophy, neuronal loss, and changes in synaptic proteins. Additionally, early activation of glial cells, predominantly astrocytes and microglia, was observed preceding neuronal degeneration, as was increased expression of key proinflammatory cytokines and downregulation of neurotrophic factors. Together, our results show that the YG8-800 mouse model exhibits a stronger phenotype than previous experimental murine models, reliably recapitulating some of the features observed in humans. Accordingly, this humanized model could represent a valuable tool for studying Friedreich ataxia molecular disease mechanisms and for preclinical evaluation of possible therapies.


Sujet(s)
Cortex cérébelleux , Modèles animaux de maladie humaine , , Ataxie de Friedreich , Souris transgéniques , Névroglie , Ataxie de Friedreich/anatomopathologie , Ataxie de Friedreich/métabolisme , Ataxie de Friedreich/génétique , Animaux , Névroglie/métabolisme , Névroglie/anatomopathologie , Cortex cérébelleux/métabolisme , Cortex cérébelleux/anatomopathologie , Souris , Protéines de liaison au fer/génétique , Protéines de liaison au fer/métabolisme , Humains , Dégénérescence nerveuse/anatomopathologie , Dégénérescence nerveuse/métabolisme , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE