Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell ; 186(4): 764-785.e21, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36803604

ABSTRACT

The choroid plexus (ChP) is the blood-cerebrospinal fluid (CSF) barrier and the primary source of CSF. Acquired hydrocephalus, caused by brain infection or hemorrhage, lacks drug treatments due to obscure pathobiology. Our integrated, multi-omic investigation of post-infectious hydrocephalus (PIH) and post-hemorrhagic hydrocephalus (PHH) models revealed that lipopolysaccharide and blood breakdown products trigger highly similar TLR4-dependent immune responses at the ChP-CSF interface. The resulting CSF "cytokine storm", elicited from peripherally derived and border-associated ChP macrophages, causes increased CSF production from ChP epithelial cells via phospho-activation of the TNF-receptor-associated kinase SPAK, which serves as a regulatory scaffold of a multi-ion transporter protein complex. Genetic or pharmacological immunomodulation prevents PIH and PHH by antagonizing SPAK-dependent CSF hypersecretion. These results reveal the ChP as a dynamic, cellularly heterogeneous tissue with highly regulated immune-secretory capacity, expand our understanding of ChP immune-epithelial cell cross talk, and reframe PIH and PHH as related neuroimmune disorders vulnerable to small molecule pharmacotherapy.


Subject(s)
Choroid Plexus , Hydrocephalus , Humans , Blood-Brain Barrier/metabolism , Brain/metabolism , Choroid Plexus/metabolism , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/immunology , Immunity, Innate , Cytokine Release Syndrome/pathology
2.
Cell ; 185(22): 4043-4045, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36306731

ABSTRACT

During neurodegenerative disease, resident CNS macrophages termed "microglia" assume a neuroprotective role and engulf toxic protein aggregates and cell debris. In this issue of Cell, two groups independently show how spleen tyrosine kinase (SYK) acts downstream of microglial surface receptors to propagate this neuroprotective program in vivo.


Subject(s)
Microglia , Neurodegenerative Diseases , Humans , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Macrophages , Syk Kinase/metabolism
3.
Immunity ; 56(5): 914-925, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37163992

ABSTRACT

Cytokines are key messengers by which immune cells communicate, and they drive many physiological processes, including immune and inflammatory responses. Early discoveries demonstrated that cytokines, such as the interleukin family members and TNF-α, regulate synaptic scaling and plasticity. Still, we continue to learn more about how these traditional immune system cytokines affect neuronal structure and function. Different cytokines shape synaptic function on multiple levels ranging from fine-tuning neurotransmission, to regulating synapse number, to impacting global neuronal networks and complex behavior. These recent findings have cultivated an exciting and growing field centered on the importance of immune system cytokines for regulating synapse and neural network structure and function. Here, we highlight the latest findings related to cytokines in the central nervous system and their regulation of synapse structure and function. Moreover, we explore how these mechanisms are becoming increasingly important to consider in diseases-especially those with a large neuroinflammatory component.


Subject(s)
Central Nervous System , Cytokines , Central Nervous System/physiology , Synapses , Neurons/physiology , Synaptic Transmission , Neuronal Plasticity/physiology
4.
Immunity ; 55(12): 2318-2335.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36379210

ABSTRACT

Microglia utilize their phagocytic activity to prune redundant synapses and refine neural circuits during precise developmental periods. However, the neuronal signals that control this phagocytic clockwork remain largely undefined. Here, we show that neuronal signal-regulatory protein alpha (SIRPα) is a permissive cue for microglial phagocytosis in the developing murine retina. Removal of neuronal, but not microglial, SIRPα reduced microglial phagocytosis, increased synpase numbers, and impaired circuit function. Conversely, prolonging neuronal SIRPα expression extended developmental microglial phagocytosis. These outcomes depended on the interaction of presynaptic SIRPα with postsynaptic CD47. Global CD47 deficiency modestly increased microglial phagocytosis, while CD47 overexpression reduced it. This effect was rescued by coexpression of neuronal SIRPα or codeletion of neuronal SIRPα and CD47. These data indicate that neuronal SIRPα regulates microglial phagocytosis by limiting microglial SIRPα access to neuronal CD47. This discovery may aid our understanding of synapse loss in neurological diseases.


Subject(s)
CD47 Antigen , Receptors, Immunologic , Mice , Animals , CD47 Antigen/metabolism , Receptors, Immunologic/metabolism , Macrophages/metabolism , Phagocytosis/physiology , Retina , Antigens, Differentiation/metabolism
5.
Immunity ; 54(11): 2444-2446, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758335

ABSTRACT

Maternal infection during pregnancy increases the offspring's risk of developing neurodevelopmental disorders. While IL-6 is involved, the mechanism by which IL-6 and other cytokines affect developing neural circuits is unknown. In this issue of Immunity, Mirabella et al. (2021) show that the pro-inflammatory cytokine IL-6 specifically increases synaptogenesis in immature excitatory neurons through downstream neuronal STAT3-dependent transcriptional regulation of Rgs4.


Subject(s)
Interleukin-6 , Neurogenesis , Cytokines/metabolism , Female , Humans , Neurons/metabolism , Pregnancy , STAT3 Transcription Factor/metabolism
6.
Immunity ; 52(1): 167-182.e7, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31883839

ABSTRACT

Multiple sclerosis (MS) is a demyelinating, autoimmune disease of the central nervous system. While work has focused on myelin and axon loss in MS, less is known about mechanisms underlying synaptic changes. Using postmortem human MS tissue, a preclinical nonhuman primate model of MS, and two rodent models of demyelinating disease, we investigated synapse changes in the visual system. Similar to other neurodegenerative diseases, microglial synaptic engulfment and profound synapse loss were observed. In mice, synapse loss occurred independently of local demyelination and neuronal degeneration but coincided with gliosis and increased complement component C3, but not C1q, at synapses. Viral overexpression of the complement inhibitor Crry at C3-bound synapses decreased microglial engulfment of synapses and protected visual function. These results indicate that microglia eliminate synapses through the alternative complement cascade in demyelinating disease and identify a strategy to prevent synapse loss that may be broadly applicable to other neurodegenerative diseases. VIDEO ABSTRACT.


Subject(s)
Complement C3/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Microglia/pathology , Multiple Sclerosis/pathology , Synapses/pathology , Thalamus/pathology , Aged , Aged, 80 and over , Animals , Callithrix , Cell Line, Tumor , Complement C3/antagonists & inhibitors , Disease Models, Animal , Female , Gliosis/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Receptors, Complement 3b/metabolism
7.
Nature ; 597(7878): 709-714, 2021 09.
Article in English | MEDLINE | ID: mdl-34497421

ABSTRACT

Multiple sclerosis (MS) lesions that do not resolve in the months after they form harbour ongoing demyelination and axon degeneration, and are identifiable in vivo by their paramagnetic rims on MRI scans1-3. Here, to define mechanisms underlying this disabling, progressive neurodegenerative state4-6 and foster development of new therapeutic agents, we used MRI-informed single-nucleus RNA sequencing to profile the edge of demyelinated white matter lesions at various stages of inflammation. We uncovered notable glial and immune cell diversity, especially at the chronically inflamed lesion edge. We define 'microglia inflamed in MS' (MIMS) and 'astrocytes inflamed in MS', glial phenotypes that demonstrate neurodegenerative programming. The MIMS transcriptional profile overlaps with that of microglia in other neurodegenerative diseases, suggesting that primary and secondary neurodegeneration share common mechanisms and could benefit from similar therapeutic approaches. We identify complement component 1q (C1q) as a critical mediator of MIMS activation, validated immunohistochemically in MS tissue, genetically by microglia-specific C1q ablation in mice with experimental autoimmune encephalomyelitis, and therapeutically by treating chronic experimental autoimmune encephalomyelitis with C1q blockade. C1q inhibition is a potential therapeutic avenue to address chronic white matter inflammation, which could be monitored by longitudinal assessment of its dynamic biomarker, paramagnetic rim lesions, using advanced MRI methods.


Subject(s)
Astrocytes/pathology , Lymphocytes/pathology , Microglia/pathology , Multiple Sclerosis/pathology , Animals , Brain/pathology , Complement C1q/antagonists & inhibitors , Complement C1q/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Inflammation/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/diagnostic imaging , RNA-Seq , Transcriptome , White Matter/pathology
8.
Nat Rev Neurosci ; 22(11): 657-673, 2021 11.
Article in English | MEDLINE | ID: mdl-34545240

ABSTRACT

Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder/physiopathology , Central Nervous System/physiology , Immunity, Cellular/physiology , Neuronal Plasticity/physiology , Schizophrenia/physiopathology , Age Factors , Animals , Autism Spectrum Disorder/immunology , Central Nervous System/cytology , Humans , Neurodevelopmental Disorders/immunology , Neurodevelopmental Disorders/physiopathology , Schizophrenia/immunology
9.
Nature ; 573(7772): 75-82, 2019 09.
Article in English | MEDLINE | ID: mdl-31316211

ABSTRACT

Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.


