bims-microg Biomed News
on Microglia in health and disease
Issue of 2024–12–15
24 papers selected by
Marcus Karlstetter, Universität zu Köln



  1. Immunity. 2024 Dec 06. pii: S1074-7613(24)00513-2. [Epub ahead of print]
      The gene inositol polyphosphate-5-phosphatase D (INPP5D), which encodes the lipid phosphatase SH2-containing inositol polyphosphate 5-phosphatase 1 (SHIP1), is associated with the risk of Alzheimer's disease (AD). How it influences microglial function and brain physiology is unclear. Here, we showed that SHIP1 was enriched in early stages of healthy brain development. By combining in vivo loss-of-function approaches and proteomics, we discovered that mice conditionally lacking microglial SHIP1 displayed increased complement and synapse loss in the early postnatal brain. SHIP1-deficient microglia showed altered transcriptional signatures and abnormal synaptic pruning that was dependent on the complement system. Mice exhibited cognitive defects in adulthood only when microglial SHIP1 was depleted early postnatally but not at later stages. Induced pluripotent stem cell (iPSC)-derived microglia lacking SHIP1 also showed increased engulfment of synaptic structures. These findings suggest that SHIP1 is essential for proper microglia-mediated synapse remodeling in the healthy developing brain. Disrupting this process has lasting behavioral effects and may be linked to vulnerability to neurodegeneration.
    Keywords:  AD risk genes; Alzheimer’s disease; INPP5D; SHIP1; cognitive dysfunction; complement; microglia; synaptic pruning
    DOI:  https://doi.org/10.1016/j.immuni.2024.11.003
  2. Cell Rep. 2024 Dec 06. pii: S2211-1247(24)01403-7. [Epub ahead of print]43(12): 115052
      How microglia digest Alzheimer's fibrillar amyloid-beta (Aβ) plaques that are too large to be phagocytosed is not well understood. Here, we show that primary microglial cells create acidic extracellular compartments, lysosomal synapses, on model plaques and digest them with exocytosed lysosomal enzymes. This mechanism, called digestive exophagy, is confirmed by electron microscopy in 5xFAD mouse brains, which shows that a lysosomal enzyme, acid phosphatase, is secreted toward the plaques in structures resembling lysosomal synapses. Signaling studies demonstrate that the PI3K-AKT pathway modulates the formation of lysosomal synapses, as inhibition of PI3K1β or AKT1/2 reduces both lysosome exocytosis and actin polymerization, both required for the formation of the compartments. Finally, we show that small fibrils of Aβ previously internalized and trafficked to lysosomes are exocytosed toward large Aβ aggregates by microglia. Thus, the release of lysosomal contents during digestive exophagy may also contribute to the spread and growth of fibrillar Aβ.
    Keywords:  5xFAD; Alzheimer’s disease; CP: Neuroscience; acid phosphatase; amyloid-beta; digestive exophagy; electron microscopy; extracellular degradation; lysosomal pH; lysosome; microglia
    DOI:  https://doi.org/10.1016/j.celrep.2024.115052
  3. Nat Rev Immunol. 2024 Dec 09.
      Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
    DOI:  https://doi.org/10.1038/s41577-024-01104-7
  4. J Neurochem. 2025 Jan;169(1): e16275
      Neuroinflammation plays an important role in the pathological cascade of Alzheimer's disease (AD) along with aggregation of extracellular amyloid-β (Aβ) plaques and intracellular aggregates of tau protein. In animal models of amyloidosis, local immune activation is centered around Aβ plaques, which are usually of uniform morphology, dependent on the transgenic model used. In postmortem human brains a diversity of Aβ plaque morphologies is seen including diffuse plaques (non-neuritic plaques, non-NP), dense-core plaques, cotton-wool plaques, and NP. In a recent study, we demonstrated that during the progression of Alzheimer's disease neuropathologic changes (ADNC), a transformation of non-NP into NP occurs which is tightly linked to the emergence of cortical, but not hippocampal neurofibrillary tangle (NFT) pathology. This highlights the central role of NP in AD pathogenesis as well as brain region-specific differences in NP formation. In order to correlate the transformation of plaque types with local immune activation, we quantified the clustering and phenotype of microglia and accumulation of astrocytes around non-NP and NP during the progression of ADNC. We hypothesize that glial clustering occurs in response to formation of neuritic dystrophy around NP. First, we show that Iba1-positive microglia preferentially cluster around NP. Utilizing microglia phenotypic markers, we furthermore demonstrate that CD68-positive phagocytic microglia show a strong preference to cluster around NP in both the hippocampus and frontal cortex. A similar preferential clustering is observed for CD11c and ferritin-positive microglia in the frontal cortex, while this preference is less pronounced in the hippocampus, highlighting differences between hippocampal and cortical Aβ plaques. Glial fibrillary acidic protein-positive astrocytes showed a clear preference for clustering around NP in both the frontal cortex and hippocampus. These data support the notion that NP are intimately associated with the neuroimmune response in AD and underscore the importance of the interplay of protein deposits and the immune system in the pathophysiology of AD.
    Keywords:  Alzheimer's disease; dystrophic neurite; microglial clustering; neuritic plaque
    DOI:  https://doi.org/10.1111/jnc.16275
  5. Adv Sci (Weinh). 2024 Dec 12. e2413451
      Parkinson's disease (PD) is characterized by the deposition of misfolded α-synuclein (α-syn) in the brain. Converging evidence indicates that the intracellular transmission and subsequent templated amplification of α-syn are involved in the onset and progression of PD. However, the molecular mechanisms underlying the cell-to-cell transmission of pathological α-syn remain poorly understood. Microglia is highly activated in the brains of PD patients. Here, it is shown that depletion of microglia slows the spread of pathological α-syn pathology in mice injected with α-syn fibrils. Microglia phagocytose α-syn fibrils and transform them into more toxic species. The phagocytosis of α-syn fibrils by microglia is partially mediated by triggering a receptor expressed on myeloid cells 2 (TREM2), a transmembrane protein expressed on the surface of microglia. The endocytosed α-syn fibrils are then cleaved by the lysosomal proteinase asparagine endopeptidase (AEP) to generate truncated α-syn 1-103 fibrils with enhanced seeding activity. Knockout of TREM2 and AEP impedes the endocytosis and cleavage of α-syn fibrils, respectively. The results demonstrate that TREM2-mediated phagocytosis of α-syn fibrils by microglia and subsequent AEP-mediated cleavage of α-syn fibrils contribute to the spread of α-syn in the brain. Blocking either of these two steps attenuates the progression of α-syn pathology.
    Keywords:  Parkinson's disease; asparagine endopeptidase; microglia; triggering receptor expressed on myeloid cells 2; α‐synuclein
    DOI:  https://doi.org/10.1002/advs.202413451
  6. Metab Brain Dis. 2024 Dec 10. 40(1): 57
      Acute liver failure results from severe hepatic injury and can lead to neurological dysfunction known as hepatic encephalopathy (HE). Thrombospondin-1 can contribute to HE by increasing cerebral edema and microglia activation in the azoxymethane (AOM) mouse model. CD47 is a receptor for TSP1 and can directly modulate inflammation in numerous disease states. However, the role of CD47 in the progression of HE is currently unknown. Therefore, the aim of this study was to assess the role of CD47 in liver and brain pathology in the AOM mouse model of HE. C57Bl/6 or CD47 knockout (CD47-/-) were administered AOM to induce acute liver failure and HE. Liver damage was evaluated by measuring serum transaminases and histological assessment. Neurological function was determined by evaluating the time taken to reach coma (loss of all reflexes), cerebral edema, and microglia activation. CD47 signaling, and downstream signaling pathways, were assessed by real-time PCR, western blotting, immunofluorescence, and immunohistochemistry. AOM-treated mice had increased expression of CD47 in the liver, cortex, hippocampus, and cerebellum when compared to vehicle-treated mice. CD47-/- AOM-treated mice had reduced liver injury and apoptosis when compared to wildtype AOM-treated mice. A slower degree of neurological decline and less cerebral edema were observed in CD47-/- mice compared to wildtype AOM-treated mice. This was associated with decreased microglia proliferation and increased SOD1 expression in CD47-/- mice compared to wildtype AOM-treated mice. These findings support that CD47 signaling exacerbates AOM-induced acute liver failure and HE by inducing hepatic cell death, cerebral edema, and microglia activation.
    Keywords:  Acute liver failure; CD47; Hepatic encephalopathy
    DOI:  https://doi.org/10.1007/s11011-024-01493-7
  7. Exp Neurol. 2024 Dec 10. pii: S0014-4886(24)00440-0. [Epub ahead of print] 115114
      Postoperative cognitive dysfunction (POCD) is a common complication following surgeries involving general anesthesia. Although the CCL5-CCR5 axis is implicated in various neurological conditions, its role in POCD remains unclear. In our POCD model, we observed an increase in CCL5 and CCR5 levels concurrent with microglial activation and significant upregulation of inflammatory cytokines IL-6 and IL-1β. Administration of MVC, a CCR5 antagonist, alleviated neuroinflammation, prevented dendritic spine loss, and improved cognitive deficits by inhibiting the CCR5/CREB/NLRP1 pathway. However, the cognitive benefits of MVC were reversed by the CREB inhibitor 666-15. Our findings highlight the potential of targeting the CCL5-CCR5 axis as a therapeutic strategy for preventing and treating POCD.
    Keywords:  CCL5-CCR5 axis; Microglia; Postoperative cognitive dysfunction (POCD)
    DOI:  https://doi.org/10.1016/j.expneurol.2024.115114
  8. Immun Inflamm Dis. 2024 Dec;12(12): e70092
       BACKGROUND: Psoriasis is recognized as a systemic disease for its accompanying comorbidities, among which psychological disorders present a high incidence rate and affect patients' life quality. Interleukin (IL)-17A is the central pathological factor in the pathogenesis and development of psoriasis.
    OBJECTIVE: To clarify if psoriasis-induced systemic IL-17A increase can mediate the neuronal inflammation and result in depressive-like symptoms.
    METHODS: Psoriasiform dermatitis model was established by imiquimod (IMQ) application on male BALB/c mice and IL-17A intervention was performed by lateral ventricular catheterization. Skin structural, histopathological characteristics, and behavioral tests were assessed. Serum IL-17A levels were detected by Enzyme-linked immunosorbent assay. mRNA expression of pro-inflammatory factors IL-1β, IL-6, and tumor necrosis factor-α (TNF-α) as well as anti-inflammatory factors IL-4 and IL-10 in the hippocampus and cortex were measured by RT-qPCR. The number of microglia and hippocampal neurons was quantified by immunofluorescent assay.
    RESULTS: IMQ treatment resulted in significant skin structural and histopathological characters of psoriasiform dermatitis with elevated serum IL-17A levels, obvious depressive-like behaviors, microglia activation with increased IL-1β, IL-6, and TNF-α expression levels in the hippocampus and cortex, and notable inhibition of hippocampal neurogenesis. While, IL-17A neutralization by intracerebroventricular injection of anti-IL-17A antibody can remarkably inhibit microglia activation and decrease the abnormally increased expression levels of IL-1β, IL-6, and TNF-α in the hippocampus and cortex of psoriasiform dermatitis mice, promote hippocampal neurogenesis, thus alleviate the depressive-like behaviors.
    CONCLUSION: In the pathological condition of psoriasis, systemic IL-17A elevation can trigger microglia activation, provoke pro-inflammation mediators to release, evoke neuroinflammation, subsequently inhibit hippocampal neurogenesis, and result in depression. IL-17A, as an important pathogenic factor in psoriasis, contributes to its critical role in mediating systemic inflammation and depression comorbidity.
    Keywords:  IL‐17A; depression; microglia; psoriasis
    DOI:  https://doi.org/10.1002/iid3.70092
  9. Front Aging Neurosci. 2024 ;16 1503336
       Introduction: During aging, many cellular processes, such as autophagic clearance, DNA repair, mitochondrial health, metabolism, nicotinamide adenine dinucleotide (NAD+) levels, and immunological responses, become compromised. Urolithin A (UA) and Nicotinamide Riboside (NR) are two naturally occurring compounds known for their anti-inflammatory and mitochondrial protective properties, yet the effects of these natural substances on microglia cells have not been thoroughly investigated. As both UA and NR are considered safe dietary supplements, it is equally important to understand their function in normal cells and in disease states.
    Methods: This study investigates the effects of UA and NR on immune signaling, mitochondrial function, and microglial activity in a human microglial cell line (HMC3).
    Results: Both UA and NR were shown to reduce DNA damage-induced cellular senescence. However, they differentially regulated gene expression related to neuroinflammation, with UA enhancing cGAS-STING pathway activation and NR displaying broader anti-inflammatory effects. Furthermore, UA and NR differently influenced mitochondrial dynamics, with both compounds improving mitochondrial respiration but exhibiting distinct effects on production of reactive oxygen species and glycolytic function.
    Discussion: These findings underscore the potential of UA and NR as therapeutic agents in managing neuroinflammation and mitochondrial dysfunction in neurodegenerative diseases.
    Keywords:  aging; innate immune signaling; microglia; mitochondrial health; nicotinamide riboside; urolithin A
    DOI:  https://doi.org/10.3389/fnagi.2024.1503336
  10. Acta Pharmacol Sin. 2024 Dec 11.
      Despite the vital role of astrocytes in preserving blood-brain barrier (BBB) integrity, their therapeutic potential as targets in ischemic stroke-induced barrier disruption remains underexplored. We previously reported externalization of phosphatidylserine (PS) on astrocytic membranes concurrent with the emergence of PS externalization in neurons. PS externalization of astrocytes induced microglial phagocytosis of astrocytes, resulting in reduced astrocyte-vascular coupling and subsequent BBB breakdown. Annexin A5 (ANXA5) belongs to the superfamily of calcium (Ca2+)- and phospholipid-binding proteins. Here, we report two X-ray structures of human ANXA5, including monomeric ANXA5 (1.42 Å) and dimeric ANXA5 (1.80 Å). Through the combination of molecular docking and functional analysis, we explored the mechanism of action of ANXA5 in stroke treatment. In addition, we observed a clear increase in therapeutic efficacy corresponding to the increased affinity of ANXA5 for PS. In summary, the phagocytosis of PS-externalized astrocytes by microglia has emerged as a critical mechanism driving BBB breakdown after ischemia. Our findings offer valuable structural insight into ANXA5 as an innovative pharmacological target for safeguarding blood-brain barrier integrity after cerebral ischemia. These insights may facilitate the development of novel PS-targeting medications aimed at achieving enhanced efficacy with minimal side effects.
    Keywords:  Annexin A5; astrocytes; blood-brain barrier; cerebral ischemia; phosphatidylserine
    DOI:  https://doi.org/10.1038/s41401-024-01432-3
  11. J Neurochem. 2025 Jan;169(1): e16276
      To date, several studies have integrated genome-wide association studies (GWAS) and expression quantitative trait loci (eQTL) data from bulk tissues to identify novel Alzheimer's disease (AD) genetic variants and susceptibility genes. However, there is highly cell-type-specific nature in different bulk eQTL data. Until now, eQTL data from different brain single cells have been reported. Therefore, integrating eQTL data from different brain single-cell types along with AD GWAS data makes biological sense for studying the potential biological explanations of AD. Here, we utilized the summary-data-based Mendelian randomization (SMR) method to integrate AD GWAS data with eQTL data from eight brain single-cell types. We identified a larger number of significant genes compared to previous SMR study based on bulk eQTL. Notably, microglia exhibited the highest number of significant genes. Moreover, we conducted validation-phase SMR analysis, single-cell analysis, protein-protein interaction (PPI), druggability evaluation, functional enrichment analyses, and colocalization analysis of the top 20 SMR significant genes in microglia. We found that most genes passed the validation and were significantly enriched in microglia. PPI analysis uncovered interactions among PICALM, BIN1, RIN3, CD2AP, CASS4, and MS4A6E. Five most significant SMR genes were further validated through colocalization analysis. RIN3 is the only significant gene across all mentioned analyses and is a novel AD susceptibility gene at the genome-wide significance level. Druggability evaluation identified KCNQ3, HLA-DQB1, and RABEP1 as known genes previously targeted for drug development in neurological disorders, suggesting their potential therapeutic relevance in AD.
    Keywords:  Alzheimer's disease; expression quantitative trait loci; genome‐wide association studies; single‐cell; summary‐data‐based Mendelian randomization
    DOI:  https://doi.org/10.1111/jnc.16276
  12. Free Radic Biol Med. 2024 Dec 04. pii: S0891-5849(24)01107-9. [Epub ahead of print]227 190-200
       BACKGROUND: The established body of knowledge attests to the pivotal influence of ANGPTL4 on lipid metabolism and vascular biology. Nevertheless, its potential implication in neurodegenerative disease remains to be fully characterized.
    METHODS: The present investigation delves into the involvement of ANGPTL4 in the pathological progression of PD, both in vitro and in vivo. PD models were induced by intraperitoneal administration of MPTP and LPS in WT and ANGPTL4-/- mice. Additionally, rANGPTL4 was administered intravenously via the tail. Primary microglia cells cultured from the SNpc and Str regions of brains were exposed to LPS to induce neuroinflammation.
    RESULTS: The observations unveiled that ANGPTL4 deficiency exacerbated behavioral aberrations, intensified dopaminergic neuron loss, and stimulated microglial activation along with p21-dependent senescence. There was an elevation in the expression of proinflammatory cytokines in the PD model. Furthermore, the administration of rANGPTL4 protein reversed the observed phenotypes in ANGPTL4-/- mice, a phenomenon further validated in LPS-induced cells. Clinical specimens also manifested diminished levels of ANGPTL4 expression in PD patients. ANGPTL4 demonstrated the ability to alleviate neuroinflammation by suppressing EIF2-JNK-mediated ER stress and eliminating senescent cells.
    CONCLUSION: Our findings posit a salutary role for ANGPTL4 in counteracting PD, rendering it a prospective therapeutic target for the development of innovative drugs aimed at treating neuroinflammation-associated neurological diseases, including PD.
