Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.781
Filter
1.
Acta Neuropathol ; 147(1): 78, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38695952

ABSTRACT

Aging is associated with cell senescence and is the major risk factor for AD. We characterized premature cell senescence in postmortem brains from non-diseased controls (NDC) and donors with Alzheimer's disease (AD) using imaging mass cytometry (IMC) and single nuclear RNA (snRNA) sequencing (> 200,000 nuclei). We found increases in numbers of glia immunostaining for galactosidase beta (> fourfold) and p16INK4A (up to twofold) with AD relative to NDC. Increased glial expression of genes related to senescence was associated with greater ß-amyloid load. Prematurely senescent microglia downregulated phagocytic pathways suggesting reduced capacity for ß-amyloid clearance. Gene set enrichment and pseudo-time trajectories described extensive DNA double-strand breaks (DSBs), mitochondrial dysfunction and ER stress associated with increased ß-amyloid leading to premature senescence in microglia. We replicated these observations with independent AD snRNA-seq datasets. Our results describe a burden of senescent glia with AD that is sufficiently high to contribute to disease progression. These findings support the hypothesis that microglia are a primary target for senolytic treatments in AD.


Subject(s)
Alzheimer Disease , Cellular Senescence , Transcriptome , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Cellular Senescence/physiology , Cellular Senescence/genetics , Aged , Male , Aged, 80 and over , Female , Microglia/pathology , Microglia/metabolism , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Neuroglia/pathology , Neuroglia/metabolism
2.
J Neurosci ; 44(20)2024 May 15.
Article in English | MEDLINE | ID: mdl-38589228

ABSTRACT

Protein misfolding, aggregation, and spread through the brain are primary drivers of neurodegenerative disease pathogenesis. Phagocytic glia are responsible for regulating the load of pathological proteins in the brain, but emerging evidence suggests that glia may also act as vectors for aggregate spread. Accumulation of protein aggregates could compromise the ability of glia to eliminate toxic materials from the brain by disrupting efficient degradation in the phagolysosomal system. A better understanding of phagocytic glial cell deficiencies in the disease state could help to identify novel therapeutic targets for multiple neurological disorders. Here, we report that mutant huntingtin (mHTT) aggregates impair glial responsiveness to injury and capacity to degrade neuronal debris in male and female adult Drosophila expressing the gene that causes Huntington's disease (HD). mHTT aggregate formation in neurons impairs engulfment and clearance of injured axons and causes accumulation of phagolysosomes in glia. Neuronal mHTT expression induces upregulation of key innate immunity and phagocytic genes, some of which were found to regulate mHTT aggregate burden in the brain. A forward genetic screen revealed Rab10 as a novel component of Draper-dependent phagocytosis that regulates mHTT aggregate transmission from neurons to glia. These data suggest that glial phagocytic defects enable engulfed mHTT aggregates to evade lysosomal degradation and acquire prion-like characteristics. Together, our findings uncover new mechanisms that enhance our understanding of the beneficial and harmful effects of phagocytic glia in HD and other neurodegenerative diseases.


Subject(s)
Disease Models, Animal , Drosophila Proteins , Drosophila , Huntingtin Protein , Huntington Disease , Neuroglia , Animals , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/genetics , Neuroglia/metabolism , Neuroglia/pathology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Female , Male , Phagocytosis/physiology , Lysosomes/metabolism , Phagosomes/metabolism , Animals, Genetically Modified , Prions/metabolism , Prions/genetics , Neurons/metabolism
3.
J Neuroimmunol ; 390: 578346, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38648696

ABSTRACT

The frequency of corticospinal tract (CST) T2/FLAIR hyperintensity in disorders with neuroglial antibodies is unclear. Herein, we retrospectively reviewed brain MRIs of 101 LGI1-antibody encephalitis patients, and observed CST hyperintensity in 30/101 (30%). It was mostly bilateral (93%), not associated with upper motor neuron signs/symptoms (7%), and frequently decreased over time (39%). In a systematic review including patients with other neuroglial antibodies, CST hyperintensity was reported in 110 with neuromyelitis optica (94%), myelin oligodendrocyte glycoprotein-associated disease (2%), Ma2-antibody (3%) and GAD65-antibody paraneoplastic neurological syndrome (1%). CST hyperintensity is not an infrequent finding in LGI1-Ab encephalitis and other disorders with neuroglial antibodies.


Subject(s)
Autoantibodies , Encephalitis , Intracellular Signaling Peptides and Proteins , Pyramidal Tracts , Humans , Autoantibodies/immunology , Autoantibodies/blood , Female , Middle Aged , Male , Retrospective Studies , Aged , Adult , Encephalitis/immunology , Encephalitis/diagnostic imaging , Pyramidal Tracts/diagnostic imaging , Pyramidal Tracts/pathology , Pyramidal Tracts/immunology , Intracellular Signaling Peptides and Proteins/immunology , Magnetic Resonance Imaging , Young Adult , Neuroglia/pathology , Neuroglia/immunology , Adolescent , Aged, 80 and over , Central Nervous System Diseases/immunology , Central Nervous System Diseases/diagnostic imaging
4.
Biomed Pharmacother ; 174: 116526, 2024 May.
Article in English | MEDLINE | ID: mdl-38574621

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a debilitating neurodegenerative disorder of the cerebellum and brainstem. Memantine has been proposed as a potential treatment for SCA1. It blocks N-methyl-D-aspartate (NMDA) receptors on neurons, reduces excitotoxicity and decreases neurodegeneration in Alzheimer models. However, in cerebellar neurodegenerative diseases, the potential value of memantine is still unclear. We investigated the effects of memantine on motor performance and synaptic transmission in the cerebellum in a mouse model where mutant ataxin 1 is specifically targeted to glia. Lentiviral vectors (LVV) were used to express mutant ataxin 1 selectively in Bergmann glia (BG). In mice transduced with the mutant ataxin 1, chronic treatment with memantine improved motor activity during initial tests, presumably due to preserved BG and Purkinje cell (PC) morphology and numbers. However, mice were unable to improve their rota rod scores during next days of training. Memantine also compromised improvement in the rota rod scores in control mice upon repetitive training. These effects may be due to the effects of memantine on plasticity (LTD suppression) and NMDA receptor modulation. Some effects of chronically administered memantine persisted even after its wash-out from brain slices. Chronic memantine reduced morphological signs of neurodegeneration in the cerebellum of SCA1 model mice. This resulted in an apparent initial reduction of ataxic phenotype, but memantine also affected cerebellar plasticity and ultimately compromised motor learning. We speculate that that clinical application of memantine in SCA1 might be hampered by its ability to suppress NMDA-dependent plasticity in cerebellar cortex.


Subject(s)
Disease Models, Animal , Memantine , Phenotype , Spinocerebellar Ataxias , Animals , Memantine/pharmacology , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/pathology , Mice , Ataxin-1/metabolism , Ataxin-1/genetics , Motor Activity/drug effects , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/metabolism , Purkinje Cells/drug effects , Purkinje Cells/pathology , Purkinje Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Neuroglia/drug effects , Neuroglia/pathology , Neuroglia/metabolism , Male , Neuronal Plasticity/drug effects
5.
J Neurosci Res ; 102(4): e25336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656664

ABSTRACT

Chronic neuroinflammation has been implicated in neurodegenerative disease pathogenesis. A key feature of neuroinflammation is neuronal loss and glial activation, including microglia and astrocytes. 4R-cembranoid (4R) is a natural compound that inhibits hippocampal pro-inflammatory cytokines and increases memory function in mice. We used the lipopolysaccharide (LPS) injection model to study the effect of 4R on neuronal density and microglia and astrocyte activation. C57BL/6J wild-type mice were injected with LPS (5 mg/kg) and 2 h later received either 4R (6 mg/kg) or vehicle. Mice were sacrificed after 72 h for analysis of brain pathology. Confocal images of brain sections immunostained for microglial, astrocyte, and neuronal markers were used to quantify cellular hippocampal phenotypes and neurons. Hippocampal lysates were used to measure the expression levels of neuronal nuclear protein (NeuN), inducible nitrous oxide synthase (iNOS), arginase-1, thrombospondin-1 (THBS1), glial cell-derived neurotrophic factor (GDNF), and orosomucoid-2 (ORM2) by western blot. iNOS and arginase-1 are widely used protein markers of pro- and anti-inflammatory microglia, respectively. GDNF promotes neuronal survival, and ORM2 and THBS1 are astrocytic proteins that regulate synaptic plasticity and inhibit microglial activation. 4R administration significantly reduced neuronal loss and the number of pro-inflammatory microglia 72 h after LPS injection. It also decreased the expression of the pro-inflammatory protein iNOS while increasing arginase-1 expression, supporting its anti-inflammatory role. The protein expression of THBS1, GDNF, and ORM2 was increased by 4R. Our data show that 4R preserves the integrity of hippocampal neurons against LPS-induced neuroinflammation in mice.


Subject(s)
Hippocampus , Lipopolysaccharides , Mice, Inbred C57BL , Neuroglia , Neurons , Animals , Lipopolysaccharides/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/drug therapy , Phenotype , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology
6.
J Neuroinflammation ; 21(1): 105, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649885

ABSTRACT

BACKGROUND: NADPH oxidase (NOX), a primary source of endothelial reactive oxygen species (ROS), is considered a key event in disrupting the integrity of the blood-retinal barrier. Abnormalities in neurovascular-coupled immune signaling herald the loss of ganglion cells in glaucoma. Persistent microglia-driven inflammation and cellular innate immune system dysregulation often lead to deteriorating retinal degeneration. However, the crosstalk between NOX and the retinal immune environment remains unresolved. Here, we investigate the interaction between oxidative stress and neuroinflammation in glaucoma by genetic defects of NOX2 or its regulation via gp91ds-tat. METHODS: Ex vivo cultures of retinal explants from wildtype C57BL/6J and Nox2 -/- mice were subjected to normal and high hydrostatic pressure (Pressure 60 mmHg) for 24 h. In vivo, high intraocular pressure (H-IOP) was induced in C57BL/6J mice for two weeks. Both Pressure 60 mmHg retinas and H-IOP mice were treated with either gp91ds-tat (a NOX2-specific inhibitor). Proteomic analysis was performed on control, H-IOP, and treatment with gp91ds-tat retinas to identify differentially expressed proteins (DEPs). The study also evaluated various glaucoma phenotypes, including IOP, retinal ganglion cell (RGC) functionality, and optic nerve (ON) degeneration. The superoxide (O2-) levels assay, blood-retinal barrier degradation, gliosis, neuroinflammation, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative PCR were performed in this study. RESULTS: We found that NOX2-specific deletion or activity inhibition effectively attenuated retinal oxidative stress, immune dysregulation, the internal blood-retinal barrier (iBRB) injury, neurovascular unit (NVU) dysfunction, RGC loss, and ON axonal degeneration following H-IOP. Mechanistically, we unveiled for the first time that NOX2-dependent ROS-driven pro-inflammatory signaling, where NOX2/ROS induces endothelium-derived endothelin-1 (ET-1) overexpression, which activates the ERK1/2 signaling pathway and mediates the shift of microglia activation to a pro-inflammatory M1 phenotype, thereby triggering a neuroinflammatory outburst. CONCLUSIONS: Collectively, we demonstrate for the first time that NOX2 deletion or gp91ds-tat inhibition attenuates iBRB injury and NVU dysfunction to rescue glaucomatous RGC loss and ON axon degeneration, which is associated with inhibition of the ET-1/ERK1/2-transduced shift of microglial cell activation toward a pro-inflammatory M1 phenotype, highlighting NOX2 as a potential target for novel neuroprotective therapies in glaucoma management.


Subject(s)
Blood-Retinal Barrier , Intraocular Pressure , Mice, Inbred C57BL , NADPH Oxidase 2 , Neuroinflammatory Diseases , Animals , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Mice , Blood-Retinal Barrier/pathology , Blood-Retinal Barrier/metabolism , Intraocular Pressure/physiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Knockout , Cell Proliferation/physiology , MAP Kinase Signaling System/physiology , Neuroglia/metabolism , Neuroglia/pathology , Ocular Hypertension/pathology , Ocular Hypertension/metabolism , Glaucoma/pathology , Glaucoma/metabolism , Oxidative Stress/physiology
7.
Alzheimers Dement ; 20(4): 2922-2942, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460121

ABSTRACT

INTRODUCTION: The BIN1 coding variant rs138047593 (K358R) is linked to Late-Onset Alzheimer's Disease (LOAD) via targeted exome sequencing. METHODS: To elucidate the functional consequences of this rare coding variant on brain amyloidosis and neuroinflammation, we generated BIN1K358R knock-in mice using CRISPR/Cas9 technology. These mice were subsequently bred with 5xFAD transgenic mice, which serve as a model for Alzheimer's pathology. RESULTS: The presence of the BIN1K358R variant leads to increased cerebral amyloid deposition, with a dampened response of astrocytes and oligodendrocytes, but not microglia, at both the cellular and transcriptional levels. This correlates with decreased neurofilament light chain in both plasma and brain tissue. Synaptic densities are significantly increased in both wild-type and 5xFAD backgrounds homozygous for the BIN1K358R variant. DISCUSSION: The BIN1 K358R variant modulates amyloid pathology in 5xFAD mice, attenuates the astrocytic and oligodendrocytic responses to amyloid plaques, decreases damage markers, and elevates synaptic densities. HIGHLIGHTS: BIN1 rs138047593 (K358R) coding variant is associated with increased risk of LOAD. BIN1 K358R variant increases amyloid plaque load in 12-month-old 5xFAD mice. BIN1 K358R variant dampens astrocytic and oligodendrocytic response to plaques. BIN1 K358R variant decreases neuronal damage in 5xFAD mice. BIN1 K358R upregulates synaptic densities and modulates synaptic transmission.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Neuroglia/pathology , Plaque, Amyloid/pathology , Humans
8.
Exp Neurol ; 376: 114756, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508482

ABSTRACT

Overexpression of the Ube3a gene and the resulting increase in Ube3a protein are linked to autism spectrum disorder (ASD). However, the cellular and molecular processes underlying Ube3a-dependent ASD remain unclear. Using both male and female mice, we find that neurons in the somatosensory cortex of the Ube3a 2× Tg ASD mouse model display reduced dendritic spine density and increased immature filopodia density. Importantly, the increased gene dosage of Ube3a in astrocytes alone is sufficient to confer alterations in neurons as immature dendritic protrusions, as observed in primary hippocampal neuron cultures. We show that Ube3a overexpression in astrocytes leads to a loss of astrocyte-derived spinogenic protein, thrombospondin-2 (TSP2), due to a suppression of TSP2 gene transcription. By neonatal intraventricular injection of astrocyte-specific virus, we demonstrate that Ube3a overexpression in astrocytes in vivo results in a reduction in dendritic spine maturation in prelimbic cortical neurons, accompanied with autistic-like behaviors in mice. These findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels. SIGNIFICANCE STATEMENT: Increased gene dosage of Ube3a is tied to autism spectrum disorders (ASDs), yet cellular and molecular alterations underlying autistic phenotypes remain unclear. We show that Ube3a overexpression leads to impaired dendritic spine maturation, resulting in reduced spine density and increased filopodia density. We find that dysregulation of spine development is not neuron autonomous, rather, it is mediated by an astrocytic mechanism. Increased gene dosage of Ube3a in astrocytes leads to reduced production of the spinogenic glycoprotein thrombospondin-2 (TSP2), leading to abnormalities in spines. Astrocyte-specific Ube3a overexpression in the brain in vivo confers dysregulated spine maturation concomitant with autistic-like behaviors in mice. These findings indicate the importance of astrocytes in aberrant neurodevelopment and brain function in Ube3a-depdendent ASD.


Subject(s)
Autism Spectrum Disorder , Dendritic Spines , Ubiquitin-Protein Ligases , Animals , Mice , Female , Dendritic Spines/pathology , Dendritic Spines/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Male , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Neurons/metabolism , Neurons/pathology , Thrombospondins/metabolism , Thrombospondins/genetics , Thrombospondins/biosynthesis , Neuroglia/metabolism , Neuroglia/pathology , Mice, Transgenic , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology , Cells, Cultured , Neurogenesis/physiology , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology
9.
Cell ; 187(8): 1990-2009.e19, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38513664

ABSTRACT

Multiple sclerosis (MS) is a neurological disease characterized by multifocal lesions and smoldering pathology. Although single-cell analyses provided insights into cytopathology, evolving cellular processes underlying MS remain poorly understood. We investigated the cellular dynamics of MS by modeling temporal and regional rates of disease progression in mouse experimental autoimmune encephalomyelitis (EAE). By performing single-cell spatial expression profiling using in situ sequencing (ISS), we annotated disease neighborhoods and found centrifugal evolution of active lesions. We demonstrated that disease-associated (DA)-glia arise independently of lesions and are dynamically induced and resolved over the disease course. Single-cell spatial mapping of human archival MS spinal cords confirmed the differential distribution of homeostatic and DA-glia, enabled deconvolution of active and inactive lesions into sub-compartments, and identified new lesion areas. By establishing a spatial resource of mouse and human MS neuropathology at a single-cell resolution, our study unveils the intricate cellular dynamics underlying MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Spinal Cord , Animals , Humans , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Mice , Single-Cell Gene Expression Analysis , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroglia/metabolism , Neuroglia/pathology
10.
Mov Disord ; 39(4): 723-728, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38357858

