Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 15.253
1.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Article En | MEDLINE | ID: mdl-38831600

PURPOSE: Radiation-induced brain injury, one of the side effects of cranial radiotherapy in tumour patients, usually results in durable and serious cognitive disorders. Microglia are important innate immune-effector cells in the central nervous system. However, the interaction between microglia and neurons in radiation-induced brain injury remains uncharacterised. METHODS AND MATERIALS: We established a microglia-neuron indirect co-culture model to assess the interaction between them. Microglia exposed to radiation were examined for pyroptosis using lactate dehydrogenase (LDH) release, Annexin V/PI staining, SYTOX staining and western blot. The role of nucleotide-binding oligomerisation domain-like receptor family pyrin domain containing 3 (NLRP3) was investigated in microglia exposed to radiation and in mouse radiation brain injury model through siRNA or inhibitor. Mini-mental state examination and cytokines in blood were performed in 23 patients who had experienced cranial irradiation. RESULTS: Microglia exerted neurotoxic features after radiation in the co-culture model. NLRP3 was up-regulated in microglia exposed to radiation, and then caspase-1 was activated. Thus, the gasdermin D protein was cleaved, and it triggered pyroptosis in microglia, which released inflammatory cytokines. Meanwhile, treatment with siRNA NLRP3 in vitro and NLRP3 inhibitor in vivo attenuated the damaged neuron cell and cognitive impairment, respectively. What is more, we found that the patients after radiation with higher IL-6 were observed to have a decreased MMSE score. CONCLUSIONS: These findings indicate that radiation-induced pyroptosis in microglia may promote radiation-induced brain injury via the secretion of neurotoxic cytokines. NLRP3 was evaluated as an important mediator in radiation-induced pyroptosis and a promising therapeutic target for radiation-induced brain injury.


Brain Injuries , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Pyroptosis/radiation effects , Pyroptosis/physiology , Microglia/metabolism , Microglia/radiation effects , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Mice , Humans , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/etiology , Male , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Coculture Techniques , Radiation Injuries/pathology , Radiation Injuries/metabolism , Female , Mice, Inbred C57BL , Middle Aged
2.
J Neuroinflammation ; 21(1): 143, 2024 May 31.
Article En | MEDLINE | ID: mdl-38822367

The dysregulation of pro- and anti-inflammatory processes in the brain has been linked to the pathogenesis of major depressive disorder (MDD), although the precise mechanisms remain unclear. In this study, we discovered that microglial conditional knockout of Pdcd4 conferred protection against LPS-induced hyperactivation of microglia and depressive-like behavior in mice. Mechanically, microglial Pdcd4 plays a role in promoting neuroinflammatory responses triggered by LPS by inhibiting Daxx-mediated PPARγ nucleus translocation, leading to the suppression of anti-inflammatory cytokine IL-10 expression. Finally, the antidepressant effect of microglial Pdcd4 knockout under LPS-challenged conditions was abolished by intracerebroventricular injection of the IL-10 neutralizing antibody IL-10Rα. Our study elucidates the distinct involvement of microglial Pdcd4 in neuroinflammation, suggesting its potential as a therapeutic target for neuroinflammation-related depression.


Co-Repressor Proteins , Interleukin-10 , Mice, Knockout , Microglia , Neuroinflammatory Diseases , PPAR gamma , Signal Transduction , Animals , Male , Mice , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/deficiency , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Depression/metabolism , Depression/etiology , Interleukin-10/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Microglia/metabolism , Microglia/drug effects , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neuroinflammatory Diseases/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Signal Transduction/physiology , Signal Transduction/drug effects
3.
Crit Rev Immunol ; 44(6): 13-25, 2024.
Article En | MEDLINE | ID: mdl-38848290

Alzheimer's disease (AD) is the most common form of dementia. Aberrant regulation of microRNAs (miRNAs) has been implicated in the pathogenesis of AD. In a large case-control study recruiting 208 patients with AD and 205 elderly control subjects, miRNA-let-7d-5p attracted our attention for its downregulated level in patients with AD. However, the biological functions of let-7d-5p in AD pathogenesis have not been investigated. This study emphasized the functions and mechanisms of let-7d-5p in the pathogenesis of AD. Mouse microglial BV2 cells treated with amyloid-ß (Aß)1-42 were used as in vitro AD inflammation models. We reported that let-7d-5p was downregulated in Aß1-42-stimulated BV2 cells, and upregulation of let-7d-5p promoted the transversion of microglial cells from Ml phenotype to M2 phenotype. Then, the binding relationship between let-7d-5p and Map3k1 was verified by luciferase reporter assays. Mechanistically, let-7d-5p could target Map3k1 3'UTR to inactivate ERK/p38 MAPK signaling. Therefore, it was suggested that let-7d-5p might be a novel modulator of microglial neuroinflammation and serve as a novel target for diagnosis and treatment of AD.


Alzheimer Disease , MAP Kinase Signaling System , MicroRNAs , Microglia , p38 Mitogen-Activated Protein Kinases , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/metabolism , Microglia/immunology , Animals , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/immunology , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/genetics , Inflammation/genetics , Inflammation/immunology , Cell Line , Amyloid beta-Peptides/metabolism
4.
Front Immunol ; 15: 1374301, 2024.
Article En | MEDLINE | ID: mdl-38835765

Background: Human immunodeficiency virus (HIV) affects nearly 40 million people globally, with roughly 80% of all people living with HIV receiving antiretroviral therapy. Antiretroviral treatment suppresses viral load in peripheral tissues but does not effectively penetrate the blood-brain barrier. Thus, viral reservoirs persist in the central nervous system and continue to produce low levels of inflammatory factors and early viral proteins, including the transactivator of transcription (Tat). HIV Tat is known to contribute to chronic neuroinflammation and synaptodendritic damage, which is associated with the development of cognitive, motor, and/or mood problems, collectively known as HIV-associated neurocognitive disorders (HAND). Cannabinoid anti-inflammatory effects are well documented, but therapeutic utility of cannabis remains limited due to its psychotropic effects, including alterations within brain regions encoding reward processing and motivation, such as the nucleus accumbens. Alternatively, inhibiting monoacylglycerol lipase (MAGL) has demonstrated therapeutic potential through interactions with the endocannabinoid system. Methods: The present study utilized a reward-related operant behavioral task to quantify motivated behavior in female Tat transgenic mice treated with vehicle or MAGL inhibitor MJN110 (1 mg/kg). Brain tissue was collected to assess dendritic injury and neuroinflammatory profiles, including dendritic microtubule-associated protein (MAP2ab) intensity, microglia density, microglia morphology, astrocyte density, astrocytic interleukin-1ß (IL-1ß) colocalization, and various lipid mediators. Results: No significant behavioral differences were observed; however, MJN110 protected against Tat-induced dendritic injury by significantly upregulating MAP2ab intensity in the nucleus accumbens and in the infralimbic cortex of Tat(+) mice. No or only minor effects were noted for Iba-1+ microglia density and/or microglia morphology. Further, Tat increased GFAP+ astrocyte density in the infralimbic cortex and GFAP+ astrocytic IL-1ß colocalization in the nucleus accumbens, with MJN110 significantly reducing these measures in Tat(+) subjects. Lastly, selected HETE-related inflammatory lipid mediators in the striatum were downregulated by chronic MJN110 treatment. Conclusions: These findings demonstrate anti-inflammatory and neuroprotective properties of MJN110 without cannabimimetic behavioral effects and suggest a promising alternative to cannabis for managing neuroinflammation.


HIV-1 , Mice, Transgenic , Monoacylglycerol Lipases , Neuroinflammatory Diseases , Animals , Mice , HIV-1/physiology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Female , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV Infections/drug therapy , Humans , Brain/drug effects , Brain/metabolism , Brain/virology , Brain/pathology , Disease Models, Animal , Microglia/drug effects , Microglia/metabolism , AIDS Dementia Complex/drug therapy
5.
Biol Direct ; 19(1): 45, 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38863009

BACKGROUND: Glioma is a common tumor that occurs in the brain and spinal cord. Hypoxia is a crucial feature of the tumor microenvironment. Tumor-associated macrophages/microglia play a crucial role in the advancement of glioma. This study aims to illuminate the detailed mechanisms by which hypoxia regulates microglia and, consequently, influences the progression of glioma. METHODS: The glioma cell viability and proliferation were analyzed by cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay. Wound healing assay and transwell assay were implemented to detect glioma cell migration and invasion, respectively. Enzyme-linked immunosorbent assay was conducted to detect protein levels in cell culture medium. The protein levels in glioma cells and tumor tissues were evaluated using western blot analysis. The histological morphology of tumor tissue was determined by hematoxylin-eosin staining. The protein expression in tumor tissues was determined using immunohistochemistry. Human glioma xenograft in nude mice was employed to test the influence of hypoxic microglia-derived interleukin-1beta (IL-1ß) and heparanase (HPSE) on glioma growth in vivo. RESULTS: Hypoxic HMC3 cells promoted proliferation, migration, and invasion abilities of U251 and U87 cells by secreting IL-1ß, which was upregulated by hypoxia-induced activation of hypoxia inducible factor-1alpha (HIF-1α). Besides, IL-1ß from HMC3 cells promoted glioma progression and caused activation of nuclear factor-κB (NF-κB) and upregulation of HPSE in vivo. We also confirmed that IL-1ß facilitated HPSE expression in U251 and U87 cells by activating NF-κB. Hypoxic HMC3 cells-secreted IL-1ß facilitated the proliferation, migration, and invasion of U251 and U87 cells via NF-κB-mediated upregulation of HPSE expression. Finally, we revealed that silencing HPSE curbed the proliferation and metastasis of glioma in mice. CONCLUSION: Hypoxia-induced activation of HIF-1α/IL-1ß axis in microglia promoted glioma progression via NF-κB-mediated upregulation of HPSE expression.


Glioma , Glucuronidase , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-1beta , Mice, Nude , Microglia , NF-kappa B , Up-Regulation , Glioma/metabolism , Glioma/genetics , Glioma/pathology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Microglia/metabolism , Animals , NF-kappa B/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Glucuronidase/metabolism , Glucuronidase/genetics , Cell Line, Tumor , Disease Progression , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation , Cell Movement , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia/genetics
6.
Nat Commun ; 15(1): 4698, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844770

Given the marginal penetration of most drugs across the blood-brain barrier, the efficacy of various agents remains limited for glioblastoma (GBM). Here we employ low-intensity pulsed ultrasound (LIPU) and intravenously administered microbubbles (MB) to open the blood-brain barrier and increase the concentration of liposomal doxorubicin and PD-1 blocking antibodies (aPD-1). We report results on a cohort of 4 GBM patients and preclinical models treated with this approach. LIPU/MB increases the concentration of doxorubicin by 2-fold and 3.9-fold in the human and murine brains two days after sonication, respectively. Similarly, LIPU/MB-mediated blood-brain barrier disruption leads to a 6-fold and a 2-fold increase in aPD-1 concentrations in murine brains and peritumoral brain regions from GBM patients treated with pembrolizumab, respectively. Doxorubicin and aPD-1 delivered with LIPU/MB upregulate major histocompatibility complex (MHC) class I and II in tumor cells. Increased brain concentrations of doxorubicin achieved by LIPU/MB elicit IFN-γ and MHC class I expression in microglia and macrophages. Doxorubicin and aPD-1 delivered with LIPU/MB results in the long-term survival of most glioma-bearing mice, which rely on myeloid cells and lymphocytes for their efficacy. Overall, this translational study supports the utility of LIPU/MB to potentiate the antitumoral activities of doxorubicin and aPD-1 for GBM.


Blood-Brain Barrier , Brain Neoplasms , Doxorubicin , Microbubbles , Programmed Cell Death 1 Receptor , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Doxorubicin/analogs & derivatives , Animals , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/drug therapy , Glioma/immunology , Glioma/pathology , Brain/metabolism , Brain/drug effects , Female , Drug Delivery Systems , Ultrasonic Waves , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioblastoma/pathology , Male , Microglia/drug effects , Microglia/metabolism , Mice, Inbred C57BL , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , Polyethylene Glycols
7.
BMC Neurol ; 24(1): 191, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38849737

BACKGROUND: Depression is a complex mood disorder whose pathogenesis involves multiple cell types and molecular pathways. The prefrontal cortex, as a key brain region for emotional regulation, plays a crucial role in depression. Microglia, as immune cells of the central nervous system, have been closely linked to the development and progression of depression through their dysfunctional states. This study aims to utilize single-cell RNA-seq technology to reveal the pathogenic mechanism of YAP1 in prefrontal cortex microglia in depression. METHODS: Firstly, we performed cell type identification and differential analysis on normal and depressed prefrontal cortex tissues by mining single-cell RNA-seq datasets from public databases. Focusing on microglia, we conducted sub-clustering, differential gene KEGG enrichment analysis, intercellular interaction analysis, and pseudotime analysis. Additionally, a cross-species analysis was performed to explore the similarities and differences between human and rhesus monkey prefrontal cortex microglia. To validate our findings, we combined bulk RNA-Seq and WGCNA analysis to reveal key genes associated with depression and verified the relationship between YAP1 and depression using clinical samples. RESULTS: Our study found significant changes in the proportion and transcriptional profiles of microglia in depressed prefrontal cortex tissues. Further analysis revealed multiple subpopulations of microglia and their associated differential genes and signaling pathways related to depression. YAP1 was identified as a key molecule contributing to the development of depression and was significantly elevated in depression patients. Moreover, the expression level of YAP1 was positively correlated with HAMD scores, suggesting its potential as a biomarker for predicting the onset of depression. CONCLUSION: This study utilized single-cell RNA-seq technology to reveal the pathogenic mechanism of YAP1 in prefrontal cortex microglia in depression, providing a new perspective for a deeper understanding of the pathophysiology of depression and identifying potential targets for developing novel treatment strategies.