    Keywords:  ANGPTL4; EIF2-JNK pathway; Microglia; PD; p21 dependent senescence
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2024.12.009
  13. Int Immunopharmacol. 2024 Dec 10. pii: S1567-5769(24)02252-5. [Epub ahead of print]145 113730
      Macrophages (MØ) participate in the induction and the control of the host's immune response in homeostasis and during inflammatory diseases. Sitagliptin is a drug that inhibits the enzyme dipeptidyl peptidase 4 (DPP-4) and, therefore, increases the bioavailability of the incretins GIP (Gastric inhibitory polypeptide) and GLP-1 (Glucagon-like polypeptide). Thus, sitagliptin has been used to treat obesity and type II diabetes and has recently been associated with anti-inflammatory effects. It is known that the drug can modulate the immune response, however, the underlying mechanisms are not yet completely elucidated, including how they interfere with the activation and function of MØ. Here, we aimed to investigate and characterize the effects of in vitro treatment with sitagliptin on MØ polarization. Bone marrow-derived MØ were differentiated with conditioned medium from the L929 cell line. For M1, MØ were stimulated with IFN-γ and LPS, and for M2, with IL-4 and IL-13 for 24 h. Sitagliptin treatment was performed during MØ polarization. Polarized MØ were assessed for M1/M2 markers, DPP-4, GLP-1 and GIP receptors, mitochondrial dynamics and phagocytosis. Sitagliptin treatment exacerbates the M2 phenotype, featured by increased expression of CD206 and ARG1 and decreased gene expression levels of TNF-α. Sitagliptin-treated M2 altered mitochondrial dynamics with reduced membrane potential and mitochondrial reactive oxygen species production. These differences were accompanied by low gene expression levels of genes related to mitofusion, suggesting that sitagliptin treatment interferes with mitochondria function in M2, and exhibited less phagocytic capacity. In summary, our data suggest that sitagliptin exacerbates M2 profile in vitro.
    Keywords:  DPP-4; GIPR; GLP-1R; Macrophages; Sitagliptin
    DOI:  https://doi.org/10.1016/j.intimp.2024.113730
  14. Int Immunopharmacol. 2024 Dec 07. pii: S1567-5769(24)02253-7. [Epub ahead of print]145 113731
      The primary objective of this study was to investigate the potential pharmacological effects of Hyperoside (Hyp) extract on chronic social defeat stress (CSDS)-induced depression-like behavior in mice. We established CSDS mice to evaluate the antidepressant effects of Hyp. Additionally, We assessed the changes in neuroinflammatory factors in the TRX1/NLRP1/Caspase-1 signaling pathway using adeno-associated virus (AAV) and BV2 microglial cells. The expression levels of TRX1 protein and BDNF also increased by Hyp, while NLRP1 and Caspase-1 a significant decrease. Additionally, Hyp was found to inhibit TRX1 ubiquitination in the microglial inflammation model. In both in vivo and in vitro experiments, it was found that Hyp significantly promotes microglial polarization towards the M2 phenotype in the hippocampus and alleviates neuroinflammation, thereby improving depression-like behavior in CSDS mice. This is associated with the regulation of TRX1 ubiquitination, which inhibits the expression levels of NLRP1 and Caspase-1 proteins.
    Keywords:  Depression; Hyperoside; Microglia polarization; Neuroinflammation; TRX1
    DOI:  https://doi.org/10.1016/j.intimp.2024.113731
  15. Brain Behav Immun. 2024 Dec 11. pii: S0889-1591(24)00742-6. [Epub ahead of print]
      Microglia, known for their diverse roles in the central nervous system, have recently been recognized for their involvement in degrading the extracellular matrix. Perineuronal nets (PNNs), a specialized form of this matrix, are crucial for stabilizing neuronal connections and constraining plasticity. Our group recently reported increased PNN densities in the ventromedial prefrontal cortex (vmPFC) of depressed individuals that died by suicide in adulthood after experiencing childhood abuse (DS-CA) compared to matched controls. To explore potential underlying mechanisms, we employed a comprehensive approach in similar postmortem vmPFC samples, combining a human matrix metalloproteinase and chemokine array, isolation of CD11b-positive microglia and enzyme-linked immunosorbent assays (ELISA). Our findings indicate a significant downregulation of matrix metalloproteinase (MMP)-9 and tissue inhibitors of metalloproteinases (TIMP)-2 in both whole vmPFC grey matter and isolated microglial cells from DS-CA samples. Furthermore, our experiments reveal that a history of child abuse is associated with diminished levels of microglial CX3CR1 and IL33R in both vmPFC whole lysate and CD11b isolated cells. However, levels of the CX3CR1 ligand, CX3CL1 (Fractalkine), did not differ between groups. While these data suggest potential long-lasting alterations in microglial markers in the vmPFC of individuals exposed to severe childhood adversity, direct functional assessments were not conducted. Nonetheless, these findings offer insight into how childhood abuse may contribute to PNN alterations via microglial-related mechanisms.
    Keywords:  Adverse Childhood Experiences; Extracellular Matrix; Humans; Matrix Metalloproteinase 9; Microglia
    DOI:  https://doi.org/10.1016/j.bbi.2024.12.013
  16. Apoptosis. 2024 Dec 10.
      Programmed cell death (PCD) has emerged as a critical regulatory mechanism in the initiation and progression of various pathological conditions. PCD in microglia, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, occurs in a variety of central nervous system (CNS) diseases. Dysregulation of microglia can lead to excessive tissue damage or neuronal death in CNS injury. Various injury stimuli trigger aberrant activation of the PCD pathway of microglia, which then further leads to inflammatory cascades that exacerbates CNS pathology in a vicious cycle. Therefore, targeting PCD in microglia is considered an important avenue for the treatment of various neurodegenerative diseases and CNS injury. In this review, we summarize the major and recent findings focusing on the mechanisms of PCD in microglia modulating functions in neurodegenerative diseases and CNS injury and provide a systematic overview of the current inhibitors targeting various PCD pathways, which may provide important therapeutic targets that merit further investigation.
    Keywords:  CNS injury; Microglia; Neurodegenerative disease; Neuroinflammation; Programmed cell death
    DOI:  https://doi.org/10.1007/s10495-024-02041-5
  17. Int Immunopharmacol. 2024 Dec 12. pii: S1567-5769(24)02308-7. [Epub ahead of print]145 113786
      Trigeminal neuralgia (TN)-related cognitive impairment is a common cause of decreased quality of life in patients and is closely associated with neuroinflammation. Although minocycline has demonstrated anti-inflammatory, analgesic, and neuroprotective functions, its role in treating TN-related cognitive impairment remains unreported. In this study, we used an in vivo TN model and an in vitro model of primary microglial neuroinflammation to investigate the potential effects of minocycline on cognitive function and microglial polarization in TN rats. Our results suggested that minocycline treatment attenuated cognitive deficits by alleviating hippocampal neuronal damage and enhancing synaptic plasticity in TN rats. Furthermore, both in vitro and in vivo assays demonstrated that minocycline polarized activated microglia to the M2 phenotype, leading to the reduction of pro-inflammatory factors, including tumor necrosis factor-α and interleukin-1, and an increase in the anti-inflammatory factors, such as interleukin-4 and interleukin-10, thereby attenuating neuroinflammation. Moreover, it was found that the TLR4/MyD88/NF-κB pathway was involved in the shift of microglia from a pro-inflammatory (M1) to an anti-inflammatory (M2). In summary, minocycline likely mediated the process of microglia polarization partly via the TLR4/MyD88/NF-κB pathway, promoting neuronal survival and restoring synaptic plasticity, thereby improving TN-related cognitive impairment.
    Keywords:  Cognitive impairment; Microglial polarization; Minocycline; Neuroinflammation; Toll-like receptor 4; Trigeminal neuralgia
    DOI:  https://doi.org/10.1016/j.intimp.2024.113786
  18. Neuroscience. 2024 Dec 11. pii: S0306-4522(24)00721-8. [Epub ahead of print]
       BACKGROUND: Microglia polarization plays a crucial role in inflammatory injury of brain following intracerebral hemorrhage (ICH). Heme oxygenase-1 (HO-1) has demonstrated protective properties against inflammation and promote hematoma clearance after ICH. The objective of this study was to explore impacts of HO-1 on microglia polarization and phagocytosis after ICH, along with the underlying mechanism.
    METHODS: ICH model was constructed in C57BL/6 mice. Neurological deficit of ICH mice was evaluated. HE detected pathological changes of mouse brain tissue. Immunofluorescence staining tested co-localization between HO-1 or NF-kB p65 and IBA1. The expressions of gene and proteins were detected by RT-qPCR and Western blot, respectively. Flow cytometry determined microglial polarization phenotype and neuron apoptosis. Cell viability of neuron was assessed by CCK-8. Red blood cells labeled by PKH-26 and co-cultured with microglia for examining microglial erythrophagocytosis.
    RESULTS: Both HO-1 and NF-kB p65 phosphorylation were elevated in brain tissues of ICH mice. ZnPP, a HO-1 inhibitor, could exacerbate microglial M1 polarization and nerve injury, as well as repress microglial erythrophagocytosis in vitro and hematoma clearance in vivo. On the contrary, Tat-NBD, a NF-kB inhibitor, greatly suppressed microglial M1 polarization, and induced M2 polarization and microglial erythrophagocytosis, thus improving nerve injury and hematoma clearance after ICH. Notably, it was observed that NF-kB p65 could be activated by ZnPP treatment, and the regulatory roles of ZnPP on microglial polarization and erythrophagocytosis after ICH in vivo and in vitro were all diminished by Tat-NBD.
    CONCLUSION: Therefore, our data demonstrated that HO-1 alleviated nerve injury and induced M2 polarization and phagocytosis of microglia after ICH via inhibiting NF-κB signaling pathway, which could provide deepen the pathological understanding of ICH and provide potential intervention targets and drug candidate for ICH.
    Keywords:  HO-1; Intracerebral hemorrhage; Microglia polarization; NF-kB; Phagocytosis
    DOI:  https://doi.org/10.1016/j.neuroscience.2024.12.020
  19. CNS Neurosci Ther. 2024 Dec;30(12): e70130
       BACKGROUND: The treatment of spinal cord injury (SCI) is usually ineffective, because neuroinflammatory secondary injury is an important cause of the continuous development of spinal cord injury, and microglial pyroptosis is an important step of neuroinflammation. Recently, Bmal1, a core component of circadian clock genes (CCGs), has been shown to play a regulatory role in various tissues and cells. However, it is still unclear whether Bmal1 regulates microglial pyroptosis after SCI.
