Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40.826
Filter
1.
Biomaterials ; 312: 122707, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121729

ABSTRACT

Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.


Subject(s)
Astrocytes , Glutamic Acid , Heterogeneous-Nuclear Ribonucleoproteins , Homeostasis , NF-E2-Related Factor 2 , Polypyrimidine Tract-Binding Protein , RNA, Small Interfering , Animals , Astrocytes/metabolism , Glutamic Acid/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , NF-E2-Related Factor 2/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , Signal Transduction , Cell Membrane/metabolism , Mice, Inbred C57BL , Male , Humans , Mitochondria/metabolism
2.
CNS Neurosci Ther ; 30(9): e70024, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218798

ABSTRACT

AIMS: Type 2 diabetes mellitus (T2DM) is related to an increased risk of postoperative cognitive dysfunction (POCD), which may be caused by neuronal hyperexcitability. Astrocyte glutamate transporter 1 (GLT-1) plays a crucial role in regulating neuron excitability. We investigated if T2DM would magnify the increased neuronal excitability induced by anesthesia/surgery (A/S) and lead to POCD in young adult mice, and if so, determined whether these effects were associated with GLT-1 expression. METHODS: T2DM model was induced by high fat diet (HFD) and injecting STZ. Then, we evaluated the spatial learning and memory of T2DM mice after A/S with the novel object recognition test (NORT) and object location test (OLT). Western blotting and immunofluorescence were used to analyze the expression levels of GLT-1 and neuronal excitability. Oxidative stress reaction and neuronal apoptosis were detected with SOD2 expression, MMP level, and Tunel staining. Hippocampal functional synaptic plasticity was assessed with long-term potentiation (LTP). In the intervention study, we overexpressed hippocampal astrocyte GLT-1 in GFAP-Cre mice. Besides, AAV-Camkllα-hM4Di-mCherry was injected to inhibit neuronal hyperexcitability in CA1 region. RESULTS: Our study found T2DM but not A/S reduced GLT-1 expression in hippocampal astrocytes. Interestingly, GLT-1 deficiency alone couldn't lead to cognitive decline, but the downregulation of GLT-1 in T2DM mice obviously enhanced increased hippocampal glutamatergic neuron excitability induced by A/S. The hyperexcitability caused neuronal apoptosis and cognitive impairment. Overexpression of GLT-1 rescued postoperative cognitive dysfunction, glutamatergic neuron hyperexcitability, oxidative stress reaction, and apoptosis in hippocampus. Moreover, chemogenetic inhibition of hippocampal glutamatergic neurons reduced oxidative stress and apoptosis and alleviated postoperative cognitive dysfunction. CONCLUSIONS: These findings suggest that the adult mice with type 2 diabetes are at an increased risk of developing POCD, perhaps due to the downregulation of GLT-1 in hippocampal astrocytes, which enhances increased glutamatergic neuron excitability induced by A/S and leads to oxidative stress reaction, and neuronal apoptosis.


Subject(s)
Astrocytes , Diabetes Mellitus, Type 2 , Down-Regulation , Excitatory Amino Acid Transporter 2 , Hippocampus , Mice, Inbred C57BL , Postoperative Cognitive Complications , Animals , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Astrocytes/metabolism , Postoperative Cognitive Complications/etiology , Postoperative Cognitive Complications/metabolism , Hippocampus/metabolism , Mice , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Male , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat/adverse effects , Mice, Transgenic
3.
FASEB J ; 38(17): e70016, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39225388

ABSTRACT

Traumatic brain injury (TBI), which is characterized by acute neurological dysfunction, is also one of the most widely recognized environmental risk factors for various neurological and psychiatric disorders. However, the role of TBI in neurological perturbation and the mechanisms underlying these disorders remain unknown. We evaluated transcriptional changes in cells of the frontal cortex after TBI by exploiting single-cell RNA sequencing (scRNA-Seq). We adopted the gene expression omnibus and scRNA-Seq to identify the mediation by secretogranin II (SCG2) of TBI-induced schizophrenia. Astrocytes are a principal source of SCG2 in the frontal cortex after TBI. Our analysis indicated that SCG2-triggered disruption of the blood-brain barrier (BBB) via the CypA-MMP-9 signaling pathway. Furthermore, astrocytic SCG2 knockout in the frontal cortex reduced BBB damage, mitigated inflammation, and inhibited schizophrenia after TBI. In conclusion, we identified the SCG2-CypA-MMP-9 signaling pathway in reactive astrocytes as a key switch in the protection of the BBB and provided a novel therapeutic avenue for treating psychiatric disorders after TBI.