Macaca mulatta , Microglia , Prefrontal Cortex , Single-Cell Analysis , YAP-Signaling Proteins , Prefrontal Cortex/metabolism , Microglia/metabolism , YAP-Signaling Proteins/metabolism , Humans , Animals , Single-Cell Analysis/methods , RNA-Seq , Depression/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Male , Female , Single-Cell Gene Expression Analysis
8.
J Neuroinflammation ; 21(1): 150, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38840206

Microglia, the brain's resident macrophages, maintain brain homeostasis and respond to injury and infection. During aging they undergo functional changes, but the underlying mechanisms and their contributions to neuroprotection versus neurodegeneration are unclear. Previous studies suggested that microglia are sex dimorphic, so we compared microglial aging in mice of both sexes. RNA-sequencing of hippocampal microglia revealed more aging-associated changes in female microglia than male microglia, and more sex differences in old microglia than young microglia. Pathway analyses and subsequent validation assays revealed a stronger AKT-mTOR-HIF1α-driven shift to glycolysis among old female microglia and indicated that C3a production and detection was elevated in old microglia, especially in females. Recombinant C3a induced AKT-mTOR-HIF1α signaling and increased the glycolytic and phagocytic activity of young microglia. Single cell analyses attributed the aging-associated sex dimorphism to more abundant disease-associated microglia (DAM) in old female mice than old male mice, and evaluation of an Alzheimer's Disease mouse model revealed that the metabolic and complement changes are also apparent in the context of neurodegenerative disease and are strongest in the neuroprotective DAM2 subset. Collectively, our data implicate autocrine C3a-C3aR signaling in metabolic reprogramming of microglia to neuroprotective DAM during aging, especially in females, and also in Alzheimer's Disease.


Aging , Microglia , Sex Characteristics , Animals , Microglia/metabolism , Female , Mice , Aging/metabolism , Aging/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Signal Transduction/physiology
9.
Alzheimers Res Ther ; 16(1): 121, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38831312

BACKGROUND: Beta-amyloid (Aß) deposition in the brain parenchyma is a crucial initiating step in the amyloid cascade hypothesis of Alzheimer's disease (AD) pathology. Furthermore, dysfunction of plaque-associated microglia, also known as disease-associated microglia (DAM) has been reported to accelerate Aß deposition and cognitive impairment. Our previous research demonstrated that intermittent hypoxia training (IHT) improved AD pathology by upregulating autophagy in DAM, thereby enhancing oligomeric Aß (oAß) clearance. Considering that oAß internalization is the initial stage of oAß clearance, this study focused on the IHT mechanism involved in upregulating Aß uptake by DAM. METHODS: IHT was administered to 8-month-old APP/PS1 mice or 6-month-old microglial vacuolar protein sorting 35 (VPS35) knockout mice in APP/PS1 background (MG VPS35 KO: APP/PS1) for 28 days. After the IHT, the spatial learning-memory capacity of the mice was assessed. Additionally, AD pathology was determined by estimating the nerve fiber and synapse density, Aß plaque deposition, and Aß load in the brain. A model of Aß-exposed microglia was constructed and treated with IHT to explore the related mechanism. Finally, triggering receptor expressed on myeloid cells 2 (TREM2) intracellular recycling and Aß internalization were measured using a fluorescence tracing technique. RESULTS: Our results showed that IHT ameliorated cognitive function and Aß pathology. In particular, IHT enhanced Aß endocytosis by augmenting the intracellular transport function of microglial TREM2, thereby contributing to Aß clearance. Furthermore, IHT specifically upregulated VPS35 in DAM, the primary cause for the enhanced intracellular recycling of TREM2. IHT lost ameliorative effect on Aß pathology in MG VPS35 KO: APP/PS1 mice brain. Lastly, the IHT mechanism of VPS35 upregulation in DAM was mediated by the transcriptional regulation of VPS35 by transcription factor EB (TFEB). CONCLUSION: IHT enhances Aß endocytosis in DAM by upregulating VPS35-dependent TREM2 recycling, thereby facilitating oAß clearance and mitigation of Aß pathology. Moreover, the transcriptional regulation of VPS35 by TFEB demonstrates a close link between endocytosis and autophagy in microglia. Our study further elucidates the IHT mechanism in improving AD pathology and provides evidence supporting the potential application of IHT as a complementary therapy for AD.


Alzheimer Disease , Amyloid beta-Peptides , Endocytosis , Membrane Glycoproteins , Microglia , Plaque, Amyloid , Receptors, Immunologic , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Microglia/metabolism , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Endocytosis/physiology , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Mice, Transgenic , Hypoxia/metabolism , Mice, Knockout , Disease Models, Animal , Male , Brain/metabolism , Brain/pathology , Mice, Inbred C57BL
10.
Inhal Toxicol ; 36(4): 261-274, 2024 Apr.
Article En | MEDLINE | ID: mdl-38836331