    METHODS: In this study, we established an in vivo mouse model of SCI using Bmal1 knockout (KO) mice and wild-type (WT) mice, and lipopolysaccharide (LPS)-induced pyroptosis in BV2 cells as an in vitro model. A series of molecular and histological methods were used to detect the level of pyroptosis and explore the regulatory mechanism in vivo and in vitro respectively.
    RESULTS: Both in vitro and in vivo results showed that Bmal1 inhibited NLRP3 inflammasome activation and microglial pyroptosis after SCI. Further analysis showed that Bmal1 inhibited pyroptosis-related proteins (NLRP3, Caspase-1, ASC, GSDMD-N) and reduced the release of IL-18 and IL-1β by inhibiting the NF-κB /MMP9 pathway. It was important that NF-κB was identified as a transcription factor that promotes the expression of MMP9, which in turn regulates microglial pyroptosis after SCI.
    CONCLUSIONS: Our study initially identified that Bmal1 regulates the NF-κB /MMP9 pathway to reduce microglial pyroptosis and thereby reduce secondary spinal cord injury, providing a new promising therapeutic target for SCI.
    Keywords:  Bmal1; MMP9; NF‐κB; pyroptosis; spinal cord injury
    DOI:  https://doi.org/10.1111/cns.70130
  20. Biomed Res Int. 2024 ;2024 8027006
      Innate immune memory or trained immunity refers to a long-lasting response of the innate immune cells against repeated exposure to the homogenous or heterogenous infectious agent. The trained immunity is induced through epigenetic modification and is characterized by the change of both intracellular immunological signaling and cellular metabolism. Recently, different groups have tried to establish protocols to generate trained innate immune cells. However, the molecular basis of innate memory induction remains poorly understood. Here, we evaluated the impact of water-soluble chitosan on the innate immune memory induction in microglial cells primed with LPS. The trained-immune response was accessed by measuring proinflammatory markers, metabolic change, and epigenetic modification. We showed that the stimulation/restimulation with LPS only caused a robust reduction of iNOS, and proinflammatory cytokines, indicating induced immune tolerance. In contrast, the treatment of chitosan induces long-lasting memory microglial cells accompanied by a high level of iNOS, increased lactate production, induced epigenetic modification, and the upregulation of proinflammatory cytokines upon further exposure to the same stimulus. These findings suggest that chitosan induces microglial-trained immunity by targeting distinct epigenetic and metabolic pathways; therefore, chitosan treatment may provide a novel approach for targeting innate immunity towards a memory-like response in an in vitro model.
    Keywords:  microglia; trained immunity; water-soluble chitosan
    DOI:  https://doi.org/10.1155/bmri/8027006
  21. Neural Regen Res. 2024 Dec 07.
      Traumatic brain injury can be categorized into primary and secondary injuries. Secondary injuries are the main cause of disability following traumatic brain injury, which involves a complex multicellular cascade. Microglia play an important role in secondary injury and can be activated in response to traumatic brain injury. In this article, we review the origin and classification of microglia as well as the dynamic changes of microglia in traumatic brain injury. We also clarify the microglial polarization pathways and the therapeutic drugs targeting activated microglia. We found that regulating the signaling pathways involved in pro-inflammatory and anti-inflammatory microglia, such as the Toll-like receptor 4 / nuclear factor-kappa B, mitogen-activated protein kinase, Janus kinase/signal transducer and activator of transcription, phosphoinositide 3-kinase/protein kinase B, Notch, and high mobility group box 1 pathways, can alleviate the inflammatory response triggered by microglia in traumatic brain injury, thereby exerting neuroprotective effects. We also reviewed the strategies developed on the basis of these pathways, such as drug and cell replacement therapies. Drugs that modulate inflammatory factors, such as rosuvastatin, have been shown to promote the polarization of anti-inflammatory microglia and reduce the inflammatory response caused by traumatic brain injury. Mesenchymal stem cells possess anti-inflammatory properties, and clinical studies have confirmed their significant efficacy and safety in patients with traumatic brain injury. Additionally, advancements in mesenchymal stem cell-delivery methods-such as combinations of novel biomaterials, genetic engineering, and mesenchymal stem cell exosome therapy-have greatly enhanced the efficiency and therapeutic effects of mesenchymal stem cells in animal models. However, numerous challenges in the application of drug and mesenchymal stem cell treatment strategies remain to be addressed. In the future, new technologies, such as single-cell RNA sequencing and transcriptome analysis, can facilitate further experimental studies. Moreover, research involving non-human primates can help translate these treatment strategies to clinical practice.