Subject(s)
Astrocytes , Blood-Brain Barrier , Brain Injuries, Traumatic , Mice, Inbred C57BL , Schizophrenia , Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Animals , Schizophrenia/metabolism , Mice , Astrocytes/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Signal Transduction , Mice, Knockout
4.
Adv Neurobiol ; 37: 221-242, 2024.
Article in English | MEDLINE | ID: mdl-39207695

ABSTRACT

Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.


Subject(s)
Learning , Memory , Neurogenesis , Neuronal Plasticity , Neurogenesis/physiology , Humans , Learning/physiology , Memory/physiology , Neuronal Plasticity/physiology , Animals , Microglia/metabolism , Microglia/physiology , Brain/physiology , Brain/metabolism , Astrocytes/metabolism , Synapses/metabolism , Synapses/physiology , Neurons/metabolism , Neurons/physiology
5.
Acta Neuropathol ; 148(1): 32, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207536

ABSTRACT

Astrocytes provide crucial support for neurons, contributing to synaptogenesis, synaptic maintenance, and neurotransmitter recycling. Under pathological conditions, deregulation of astrocytes contributes to neurodegenerative diseases such as Alzheimer's disease (AD). While most research in this field has focused on protein-coding genes, non-coding RNAs, particularly long non-coding RNAs (lncRNAs), have emerged as significant regulatory molecules. In this study, we identified the lncRNA PRDM16-DT as highly enriched in the human brain, where it is almost exclusively expressed in astrocytes. PRDM16-DT and its murine homolog, Prdm16os, are downregulated in the brains of AD patients and in AD models. In line with this, knockdown of PRDM16-DT and Prdm16os revealed its critical role in maintaining astrocyte homeostasis and supporting neuronal function by regulating genes essential for glutamate uptake, lactate release, and neuronal spine density through interactions with the RE1-Silencing Transcription factor (Rest) and Polycomb Repressive Complex 2 (PRC2). Notably, CRISPR-mediated overexpression of Prdm16os mitigated functional deficits in astrocytes induced by stimuli linked to AD pathogenesis. These findings underscore the importance of PRDM16-DT in astrocyte function and its potential as a novel therapeutic target for neurodegenerative disorders characterized by astrocyte dysfunction.


Subject(s)
Alzheimer Disease , Astrocytes , DNA-Binding Proteins , RNA, Long Noncoding , Transcription Factors , Astrocytes/metabolism , Astrocytes/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Humans , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Male , Brain/metabolism , Brain/pathology , Neurons/metabolism , Neurons/pathology , Mice, Inbred C57BL
6.
Sci Rep ; 14(1): 20180, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215144

ABSTRACT

Spinal cord injury (SCI) results in irreversible neurological impairment. After SCI, Ferritinophagy-induced free iron released from ferritin can lead to extensive lipid peroxidation and aggravate neurological damage. NRF2/HO-1 pathway is to endow cells with a protective effect against oxidative stress, and it plays an important role in the transcriptional activation of a series of antioxidant and detoxification genes. UAMC-3203 is a ferrostatin-1(Fer-1) analogue with better solubility and stability, which can more effectively inhibit ferroptosis after SCI. A rat SCI model was constructed, and the recovery of motor function was observed after treatment with UAMC-3203. ELISA was employed to assess the impact of UAMC-3203 on inflammation-related factors, while immunofluorescence was utilized to investigate the influence of UAMC-3203 on neuronal count as well as the activation of astrocytes and microglia/macrophages. Malondialdehyde (MDA) were detected to reflect the level of oxidation products. Western blot analysis was used to measure the level of ferroptosis markers and the expression of NRF2/HO-1. Our findings demonstrate that UAMC-3203 inhibits the production of reactive oxygen species (ROS) and lipid peroxides, preventing ferroptosis and reducing neuronal degeneration. Additionally, UAMC-3203 suppresses astrocyte proliferation and microglia/macrophage activation, as well as the release of ferroptosis-related inflammatory factors. These combined effects contribute to the preservation of spinal cord tissue and the facilitation of motor function recovery. UAMC-3203 maybe inhibit ferroptosis after SCI to promote functional recovery.


Subject(s)
Ferroptosis , NF-E2-Related Factor 2 , Recovery of Function , Spinal Cord Injuries , Animals , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Ferroptosis/drug effects , Rats , Recovery of Function/drug effects , NF-E2-Related Factor 2/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Disease Models, Animal , Male , Cyclohexylamines/pharmacology , Oxidative Stress/drug effects , Phenylenediamines/pharmacology , Astrocytes/metabolism , Astrocytes/drug effects , Microglia/metabolism , Microglia/drug effects , Lipid Peroxidation/drug effects , Neurons/metabolism , Neurons/drug effects , Macrophages/metabolism , Macrophages/drug effects , Heme Oxygenase (Decyclizing)
7.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201299

ABSTRACT

The receptor-receptor interaction (RRI) of G protein-coupled receptors (GPCRs) leads to new functional entities that are conceptually distinct from the simple addition of signals mediated by the activation of the receptors that form the heteromers. Focusing on astrocytes, there is evidence for the existence of inhibitory and facilitatory RRIs, including the heteromers formed by the adenosine A2A and the dopamine D2 receptors, by A2A and the oxytocin receptor (OTR), and the D2-OTR heteromers. The possible involvement of these receptors in mosaicism has never been investigated in striatal astrocytes. By biophysical and functional approaches, we focused our attention on the existence of an A2A-D2-OTR high-order receptor complex and its role in modulating cytosolic calcium levels and endogenous glutamate release, when striatal astrocyte processes were stimulated with 4-aminopyridine. Functional data indicate a permissive role of OTR on dopamine signaling in the regulation of the glutamatergic transmission, and an inhibitory control mediated by A2A on both the D2-mediated signaling and on the OTR-facilitating effect on D2. Imaging biochemical and bioinformatic evidence confirmed the existence of the A2A-D2-OTR complex and its ternary structure in the membrane. In conclusion, the D2 receptor appears to be a hotspot in the control of the glutamate release from the astrocytic processes and may contribute to the regulation and integration of different neurotransmitter-mediated signaling in the striatum by the A2A-D2-OTR heterotrimers. Considering the possible selectivity of allosteric interventions on GPCRs organized as receptor mosaics, A2A-D2-OTR heterotrimers may offer selective pharmacological targets in neuropsychiatric disorders and neurodegenerative diseases.


Subject(s)
Astrocytes , Corpus Striatum , Dopamine , Receptor, Adenosine A2A , Receptors, Dopamine D2 , Signal Transduction , Astrocytes/metabolism , Animals , Receptor, Adenosine A2A/metabolism , Corpus Striatum/metabolism , Corpus Striatum/cytology , Receptors, Dopamine D2/metabolism , Dopamine/metabolism , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/genetics , Humans , Calcium/metabolism , Glutamic Acid/metabolism , Mice
8.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201378

ABSTRACT

Endoplasmic reticulum (ER) stress is a significant player in the pathophysiology of various neurodegenerative and neuropsychiatric disorders. Despite the established link between ER stress and inflammatory pathways, there remains a need for deeper exploration of the specific cellular mechanisms underlying ER stress-mediated neuroinflammation. This study aimed to investigate how the severity of ER stress (triggered by different concentrations of tunicamycin) can impact the release of proinflammatory cytokines IL-6 and IL-8 from astrocytes and microglia, comparing the effects with those induced by well-known immunostimulants-tumor necrosis factor alpha (TNF-α) or lipopolysaccharide (LPS). Mild ER stress has a distinct effect on the cytokine release compared to more intense stress levels, i.e., diminished IL-6 production was accompanied by an increase in IL-8 level, which was significantly more pronounced in astrocytes than in microglia. On the contrary, prolonged or more severe ER stress induced inflammation in glial cells, leading to a time- and concentration-dependent buildup of proinflammatory IL-6, but unlike inflammatory agents, an ER stress inducer diminished IL-8 secretions by glial cells. The differences could hold importance in identifying ER stress markers as potential drug targets for the treatment of neurodegenerative diseases or mood disorders, yet this requires confirmation in more complex animal studies.