OBJECTIVE: Our work is focused on tungsten, considered as an emerging contaminant. Its environmental dispersion is partly due to mining and military activities. Exposure scenario can also be occupational, in areas such as the hard metal industry and specific nuclear facilities. Our study investigated the cerebral effects induced by the inhalation of tungsten particles. METHODS: Inhalation exposure campaigns were carried out at two different concentrations (5 and 80 mg/m3) in single and repeated modes (4 consecutive days) in adult rats within a nose-only inhalation chamber. Processes involved in brain toxicity were investigated 24 h after exposure. RESULTS AND DISCUSSION: Site-specific effects in terms of neuroanatomy and concentration-dependent changes in specific cellular actors were observed. Results obtained in the olfactory bulb suggest a potential early effect on the survival of microglial cells. Depending on the mode of exposure, these cells showed a decrease in density accompanied by an increase in an apoptotic marker. An abnormal phenotype of the nuclei of mature neurons, suggesting neuronal suffering, was also observed in the frontal cortex, and can be linked to the involvement of oxidative stress. The differential effects observed according to exposure patterns could involve two components: local (brain-specific) and/or systemic. Indeed, tungsten, in addition to being found in the lungs and kidneys, was present in the brain of animals exposed to the high concentration. CONCLUSION: Our data question the perceived innocuity of tungsten relative to other metals and raise hypotheses regarding possible adaptive or neurotoxic mechanisms that could ultimately alter neuronal integrity.


Brain , Inhalation Exposure , Rats, Wistar , Tungsten , Animals , Tungsten/toxicity , Male , Inhalation Exposure/adverse effects , Brain/drug effects , Brain/metabolism , Rats , Biomarkers/metabolism , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Lung/drug effects , Lung/metabolism , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Apoptosis/drug effects , Oxidative Stress/drug effects
11.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 66-72, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38836680

This study aimed to investigate the mechanism of the effect of TREM2 on cognitive function in autistic mice. TREM2 overexpression and knockdown viruses were given to autism spectrum disorder (ASD) mice and BV2 microglia cell line. To assess cognitive performance, all groups of mice took part in the open field, new object recognition, Morris water maze, and three-box social experiments. Double immunofluorescence labeling demonstrated co-localization of LC3II and NeuN. Proteins from the PI3K/Akt/mTOR pathway were identified. In vivo, behavior studies revealed that TREM2 could successfully improve ASD mice's social interaction and cognitive performance. Besides, we discovered that TREM2 could increase autophagy in ASD mice. In vitro, overexpressing TREM2 reduced the expression of PI3K/AKT/mTOR pathway proteins, whereas knocking down TREM2 increased the expression of PI3K/AKT/mTOR pathway proteins. In conclusion, TREM2 could inhibit PI3K/Akt/mTOR signaling pathway, enhance autophagy, and improve the social communication ability and cognitive function of ASD mice.


Autophagy , Cognition , Membrane Glycoproteins , Microglia , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Receptors, Immunologic , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Cognition/physiology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Microglia/metabolism , Male , Autistic Disorder/metabolism , Autistic Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/psychology , Disease Models, Animal , Behavior, Animal , Cell Line , Mice, Inbred C57BL , Social Behavior
12.
J Neurosci Res ; 102(6): e25358, 2024 Jun.
Article En | MEDLINE | ID: mdl-38859672

Neuroinflammation caused by excessive microglial activation plays a key role in the pathogenesis of ischemic stroke. Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive neuromodulatory technique that has recently been reported to regulate microglial functions and exert anti-inflammatory effects. The intermittent burst stimulation (iTBS) regimen in rTMS improves neuronal excitability. However, whether iTBS exerts its anti-inflammatory effects by stimulating neurons and thereby modulating microglial polarization remains unclear. Motor function was assessed after 1 week of rTMS (iTBS regimen) treatment in adult male mice with occlusion/reperfusion of the middle cerebral artery (MCAO/r) injury. We also investigated the molecular biological alterations associated with microglial polarization using a cell proliferation assay, multiplex cytokine bioassays, and immunofluorescence staining. iTBS regimen can improve balance and motor coordination function, increase spontaneous movement, and improve walking function in mice with early cerebral ischemia injury. Expression levels of IL-1ß, TNF-α, and IL-10 increased significantly in mice with MCAO injury. Especially, rTMS significantly increased the number of proliferating cells in the infarcted cortex. The fluorescence intensity of MAP2 in the peri-infarct area of MCAO injured mice was low, but the signal was broader. Compared with MCAO group, the fluorescence intensity of MAP2 in rTMS group was significantly increased. rTMS inhibited pro-inflammatory M1 activation (Iba1+/CD86+) and improved anti-inflammatory M2 activation (Iba1+/CD206+) in the peri-infarct zone, thus significantly changing the phenotypic ratio M1/M2. rTMS improves motor dysfunction and neuroinflammation after cerebral I/R injury in mice by regulating microglial polarization.


