Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.478
Filter
1.
Invest Ophthalmol Vis Sci ; 65(6): 17, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38861275

ABSTRACT

Purpose: N6-methyladenosine (m6A) methylation is a chemical modification that occurs on RNA molecules, where the hydrogen atom of adenine (A) nucleotides is replaced by a methyl group, forming N6-methyladenosine. This modification is a dynamic and reversible process that plays a crucial role in regulating various biological processes, including RNA stability, transport, translation, and degradation. Currently, there is a lack of research on the role of m6A modifications in maintaining the characteristics of RPE cells. m6A readers play a crucial role in executing the functions of m6A modifications, which prompted our investigation into their regulatory roles in the RPE. Methods: Phagocytosis assays, immunofluorescence staining, flow cytometry experiments, ß-galactosidase staining, and RNA sequencing (RNA-seq) were conducted to assess the functional and cellular characteristics changes in retinal pigment epithelium (RPE) cells following short-hairpin RNA-mediated knockdown of insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2). RNA-seq and ultraviolet crosslinking immunoprecipitation with high-throughput sequencing (HITS-CLIP) were employed to identify the target genes regulated by IGF2BP2. adeno-associated virus (AAV) subretinal injection was performed in 6- to 8-week-old C57 mice to reduce IGF2BP2 expression in the RPE, and the impact of IGF2BP2 knockdown on mouse visual function was assessed using immunofluorescence, quantitative real-time PCR, optical coherence tomography, and electroretinography. Results: IGF2BP2 was found to have a pronounced effect on RPE phagocytosis. Subsequent in-depth exploration revealed that IGF2BP2 modulates the mRNA stability of PAX6 and OTX2, and the loss of IGF2BP2 induces inflammatory and aging phenotypes in RPE cells. IGF2BP2 knockdown impaired RPE function, leading to retinal dysfunction in vivo. Conclusions: Our data suggest a crucial role of IGF2BP2 as an m6A reader in maintaining RPE homeostasis by regulating the stability of PAX6 and OTX2, making it a potential target for preventing the occurrence of retinal diseases related to RPE malfunction.


Subject(s)
Homeostasis , Mice, Inbred C57BL , Otx Transcription Factors , PAX6 Transcription Factor , RNA-Binding Proteins , Retinal Pigment Epithelium , Retinal Pigment Epithelium/metabolism , Animals , Mice , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Homeostasis/physiology , Otx Transcription Factors/metabolism , Otx Transcription Factors/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Phagocytosis/physiology , Flow Cytometry , Gene Expression Regulation/physiology , Tomography, Optical Coherence , Electroretinography , Cells, Cultured
2.
Glia ; 72(8): 1484-1500, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780213

ABSTRACT

Microglia are innate immune cells in the brain and show exceptional heterogeneity. They are key players in brain physiological development regulating synaptic plasticity and shaping neuronal networks. In pathological disease states, microglia-induced synaptic pruning mediates synaptic loss and targeting microglia was proposed as a promising therapeutic strategy. However, the effect of microglia depletion and subsequent repopulation on dendritic spine density and neuronal function in the adult brain is largely unknown. In this study, we investigated whether pharmacological microglia depletion affects dendritic spine density after long-term permanent microglia depletion and after short-term microglia depletion with subsequent repopulation. Long-term microglia depletion using colony-stimulating-factor-1 receptor (CSF1-R) inhibitor PLX5622 resulted in increased overall spine density, especially of mushroom spines, and increased excitatory postsynaptic current amplitudes. Short-term PLX5622 treatment with subsequent repopulation of microglia had an opposite effect resulting in activated microglia with increased synaptic phagocytosis and consequently decreased spine density and reduced excitatory neurotransmission, while Barnes maze and elevated plus maze testing was unaffected. Moreover, RNA sequencing data of isolated repopulated microglia showed an activated and proinflammatory phenotype. Long-term microglia depletion might be a promising therapeutic strategy in neurological diseases with pathological microglial activation, synaptic pruning, and synapse loss. However, repopulation after depletion induces activated microglia and results in a decrease of dendritic spines possibly limiting the therapeutic application of microglia depletion. Instead, persistent modulation of pathological microglia activity might be beneficial in controlling synaptic damage.