Subject(s)
Astrocytes , Endoplasmic Reticulum Stress , Interleukin-6 , Interleukin-8 , Neuroglia , Endoplasmic Reticulum Stress/drug effects , Humans , Interleukin-8/metabolism , Interleukin-6/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Neuroglia/metabolism , Neuroglia/drug effects , Microglia/metabolism , Microglia/drug effects , Lipopolysaccharides/pharmacology , Tunicamycin/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured
9.
Int J Mol Sci ; 25(16)2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39201439

ABSTRACT

Cerebral small vessel disease (CSVD) is a group of pathologies that affect the cerebral blood vessels. CSVD accounts for 25% of strokes and contributes to 45% of dementia. However, the pathogenesis of CSVD remains unclear, involving a variety of complex mechanisms. CSVD may result from dysfunction in the glymphatic system (GS). The GS contains aquaporin-4 (AQP-4), which is in the perivascular space, at the endfeet of the astrocyte. The GS contributes to the removal of waste products from the central nervous system, occupying perivascular spaces and regulating the exchange and movement of cerebrospinal fluid and interstitial fluid. The GS involves astrocytes and aquaporin channels, which are components of the blood-brain barrier, and problems with them may constitute the pathogenesis of CSVD. Vascular risk factors, including diabetes, dilate the perivascular space, disrupting the glymphatic system and the active regulation of AQP-4. CSVD exacerbation due to disorders of the GS is associated with multiple vasculopathies. Dysfunction of the glymphatic system and AQP-4 interferes with the functioning of the blood-brain barrier, which exacerbates CSVD. In a long-term follow-up of CSVD patients with microbleeds, lacunar infarcts, and white matter hyperintensity, several vascular risk factors, including hypertension, increased the risk of ischemic stroke. Dysfunction of the GS may be the cause of CSVD; however, the underlying treatment needs to be studied further.


Subject(s)
Aquaporin 4 , Blood-Brain Barrier , Cerebral Small Vessel Diseases , Glymphatic System , Cerebral Small Vessel Diseases/metabolism , Cerebral Small Vessel Diseases/pathology , Cerebral Small Vessel Diseases/etiology , Humans , Glymphatic System/metabolism , Glymphatic System/pathology , Aquaporin 4/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Risk Factors
10.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201471

ABSTRACT

This study asked whether the P2X7 receptor was necessary and sufficient to trigger astrocyte polarization into neuroinflammatory activation states. Intravitreal injection of agonist BzATP increased gene expression of pan-astrocyte activation markers Gfap, Steap4, and Vim and A1-type astrocyte activation markers C3, Serping1, and H2T23, but also the Cd14 and Ptx3 genes usually associated with the A2-type astrocyte activation state and Tnfa, IL1a, and C1qa, assumed to be upstream of astrocyte activation in microglia. Correlation analysis of gene expression suggested the P2X7 receptor induced a mixed A1/A2-astrocyte activation state, although A1-state genes like C3 increased the most. A similar pattern of mixed glial activation genes occurred one day after intraocular pressure (IOP) was elevated in wild-type mice, but not in P2X7-/- mice, suggesting the P2X7 receptor is necessary for the glial activation that accompanies IOP elevation. In summary, this study suggests stimulation of the P2X7R is necessary and sufficient to trigger the astrocyte activation in the retina following IOP elevation, with a rise in markers for pan-, A1-, and A2-type astrocyte activation. The P2X7 receptor is expressed on microglia, optic nerve head astrocytes, and retinal ganglion cells (RGCs) in the retina, and can be stimulated by the mechanosensitive release of ATP that accompanies IOP elevation. Whether the P2X7 receptor connects this mechanosensitive ATP release to microglial and astrocyte polarization in glaucoma remains to be determined.


Subject(s)
Adenosine Triphosphate , Astrocytes , Receptors, Purinergic P2X7 , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Animals , Astrocytes/metabolism , Mice , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/analogs & derivatives , Mice, Knockout , Mice, Inbred C57BL , Intraocular Pressure , Biomarkers , Male , Retina/metabolism , Microglia/metabolism
11.
Sci Rep ; 14(1): 19035, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152163

ABSTRACT

Glial scar formation represents a fundamental response to central nervous system (CNS) injuries. It is mainly characterized by a well-defined spatial rearrangement of reactive astrocytes and microglia. The mechanisms underlying glial scar formation have been extensively studied, yet quantitative descriptors of the spatial arrangement of reactive glial cells remain limited. Here, we present a novel approach using point pattern analysis (PPA) and topological data analysis (TDA) to quantify spatial patterns of reactive glial cells after experimental ischemic stroke in mice. We provide open and reproducible tools using R and Julia to quantify spatial intensity, cell covariance and conditional distribution, cell-to-cell interactions, and short/long-scale arrangement, which collectively disentangle the arrangement patterns of the glial scar. This approach unravels a substantial divergence in the distribution of GFAP+ and IBA1+ cells after injury that conventional analysis methods cannot fully characterize. PPA and TDA are valuable tools for studying the complex spatial arrangement of reactive glia and other nervous cells following CNS injuries and have potential applications for evaluating glial-targeted restorative therapies.