Ischemic Stroke , Microglia , Transcranial Magnetic Stimulation , Animals , Male , Transcranial Magnetic Stimulation/methods , Ischemic Stroke/therapy , Ischemic Stroke/physiopathology , Mice , Microglia/metabolism , Mice, Inbred C57BL , Infarction, Middle Cerebral Artery/therapy , Motor Activity/physiology , Neuronal Plasticity/physiology
13.
Nat Commun ; 15(1): 5031, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38866759

Alzheimer's disease (AD) is a brain network disorder where pathological proteins accumulate through networks and drive cognitive decline. Yet, the role of network connectivity in facilitating this accumulation remains unclear. Using in-vivo multimodal imaging, we show that the distribution of tau and reactive microglia in humans follows spatial patterns of connectivity variation, the so-called gradients of brain organization. Notably, less distinct connectivity patterns ("gradient contraction") are associated with cognitive decline in regions with greater tau, suggesting an interaction between reduced network differentiation and tau on cognition. Furthermore, by modeling tau in subject-specific gradient space, we demonstrate that tau accumulation in the frontoparietal and temporo-occipital cortices is associated with greater baseline tau within their functionally and structurally connected hubs, respectively. Our work unveils a role for both functional and structural brain organization in pathology accumulation in AD, and supports subject-specific gradient space as a promising tool to map disease progression.


Alzheimer Disease , Brain , Magnetic Resonance Imaging , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , tau Proteins/metabolism , Male , Female , Aged , Brain/metabolism , Brain/diagnostic imaging , Brain/pathology , Microglia/metabolism , Microglia/pathology , Aged, 80 and over , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/diagnostic imaging , Middle Aged , Nerve Net/metabolism , Nerve Net/pathology , Nerve Net/diagnostic imaging , Brain Mapping/methods
14.
Nat Commun ; 15(1): 5013, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38866782

Multiple sclerosis (MS) is characterized by heterogeneity in disease course and prediction of long-term outcome remains a major challenge. Here, we investigate five myeloid markers - CHIT1, CHI3L1, sTREM2, GPNMB and CCL18 - in the cerebrospinal fluid (CSF) at diagnostic lumbar puncture in a longitudinal cohort of 192 MS patients. Through mixed-effects and machine learning models, we show that CHIT1 is a robust predictor for faster disability progression. Integrative analysis of 11 CSF and 26 central nervous system (CNS) parenchyma single-cell/nucleus RNA sequencing samples reveals CHIT1 to be predominantly expressed by microglia located in active MS lesions and enriched for lipid metabolism pathways. Furthermore, we find CHIT1 expression to accompany the transition from a homeostatic towards a more activated, MS-associated cell state in microglia. Neuropathological evaluation in post-mortem tissue from 12 MS patients confirms CHIT1 production by lipid-laden phagocytes in actively demyelinating lesions, already in early disease stages. Altogether, we provide a rationale for CHIT1 as an early biomarker for faster disability progression in MS.


Biomarkers , Disease Progression , Microglia , Multiple Sclerosis , Humans , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/diagnosis , Multiple Sclerosis/pathology , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Female , Male , Adult , Middle Aged , Hexosaminidases/metabolism , Hexosaminidases/genetics , Hexosaminidases/cerebrospinal fluid , Longitudinal Studies , Chitinase-3-Like Protein 1/cerebrospinal fluid , Chitinase-3-Like Protein 1/metabolism , Chitinase-3-Like Protein 1/genetics
15.
CNS Neurosci Ther ; 30(6): e14794, 2024 Jun.
Article En | MEDLINE | ID: mdl-38867379

BACKGROUND: Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS: This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS: Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS: The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.


Brain Injuries , Microglia , Radiation Injuries , Microglia/radiation effects , Microglia/metabolism , Animals , Humans , Radiation Injuries/metabolism , Radiation Injuries/therapy , Brain Injuries/etiology , Brain Injuries/metabolism , Neuroinflammatory Diseases/etiology
16.
Sci Rep ; 14(1): 13430, 2024 06 11.
Article En | MEDLINE | ID: mdl-38862696

Previous studies have shown that scutellarin inhibits the excessive activation of microglia, reduces neuronal apoptosis, and exerts neuroprotective effects. However, whether scutellarin regulates activated microglia-mediated neuronal apoptosis and its mechanisms remains unclear. This study aimed to investigate whether scutellarin can attenuate PC12 cell apoptosis induced by activated microglia via the JAK2/STAT3 signalling pathway. Microglia were cultured in oxygen-glucose deprivation (OGD) medium, which acted as a conditioning medium (CM) to activate PC12 cells, to investigate the expression of apoptosis and JAK2/STAT3 signalling-related proteins. We observed that PC12 cells apoptosis in CM was significantly increased, the expression and fluorescence intensity of the pro-apoptotic protein Bax and apoptosis-related protein cleaved caspase-3 were increased, and expression of the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2) was decreased. Phosphorylation levels and fluorescence intensity of the JAK2/STAT3 signalling pathway-related proteins JAK2 and STAT3 decreased. After treatment with scutellarin, PC12 cells apoptosis as well as cleaved caspase-3 and Bax protein expression and fluorescence intensity decreased. The expression and fluorescence intensity of Bcl-2, phosphorylated JAK2, and STAT3 increased. AG490, a specific inhibitor of the JAK2/STAT3 signalling pathway, was used. Our findings suggest that AG490 attenuates the effects of scutellarin. Our study revealed that scutellarin inhibited OGD-activated microglia-mediated PC12 cells apoptosis which was regulated via the JAK2/STAT3 signalling pathway.


Apigenin , Apoptosis , Glucuronates , Janus Kinase 2 , Microglia , STAT3 Transcription Factor , Signal Transduction , Animals , Apigenin/pharmacology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Glucuronates/pharmacology , PC12 Cells , Apoptosis/drug effects , Microglia/drug effects , Microglia/metabolism , Signal Transduction/drug effects , Rats , Mice , Caspase 3/metabolism , Glucose/metabolism , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , bcl-2-Associated X Protein/metabolism , Tyrphostins/pharmacology
17.
Sci Transl Med ; 16(751): eadj9672, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38865481

Cancer treatment with anti-PD-1 immunotherapy can cause central nervous system immune-related adverse events (CNS-irAEs). The role of microglia in anti-PD-1 immunotherapy-induced CNS-irAEs is unclear. We found that anti-PD-1 treatment of mice caused morphological signs of activation and major histocompatibility complex (MHC) class II up-regulation on microglia. Functionally, anti-PD-1 treatment induced neurocognitive deficits in mice, independent of T cells, B cells, and natural killer cells. Instead, we found that microglia mediated these CNS-irAEs. Single-cell RNA sequencing revealed major transcriptional changes in microglia upon anti-PD-1 treatment. The anti-PD-1 effects were mediated by anti-PD-1 antibodies interacting directly with microglia and were not secondary to peripheral T cell activation. Using a proteomics approach, we identified spleen tyrosine kinase (Syk) as a potential target in activated microglia upon anti-PD-1 treatment. Syk inhibition reduced microglia activation and improved neurocognitive function without impairing anti-melanoma effects. Moreover, we analyzed CNS tissue from a patient cohort that had received anti-PD-1 treatment. Imaging mass cytometry revealed that anti-PD-1 treatment of patients was associated with increased surface marker expression indicative of microglia activation. In summary, we identified a disease-promoting role for microglia in CNS-irAEs driven by Syk and provide an inhibitor-based approach to interfere with this complication after anti-PD-1 immunotherapy.


Central Nervous System , Immunotherapy , Microglia , Programmed Cell Death 1 Receptor , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Immunotherapy/adverse effects , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Humans , Central Nervous System/pathology , Central Nervous System/drug effects , Mice, Inbred C57BL , Syk Kinase/metabolism , Mice
18.
Sci Transl Med ; 16(751): eadi3259, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38865485

Robust structural remodeling and synaptic plasticity occurs within spinal autonomic circuitry after severe high-level spinal cord injury (SCI). As a result, normally innocuous visceral or somatic stimuli elicit uncontrolled activation of spinal sympathetic reflexes that contribute to systemic disease and organ-specific pathology. How hyperexcitable sympathetic circuitry forms is unknown, but local cues from neighboring glia likely help mold these maladaptive neuronal networks. Here, we used a mouse model of SCI to show that microglia surrounded active glutamatergic interneurons and subsequently coordinated multi-segmental excitatory synaptogenesis and expansion of sympathetic networks that control immune, neuroendocrine, and cardiovascular functions. Depleting microglia during critical periods of circuit remodeling after SCI prevented maladaptive synaptic and structural plasticity in autonomic networks, decreased the frequency and severity of autonomic dysreflexia, and prevented SCI-induced immunosuppression. Forced turnover of microglia in microglia-depleted mice restored structural and functional indices of pathological dysautonomia, providing further evidence that microglia are key effectors of autonomic plasticity. Additional data show that microglia-dependent autonomic plasticity required expression of triggering receptor expressed on myeloid cells 2 (Trem2) and α2δ-1-dependent synaptogenesis. These data suggest that microglia are primary effectors of autonomic neuroplasticity and dysautonomia after SCI in mice. Manipulating microglia may be a strategy to limit autonomic complications after SCI or other forms of neurologic disease.


Microglia , Neuronal Plasticity , Spinal Cord Injuries , Animals , Microglia/pathology , Microglia/metabolism , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/pathology , Mice , Receptors, Immunologic/metabolism , Membrane Glycoproteins/metabolism , Autonomic Nervous System/physiopathology , Mice, Inbred C57BL , Synapses/metabolism , Interneurons/metabolism
19.
J Neuroinflammation ; 21(1): 148, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38840180

BACKGROUND: White matter injury (WMI) represents a significant etiological factor contributing to neurological impairment subsequent to Traumatic Brain Injury (TBI). CD36 receptors are recognized as pivotal participants in the pathogenesis of neurological disorders, including stroke and spinal cord injury. Furthermore, dynamic fluctuations in the phenotypic polarization of microglial cells have been intimately associated with the regenerative processes within the injured tissue following TBI. Nevertheless, there is a paucity of research addressing the impact of CD36 receptors on WMI and microglial polarization. This investigation aims to elucidate the functional role and mechanistic underpinnings of CD36 in modulating microglial polarization and WMI following TBI. METHODS: TBI models were induced in murine subjects via controlled cortical impact (CCI). The spatiotemporal patterns of CD36 expression were examined through quantitative polymerase chain reaction (qPCR), Western blot analysis, and immunofluorescence staining. The extent of white matter injury was assessed via transmission electron microscopy, Luxol Fast Blue (LFB) staining, and immunofluorescence staining. Transcriptome sequencing was employed to dissect the molecular mechanisms underlying CD36 down-regulation and its influence on white matter damage. Microglial polarization status was ascertained using qPCR, Western blot analysis, and immunofluorescence staining. In vitro, a Transwell co-culture system was employed to investigate the impact of CD36-dependent microglial polarization on oligodendrocytes subjected to oxygen-glucose deprivation (OGD). RESULTS: Western blot and qPCR analyses revealed that CD36 expression reached its zenith at 7 days post-TBI and remained sustained at this level thereafter. Immunofluorescence staining exhibited robust CD36 expression in astrocytes and microglia following TBI. Genetic deletion of CD36 ameliorated TBI-induced white matter injury, as evidenced by a reduced SMI-32/MBP ratio and G-ratio. Transcriptome sequencing unveiled differentially expressed genes enriched in processes linked to microglial activation, regulation of neuroinflammation, and the TNF signaling pathway. Additionally, bioinformatics analysis pinpointed the Traf5-p38 axis as a critical signaling pathway. In vivo and in vitro experiments indicated that inhibition of the CD36-Traf5-MAPK axis curtailed microglial polarization toward the pro-inflammatory phenotype. In a Transwell co-culture system, BV2 cells treated with LPS + IFN-γ exacerbated the damage of post-OGD oligodendrocytes, which could be rectified through CD36 knockdown in BV2 cells. CONCLUSIONS: This study illuminates that the suppression of CD36 mitigates WMI by constraining microglial polarization towards the pro-inflammatory phenotype through the down-regulation of the Traf5-MAPK signaling pathway. Our findings present a potential therapeutic strategy for averting neuroinflammatory responses and ensuing WMI damage resulting from TBI.


CD36 Antigens , Mice, Inbred C57BL , Microglia , Animals , Microglia/metabolism , Microglia/pathology , Mice , CD36 Antigens/metabolism , CD36 Antigens/genetics , Mice, Knockout , White Matter/pathology , White Matter/metabolism , MAP Kinase Signaling System/physiology , Male , Cell Polarity/physiology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Signal Transduction/physiology
20.
J Cell Mol Med ; 28(10): e18396, 2024 May.
Article En | MEDLINE | ID: mdl-38801304

Previous studies have found that ferroptosis plays an important role in a variety of neurological diseases. However, the precise role of ferroptosis in the multiple sclerosis patients remains uncertain. We defined and validated a computational metric of ferroptosis levels. The ferroptosis scores were computed using the AUCell method, which reflects the enrichment scores of ferroptosis-related genes through gene ranking. The reliability of the ferroptosis score was assessed using various methods, involving cells induced to undergo ferroptosis by six different ferroptosis inducers. Through a comprehensive approach integrating snRNA-seq, spatial transcriptomics, and spatial proteomics data, we explored the role of ferroptosis in multiple sclerosis. Our findings revealed that among seven sampling regions of different white matter lesions, the edges of active lesions exhibited the highest ferroptosis score, which was associated with activation of the phagocyte system. Remyelination lesions exhibit the lowest ferroptosis score. In the cortex, ferroptosis score were elevated in neurons, relevant to a variety of neurodegenerative disease-related pathways. Spatial transcriptomics demonstrated a significant co-localization among ferroptosis score, neurodegeneration and microglia, which was verified by spatial proteomics. Furthermore, we established a diagnostic model of multiple sclerosis based on 24 ferroptosis-related genes in the peripheral blood. Ferroptosis might exhibits a dual role in the context of multiple sclerosis, relevant to both neuroimmunity and neurodegeneration, thereby presenting a promising and novel therapeutic target. Ferroptosis-related genes in the blood that could potentially serve as diagnostic and prognostic markers for multiple sclerosis.


Ferroptosis , Multiple Sclerosis , Proteomics , Ferroptosis/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Humans , Proteomics/methods , Transcriptome , Microglia/metabolism , Microglia/pathology , Gene Expression Profiling , Computational Biology/methods , Neurons/metabolism , Neurons/pathology , Multiomics
...