Subject(s)
Cell Lineage , Multiple Sclerosis/pathology , Neurons/pathology , Adult , Animals , Astrocytes/metabolism , Astrocytes/pathology , Autopsy , Cryopreservation , Female , Homeodomain Proteins/metabolism , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Microglia/metabolism , Microglia/pathology , Middle Aged , Multiple Sclerosis/genetics , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phagocytosis , RNA, Small Nuclear/analysis , RNA, Small Nuclear/genetics , RNA-Seq , Transcriptome/genetics
10.
Trends Immunol ; 42(3): 228-247, 2021 03.
Article in English | MEDLINE | ID: mdl-33593693

ABSTRACT

Glial subtype diversity is an emerging topic in neurobiology and immune-mediated neurological diseases such as multiple sclerosis (MS). We discuss recent conceptual and technological advances that allow a better understanding of the transcriptomic and functional heterogeneity of oligodendrocytes (OLs), astrocytes, and microglial cells under inflammatory-demyelinating conditions. Recent single cell transcriptomic studies suggest the occurrence of novel homeostatic and reactive glial subtypes and provide insight into the molecular events during disease progression. Multiplexed RNA in situ hybridization has enabled 'mapping back' dysregulated gene expression to glial subtypes within the MS lesion microenvironment. These findings suggest novel homeostatic and reactive glial-cell-type functions both in immune-related processes and neuroprotection relevant to understanding the pathology of MS.


Subject(s)
Multiple Sclerosis , Astrocytes , Humans , Microglia , Neuroglia , Oligodendroglia
11.
Immunity ; 42(4): 600-2, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902477

ABSTRACT

Mutations in methyl-CpG-binding protein 2 (MECP2) underlie most cases of Rett Syndrome, a neurodevelopmental disorder with neurological and somatic impairments. In this issue of Immunity, Cronk et al. (2015) find that macrophages in MeCP2-deficient mice are abnormal in number, as well as in glucocorticoid, hypoxia, and inflammatory responses.


Subject(s)
CpG Islands/immunology , Epigenesis, Genetic , Macrophages, Peritoneal/immunology , Methyl-CpG-Binding Protein 2/immunology , Microglia/immunology , Rett Syndrome/immunology , Animals , Female , Humans , Male
12.
J Neurosci ; 42(32): 6171-6185, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35790400

ABSTRACT

Interferon regulatory factor 8 (IRF8) is a transcription factor necessary for the maturation of microglia, as well as other peripheral immune cells. It also regulates the transition of microglia and other immune cells to a pro-inflammatory phenotype. Irf8 is also a known risk gene for multiple sclerosis and lupus, and it has recently been shown to be downregulated in schizophrenia. While most studies have focused on IRF8-dependent regulation of immune cell function, little is known about how it impacts neural circuits. Here, we show by RNAseq from Irf8 -/- male and female mouse brains that several genes involved in regulation of neural activity are dysregulated. We then show that these molecular changes are reflected in heightened neural excitability and a profound increase in susceptibility to lethal seizures in male and female Irf8 -/- mice. Finally, we identify that TNF-α is elevated specifically in microglia in the CNS, and genetic or acute pharmacological blockade of TNF-α in the Irf8 -/- CNS rescued the seizure phenotype. These results provide important insights into the consequences of IRF8 signaling and TNF-α on neural circuits. Our data further suggest that neuronal function is impacted by loss of IRF8, a factor involved in neuropsychiatric and neurodegenerative diseases.SIGNIFICANCE STATEMENT Here, we identify a previously unknown and key role for interferon regulator factor 8 (IRF8) in regulating neural excitability and seizures. We further determine that these effects on neural circuits are through elevated TNF-α in the CNS. As IRF8 has most widely been studied in the context of regulating the development and inflammatory signaling in microglia and other immune cells, we have uncovered a novel function. Further, IRF8 is a risk gene for multiple sclerosis and lupus, IRF8 is dysregulated in schizophrenia, and elevated TNF-α has been identified in a multitude of neurologic conditions. Thus, elucidating these IRF8 and TNF-α-dependent effects on brain circuit function has profound implications for understanding underlying, therapeutically relevant mechanisms of disease.


Subject(s)
Interferon Regulatory Factors/metabolism , Seizures/metabolism , Tumor Necrosis Factor-alpha , Animals , Female , Interferon Regulatory Factors/genetics , Male , Mice , Multiple Sclerosis/pathology , Seizures/pathology , Tumor Necrosis Factor-alpha/metabolism
13.
Glia ; 70(10): 1850-1863, 2022 10.
Article in English | MEDLINE | ID: mdl-35635122

ABSTRACT

Microglia are myeloid cells of the central nervous system that perform tasks essential for brain development, neural circuit homeostasis, and neural disease. Microglia react to inflammatory stimuli by upregulating inflammatory signaling through several different immune cell receptors such as the Toll-like receptor 4 (TLR4), which signals to several downstream effectors including transforming growth factor beta-activated kinase 1 (TAK1). Here, we show that TAK1 levels are regulated by CPEB1, a sequence-specific RNA binding protein that controls translation as well as RNA splicing and alternative poly(A) site selection in microglia. Lipopolysaccharide (LPS) binds the TLR4 receptor, which in CPEB1-deficient mice leads to elevated expression of ionized calcium binding adaptor molecule 1 (Iba1), a microglial protein that increases with inflammation, and increased levels of the cytokine IL6. This LPS-induced IL6 response is blocked by inhibitors of JNK, p38, ERK, NFκB, and TAK1. In contrast, phagocytosis, which is elevated in CPEB1-deficient microglia, is unaffected by LPS treatment or ERK inhibition, but is blocked by TAK1 inhibition. These data indicate that CPEB1 regulates microglial inflammatory responses and phagocytosis. RNA-seq indicates that these changes in inflammation and phagocytosis are accompanied by changes in RNA levels, splicing, and alternative poly(A) site selection. Thus, CPEB1 regulation of RNA expression plays a role in microglial function.


Subject(s)
Microglia , Phagocytosis , Polyadenylation , Transcription Factors , mRNA Cleavage and Polyadenylation Factors , Animals , Inflammation/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Mice , Microglia/metabolism , RNA/metabolism , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
14.
J Neurosci ; 40(34): 6503-6521, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32661024

ABSTRACT

Microglia, a resident CNS macrophage, are dynamic cells, constantly extending and retracting their processes as they contact and functionally regulate neurons and other glial cells. There is far less known about microglia-vascular interactions, particularly under healthy steady-state conditions. Here, we use the male and female mouse cerebral cortex to show that a higher percentage of microglia associate with the vasculature during the first week of postnatal development compared with older ages and that the timing of these associations is dependent on the fractalkine receptor (CX3CR1). Similar developmental microglia-vascular associations were detected in the human brain. Using live imaging in mice, we found that juxtavascular microglia migrated when microglia are actively colonizing the cortex and became stationary by adulthood to occupy the same vascular space for nearly 2 months. Further, juxtavascular microglia at all ages associate with vascular areas void of astrocyte endfeet, and the developmental shift in microglial migratory behavior along vessels corresponded to when astrocyte endfeet more fully ensheath vessels. Together, our data provide a comprehensive assessment of microglia-vascular interactions. They support a mechanism by which microglia use the vasculature to migrate within the developing brain parenchyma. This migration becomes restricted on the arrival of astrocyte endfeet such that juxtavascular microglia become highly stationary and stable in the mature cortex.SIGNIFICANCE STATEMENT We report the first extensive analysis of juxtavascular microglia in the healthy, developing, and adult brain. Live imaging revealed that juxtavascular microglia within the cortex are highly motile and migrate along vessels as they are colonizing cortical regions. Using confocal, expansion, super-resolution, and electron microscopy, we determined that microglia associate with the vasculature at all ages in areas lacking full astrocyte endfoot coverage and motility of juxtavascular microglia ceases as astrocyte endfeet more fully ensheath the vasculature. Our data lay the fundamental groundwork to investigate microglia-astrocyte cross talk and juxtavascular microglial function in the healthy and diseased brain. They further provide a potential mechanism by which vascular interactions facilitate microglial colonization of the brain to later regulate neural circuit development.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/growth & development , Microglia/physiology , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Capillaries/growth & development , Capillaries/ultrastructure , Cerebral Cortex/ultrastructure , Female , Humans , Male , Mice, Inbred C57BL , Microglia/ultrastructure , Somatosensory Cortex/metabolism
15.
J Neurosci ; 39(11): 2025-2040, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30647151

ABSTRACT

Microglia play important roles in shaping the developing CNS, and at early stages they have been proposed to regulate progenitor proliferation, differentiation, and neuronal survival. However, these studies reveal contradictory outcomes, highlighting the complexity of these cell-cell interactions. Here, we investigate microglia function during embryonic mouse retina development, where only microglia, progenitors, and neurons are present. In both sexes, we determine that microglia primarily interact with retinal neurons and find that depletion of microglia via conditional KO of the Csf1 receptor results in increased density of retinal ganglion cells (RGCs). Pharmacological inhibition of microglia also results in an increase in RGCs, with no effect on retinal progenitor proliferation, RGC genesis, or apoptosis. We show that microglia in the embryonic retina are enriched for phagocytic markers and observe engulfment of nonapoptotic Brn3-labeled RGCs. We investigate the molecular pathways that can mediate cell engulfment by microglia and find selective downregulation of complement pathway components with microglia inhibition, and further show that C1q protein marks a subset of RGCs in the embryonic retina. KO of complement receptor 3 (CR3; Itgam), which is only expressed by microglia, results in increased RGC density, similar to what we observed after depletion or inhibition of microglia. Thus, our data suggest that microglia regulate neuron elimination in the embryonic mouse retina by complement-mediated phagocytosis of non-apoptotic newborn RGCs.SIGNIFICANCE STATEMENT Microglia are emerging as active and important participants in regulating neuron number in development, during adult neurogenesis, and following stem cell therapies. However, their role in these contexts and the mechanisms involved are not fully defined. Using a well-characterized in vivo system, we provide evidence that microglia regulate neuronal elimination by complement-mediated engulfment of nonapoptotic neurons. This work provides a significant advancement of the field by defining in vivo molecular mechanisms for microglia-mediated cell elimination. Our data add to a growing body of evidence that microglia are essential for proper nervous system development. In addition, we elucidate microglia function in the developing retina, which may shed light on microglia involvement in the context of retinal injury and disease.


Subject(s)
Complement System Proteins/physiology , Microglia/physiology , Phagocytosis/physiology , Retina/growth & development , Retinal Ganglion Cells/physiology , Animals , Cell Count , Female , Macrophage Colony-Stimulating Factor/genetics , Male , Mice, Knockout
16.
J Neuroinflammation ; 17(1): 296, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33036616

ABSTRACT

BACKGROUND: Chronic alcohol consumption is associated with neuroinflammation, neuronal damage, and behavioral alterations including addiction. Alcohol-induced neuroinflammation is characterized by increased expression of proinflammatory cytokines (including TNFα, IL-1ß, and CCL2) and microglial activation. We hypothesized chronic alcohol consumption results in peripheral immune cell infiltration to the CNS. Since chemotaxis through the CCL2-CCR2 signaling axis is critical for macrophage recruitment peripherally and centrally, we further hypothesized that blockade of CCL2 signaling using the dual CCR2/5 inhibitor cenicriviroc (CVC) would prevent alcohol-induced CNS infiltration of peripheral macrophages and alter the neuroinflammatory state in the brain after chronic alcohol consumption. METHODS: C57BL/6J female mice were fed an isocaloric or 5% (v/v) ethanol Lieber DeCarli diet for 6 weeks. Some mice received daily injections of CVC. Microglia and infiltrating macrophages were characterized and quantified by flow cytometry and visualized using CX3CR1eGFP/+ CCR2RFP/+ reporter mice. The effect of ethanol and CVC treatment on the expression of inflammatory genes was evaluated in various regions of the brain, using a Nanostring nCounter inflammation panel. Microglia activation was analyzed by immunofluorescence. CVC-treated and untreated mice were presented with the two-bottle choice test. RESULTS: Chronic alcohol consumption induced microglia activation and peripheral macrophage infiltration in the CNS, particularly in the hippocampus. Treatment with CVC abrogated ethanol-induced recruitment of peripheral macrophages and partially reversed microglia activation. Furthermore, the expression of proinflammatory markers was upregulated by chronic alcohol consumption in various regions of the brain, including the cortex, hippocampus, and cerebellum. Inhibition of CCR2/5 decreased alcohol-mediated expression of inflammatory markers. Finally, microglia function was impaired by chronic alcohol consumption and restored by CVC treatment. CVC treatment did not change the ethanol consumption or preference of mice in the two-bottle choice test. CONCLUSIONS: Together, our data establish that chronic alcohol consumption promotes the recruitment of peripheral macrophages into the CNS and microglia alterations through the CCR2/5 axis. Therefore, further exploration of the CCR2/5 axis as a modulator of neuroinflammation may offer a potential therapeutic approach for the treatment of alcohol-associated neuroinflammation.


Subject(s)
Brain/metabolism , Ethanol/toxicity , Macrophages/metabolism , Microglia/metabolism , Receptors, CCR2/metabolism , Receptors, CCR5/metabolism , Animals , Brain/drug effects , CCR5 Receptor Antagonists/pharmacology , Ethanol/administration & dosage , Female , Imidazoles/pharmacology , Inflammation Mediators/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Receptors, CCR2/antagonists & inhibitors , Sulfoxides/pharmacology
17.
Glia ; 67(7): 1374-1384, 2019 07.
Article in English | MEDLINE | ID: mdl-30861188

ABSTRACT

It is now well-established that the macrophage and microglial response to CNS demyelination influences remyelination by removing myelin debris and secreting a variety of signaling molecules that influence the behaviour of oligodendrocyte progenitor cells (OPCs). Previous studies have shown that changes in microglia contribute to the age-related decline in the efficiency of remyelination. In this study, we show that microglia increase their expression of the proteoglycan NG2 with age, and that this is associated with an altered micro-niche generated by aged, but not young, microglia that can divert the differentiation OPCs from oligodendrocytes into astrocytes in vitro. We further show that these changes in ageing microglia are generated by exposure to high levels of TGFß. Thus, our findings suggest that the rising levels of circulating TGFß known to occur with ageing contribute to the age-related decline in remyelination by impairing the ability of microglia to promote oligodendrocyte differentiation from OPCs, and therefore could be a potential therapeutic target to promote remyelination.


Subject(s)
Cellular Senescence/physiology , Microglia/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Transforming Growth Factor beta/pharmacology , Age Factors , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Cellular Senescence/drug effects , Central Nervous System/drug effects , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Microglia/drug effects , Oligodendrocyte Precursor Cells/drug effects , Oligodendroglia/drug effects , Rats , Rats, Sprague-Dawley
18.
Ann N Y Acad Sci ; 1533(1): 38-50, 2024 03.
Article in English | MEDLINE | ID: mdl-38294960

ABSTRACT

It has been more than a century since Pío del Río-Hortega first characterized microglia in histological stains of brain tissue. Since then, significant advances have been made in understanding the role of these resident central nervous system (CNS) macrophages. In particular, it is now known that microglia can sense neural activity and modulate neuronal circuits accordingly. We review the mechanisms by which microglia detect changes in neural activity to then modulate synapse numbers in the developing and mature CNS. This includes responses to both spontaneous and experience-driven neural activity. We further discuss activity-dependent mechanisms by which microglia regulate synaptic function and neural circuit excitability. Together, our discussion provides a comprehensive review of the activity-dependent functions of microglia within neural circuits in the healthy CNS, and highlights exciting new open questions related to understanding more fully microglia as key components and regulators of neural circuits.


Subject(s)
Central Nervous System , Microglia , Humans , Neurons/physiology , Synapses/physiology , Macrophages
19.
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
20.
bioRxiv ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38370645

ABSTRACT

Astrocytes and microglia are emerging key regulators of activity-dependent synapse remodeling that engulf and remove synapses in response to changes in neural activity. Yet, the degree to which these cells communicate to coordinate this process remains an open question. Here, we use whisker removal in postnatal mice to induce activity-dependent synapse removal in the barrel cortex. We show that astrocytes do not engulf synapses in this paradigm. Instead, astrocytes reduce their contact with synapses prior to microglia-mediated synapse engulfment. We further show that reduced astrocyte-contact with synapses is dependent on microglial CX3CL1-CX3CR1 signaling and release of Wnts from microglia following whisker removal. These results demonstrate an activity-dependent mechanism by which microglia instruct astrocyte-synapse interactions, which then provides a permissive environment for microglia to remove synapses. We further show that this mechanism is critical to remodel synapses in a changing sensory environment and this signaling is upregulated in several disease contexts.

SELECTION OF CITATIONS
SEARCH DETAIL