Subject(s)
Astrocytes , Cicatrix , Neuroglia , Animals , Mice , Cicatrix/pathology , Neuroglia/pathology , Astrocytes/pathology , Microglia/pathology , Ischemic Stroke/pathology , Data Analysis , Disease Models, Animal , Male , Glial Fibrillary Acidic Protein/metabolism , Mice, Inbred C57BL
12.
Transl Psychiatry ; 14(1): 323, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107272

ABSTRACT

This study investigates the cellular origin and tissue heterogeneity in bipolar disorder (BD) by integrating multiomics data. Four distinct datasets were employed, including single-cell RNA sequencing (scRNA-seq) data (embryonic and fetal brain, n = 8, 1,266 cells), BD Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) data (adult brain, n = 210), BD bulk RNA-seq data (adult brain, n = 314), and BD genome-wide association study (GWAS) summary data (n = 413,466). The integration of scRNA-seq data with multiomics data relevant to BD was accomplished using the single-cell disease relevance score (scDRS) algorithm. We have identified a novel brain cell cluster named ADCY1, which exhibits distinct genetic characteristics. From a high-resolution genetic perspective, glial cells emerge as the primary cytopathology associated with BD. Specifically, astrocytes were significantly related to BD at the RNA-seq level, while microglia showed a strong association with BD across multiple panels, including the transcriptome-wide association study (TWAS), ATAC-seq, and RNA-seq. Additionally, oligodendrocyte precursor cells displayed a significant association with BD in both ATAC-seq and RNA-seq panel. Notably, our investigation of brain regions affected by BD revealed significant associations between BD and all three types of glial cells in the dorsolateral prefrontal cortex (DLPFC). Through comprehensive analyses, we identified several BD-associated genes, including CRMP1, SYT4, UCHL1, and ZBTB18. In conclusion, our findings suggest that glial cells, particularly in specific brain regions such as the DLPFC, may play a significant role in the pathogenesis of BD. The integration of multiomics data has provided valuable insights into the etiology of BD, shedding light on potential mechanisms underlying this complex psychiatric disorder.


Subject(s)
Bipolar Disorder , Brain , Genome-Wide Association Study , Single-Cell Analysis , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Humans , Brain/pathology , Brain/metabolism , Astrocytes/metabolism , Microglia/metabolism , Microglia/pathology , Sequence Analysis, RNA , Adult , Transcriptome , Multiomics
13.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39129363

ABSTRACT

Understanding the intracellular dynamics of brain cells entails performing three-dimensional molecular simulations incorporating ultrastructural models that can capture cellular membrane geometries at nanometer scales. While there is an abundance of neuronal morphologies available online, e.g. from NeuroMorpho.Org, converting those fairly abstract point-and-diameter representations into geometrically realistic and simulation-ready, i.e. watertight, manifolds is challenging. Many neuronal mesh reconstruction methods have been proposed; however, their resulting meshes are either biologically unplausible or non-watertight. We present an effective and unconditionally robust method capable of generating geometrically realistic and watertight surface manifolds of spiny cortical neurons from their morphological descriptions. The robustness of our method is assessed based on a mixed dataset of cortical neurons with a wide variety of morphological classes. The implementation is seamlessly extended and applied to synthetic astrocytic morphologies that are also plausibly biological in detail. Resulting meshes are ultimately used to create volumetric meshes with tetrahedral domains to perform scalable in silico reaction-diffusion simulations for revealing cellular structure-function relationships. Availability and implementation: Our method is implemented in NeuroMorphoVis, a neuroscience-specific open source Blender add-on, making it freely accessible for neuroscience researchers.