ABSTRACT

BACKGROUND: The architecture and composition of glial (GCI) and neuronal (NCI) α-synuclein inclusions observed in multiple system atrophy (MSA) remain to be precisely defined to better understand the disease. METHODS: Here, we used stochastic optical reconstruction microscopy (STORM) to characterize the nanoscale organization of glial (GCI) and neuronal (NCI) α-synuclein inclusions in cryopreserved brain sections from MSA patients. RESULTS: STORM revealed a dense cross-linked internal structure of α-synuclein in all GCI and NCI. The internal architecture of hyperphosphorylated α-synuclein (p-αSyn) inclusions was similar in glial and neuronal cells, suggesting a common aggregation mechanism. A similar sequence of p-αSyn stepwise intracellular aggregation was defined in oligodendrocytes and neurons, starting from the perinuclear area and growing inside the cells. Consistent with this hypothesis, we found a higher mitochondrial density in GCI and NCI compared to oligodendrocytes and neurons from unaffected donors (P < 0.01), suggesting an active recruitment of the organelles during the aggregation process. CONCLUSIONS: These first STORM images of GCI and NCI suggest stepwise α-synuclein aggregation in MSA. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Inclusion Bodies , Multiple System Atrophy , Neurons , alpha-Synuclein , Humans , Multiple System Atrophy/pathology , Multiple System Atrophy/metabolism , alpha-Synuclein/metabolism , Inclusion Bodies/pathology , Inclusion Bodies/metabolism , Neurons/metabolism , Neurons/pathology , Female , Aged , Male , Middle Aged , Brain/pathology , Brain/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/pathology , Oligodendroglia/metabolism , Microscopy/methods
11.
Exp Neurol ; 374: 114716, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38331161

ABSTRACT

SOD1 gene is associated with progressive motor neuron degeneration in the familiar forms of amyotrophic lateral sclerosis. Although studies on mutant human SOD1 transgenic rodent models have provided important insights into disease pathogenesis, they have not led to the discovery of early biomarkers or effective therapies in human disease. The recent generation of a transgenic swine model expressing the human pathological hSOD1G93A gene, which recapitulates the course of human disease, represents an interesting tool for the identification of early disease mechanisms and diagnostic biomarkers. Here, we analyze the activation state of CNS cells in transgenic pigs during the disease course and investigate whether changes in neuronal and glial cell activation state can be reflected by the amount of extracellular vesicles they release in biological fluids. To assess the activation state of neural cells, we performed a biochemical characterization of neurons and glial cells in the spinal cords of hSOD1G93A pigs during the disease course. Quantification of EVs of CNS cell origin was performed in cerebrospinal fluid and plasma of transgenic pigs at different disease stages by Western blot and peptide microarray analyses. We report an early activation of oligodendrocytes in hSOD1G93A transgenic tissue followed by astrocyte and microglia activation, especially in animals with motor symptoms. At late asymptomatic stage, EV production from astrocytes and microglia is increased in the cerebrospinal fluid, but not in the plasma, of transgenic pigs reflecting donor cell activation in the spinal cord. Estimation of EV production by biochemical analyses is corroborated by direct quantification of neuron- and microglia-derived EVs in the cerebrospinal fluid by a Membrane Sensing Peptide enabled on-chip analysis that provides fast results and low sample consumption. Collectively, our data indicate that alteration in astrocytic EV production precedes the onset of disease symptoms in the hSODG93A swine model, mirroring donor cell activation in the spinal cord, and suggest that EV measurements from the cells first activated in the ALS pig model, i.e. OPCs, may further improve early disease detection.


Subject(s)
Amyotrophic Lateral Sclerosis , Extracellular Vesicles , Mice , Animals , Humans , Swine , Superoxide Dismutase-1/genetics , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Mice, Transgenic , Amyotrophic Lateral Sclerosis/pathology , Spinal Cord/pathology , Neuroglia/pathology , Biomarkers/metabolism , Peptides/metabolism , Disease Models, Animal
12.
Methods Mol Biol ; 2764: 165-176, 2024.
Article in English | MEDLINE | ID: mdl-38393595

ABSTRACT

The macro-metastasis/organ parenchyma interface (MMPI) is gaining increasing significance due to its prognostic relevance for cancer (brain) metastasis. We have developed an organotypic 3D ex vivo co-culture model that mimics the MMPI and allows us to evaluate the histopathological growth pattern (HGP) and infiltration grade of the tumor cells into the neighboring brain tissue and to study the interactions of cancer and glial cells ex vivo. This system consists of a murine brain slice and a 3D tumor plug that can be co-cultured for several days. After slicing the brain of 5- to 8-day-old mice, a Matrigel plug containing fluorescent-labelled tumor cells is placed next to it, so that tumor cells in the 3D plug and glial cells in the brain slice can interact at the interface for up to 96 h. To facilitate the positioning of the co-culture and increase the reproducibility of the model, a brain spacer can be used. The HGP and infiltration of the tumor cells into the brain slice as well as the activation of the glial cells can be assessed by live and/or confocal microscopy after immunofluorescence staining of microglia and/or astrocytes. Alternatively, the co-culture can also be used for other purposes, such as RNA analysis. This organotypic 3D ex vivo co-culture offers a perfect tool for preliminary screenings before in vivo experiments and reduces the number of animals, thus contributing to the 3R concept as a central precept in preclinical research.


Subject(s)
Brain Neoplasms , Neuroglia , Mice , Animals , Coculture Techniques , Reproducibility of Results , Neuroglia/pathology , Brain Neoplasms/pathology , Brain/pathology , Organ Culture Techniques
13.
Mol Neurobiol ; 61(4): 2081-2098, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37840070

ABSTRACT

The gastrointestinal tract is the largest immune organ and it receives dense innervation from intrinsic (enteric) and extrinsic (sympathetic, parasympathetic, and somatosensory) neurons. The immune and neural systems of the gut communicate with each other and their interactions shape gut defensive mechanisms and neural-controlled gut functions such as motility and secretion. Changes in neuroimmune interactions play central roles in the pathogenesis of diseases such as Parkinson's disease (PD), which is a multicentric disorder that is heterogeneous in its manifestation and pathogenesis. Non-motor and premotor symptoms of PD are common in the gastrointestinal tract and the gut is considered a potential initiation site for PD in some cases. How the enteric nervous system and neuroimmune signaling contribute to PD disease progression is an emerging area of interest. This review focuses on intestinal neuroimmune loops such as the neuroepithelial unit, enteric glial cells and their immunomodulatory effects, anti-inflammatory cholinergic signaling and the relationship between myenteric neurons and muscularis macrophages, and the role of α-synuclein in gut immunity. Special consideration is given to the discussion of intestinal neuroimmune connectomes during PD and their possible implications for various aspects of the disease.


Subject(s)
Connectome , Enteric Nervous System , Parkinson Disease , Humans , Parkinson Disease/pathology , Enteric Nervous System/pathology , alpha-Synuclein , Neurons/pathology , Neuroglia/pathology
14.
Cell Prolif ; 57(1): e13536, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37551711

ABSTRACT

Enteric glial cells in the enteric nervous system are critical for the regulation of gastrointestinal homeostasis. Increasing evidence suggests two-way communication between enteric glial cells and both enteric neurons and immune cells. These interactions may be important in the pathogenesis of Crohn's disease (CD), a chronic relapsing disease characterized by a dysregulated immune response. Structural abnormalities in glial cells have been identified in CD. Furthermore, classical inflammatory pathways associated with CD (e.g., the nuclear factor kappa-B pathway) function in enteric glial cells. However, the specific mechanisms by which enteric glial cells contribute to CD have not been summarized in detail. In this review, we describe the possible roles of enteric glial cells in the pathogenesis of CD, including the roles of glia-immune interactions, neuronal modulation, neural plasticity, and barrier integrity. Additionally, the implications for the development of therapeutic strategies for CD based on enteric glial cell-mediated pathogenic processes are discussed.


Subject(s)
Crohn Disease , Enteric Nervous System , Humans , Crohn Disease/metabolism , Neuroglia/pathology , Neurons/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology
15.
Curr Neuropharmacol ; 22(2): 204-216, 2024.
Article in English | MEDLINE | ID: mdl-37581321

ABSTRACT

Pathological pain imposes a huge burden on the economy and the lives of patients. At present, drugs used for the treatment of pathological pain have only modest efficacy and are also plagued by adverse effects and risk for misuse and abuse. Therefore, understanding the mechanisms of pathological pain is essential for the development of novel analgesics. Several lines of evidence indicate that interleukin-17 (IL-17) is upregulated in rodent models of pathological pain in the periphery and central nervous system. Besides, the administration of IL-17 antibody alleviated pathological pain. Moreover, IL-17 administration led to mechanical allodynia which was alleviated by the IL-17 antibody. In this review, we summarized and discussed the therapeutic potential of targeting IL-17 for pathological pain. The upregulation of IL-17 promoted the development of pathological pain by promoting neuroinflammation, enhancing the excitability of dorsal root ganglion neurons, and promoting the communication of glial cells and neurons in the spinal cord. In general, the existing research shows that IL-17 is an attractive therapeutic target for pathologic pain, but the underlying mechanisms still need to be investigated.


Subject(s)
Interleukin-17 , Pain , Rats , Animals , Humans , Rats, Sprague-Dawley , Pain/drug therapy , Pain/pathology , Hyperalgesia/pathology , Neuroglia/pathology
16.
J Neurosci Methods ; 402: 110029, 2024 02.
Article in English | MEDLINE | ID: mdl-38042304

ABSTRACT

BACKGROUND: The blood-brain barrier (BBB) is a specialized layer between blood vessels and tissue in the brain, which is comprised of a neuro-glia-vascular (NGV) unit, thus play a vital role in various brain diseases. NEW METHOD: We developed the in vitro NGV units by co-culturing brain microvascular endothelial cells (BMECs; bEnd.3) and primary neural stem cells extracted from subventricular zone of adult mice. This approach was designed to mimic the RNA profile conditions found in the microvessels of a mouse brain and confirmed through various comparative transcriptome analyses. RESULTS: Optimal NGV unit development was achieved by adjusting cell density-dependent co-culture ratios. Specifically, the morphogenic development and neuronal association of astrocyte endfeet were well observed in the contact region with BMECs in the NGV unit. Through transcriptome analysis, we compared co-cultured bEnd.3/NSCs with monocultured bEnd.3 or NSCs and additionally compared them with previously reported mouse brain vascular tissue to show that this NGV unit model is a suitable in vitro model for neurological disease such as Alzheimer's disease (AD). COMPARISON WITH EXISTING METHOD(S): This in vitro NGV unit was formed from neural stem cells and vascular cells in the brain of adult mice, not embryos. It is very useful for studying brain disease mechanisms by identifying proteins and genes associated with diseases progress. CONCLUSIONS: We suggest that this simple in vitro NGV model is appropriate to investigate the relationship between BBB changes and pathological factors in the fields of neurovascular biology and cerebrovascular diseases including AD.


Subject(s)
Neural Stem Cells , Animals , Mice , Alzheimer Disease/pathology , Blood-Brain Barrier/physiology , Brain , Coculture Techniques , Endothelial Cells/physiology , Gene Expression Profiling , Neuroglia/pathology
17.
Adv Mater ; 36(3): e2307747, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37990971

ABSTRACT

Current treatments for modulating the glial-mediated inflammatory response after spinal cord injury (SCI) have limited ability to improve recovery. This is quite likely due to the lack of a selective therapeutic approach acting on microgliosis and astrocytosis, the glia components most involved after trauma, while maximizing efficacy and minimizing side effects. A new nanogel that can selectively release active compounds in microglial cells and astrocytes is developed and characterized. The degree of selectivity and subcellular distribution of the nanogel is evaluated by applying an innovative super-resolution microscopy technique, expansion microscopy. Two different administration schemes are then tested in a SCI mouse model: in an early phase, the nanogel loaded with Rolipram, an anti-inflammatory drug, achieves significant improvement in the animal's motor performance due to the increased recruitment of microglia and macrophages that are able to localize the lesion. Treatment in the late phase, however, gives opposite results, with worse motor recovery because of the widespread degeneration. These findings demonstrate that the nanovector can be selective and functional in the treatment of the glial component in different phases of SCI. They also open a new therapeutic scenario for tackling glia-mediated inflammation after neurodegenerative events in the central nervous system.


Subject(s)
Polyethylene Glycols , Polyethyleneimine , Spinal Cord Injuries , Mice , Animals , Nanogels/therapeutic use , Spinal Cord Injuries/pathology , Neuroglia/pathology , Microglia
18.
Neurol Sci ; 45(5): 1875-1883, 2024 May.
Article in English | MEDLINE | ID: mdl-38133856

ABSTRACT

Alzheimer's disease (AD) is the prevailing type of dementia in the elderly, yet a comprehensive comprehension of its precise underlying mechanisms remains elusive. The investigation of the involvement of cerebral small veins in the advancement of AD has yet to be sufficiently explored in previous studies, primarily due to constraints associated with pathological staining techniques. However, recent research has provided valuable insights into multiple pathophysiological occurrences concerning cerebral small veins in AD, which may manifest sequentially, concurrently, or in a self-perpetuating manner. These events are presumed to be among the initial processes in the disease's progression. The impact of cerebral small vein loss on amyloid beta (Aß) clearance through the glial lymphatic system is noteworthy. There exists a potential interdependence between collagen deposition and Aß deposition in cerebral small veins. The compromised functionality of cerebral small veins can result in decreased cerebral perfusion pressure, potentially leading to cerebral tissue ischemia and edema. Additionally, the reduction of cerebral small veins may facilitate the infiltration of inflammatory factors into the brain parenchyma, thereby eliciting neuroinflammatory responses. Susceptibility-weighted imaging (SWI) is a valuable modality for the efficient assessment of cerebral small veins, precisely the deep medullary vein (DMV), and holds promise for the identification of precise and reliable imaging biomarkers for AD. This review presents a comprehensive overview of the current advancements and obstacles to the impairment of cerebral small veins in AD. Additionally, we emphasize future research avenues and the importance of conducting further investigations in this domain.


Subject(s)
Alzheimer Disease , Humans , Aged , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Neuroglia/pathology
19.
Int J Cancer ; 154(7): 1298-1308, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38146864

ABSTRACT

Here, we describe a blood test for the detection of glial malignancies (GLI-M) based on the identification of circulating glial cells (CGCs). The test is highly specific for GLI-M and can detect multiple grades (II-IV) and subtypes including gliomas, astrocytomas, oligodendrogliomas, oligoastrocytomas and glioblastomas, irrespective of gender and age. Analytical validation of the test was performed as per Clinical and Laboratory Standards Institute (CLSI) guidelines. Real-world performance characteristics of the test were evaluated in four clinical (observational) studies. The test has high analytical sensitivity (95%), specificity (100%) and precision (coefficient of variation [CV] = 13.7% for repeatability and CV = 23.5% for within laboratory precision, both at the detection threshold) and is not prone to interference from common drugs and serum factors. The ability of the test to detect and differentiate GLI-M from non-malignant brain tumours (NBT), brain metastases from primary epithelial malignancies (EPI-M) and healthy individual donors (HD) was evaluated in four clinical cohorts. Across these clinical studies, the test showed 99.35% sensitivity (95% confidence interval [CI]: 96.44%-99.98%) and 100% specificity (95% CI: 99.37%-100%). The performance characteristics of this test support its clinical utility for diagnostic triaging of individuals presenting with intracranial space-occupying lesions (ICSOL).


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Oligodendroglioma , Humans , Astrocytoma/diagnosis , Astrocytoma/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Glioma/pathology , Neuroglia/pathology , Oligodendroglioma/diagnosis , Oligodendroglioma/pathology , Observational Studies as Topic
20.
Mol Neurodegener ; 18(1): 75, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37858176

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of both upper and lower motor neurons, resulting in muscle weakness, atrophy, paralysis, and eventually death. Motor cortical hyperexcitability is a common phenomenon observed at the presymptomatic stage of ALS. Both cell-autonomous (the intrinsic properties of motor neurons) and non-cell-autonomous mechanisms (cells other than motor neurons) are believed to contribute to cortical hyperexcitability. Decoding the pathological relevance of these dynamic changes in motor neurons and glial cells has remained a major challenge. This review summarizes the evidence of cortical hyperexcitability from both clinical and preclinical research, as well as the underlying mechanisms. We discuss the potential role of glial cells, particularly microglia, in regulating abnormal neuronal activity during the disease progression. Identifying early changes such as neuronal hyperexcitability in the motor system may provide new insights for earlier diagnosis of ALS and reveal novel targets to halt the disease progression.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Neuroglia/pathology , Microglia/pathology , Disease Progression
SELECTION OF CITATIONS
SEARCH DETAIL
...