Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Trends Immunol ; 45(5): 358-370, 2024 May.
Article En | MEDLINE | ID: mdl-38658221

Microglia are brain-resident macrophages that play key roles in brain development and experience dependent plasticity. In this review we discuss recent findings regarding the molecular mechanisms through which mammalian microglia sense the unique molecular patterns of the homeostatic brain. We propose that microglial function is acutely controlled in response to 'brain-associated molecular patterns' (BAMPs) that function as indicators of neuronal activity and neural circuit remodeling. A further layer of regulation comes from instructive cytokine cues that define unique microglial functional states. A systematic investigation of the receptors and signaling pathways that mediate these two regulatory axes may begin to define a functional code for microglia-neuron interactions.


Brain , Microglia , Signal Transduction , Microglia/immunology , Microglia/metabolism , Humans , Animals , Brain/physiology , Cytokines/metabolism , Neurons/metabolism , Neurons/physiology , Neuronal Plasticity , Homeostasis
2.
Cell ; 187(8): 1936-1954.e24, 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38490196

Microglia are brain-resident macrophages that shape neural circuit development and are implicated in neurodevelopmental diseases. Multiple microglial transcriptional states have been defined, but their functional significance is unclear. Here, we identify a type I interferon (IFN-I)-responsive microglial state in the developing somatosensory cortex (postnatal day 5) that is actively engulfing whole neurons. This population expands during cortical remodeling induced by partial whisker deprivation. Global or microglial-specific loss of the IFN-I receptor resulted in microglia with phagolysosomal dysfunction and an accumulation of neurons with nuclear DNA damage. IFN-I gain of function increased neuronal engulfment by microglia in both mouse and zebrafish and restricted the accumulation of DNA-damaged neurons. Finally, IFN-I deficiency resulted in excess cortical excitatory neurons and tactile hypersensitivity. These data define a role for neuron-engulfing microglia during a critical window of brain development and reveal homeostatic functions of a canonical antiviral signaling pathway in the brain.


Brain , Interferon Type I , Microglia , Animals , Mice , Interferon Type I/metabolism , Microglia/metabolism , Neurons/metabolism , Zebrafish , Brain/cytology , Brain/growth & development
3.
J Neurosci ; 43(50): 8621-8636, 2023 12 13.
Article En | MEDLINE | ID: mdl-37845031

Astrocytes play essential roles in the developing nervous system, including supporting synapse function. These astrocyte support functions emerge coincident with brain maturation and may be tailored in a region-specific manner. For example, gray matter astrocytes have elaborate synapse-associated processes and are morphologically and molecularly distinct from white matter astrocytes. This raises the question of whether there are unique environmental cues that promote gray matter astrocyte identity and synaptogenic function. We previously identified adrenergic receptors as preferentially enriched in developing gray versus white matter astrocytes, suggesting that noradrenergic signaling could be a cue that promotes the functional maturation of gray matter astrocytes. We first characterized noradrenergic projections during postnatal brain development in mouse and human, finding that process density was higher in the gray matter and increased concurrently with astrocyte maturation. RNA sequencing revealed that astrocytes in both species expressed α- and ß-adrenergic receptors. We found that stimulation of ß-adrenergic receptors increased primary branching of rodent astrocytes in vitro Conversely, astrocyte-conditional knockout of the ß1-adrenergic receptor reduced the size of gray matter astrocytes and led to dysregulated sensorimotor integration in female mice. These studies suggest that adrenergic signaling to developing astrocytes impacts their morphology and has implications for adult behavior, particularly in female animals. More broadly, they demonstrate a mechanism through which environmental cues impact astrocyte development. Given the key roles of norepinephrine in brain states, such as arousal, stress, and learning, these findings could prompt further inquiry into how developmental stressors impact astrocyte development and adult brain function.SIGNIFICANCE STATEMENT This study demonstrates a role for noradrenergic signaling in the development of gray matter astrocytes. We provide new evidence that the ß1-adrenergic receptor is robustly expressed by both mouse and human astrocytes, and that conditional KO of the ß1-adrenergic receptor from female mouse astrocytes impairs gray matter astrocyte maturation. Moreover, female conditional KO mice exhibit behavioral deficits in two paradigms that test sensorimotor function. Given the emerging interest in moving beyond RNA sequencing to probe specific pathways that underlie astrocyte heterogeneity, this study provides a foundation for future investigation into the effect of noradrenergic signaling on astrocyte functions in conditions where noradrenergic signaling is altered, such as stress, arousal, and learning.


Adrenergic Agents , Astrocytes , Humans , Mice , Animals , Female , Adrenergic Agents/metabolism , Astrocytes/metabolism , Signal Transduction , Norepinephrine/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic
4.
bioRxiv ; 2023 Mar 17.
Article En | MEDLINE | ID: mdl-36993292

The innate immune system plays essential roles in brain synaptic development, and immune dysregulation is implicated in neurodevelopmental diseases. Here we show that a subset of innate lymphocytes (group 2 innate lymphoid cells, ILC2s) is required for cortical inhibitory synapse maturation and adult social behavior. ILC2s expanded in the developing meninges and produced a surge of their canonical cytokine Interleukin-13 (IL-13) between postnatal days 5-15. Loss of ILC2s decreased cortical inhibitory synapse numbers in the postnatal period where as ILC2 transplant was sufficient to increase inhibitory synapse numbers. Deletion of the IL-4/IL-13 receptor (Il4ra) from inhibitory neurons phenocopied the reduction inhibitory synapses. Both ILC2 deficient and neuronal Il4ra deficient animals had similar and selective impairments in adult social behavior. These data define a type 2 immune circuit in early life that shapes adult brain function.

5.
bioRxiv ; 2023 Mar 20.
Article En | MEDLINE | ID: mdl-35233577

Microglia are brain resident phagocytes that can engulf synaptic components and extracellular matrix as well as whole neurons. However, whether there are unique molecular mechanisms that regulate these distinct phagocytic states is unknown. Here we define a molecularly distinct microglial subset whose function is to engulf neurons in the developing brain. We transcriptomically identified a cluster of Type I interferon (IFN-I) responsive microglia that expanded 20-fold in the postnatal day 5 somatosensory cortex after partial whisker deprivation, a stressor that accelerates neural circuit remodeling. In situ, IFN-I responsive microglia were highly phagocytic and actively engulfed whole neurons. Conditional deletion of IFN-I signaling (Ifnar1fl/fl) in microglia but not neurons resulted in dysmorphic microglia with stalled phagocytosis and an accumulation of neurons with double strand DNA breaks, a marker of cell stress. Conversely, exogenous IFN-I was sufficient to drive neuronal engulfment by microglia and restrict the accumulation of damaged neurons. IFN-I deficient mice had excess excitatory neurons in the developing somatosensory cortex as well as tactile hypersensitivity to whisker stimulation. These data define a molecular mechanism through which microglia engulf neurons during a critical window of brain development. More broadly, they reveal key homeostatic roles of a canonical antiviral signaling pathway in brain development.

6.
J Exp Med ; 220(2)2023 02 06.
Article En | MEDLINE | ID: mdl-36520518

Microglia are critical regulators of brain development that engulf synaptic proteins during postnatal synapse remodeling. However, the mechanisms through which microglia sense the brain environment are not well defined. Here, we characterized the regulatory program downstream of interleukin-33 (IL-33), a cytokine that promotes microglial synapse remodeling. Exposing the developing brain to a supraphysiological dose of IL-33 altered the microglial enhancer landscape and increased binding of stimulus-dependent transcription factors including AP-1/FOS. This induced a gene expression program enriched for the expression of pattern recognition receptors, including the scavenger receptor MARCO. CNS-specific deletion of IL-33 led to increased excitatory/inhibitory synaptic balance, spontaneous absence-like epileptiform activity in juvenile mice, and increased seizure susceptibility in response to chemoconvulsants. We found that MARCO promoted synapse engulfment, and Marco-deficient animals had excess thalamic excitatory synapses and increased seizure susceptibility. Taken together, these data define coordinated epigenetic and functional changes in microglia and uncover pattern recognition receptors as potential regulators of postnatal synaptic refinement.


Interleukin-33 , Microglia , Animals , Mice , Microglia/metabolism , Interleukin-33/metabolism , Synapses/metabolism , Brain/metabolism , Seizures/metabolism , Mice, Inbred C57BL
7.
Sci Transl Med ; 14(652): eabj4310, 2022 07 06.
Article En | MEDLINE | ID: mdl-35857628

Inflammatory processes induced by brain injury are important for recovery; however, when uncontrolled, inflammation can be deleterious, likely explaining why most anti-inflammatory treatments have failed to improve neurological outcomes after brain injury in clinical trials. In the thalamus, chronic activation of glial cells, a proxy of inflammation, has been suggested as an indicator of increased seizure risk and cognitive deficits that develop after cortical injury. Furthermore, lesions in the thalamus, more than other brain regions, have been reported in patients with viral infections associated with neurological deficits, such as SARS-CoV-2. However, the extent to which thalamic inflammation is a driver or by-product of neurological deficits remains unknown. Here, we found that thalamic inflammation in mice was sufficient to phenocopy the cellular and circuit hyperexcitability, enhanced seizure risk, and disruptions in cortical rhythms that develop after cortical injury. In our model, down-regulation of the GABA transporter GAT-3 in thalamic astrocytes mediated this neurological dysfunction. In addition, GAT-3 was decreased in regions of thalamic reactive astrocytes in mouse models of cortical injury. Enhancing GAT-3 in thalamic astrocytes prevented seizure risk, restored cortical states, and was protective against severe chemoconvulsant-induced seizures and mortality in a mouse model of traumatic brain injury, emphasizing the potential of therapeutically targeting this pathway. Together, our results identified a potential therapeutic target for reducing negative outcomes after brain injury.


Brain Injuries , COVID-19 , Animals , Astrocytes/metabolism , Disease Models, Animal , GABA Plasma Membrane Transport Proteins/metabolism , Inflammation/pathology , Mice , Polymers , Rodentia/metabolism , SARS-CoV-2 , Seizures , Thalamus/metabolism , Thalamus/pathology
8.
Nat Commun ; 12(1): 5916, 2021 10 08.
Article En | MEDLINE | ID: mdl-34625548

Microglia are brain resident macrophages that play vital roles in central nervous system (CNS) development, homeostasis, and pathology. Microglia both remodel synapses and engulf apoptotic cell corpses during development, but whether unique molecular programs regulate these distinct phagocytic functions is unknown. Here we identify a molecularly distinct microglial subset in the synapse rich regions of the zebrafish (Danio rerio) brain. We found that ramified microglia increased in synaptic regions of the midbrain and hindbrain between 7 and 28 days post fertilization. In contrast, microglia in the optic tectum were ameboid and clustered around neurogenic zones. Using single-cell mRNA sequencing combined with metadata from regional bulk sequencing, we identified synaptic-region associated microglia (SAMs) that were highly enriched in the hindbrain and expressed multiple candidate synapse modulating genes, including genes in the complement pathway. In contrast, neurogenic associated microglia (NAMs) were enriched in the optic tectum, had active cathepsin activity, and preferentially engulfed neuronal corpses. These data reveal that molecularly distinct phagocytic programs mediate synaptic remodeling and cell engulfment, and establish the zebrafish hindbrain as a model for investigating microglial-synapse interactions.


Mesencephalon/cytology , Microglia/cytology , Neurogenesis/genetics , Rhombencephalon/cytology , Superior Colliculi/cytology , Transcriptome , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Cathepsin B/genetics , Cathepsin B/immunology , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mesencephalon/growth & development , Mesencephalon/immunology , Microglia/immunology , Neurogenesis/immunology , Neurons/cytology , Neurons/immunology , Phagocytosis , Rhombencephalon/growth & development , Rhombencephalon/immunology , Single-Cell Analysis , Superior Colliculi/growth & development , Superior Colliculi/immunology , Synapses/immunology , Synapses/metabolism , Synapses/ultrastructure , Zebrafish , Zebrafish Proteins/immunology
9.
Elife ; 102021 10 15.
Article En | MEDLINE | ID: mdl-34652270

Peripheral nerve injury-induced neuropathic pain is a chronic and debilitating condition characterized by mechanical hypersensitivity. We previously identified microglial activation via release of colony-stimulating factor 1 (CSF1) from injured sensory neurons as a mechanism contributing to nerve injury-induced pain. Here, we show that intrathecal administration of CSF1, even in the absence of injury, is sufficient to induce pain behavior, but only in male mice. Transcriptional profiling and morphologic analyses after intrathecal CSF1 showed robust immune activation in male but not female microglia. CSF1 also induced marked expansion of lymphocytes within the spinal cord meninges, with preferential expansion of regulatory T-cells (Tregs) in female mice. Consistent with the hypothesis that Tregs actively suppress microglial activation in females, Treg deficient (Foxp3DTR) female mice showed increased CSF1-induced microglial activation and pain hypersensitivity equivalent to males. We conclude that sexual dimorphism in the contribution of microglia to pain results from Treg-mediated suppression of microglial activation and pain hypersensitivity in female mice.


Macrophage Colony-Stimulating Factor/genetics , Microglia/metabolism , Neuralgia/genetics , T-Lymphocytes, Regulatory/physiology , Animals , Female , Injections, Spinal , Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Sex Factors
11.
ACS Sens ; 6(6): 2168-2180, 2021 06 25.
Article En | MEDLINE | ID: mdl-34102054

Lysosomes are important sites for macromolecular degradation, defined by an acidic lumenal pH of ∼4.5. To better understand lysosomal pH, we designed a novel, genetically encoded, fluorescent protein (FP)-based pH biosensor called Fluorescence Indicator REporting pH in Lysosomes (FIRE-pHLy). This biosensor was targeted to lysosomes with lysosomal-associated membrane protein 1 (LAMP1) and reported lumenal pH between 3.5 and 6.0 with monomeric teal fluorescent protein 1 (mTFP1), a bright cyan pH-sensitive FP variant with a pKa of 4.3. Ratiometric quantification was enabled with cytosolically oriented mCherry using high-content quantitative imaging. We expressed FIRE-pHLy in several cellular models and quantified the alkalinizing response to bafilomycin A1, a specific V-ATPase inhibitor. In summary, we have engineered FIRE-pHLy, a specific, robust, and versatile lysosomal pH biosensor, that has broad applications for investigating pH dynamics in aging- and lysosome-related diseases, as well as in lysosome-based drug discovery.


Biosensing Techniques , Lysosomes , Green Fluorescent Proteins , Hydrogen-Ion Concentration
12.
Immunity ; 54(2): 211-224, 2021 02 09.
Article En | MEDLINE | ID: mdl-33567261

Astrocytes play both physiological and pathological roles in maintaining central nervous system (CNS) function. Here, we review the varied functions of astrocytes and how these might change in subsets of reactive astrocytes. We review the current understanding of astrocyte interactions with microglia and the vasculature and protective barriers in the central nervous system as well as highlight recent insights into physiologic and reactive astrocyte sub-states identified by transcriptional profiling. Our goal is to stimulate inquiry into how these molecular identifiers link to specific functional changes in astrocytes and to define the implications of these heterogeneous molecular and functional changes in brain function and pathology. Defining these complex interactions has the potential to yield new therapies in CNS injury, infection, and disease.


Astrocytes/immunology , Brain/pathology , Central Nervous System Diseases/immunology , Infections/immunology , Inflammation/immunology , Animals , Cell Communication , Humans , Immunity, Cellular , Neuroimmunomodulation
13.
Nat Neurosci ; 24(3): 312-325, 2021 03.
Article En | MEDLINE | ID: mdl-33589835

Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.


Aging/pathology , Astrocytes/pathology , Brain/pathology , Spinal Cord/pathology , Animals , Brain Diseases/pathology , Brain Injuries/pathology , Humans , Spinal Cord Injuries/pathology
14.
Nat Neurosci ; 23(11): 1444-1452, 2020 11.
Article En | MEDLINE | ID: mdl-32929245

The ventral hippocampus (vHPC) is a critical hub in networks that process emotional information. While recent studies have indicated that ventral CA1 (vCA1) projection neurons are functionally dissociable, the basic principles of how the inputs and outputs of vCA1 are organized remain unclear. Here, we used viral and sequencing approaches to define the logic of the extended vCA1 circuit. Using high-throughput sequencing of genetically barcoded neurons (MAPseq) to map the axonal projections of thousands of vCA1 neurons, we identify a population of neurons that simultaneously broadcast information to multiple areas known to regulate the stress axis and approach-avoidance behavior. Through molecular profiling and viral input-output tracing of vCA1 projection neurons, we show how neurons with distinct projection targets may differ in their inputs and transcriptional signatures. These studies reveal new organizational principles of vCA1 that may underlie its functional heterogeneity.


CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Brain/cytology , Brain/metabolism , Female , Gene Expression Profiling , Male , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/metabolism
15.
Trends Immunol ; 41(9): 753-755, 2020 09.
Article En | MEDLINE | ID: mdl-32800455

Huang et al. have found that deletion of astrocyte lineage-specifying transcription factor NFIA from mature astrocytes alters astrocyte morphology, molecular identity, and synaptic-support capacity in a region-specific manner. We discuss the implications of these findings in light of emerging roles for astrocytes in immune cell crosstalk.


Astrocytes , Humans
16.
Cell ; 182(2): 388-403.e15, 2020 07 23.
Article En | MEDLINE | ID: mdl-32615087

Synapse remodeling is essential to encode experiences into neuronal circuits. Here, we define a molecular interaction between neurons and microglia that drives experience-dependent synapse remodeling in the hippocampus. We find that the cytokine interleukin-33 (IL-33) is expressed by adult hippocampal neurons in an experience-dependent manner and defines a neuronal subset primed for synaptic plasticity. Loss of neuronal IL-33 or the microglial IL-33 receptor leads to impaired spine plasticity, reduced newborn neuron integration, and diminished precision of remote fear memories. Memory precision and neuronal IL-33 are decreased in aged mice, and IL-33 gain of function mitigates age-related decreases in spine plasticity. We find that neuronal IL-33 instructs microglial engulfment of the extracellular matrix (ECM) and that its loss leads to impaired ECM engulfment and a concomitant accumulation of ECM proteins in contact with synapses. These data define a cellular mechanism through which microglia regulate experience-dependent synapse remodeling and promote memory consolidation.


Extracellular Matrix/metabolism , Microglia/physiology , Neuronal Plasticity/physiology , Aging , Animals , Fear , Gene Expression Regulation , Hippocampus/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Signal Transduction
17.
Trends Neurosci ; 43(3): 144-154, 2020 03.
Article En | MEDLINE | ID: mdl-32044129

Healthy central nervous system (CNS) development and function require an intricate and balanced bidirectional communication between neurons and glia cells. In this review, we discuss the complementary roles of astrocytes and microglia in building the brain, including in the formation and refinement of synapses. We discuss recent evidence demonstrating how these interactions are coordinated in the transition from healthy physiology towards disease and discuss known and potential molecular mechanisms that mediate this cellular crosstalk.


Astrocytes , Microglia , Humans , Neuroglia , Neurons , Synapses
18.
Immunity ; 50(3): 707-722.e6, 2019 03 19.
Article En | MEDLINE | ID: mdl-30824323

Type 2 lymphocytes promote both physiologic tissue remodeling and allergic pathology, yet their physical tissue niches are poorly described. Here, we used quantitative imaging to define the tissue niches of group 2 innate lymphoid cells (ILC2s), which are critical instigators of type 2 immunity. We identified a dominant adventitial niche around lung bronchi and larger vessels in multiple tissues, where ILC2s localized with subsets of dendritic and regulatory T cells. However, ILC2s were most intimately associated with adventitial stromal cells (ASCs), a mesenchymal fibroblast-like subset that expresses interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP). In vitro, ASCs produced TSLP that supported ILC2 accumulation and activation. ILC2s and IL-13 drove reciprocal ASC expansion and IL-33 expression. During helminth infection, ASC depletion impaired lung ILC2 and Th2 cell accumulation and function, which are in part dependent on ASC-derived IL-33. These data indicate that adventitial niches are conserved sites where ASCs regulate type 2 lymphocyte expansion and function.


Immunity, Innate/immunology , Lymphocytes/immunology , Stromal Cells/immunology , Animals , Bronchi/immunology , Cytokines/immunology , Interleukin-13/immunology , Interleukin-33/immunology , Mice , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Thymic Stromal Lymphopoietin
19.
Immunity ; 50(1): 11-13, 2019 01 15.
Article En | MEDLINE | ID: mdl-30650371

Microglia actively shape the developing brain, but their transcriptional diversity is not well understood. Complementary studies by Hammond et al. (2018) and Li et al. (2019) characterize the microglial transcriptome at single cell resolution, highlighting their diversity during development, aging, and pathology.


Microglia , RNA , Brain , Myeloid Cells , Sequence Analysis, RNA
20.
Nat Neurosci ; 21(9): 1171-1184, 2018 09.
Article En | MEDLINE | ID: mdl-30154505

It is widely assumed that cells must be physically isolated to study their molecular profiles. However, intact tissue samples naturally exhibit variation in cellular composition, which drives covariation of cell-class-specific molecular features. By analyzing transcriptional covariation in 7,221 intact CNS samples from 840 neurotypical individuals, representing billions of cells, we reveal the core transcriptional identities of major CNS cell classes in humans. By modeling intact CNS transcriptomes as a function of variation in cellular composition, we identify cell-class-specific transcriptional differences in Alzheimer's disease, among brain regions, and between species. Among these, we show that PMP2 is expressed by human but not mouse astrocytes and significantly increases mouse astrocyte size upon ectopic expression in vivo, causing them to more closely resemble their human counterparts. Our work is available as an online resource ( http://oldhamlab.ctec.ucsf.edu/ ) and provides a generalizable strategy for determining the core molecular features of cellular identity in intact biological systems.


Central Nervous System/cytology , Transcription, Genetic/physiology , Alzheimer Disease/genetics , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Brain/cytology , Brain/metabolism , Cell Size , Databases, Genetic , Gene Expression Profiling , Humans , Mice , Models, Genetic , Myelin P2 Protein/biosynthesis , Myelin P2 Protein/genetics , Sequence Analysis, RNA , Transcriptome
...