Subject(s)
Computer Simulation , Neurons , Neurons/ultrastructure , Neurons/cytology , Models, Neurological , Humans , Animals , Astrocytes/cytology , Astrocytes/ultrastructure
14.
Biol Res ; 57(1): 53, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135103

ABSTRACT

BACKGROUND: As a common disabling disease, irreversible neuronal death due to spinal cord injury (SCI) is the root cause of functional impairment; however, the capacity for neuronal regeneration in the developing spinal cord tissue is limited. Therefore, there is an urgent need to investigate how defective neurons can be replenished and functionally integrated by neural regeneration; the reprogramming of intrinsic cells into functional neurons may represent an ideal solution. METHODS: A mouse model of transection SCI was prepared by forceps clamping, and an adeno-associated virus (AAV) carrying the transcription factors NeuroD1 and Neurogenin-2(Ngn2) was injected in situ into the spinal cord to specifically overexpress these transcription factors in astrocytes close to the injury site. 5-bromo-2´-deoxyuridine (BrdU) was subsequently injected intraperitoneally to continuously track cell regeneration, neuroblasts and immature neurons marker expression, neuronal regeneration, and glial scar regeneration. In addition, immunoprotein blotting was used to measure the levels of transforming growth factor-ß (TGF-ß) pathway-related protein expression. We also evaluated motor function, sensory function, and the integrity of the blood-spinal cord barrier(BSCB). RESULTS: The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord was achieved by specific AAV vectors. This intervention led to a significant increase in cell regeneration and the proportion of cells with neuroblasts and immature neurons cell properties at the injury site(p < 0.0001). Immunofluorescence staining identified astrocytes with neuroblasts and immature neurons cell properties at the site of injury while neuronal marker-specific staining revealed an increased number of mature astrocytes at the injury site. Behavioral assessments showed that the intervention did not improve The BMS (Basso mouse scale) score (p = 0.0726) and gait (p > 0.05), although the treated mice had more sensory sensitivity and greater voluntary motor ability in open field than the non-intervention mice. We observed significant repair of the BSCB at the center of the injury site (p < 0.0001) and a significant improvement in glial scar proliferation. Electrophysiological assessments revealed a significant improvement in spinal nerve conduction (p < 0.0001) while immunostaining revealed that the levels of TGF-ß protein at the site of injury in the intervention group were lower than control group (p = 0.0034); in addition, P70 s6 and PP2A related to the TGF-ß pathway showed ascending trend (p = 0.0036, p = 0.0152 respectively). CONCLUSIONS: The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord after spinal cord injury can reprogram astrocytes into neurons and significantly enhance cell regeneration at the injury site. The reprogramming of astrocytes can lead to tissue repair, thus improving the reduced threshold and increasing voluntary movements. This strategy can also improve the integrity of the blood-spinal cord barrier and enhance nerve conduction function. However, the simple reprogramming of astrocytes cannot lead to significant improvements in the striding function of the lower limbs.


Subject(s)
Astrocytes , Basic Helix-Loop-Helix Transcription Factors , Disease Models, Animal , Nerve Tissue Proteins , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Spinal Cord Injuries/physiopathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Astrocytes/physiology , Nerve Tissue Proteins/metabolism , Mice , Nerve Regeneration/physiology , Neurons , Female , Mice, Inbred C57BL , Spinal Cord/metabolism
15.
Front Immunol ; 15: 1418422, 2024.
Article in English | MEDLINE | ID: mdl-39136022

ABSTRACT

Introduction: Alzheimer pathology (AD) is characterized by the deposition of amyloid beta (Aß) and chronic neuroinflammation, with the NLRP3 inflammasome playing a significant role. This study demonstrated that the OCD drug fluvoxamine maleate (FXN) can potently ameliorate AD pathology in 5XFAD mice by promoting autophagy-mediated clearance of Aß and inhibiting the NLRP3 inflammasome. Methods: We used mice primary astrocytes to establish the mechanism of action of FXN against NLRP3 inflammasome by using various techniques like ELISA, Western blotting, confocal microscopy, Immunofluorescence, etc. The anti-AD activity of FXN was validated in transgenic 5XFAD mice following two months of treatment. This was followed by behavior analysis, examination of inflammatory and autophagy proteins and immunohistochemistry analysis for Aß load in the hippocampi. Results: Our data showed that FXN, at a low concentration of 78 nM, induces autophagy to inhibit NF-κB and the NLRP3 inflammasome, apart from directly inhibiting NLRP3 inflammasome in primary astrocytes. FXN activated the PRKAA2 pathway through CAMKK2 signaling, leading to autophagy induction. It inhibited the ATP-mediated NLRP3 inflammasome activation by promoting the autophagic degradation of NF-κB, resulting in the downregulation of pro-IL-1ß and NLRP3. The anti-NLRP3 inflammasome effect of FXN was reversed when autophagy was inhibited by either genetic knockdown of the PRKAA2 pathway or pharmacological inhibition with bafilomycin A1. Furthermore, FXN treatment led to improved AD pathology in 5XFAD mice, resulting in significant improvements in various behavioral parameters such as working memory and neuromuscular coordination, making their behavior more similar to that of wild-type animals. FXN improved behavior in 5XFAD mice by clearing the Aß deposits from the hippocampi and significantly reducing multiple inflammatory proteins, including NF-κB, GFAP, IBA1, IL-1ß, TNF-α, and IL-6, which are associated with NF-κB and NLRP3 inflammasome in the brain. Moreover, these changes were accompanied by increased expression of autophagic proteins. Discussion: Our data suggest that FXN ameliorates AD pathology, by simultaneously targeting two key pathological features: Aß deposits and neuroinflammation. As an already approved drug, FXN holds potential as a candidate for human studies against AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Astrocytes , Autophagy , Disease Models, Animal , Fluvoxamine , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Mice , Fluvoxamine/pharmacology , Fluvoxamine/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Autophagy/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects
16.
Methods Mol Biol ; 2831: 283-299, 2024.
Article in English | MEDLINE | ID: mdl-39134857