    DOI:  https://doi.org/10.4103/NRR.NRR-D-24-00810
  22. Mol Neurobiol. 2024 Dec 13.
      Subarachnoid hemorrhage (SAH), a subtype of hemorrhagic stroke primarily resulting from the rupture of intracranial aneurysms, remains a significant contributor to disability and mortality, notwithstanding advancements in treatment. This study investigates the neuroprotective effects of pioglitazone in SAH, focusing on the PPAR-γ pathway and its potential role in mitigating early brain injury (EBI) following SAH. Neuroprotective efficacy was assessed through neurobehavioral assessment, brain water content analysis, TUNEL, immunofluorescence, western blotting, and inflammatory factor assay. Results indicate that pioglitazone treatment effectively mitigated brain edema, reduced neuronal death, and enhanced short-term neurobehavioral function in SAH-afflicted rats. Furthermore, pioglitazone demonstrated sustained improvements in long-term neurobehavioral function and decreased neuronal loss post-SAH. Mechanistically, SAH induced the polarization of microglia towards the M1 phenotype and the release of pro-inflammatory cytokines. Conversely, pioglitazone treatment predominantly shifted microglia polarization towards the M2 phenotype, eliciting a notable release of anti-inflammatory cytokines. Notably, the positive effects of pioglitazone were nullified by the PPAR-γ inhibitor T0070907. In conclusion, our findings suggest that pioglitazone may alleviate neuroinflammation by modulating microglia M1/M2 polarization through the PPAR-γ pathway, thereby conferring neuroprotection against SAH injury and positing itself as a potential therapeutic agent for SAH treatment.
    Keywords:  Early brain injury; Microglial polarization; Neuroinflammation; PPAR-γ; Subarachnoid hemorrhage
    DOI:  https://doi.org/10.1007/s12035-024-04664-w
  23. Neural Regen Res. 2024 Dec 07.
      Brain organoids encompass a large collection of in vitro stem cell-derived 3D culture systems that aim to recapitulate multiple aspects of in vivo brain development and function. First, this review provides a brief introduction to the current state-of-the-art for neuro-ectoderm brain organoid development, emphasizing their biggest advantages in comparison with classical two-dimensional cell cultures and animal models. However, despite their usefulness for developmental studies, a major limitation for most brain organoid models is the absence of contributing cell types from endodermal and mesodermal origin. As such, current research is highly investing towards the incorporation of a functional vasculature and the microglial immune component. In this review, we will specifically focus on the development of immune-competent brain organoids. By summarizing the different approaches applied to incorporate microglia, it is highlighted that immune-competent brain organoids are not only important for studying neuronal network formation, but also offer a clear future as a new tool to study inflammatory responses in vitro in 3D in a brain-like environment. Therefore, our main focus here is to provide a comprehensive overview of assays to measure microglial phenotype and function within brain organoids, with an outlook on how these findings could better understand neuronal network development or restoration, as well as the influence of physical stress on microglia-containing brain organoids. Finally, we would like to stress that even though the development of immune-competent brain organoids has largely evolved over the past decade, their full potential as a pre-clinical tool to study novel therapeutic approaches to halt or reduce inflammation-mediated neurodegeneration still needs to be explored and validated.
    DOI:  https://doi.org/10.4103/NRR.NRR-D-24-00921
  24. Neuroscience. 2024 Dec 07. pii: S0306-4522(24)00712-7. [Epub ahead of print]
      Ginsenoside Rg1 (Rg1) has been shown to treat a variety of human diseases, including Alzheimer's disease (AD). However, its mechanism in AD needs further investigation. Microglial cells (BV2) were treated with Aβ1-42 to induce AD cell models. Cell viability and apoptosis were tested by cell counting kit 8 assay and flow cytometry. The protein levels of GATA-binding protein 4 (GATA4), phosphodiesterase 4A (PDE4A), autophagy-related markers, M1/M2 polarization-related markers and PI3K/AKT-related markers were detected by western blot. Inflammation factors were detected by ELISA. Jaspar and dual-luciferase reporter assay were used to evaluate the interaction between GATA4 and PDE4A. Our results showed that Rg1 promoted viability and autophagy, while suppressed apoptosis and inflammation in Aβ1-42-induced BV2 cells. Rg1 reduced GATA4 protein expression, and GATA4 upregulation reversed the regulation of Rg1 on Aβ1-42-induced BV2 cell injury. GATA4 interacted with PDE4A, and GATA4 facilitated Aβ1-42-induced BV2 cell injury by increasing PDE4A expression. Besides, GATA4 knockdown reduced PDE4A protein expression and inactivated PI3K/AKT axis, while these effects were abolished by PDE4A overexpression. In conclusion, our data suggested that Ginsenoside Rg1 inhibited microglial cell apoptosis and inflammation to attenuate AD progression by regulating the GATA4/PDE4A/PI3K/AKT axis.
    Keywords:  Alzheimer’s disease; GATA4; Ginsenoside Rg1; PDE4A
    DOI:  https://doi.org/10.1016/j.neuroscience.2024.12.011