Subject(s)
Brain , Dendritic Spines , Mice, Inbred C57BL , Microglia , Animals , Microglia/drug effects , Microglia/metabolism , Dendritic Spines/drug effects , Male , Mice , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Phagocytosis/physiology , Phagocytosis/drug effects , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Mice, Transgenic , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Organic Chemicals
3.
Biochem Pharmacol ; 225: 116281, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744379

ABSTRACT

Efferocytosis of massive non-viable germ cells by Sertoli cells (SCs), the specialized phagocytes, is essential for maintaining testis homeostasis. What elusive is the contribution of mitochondrial metabolism to this energy-consuming process, as SC has a preference of aerobic glycolysis. All-trans retinoic acid (ATRA, hereafter referred to as RA) is a well-known morphogen that primarily acts through the nuclear RA receptor (RAR). It sustains SC blood-testisbarrier integrity, and it's SC-derived RA sets the timing of meiotic commitment. In this study, we revisited RA in SC biology, from the perspective of SC-mediated efferocytosis. We provide evidence that RA induces transcriptional programming of multiple regulators involved in efferocytosis, which thereby represses SC-mediated efferocytosis, via a RAR-independent mechanism, as blocking pan-RAR activity fails to rescue RA-induced defective efferocytosis. RA-treated SCs exhibit alternations in mitochondrial dynamics and metabolism, and the hindered efferocytosis can be rescued by stimulating mitochondrial OXPHOS via pharmacological targeting of AMPK and PDK. We thus prefer to propose a signaling axis of RA-mitochondrial metabolism-efferocytosis. Our study uncovers a hitherto unappreciated role of RA in SC biology and tiers mitochondria metabolism to SC-mediated efferocytosis, contributing a deeper understanding of SC in male reproduction.


Subject(s)
Mitochondria , Phagocytosis , Sertoli Cells , Tretinoin , Sertoli Cells/metabolism , Sertoli Cells/drug effects , Male , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Tretinoin/pharmacology , Tretinoin/metabolism , Phagocytosis/drug effects , Phagocytosis/physiology , Receptors, Retinoic Acid/metabolism , Mice , Efferocytosis
4.
Exp Eye Res ; 244: 109935, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763352

ABSTRACT

Müller glia and microglia are capable of phagocytosing fragments of retinal cells in response to retinal injury or degeneration. However, the direct evidence for their mutual interactions between Müller glia and microglia in the progression of retinal degeneration (RD) remains largely unclear. This study aims to construct a progressive RD mouse model and investigate the activated pattern of Müller glia and the interplay between Müller glia and microglia in the early stage or progression of RD. A Prohibitin 2 (Phb2) photoreceptor-specific knockout (RKO) mouse model was generated by crossing Phb2flox/flox mice with Rhodopsin-Cre mice. Optical Coherence Tomography (OCT), histological staining, and Electroretinography (ERG) assessed retinal structure and function, and RKO mice exhibited progressive RD from six weeks of age. In detail, six-week-old RKO mice showed no significant retinal impairment, but severe vision dysfunction and retina thinning were shown in ten-week-old RKO mice. Furthermore, RKO mice were sensitive to Light Damage (LD) and showed severe RD at an early age after light exposure. Bulk retina RNA-seq analysis from six-week-old control (Ctrl) and RKO mice showed reactive retinal glia in RKO mice. The activated pattern of Müller glia and the interplay between Müller glia and microglia was visualized by immunohistology and 3D reconstruction. In six-week-old RKO mice or light-exposed Ctrl mice, Müller glia were initially activated at the edge of the retina. Moreover, in ten-week-old RKO mice or light-exposed six-week-old RKO mice with severe photoreceptor degeneration, abundant Müller glia were activated across the whole retinas. With the progression of RD, phagocytosis of microglia debris by activated Müller glia were remarkably increased. Altogether, our study establishes a Phb2 photoreceptor-specific knockout mouse model, which is a novel mouse model of RD and can well demonstrate the phenotype of progressive RD. We also report that Müller glia in the peripheral retina is more sensitive to the early damage of photoreceptors. Our study provides more direct evidence for Müller glia engulfing microglia debris in the progression of RD due to photoreceptor Phb2 deficiency.


Subject(s)
Disease Models, Animal , Electroretinography , Ependymoglial Cells , Mice, Knockout , Microglia , Photoreceptor Cells, Vertebrate , Prohibitins , Repressor Proteins , Retinal Degeneration , Tomography, Optical Coherence , Animals , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Microglia/metabolism , Microglia/pathology , Mice , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/deficiency , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/metabolism , Mice, Inbred C57BL , Phagocytosis/physiology
5.
Neuropathol Appl Neurobiol ; 50(3): e12982, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38742276

ABSTRACT

AIMS: Perineuronal nets (PNNs) are an extracellular matrix structure that encases excitable neurons. PNNs play a role in neuroprotection against oxidative stress. Oxidative stress within motor neurons can trigger neuronal death, which has been implicated in amyotrophic lateral sclerosis (ALS). We investigated the spatio-temporal timeline of PNN breakdown and the contributing cellular factors in the SOD1G93A strain, a fast-onset ALS mouse model. METHODS: This was conducted at the presymptomatic (P30), onset (P70), mid-stage (P130), and end-stage disease (P150) using immunofluorescent microscopy, as this characterisation has not been conducted in the SOD1G93A strain. RESULTS: We observed a significant breakdown of PNNs around α-motor neurons in the ventral horn of onset and mid-stage disease SOD1G93A mice compared with wild-type controls. This was observed with increased numbers of microglia expressing matrix metallopeptidase-9 (MMP-9), an endopeptidase that degrades PNNs. Microglia also engulfed PNN components in the SOD1G93A mouse. Further increases in microglia and astrocyte number, MMP-9 expression, and engulfment of PNN components by glia were observed in mid-stage SOD1G93A mice. This was observed with increased expression of fractalkine, a signal for microglia engulfment, within α-motor neurons of SOD1G93A mice. Following PNN breakdown, α-motor neurons of onset and mid-stage SOD1G93A mice showed increased expression of 3-nitrotyrosine, a marker for protein oxidation, which could render them vulnerable to death. CONCLUSIONS: Our observations suggest that increased numbers of MMP-9 expressing glia and their subsequent engulfment of PNNs around α-motor neurons render these neurons sensitive to oxidative damage and eventual death in the SOD1G93A ALS model mouse.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , Matrix Metalloproteinase 9 , Microglia , Phagocytosis , Superoxide Dismutase-1 , Animals , Mice , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Matrix Metalloproteinase 9/metabolism , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Motor Neurons/pathology , Motor Neurons/metabolism , Phagocytosis/physiology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
6.
Med Sci (Paris) ; 40(5): 428-436, 2024 May.
Article in French | MEDLINE | ID: mdl-38819278

ABSTRACT

The resolution of inflammation is an active process leading to the restoration of tissue homeostasis. A critical step in the initiation of this process is the elimination of apoptotic immune cells by macrophages. This well-organized process, called efferocytosis, involves four different steps, namely the attraction of macrophages to the site where the cells die, the recognition of apoptotic cells, their internalization and their digestion leading to the activation of different metabolic pathways. All these steps are responsible for the reprogramming of macrophages towards a pro-resolving profile. Efferocytic macrophages produce several factors involved in the resolution of inflammation. These factors include lipids (i.e., specialized pro-resolving mediators such as lipoxins), and proteins (e.g., IL-10 or TGF-ß). Here, we describe the different steps of efferocytosis and the mechanisms responsible for both macrophage reprogramming and the release of pro-resolving factors. These factors may represent a new therapeutic approach, called resolution therapy.


Title: « Fort comme la mort ¼,* où comment l'efferocytose contrôle la résolution de l'inflammation. Abstract: L'arrêt de la réponse inflammatoire, ou résolution de l'inflammation, est considéré aujourd'hui comme un processus actif lié à la production (ou à la libération) de composés anti-inflammatoires aussi appelés composés pro-résolutifs. L'évènement permettant d'enclencher la résolution de l'inflammation est l'élimination des cellules immunitaires apoptotiques par les macrophages, un processus nommé efferocytose, dont l'altération est à l'origine de différentes maladies. Dans cette synthèse, nous décrivons les étapes de cette efferocytose et les mécanismes qui en résultent et permettent de stopper l'inflammation. Nous évoquerons également de nouvelles pistes thérapeutiques fondées sur les facteurs pro-résolutifs : la thérapie résolutive.


Subject(s)
Apoptosis , Inflammation , Macrophages , Phagocytosis , Macrophages/immunology , Macrophages/physiology , Inflammation/pathology , Humans , Phagocytosis/physiology , Animals , Apoptosis/physiology
7.
CNS Neurosci Ther ; 30(5): e14742, 2024 05.
Article in English | MEDLINE | ID: mdl-38715283

ABSTRACT

BACKGROUND: Adenosine A3 receptor (ADORA3) belongs to the adenosine receptor families and the role of ADORA3 in vascular dementia (VaD) is largely unexplored. The present study sought to determine the therapeutic role of ADORA3 antagonist in a mouse model of VaD. METHODS: The GSE122063 dataset was selected to screen the differential expression genes and pathways between VaD patients and controls. A mouse model of bilateral carotid artery stenosis (BCAS) was established. The cognitive functions were examined by the novel object recognition test, Y maze test, and fear of conditioning test. The white matter injury (WMI) was examined by 9.4 T MRI, western blot, and immunofluorescence staining. The mechanisms of ADORA3-regulated phagocytosis by microglia were examined using qPCR, western blot, dual immunofluorescence staining, and flow cytometry. RESULTS: The expression of ADORA3 was elevated in brain tissues of VaD patients and ADORA3 was indicated as a key gene for VaD in the GSE122063. In BCAS mice, the expression of ADORA3 was predominantly elevated in microglia in the corpus callosum. ADORA3 antagonist promotes microglial phagocytosis to myelin debris by facilitating cAMP/PKA/p-CREB pathway and thereby ameliorates WMI and cognitive impairment in BCAS mice. The therapeutic effect of ADORA3 antagonist was partially reversed by the inhibition of the cAMP/PKA pathway. CONCLUSIONS: ADORA3 antagonist alleviates chronic ischemic WMI by modulating myelin clearance of microglia, which may be a potential therapeutic target for the treatment of VaD.


Subject(s)
Dementia, Vascular , Mice, Inbred C57BL , Microglia , Phagocytosis , Receptor, Adenosine A3 , Animals , Humans , Male , Mice , Brain Ischemia/metabolism , Brain Ischemia/pathology , Carotid Stenosis , Dementia, Vascular/pathology , Dementia, Vascular/metabolism , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Organic Chemicals , Phagocytosis/drug effects , Phagocytosis/physiology , Receptor, Adenosine A3/metabolism , Receptor, Adenosine A3/genetics , White Matter/pathology , White Matter/metabolism , White Matter/drug effects
8.
Brain Behav Immun ; 119: 454-464, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38642614

ABSTRACT

BACKGROUND: Both functional brain imaging studies and autopsy reports have indicated the presence of synaptic loss in the brains of depressed patients. The activated microglia may dysfunctionally engulf neuronal synapses, leading to synaptic loss and behavioral impairments in depression. However, the mechanisms of microglial-synaptic interaction under depressive conditions remain unclear. METHODS: We utilized lipopolysaccharide (LPS) to induce a mouse model of depression, examining the effects of LPS on behaviors, synapses, microglia, microglial phagocytosis of synapses, and the C1q/C3-CR3 complement signaling pathway. Additionally, a C1q neutralizing antibody was employed to inhibit the C1q/C3-CR3 signaling pathway and assess its impact on microglial phagocytosis of synapses and behaviors in the mice. RESULTS: LPS administration resulted in depressive and anxiety-like behaviors, synaptic loss, and abnormal microglial phagocytosis of synapses in the hippocampal dentate gyrus (DG) of mice. We found that the C1q/C3-CR3 signaling pathway plays a crucial role in this abnormal microglial activity. Treatment with the C1q neutralizing antibody moderated the C1q/C3-CR3 pathway, leading to a decrease in abnormal microglial phagocytosis, reduced synaptic loss, and improved behavioral impairments in the mice. CONCLUSIONS: The study suggests that the C1q/C3-CR3 complement signaling pathway, which mediates abnormal microglial phagocytosis of synapses, presents a novel potential therapeutic target for depression treatment.


Subject(s)
Complement C1q , Complement C3 , Depression , Disease Models, Animal , Microglia , Phagocytosis , Signal Transduction , Synapses , Animals , Complement C1q/metabolism , Microglia/metabolism , Synapses/metabolism , Mice , Signal Transduction/physiology , Depression/metabolism , Phagocytosis/physiology , Complement C3/metabolism , Male , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL
9.
Dev Cell ; 59(10): 1284-1301.e8, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38569551

ABSTRACT

Macrophages constitute the first defense line against the non-self, but their ability to remodel their environment in organ development/homeostasis is starting to be appreciated. Early-wave macrophages (EMs), produced from hematopoietic stem cell (HSC)-independent progenitors, seed the mammalian fetal liver niche wherein HSCs expand and differentiate. The involvement of niche defects in myeloid malignancies led us to identify the cues controlling HSCs. In Drosophila, HSC-independent EMs also colonize the larva when late hematopoiesis occurs. The evolutionarily conserved immune system allowed us to investigate whether/how EMs modulate late hematopoiesis in two models. We show that loss of EMs in Drosophila and mice accelerates late hematopoiesis, which does not correlate with inflammation and does not rely on macrophage phagocytic ability. Rather, EM-derived extracellular matrix components underlie late hematopoiesis acceleration. This demonstrates a developmental role for EMs.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Macrophages , Animals , Hematopoiesis/physiology , Macrophages/metabolism , Mice , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Phagocytosis/physiology , Drosophila melanogaster , Extracellular Matrix/metabolism , Drosophila , Cell Differentiation
10.
Brain Behav Immun ; 119: 416-430, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636563

ABSTRACT

The role of microglia in triggering the blood-brain barrier (BBB) impairment and white matter damage after chronic cerebral hypoperfusion is unclear. Here we demonstrated that the vessel-adjacent microglia were specifically activated by the leakage of plasma low-density lipoprotein (LDL), which led to BBB breakdown and ischemic demyelination. Interestingly, we found that LDL stimulation enhanced microglial phagocytosis, causing excessive engulfment of myelin debris and resulting in an overwhelming lipid burden in microglia. Surprisingly, these lipid-laden microglia exhibited a suppressed profile of inflammatory response and compromised pro-regenerative properties. Microglia-specific knockdown of LDLR or systematic medication lowering circulating LDL-C showed protective effects against ischemic demyelination. Overall, our findings demonstrated that LDL-stimulated vessel-adjacent microglia possess a disease-specific molecular signature, characterized by suppressed regenerative properties, which is associated with the propagation of demyelination during ischemic white matter damage.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Lipoproteins, LDL , Microglia , White Matter , Microglia/metabolism , Animals , White Matter/metabolism , White Matter/pathology , Mice , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Brain Ischemia/metabolism , Blood-Brain Barrier/metabolism , Male , Mice, Inbred C57BL , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Phagocytosis/physiology , Myelin Sheath/metabolism
11.
Curr Opin Neurobiol ; 86: 102877, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631077

ABSTRACT

Microglia are tissue-resident macrophages and professional phagocytes of the central nervous system (CNS). In development, microglia-mediated phagocytosis is important for sculpting the cellular architecture. This includes the engulfment of dead/dying cells, pruning extranumerary synapses and axons, and phagocytosing fragments of myelin sheaths. Intriguingly, these developmental phagocytic mechanisms by which microglia sculpt the CNS are now appreciated as important for eliminating synapses, myelin, and proteins during neurodegeneration. Here, we discuss parallels between neurodevelopment and neurodegeneration, which highlights how development is informing disease. We further discuss recent advances and challenges towards therapeutically targeting these phagocytic pathways and how we can leverage development to overcome these challenges.


Subject(s)
Microglia , Phagocytosis , Humans , Microglia/physiology , Microglia/pathology , Animals , Phagocytosis/physiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/physiopathology , Myelin Sheath/physiology , Central Nervous System/pathology
12.
Neuroscience ; 547: 1-16, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38570063

ABSTRACT

After spinal cord injury (SCI), the accumulation of myelin debris can serve as proinflammatory agents, hindering axon regrowth and exacerbating damage. While astrocytes have been implicated in the phagocytosis of myelin debris, the impact of this process on the phenotypic transformation of astrocytes and their characteristics following SCI in rats is not well understood. Here, we demonstrated that the conditioned medium of myelin debris can trigger apoptosis in rat primary astrocytes in vitro. Using a compressional SCI model in rats, we observed that astrocytes can engulf myelin debris through ATP-binding cassette transporter sub-family A member 1 (ABCA1), and these engulfed cells tend to transform into A1 astrocytes, as indicated by C3 expression. At 4 days post-injury (dpi), astrocytes rapidly transitioned into A1 astrocytes and maintained this phenotype from 4 to 28 dpi, while A2 astrocytes, characterized by S100, were only detected at 14 and 28 dpi. Reactive astrocytes, identified by Nestin, emerged at 4 and 7 dpi, whereas scar-forming astrocytes, marked by N-cadherin, were evident at 14 and 28 dpi. This study illustrates the distribution patterns of astrocyte subtypes and the potential interplay between astrocytes and myelin debris after SCI in rats. We emphasize that myelin debris can induce astrocyte apoptosis in vitro and promote the transformation of astrocytes into A1 astrocytes in vivo. These two classification methods are not mutually exclusive, but rather complementary.


Subject(s)
Astrocytes , Myelin Sheath , Spinal Cord Injuries , Animals , Female , Rats , Apoptosis/physiology , Astrocytes/metabolism , Astrocytes/pathology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Myelin Sheath/pathology , Myelin Sheath/metabolism , Phagocytosis/physiology , Phenotype , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/metabolism
13.
Neuron ; 112(8): 1205-1207, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38608706

ABSTRACT

Removal of toxic debris that can hinder brain function is performed primarily by microglia, the brain's professional phagocytes. A recent study in Cell1 identified that viral response interferons are required for priming microglia, ensuring competent phagocytosis and proper circuit wiring.


Subject(s)
Interferons , Microglia , Microglia/physiology , Phagocytosis/physiology , Brain
14.
Glia ; 72(7): 1319-1339, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38577970

ABSTRACT

Neuroinflammation and chronic activation of microglial cells are the prominent features of amyotrophic lateral sclerosis (ALS) pathology. While alterations in the mRNA profile of diseased microglia have been well documented, the actual microglia proteome remains poorly characterized. Here we performed a functional characterization together with proteome analyses of microglial cells at different stages of disease in the SOD1-G93A model of ALS. Functional analyses of microglia derived from the lumbar spinal cord of symptomatic mice revealed: (i) remarkably high mitotic index (close to 100% cells are Ki67+) (ii) significant decrease in phagocytic capacity when compared to age-matched control microglia, and (iii) diminished response to innate immune challenges in vitro and in vivo. Proteome analysis revealed a development of two distinct molecular signatures at early and advanced stages of disease. While at early stages of disease, we identified several proteins implicated in microglia immune functions such as GPNMB, HMBOX1, at advanced stages of disease microglia signature at protein level was characterized with a robust upregulation of several unconventional proteins including rootletin, major vaults proteins and STK38. Upregulation of GPNMB and rootletin has been also found in the spinal cord samples of sporadic ALS. Remarkably, the top biological functions of microglia, in particular in the advanced disease, were not related to immunity/immune response, but were highly enriched in terms linked to RNA metabolism. Together, our results suggest that, over the course of disease, chronically activated microglia develop unconventional protein signatures and gradually lose their immune identity ultimately turning into functionally inefficient immune cells.


Subject(s)
Amyotrophic Lateral Sclerosis , Mice, Transgenic , Microglia , Proteome , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/genetics , Microglia/metabolism , Microglia/immunology , Animals , Proteome/metabolism , Mice , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/immunology , Disease Models, Animal , Phagocytosis/physiology , Humans , Female , Mice, Inbred C57BL , Male
15.
J Neurosci ; 44(20)2024 May 15.
Article in English | MEDLINE | ID: mdl-38589228

ABSTRACT

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


Subject(s)
Disease Models, Animal , Drosophila Proteins , Drosophila , Huntingtin Protein , Huntington Disease , Neuroglia , Animals , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/genetics , Neuroglia/metabolism , Neuroglia/pathology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Female , Male , Phagocytosis/physiology , Lysosomes/metabolism , Phagosomes/metabolism , Animals, Genetically Modified , Prions/metabolism , Prions/genetics , Neurons/metabolism
16.
Methods Mol Biol ; 2761: 231-243, 2024.
Article in English | MEDLINE | ID: mdl-38427240

ABSTRACT

Microglia are scavengers of the brain environment that clear dead cells, debris, and microbes. In Alzheimer's disease, microglia get activated to phagocytose damaged neurons, extracellular Amyoid-ß, and Tau deposits. Several Tau internalization mechanisms of microglia have been studied which include phagocytosis, pinocytosis, and receptor-mediated endocytosis. In this chapter, we have visualized microglial phagocytic structures that are actin-rich cup-like extensions, which surrounds extracellular Tau species by wide-field fluorescence and confocal microscopy. We have shown the association of filamentous actin in Tau phagocytosis along the assembly of LC-3 molecules to phagosomes. The 3-dimensional, orthogonal and gallery wise representation of these phagocytic structures provides an overview of the phagocytic mechanism of extracellular Tau by microglia.


Subject(s)
Alzheimer Disease , Microglia , Humans , Microglia/metabolism , Actins , Phagocytosis/physiology , Biological Transport , Amyloid beta-Peptides/metabolism
17.
Neurochem Res ; 49(7): 1782-1793, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555337

ABSTRACT

Convulsive status epilepticus (CSE) is a common critical neurological condition that can lead to irreversible hippocampal neuron damage and cognitive dysfunction. Multiple studies have demonstrated the critical roles that long non-coding RNA Mir155hg plays in a variety of diseases. However, less is known about the function and mechanism of Mir155hg in CSE. Here we investigate and elucidate the mechanism underlying the contribution of Mir155hg to CSE-induced hippocampal neuron injury. By applying high-throughput sequencing, we examined the expression of differentially expressed genes in normal and CSE rats. Subsequent RT-qPCR enabled us to measure the level of Mir155hg in rat hippocampal tissue. Targeted knockdown of Mir155hg was achieved by the AAV9 virus. Additionally, we utilized HE and Tunel staining to evaluate neuronal injury. Immunofluorescence (IF), Golgi staining, and brain path clamping were also used to detect the synaptic plasticity of hippocampal neurons. Finally, through IF staining and Sholl analysis, we assessed the degree of microglial phagocytic function. It was found that the expression of Mir155hg was elevated in CSE rats. HE and Tunel staining results showed that Mir155hg knockdown suppressed the hippocampal neuron loss and apoptosis followed CSE. IF, Golgi staining and brain path clamp data found that Mir155hg knockdown enhanced neuronal synaptic plasticity. The results from IF staining and Sholl analysis showed that Mir155hg knockdown enhanced microglial phagocytosis. Our findings suggest that Mir155hg promotes CSE-induced hippocampal neuron injury by inhibiting microglial phagocytosis.


Subject(s)
Hippocampus , MicroRNAs , Microglia , Neurons , Phagocytosis , Rats, Sprague-Dawley , Status Epilepticus , Animals , Status Epilepticus/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Hippocampus/metabolism , Hippocampus/pathology , Microglia/metabolism , Neurons/metabolism , Male , Phagocytosis/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Apoptosis/physiology , Neuronal Plasticity/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
18.
Cell Biochem Funct ; 42(2): e3972, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38500392

ABSTRACT

Cell death and the efficient removal of dead cells are two basic mechanisms that maintain homeostasis in multicellular organisms. efferocytosis, which includes four steps recruitment, recognition, binding and signaling, and engulfment. Effectively and quickly removes apoptotic cells from the body. Any alteration in efferocytosis can lead to several diseases, including autoimmune and inflammatory conditions, atherosclerosis, and cancer. A wide range of dietary components affects apoptosis and, subsequently, efferocytosis. Some vitamins, including fat-soluble vitamins, affect different stages of efferocytosis. Among other things, by affecting macrophages, they are effective in the apoptotic cleansing of cells. Also, polyphenols indirectly intervene in efferocytosis through their effect on apoptosis. Considering that there are limited articles on the effect of nutrition on efferocytosis, in this article we will examine the effect of some dietary components on efferocytosis.


Subject(s)
Efferocytosis , Phagocytosis , Phagocytosis/physiology , Macrophages/metabolism , Apoptosis , Vitamins/pharmacology , Vitamins/metabolism
19.
Chem Commun (Camb) ; 60(21): 2930-2933, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38372418

ABSTRACT

Pseudaminic acid (Pse) on pathogenic bacteria exopolysaccharide engages with the sialic acid-binding immunoglobulin-type lectin (Siglec)-10 receptor on macrophages via the critical 7-N-acetyl group. This binding stimulates macrophages to secrete interleukin 10 that suppresses phagocytosis against bacteria, but can be reverted by blocking Pse-Siglec-10 interaction with Pse-binding protein as a promising therapy.


Subject(s)
Interleukin-10 , Macrophages , Sugar Acids , Interleukin-10/metabolism , Macrophages/metabolism , Phagocytosis/physiology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
20.
Dev Cell ; 59(7): 853-868.e7, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38359833

ABSTRACT

Phagocytes remove dead and dying cells by engaging "eat-me" ligands such as phosphatidylserine (PtdSer) on the surface of apoptotic targets. However, PtdSer is obscured by the bulky exofacial glycocalyx, which also exposes ligands that activate "don't-eat-me" receptors such as Siglecs. Clearly, unshielding the juxtamembrane "eat-me" ligands is required for the successful engulfment of apoptotic cells, but the mechanisms underlying this process have not been described. Using human and murine cells, we find that apoptosis-induced retraction and weakening of the cytoskeleton that anchors transmembrane proteins cause an inhomogeneous redistribution of the glycocalyx: actin-depleted blebs emerge, lacking the glycocalyx, while the rest of the apoptotic cell body retains sufficient actin to tether the glycocalyx in place. Thus, apoptotic blebs can be engaged by phagocytes and are targeted for engulfment. Therefore, in cells with an elaborate glycocalyx, such as mucinous cancer cells, this "don't-come-close-to-me" barrier must be removed to enable clearance by phagocytosis.


Subject(s)
Actins , Glycocalyx , Animals , Humans , Mice , Glycocalyx/metabolism , Actins/metabolism , Phagocytes , Phagocytosis/physiology , Ligands , Apoptosis/physiology , Phosphatidylserines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...