ABSTRACT

Mosaic Analysis with Double Markers (MADM) is a powerful genetic method typically used for lineage tracing and to disentangle cell autonomous and tissue-wide roles of candidate genes with single cell resolution. Given the relatively sparse labeling, depending on which of the 19 MADM chromosomes one chooses, the MADM approach represents the perfect opportunity for cell morphology analysis. Various MADM studies include reports of morphological anomalies and phenotypes in the central nervous system (CNS). MADM for any candidate gene can easily incorporate morphological analysis within the experimental workflow. Here, we describe the methods of morphological cell analysis which we developed in the course of diverse recent MADM studies. This chapter will specifically focus on methods to quantify aspects of the morphology of neurons and astrocytes within the CNS, but these methods can broadly be applied to any MADM-labeled cells throughout the entire organism. We will cover two analyses-soma volume and dendrite characterization-of physical characteristics of pyramidal neurons in the somatosensory cortex, and two analyses-volume and Sholl analysis-of astrocyte morphology.


Subject(s)
Astrocytes , Neuroglia , Neurons , Animals , Neurons/cytology , Neurons/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Mice , Mosaicism , Biomarkers , Dendrites/metabolism , Somatosensory Cortex/cytology
17.
J Med Virol ; 96(8): e29854, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135475

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) has a high mortality rate compared to other infectious diseases. SFTS is particularly associated with a high risk of mortality in immunocompromised individuals, while most patients who die of SFTS exhibit symptoms of severe encephalitis before death. However, the region of brain damage and mechanisms by which the SFTS virus (SFTSV) causes encephalitis remains unknown. Here, we revealed that SFTSV infects the brainstem and spinal cord, which are regions of the brain associated with respiratory function, and motor nerves in IFNAR1-/- mice. Further, we show that A1-reactive astrocytes are activated, causing nerve cell death, in infected mice. Primary astrocytes of SFTSV-infected IFNAR1-/- mice also induced neuronal cell death through the activation of A1-reactive astrocytes. Herein, we showed that SFTSV induces fatal neuroinflammation in the brain regions important for respiratory function and motor nerve, which may underlie mortality in SFTS patients. This study provides new insights for the treatment of SFTS, for which there is currently no therapeutic approach.


Subject(s)
Astrocytes , Bunyaviridae Infections , Mice, Knockout , Phlebovirus , Receptor, Interferon alpha-beta , Animals , Astrocytes/virology , Astrocytes/pathology , Mice , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/deficiency , Phlebovirus/genetics , Phlebovirus/physiology , Phlebovirus/pathogenicity , Bunyaviridae Infections/virology , Bunyaviridae Infections/pathology , Bunyaviridae Infections/immunology , Brain/virology , Brain/pathology , Brain/immunology , Spinal Cord/virology , Spinal Cord/pathology , Disease Models, Animal , Neurons/virology , Neurons/pathology , Mice, Inbred C57BL , Brain Stem/virology , Brain Stem/pathology , Cell Death
18.
Biol Res ; 57(1): 54, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143594

ABSTRACT

Brain damage triggers diverse cellular and molecular events, with astrocytes playing a crucial role in activating local neuroprotective and reparative signaling within damaged neuronal circuits. Here, we investigated reactive astrocytes using a multidimensional approach to categorize their responses into different subtypes based on morphology. This approach utilized the StarTrack lineage tracer, single-cell imaging reconstruction and multivariate data analysis. Our findings identified three profiles of reactive astrocyte responses, categorized by their effects on cell size- and shape- related morphological parameters: "moderate", "strong," and "very strong". We also examined the heterogeneity of astrocyte reactivity, focusing on spatial and clonal distribution. Our research revealed a notable enrichment of protoplasmic and fibrous astrocytes within the "strong" and "very strong" response subtypes. Overall, our study contributes to a better understanding of astrocyte heterogeneity in response to an injury. By characterizing the diverse reactive responses among astrocyte subpopulations, we provide insights that could guide future research aimed at identifying novel therapeutic targets to mitigate brain damage and promote neural repair.


Subject(s)
Astrocytes , Astrocytes/physiology , Animals , Mice , Cell Lineage/physiology , Cluster Analysis , Single-Cell Analysis
19.
Cell Death Dis ; 15(8): 592, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147758

ABSTRACT

Smac mimetic compounds (SMCs) are small molecule drugs that sensitize cancer cells to TNF-α-induced cell death and have multiple immunostimulatory effects through alterations in NF-κB signaling. The combination of SMCs with immunotherapies has been reported to result in durable cures of up to 40% in syngeneic, orthotopic murine glioblastoma (GBM) models. Herein, we find that SMC resistance is not due to a cell-intrinsic mechanism of resistance. We thus evaluated the contribution of GBM and brain stromal components to identify parameters leading to SMC efficacy and resistance. The common physiological features of GBM tumors, such as hypoxia, hyaluronic acid, and glucose deprivation were found not to play a significant role in SMC efficacy. SMCs induced the death of microglia and macrophages, which are the major immune infiltrates in the tumor microenvironment. This death of microglia and macrophages then enhances the ability of SMCs to induce GBM cell death. Conversely, astrocytes promoted GBM cell growth and abrogated the ability of SMCs to induce death of GBM cells. The astrocyte-mediated resistance can be overcome in the presence of exogenous TNF-α. Overall, our results highlight that SMCs can induce death of microglia and macrophages, which then provides a source of death ligands for GBM cells, and that the targeting of astrocytes is a potential mechanism for overcoming SMC resistance for the treatment of GBM.


Subject(s)
Astrocytes , Glioblastoma , Tumor Microenvironment , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/immunology , Tumor Microenvironment/drug effects , Animals , Humans , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Mice , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Tumor Necrosis Factor-alpha/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Inflammation/pathology , Inflammation/drug therapy , Mitochondrial Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL
20.
J Cell Mol Med ; 28(16): e70004, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159174

ABSTRACT

Ischemia and hypoxia activate astrocytes into reactive types A1 and A2, which play roles in damage and protection, respectively. However, the function and mechanism of A1 and A2 astrocyte exosomes are unknown. After astrocyte exosomes were injected into the lateral ventricle, infarct volume, damage to the blood-brain barrier (BBB), apoptosis and the expression of microglia-related proteins were measured. The dual luciferase reporter assay was used to detect the target genes of miR-628, and overexpressing A2-Exos overexpressed and knocked down miR-628 were constructed. qRT-PCR, western blotting and immunofluorescence staining were subsequently performed. A2-Exos obviously reduced the infarct volume, damage to the BBB and apoptosis and promoted M2 microglial polarization. RT-PCR showed that miR-628 was highly expressed in A2-Exos. Dual luciferase reporter assays revealed that NLRP3, S1PR3 and IRF5 are target genes of miR-628. After miR-628 was overexpressed or knocked down, the protective effects of A2-Exos increased or decreased, respectively. A2-Exos reduced pyroptosis and BBB damage and promoted M2 microglial polarization through the inhibition of NLRP3, S1PR3 and IRF5 via the delivery of miR-628. This study explored the mechanism of action of A2-Exos and provided new therapeutic targets and concepts for treating cerebral ischemia.


Subject(s)
Astrocytes , Blood-Brain Barrier , Brain Ischemia , Exosomes , MicroRNAs , Reperfusion Injury , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Astrocytes/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Exosomes/metabolism , Brain Ischemia/metabolism , Brain Ischemia/genetics , Brain Ischemia/therapy , Brain Ischemia/pathology , Blood-Brain Barrier/metabolism , Male , Apoptosis/genetics , Microglia/metabolism , Microglia/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL