Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.023
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 92: 435-464, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37018845

ABSTRACT

The polyamines putrescine, spermidine, and spermine are abundant polycations of vital importance in mammalian cells. Their cellular levels are tightly regulated by degradation and synthesis, as well as by uptake and export. Here, we discuss the delicate balance between the neuroprotective and neurotoxic effects of polyamines in the context of Parkinson's disease (PD). Polyamine levels decline with aging and are altered in patients with PD, whereas recent mechanistic studies on ATP13A2 (PARK9) demonstrated a driving role of a disturbed polyamine homeostasis in PD. Polyamines affect pathways in PD pathogenesis, such as α-synuclein aggregation, and influence PD-related processes like autophagy, heavy metal toxicity, oxidative stress, neuroinflammation, and lysosomal/mitochondrial dysfunction. We formulate outstanding research questions regarding the role of polyamines in PD, their potential as PD biomarkers, and possible therapeutic strategies for PD targeting polyamine homeostasis.


Subject(s)
Parkinson Disease , Parkinsonian Disorders , Animals , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Polyamines/metabolism , Neuroprotection , Spermidine/metabolism , Mammals/metabolism
2.
Cell ; 186(1): 194-208.e18, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36580914

ABSTRACT

The diversity and complex organization of cells in the brain have hindered systematic characterization of age-related changes in its cellular and molecular architecture, limiting our ability to understand the mechanisms underlying its functional decline during aging. Here, we generated a high-resolution cell atlas of brain aging within the frontal cortex and striatum using spatially resolved single-cell transcriptomics and quantified changes in gene expression and spatial organization of major cell types in these regions over the mouse lifespan. We observed substantially more pronounced changes in cell state, gene expression, and spatial organization of non-neuronal cells over neurons. Our data revealed molecular and spatial signatures of glial and immune cell activation during aging, particularly enriched in the subcortical white matter, and identified both similarities and notable differences in cell-activation patterns induced by aging and systemic inflammatory challenge. These results provide critical insights into age-related decline and inflammation in the brain.


Subject(s)
Aging , White Matter , Mice , Animals , Aging/genetics , Brain/metabolism , Neuroglia , Longevity , Transcriptome , Single-Cell Analysis
3.
Cell ; 184(21): 5465-5481.e16, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34582787

ABSTRACT

In vivo cell fate conversions have emerged as potential regeneration-based therapeutics for injury and disease. Recent studies reported that ectopic expression or knockdown of certain factors can convert resident astrocytes into functional neurons with high efficiency, region specificity, and precise connectivity. However, using stringent lineage tracing in the mouse brain, we show that the presumed astrocyte-converted neurons are actually endogenous neurons. AAV-mediated co-expression of NEUROD1 and a reporter specifically and efficiently induces reporter-labeled neurons. However, these neurons cannot be traced retrospectively to quiescent or reactive astrocytes using lineage-mapping strategies. Instead, through a retrograde labeling approach, our results reveal that endogenous neurons are the source for these viral-reporter-labeled neurons. Similarly, despite efficient knockdown of PTBP1 in vivo, genetically traced resident astrocytes were not converted into neurons. Together, our results highlight the requirement of lineage-tracing strategies, which should be broadly applied to studies of cell fate conversions in vivo.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Cell Lineage , Neurons/cytology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/pathology , Brain Injuries/pathology , Cell Line, Tumor , Cellular Reprogramming , Dependovirus/metabolism , Down-Regulation , Gene Expression Regulation , Genes, Reporter , Glial Fibrillary Acidic Protein/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homeodomain Proteins/metabolism , Humans , Integrases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
4.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32702314

ABSTRACT

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Subject(s)
Alzheimer Disease/pathology , Sequence Analysis, DNA/methods , Transcriptome , Alzheimer Disease/genetics , Amyloid/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Lysosomes/genetics , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oxidative Stress/genetics
5.
Cell ; 181(4): 784-799.e19, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32413299

ABSTRACT

Swelling of the brain or spinal cord (CNS edema) affects millions of people every year. All potential pharmacological interventions have failed in clinical trials, meaning that symptom management is the only treatment option. The water channel protein aquaporin-4 (AQP4) is expressed in astrocytes and mediates water flux across the blood-brain and blood-spinal cord barriers. Here we show that AQP4 cell-surface abundance increases in response to hypoxia-induced cell swelling in a calmodulin-dependent manner. Calmodulin directly binds the AQP4 carboxyl terminus, causing a specific conformational change and driving AQP4 cell-surface localization. Inhibition of calmodulin in a rat spinal cord injury model with the licensed drug trifluoperazine inhibited AQP4 localization to the blood-spinal cord barrier, ablated CNS edema, and led to accelerated functional recovery compared with untreated animals. We propose that targeting the mechanism of calmodulin-mediated cell-surface localization of AQP4 is a viable strategy for development of CNS edema therapies.


Subject(s)
Aquaporin 4/metabolism , Edema/metabolism , Edema/therapy , Animals , Aquaporin 4/physiology , Astrocytes/metabolism , Brain/metabolism , Brain Edema/metabolism , Calmodulin/metabolism , Central Nervous System/metabolism , Edema/physiopathology , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Trifluoperazine/pharmacology
6.
Cell ; 180(5): 833-846.e16, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142677

ABSTRACT

Cognitive dysfunction and reactive microglia are hallmarks of traumatic brain injury (TBI), yet whether these cells contribute to cognitive deficits and secondary inflammatory pathology remains poorly understood. Here, we show that removal of microglia from the mouse brain has little effect on the outcome of TBI, but inducing the turnover of these cells through either pharmacologic or genetic approaches can yield a neuroprotective microglial phenotype that profoundly aids recovery. The beneficial effects of these repopulating microglia are critically dependent on interleukin-6 (IL-6) trans-signaling via the soluble IL-6 receptor (IL-6R) and robustly support adult neurogenesis, specifically by augmenting the survival of newborn neurons that directly support cognitive function. We conclude that microglia in the mammalian brain can be manipulated to adopt a neuroprotective and pro-regenerative phenotype that can aid repair and alleviate the cognitive deficits arising from brain injury.


Subject(s)
Brain Injuries, Traumatic/therapy , Interleukin-6/genetics , Receptors, Interleukin-6/genetics , Regeneration/genetics , Animals , Brain/growth & development , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/therapy , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Signal Transduction/genetics
7.
Cell ; 177(5): 1280-1292.e20, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31031006

ABSTRACT

Hyperactivity and disturbances of attention are common behavioral disorders whose underlying cellular and neural circuit causes are not understood. We report the discovery that striatal astrocytes drive such phenotypes through a hitherto unknown synaptic mechanism. We found that striatal medium spiny neurons (MSNs) triggered astrocyte signaling via γ-aminobutyric acid B (GABAB) receptors. Selective chemogenetic activation of this pathway in striatal astrocytes in vivo resulted in acute behavioral hyperactivity and disrupted attention. Such responses also resulted in upregulation of the synaptogenic cue thrombospondin-1 (TSP1) in astrocytes, increased excitatory synapses, enhanced corticostriatal synaptic transmission, and increased MSN action potential firing in vivo. All of these changes were reversed by blocking TSP1 effects. Our data identify a form of bidirectional neuron-astrocyte communication and demonstrate that acute reactivation of a single latent astrocyte synaptogenic cue alters striatal circuits controlling behavior, revealing astrocytes and the TSP1 pathway as therapeutic targets in hyperactivity, attention deficit, and related psychiatric disorders.


Subject(s)
Astrocytes/metabolism , Attention Deficit Disorder with Hyperactivity/metabolism , Behavior, Animal , Cell Communication , Neurons/metabolism , Signal Transduction , Synapses/metabolism , Animals , Astrocytes/pathology , Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/pathology , Attention Deficit Disorder with Hyperactivity/physiopathology , Female , Male , Mice , Mice, Transgenic , Neurons/pathology , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Synapses/genetics , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
8.
Cell ; 176(5): 1143-1157.e13, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30794775

ABSTRACT

We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.


Subject(s)
Brain Injuries, Traumatic/therapy , Receptors, CCR5/metabolism , Stroke/therapy , Aged , Aged, 80 and over , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Middle Aged , Motor Cortex/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, CCR5/physiology , Stroke Rehabilitation/methods
9.
Cell ; 176(1-2): 43-55.e13, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30528430

ABSTRACT

Chemotherapy results in a frequent yet poorly understood syndrome of long-term neurological deficits. Neural precursor cell dysfunction and white matter dysfunction are thought to contribute to this debilitating syndrome. Here, we demonstrate persistent depletion of oligodendrocyte lineage cells in humans who received chemotherapy. Developing a mouse model of methotrexate chemotherapy-induced neurological dysfunction, we find a similar depletion of white matter OPCs, increased but incomplete OPC differentiation, and a persistent deficit in myelination. OPCs from chemotherapy-naive mice similarly exhibit increased differentiation when transplanted into the microenvironment of previously methotrexate-exposed brains, indicating an underlying microenvironmental perturbation. Methotrexate results in persistent activation of microglia and subsequent astrocyte activation that is dependent on inflammatory microglia. Microglial depletion normalizes oligodendroglial lineage dynamics, myelin microstructure, and cognitive behavior after methotrexate chemotherapy. These findings indicate that methotrexate chemotherapy exposure is associated with persistent tri-glial dysregulation and identify inflammatory microglia as a therapeutic target to abrogate chemotherapy-related cognitive impairment. VIDEO ABSTRACT.


Subject(s)
Cognitive Dysfunction/chemically induced , Methotrexate/adverse effects , Oligodendroglia/drug effects , Animals , Brain/metabolism , Cell Differentiation , Cell Lineage , Cognitive Dysfunction/metabolism , Disease Models, Animal , Drug Therapy , Drug-Related Side Effects and Adverse Reactions , Humans , Methotrexate/pharmacology , Mice , Microglia/metabolism , Myelin Sheath/metabolism , Nerve Fibers, Myelinated , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/drug effects , Oligodendroglia/metabolism , White Matter/metabolism
10.
Cell ; 176(3): 581-596.e18, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661753

ABSTRACT

Genome-wide studies have identified genetic variants linked to neurologic diseases. Environmental factors also play important roles, but no methods are available for their comprehensive investigation. We developed an approach that combines genomic data, screens in a novel zebrafish model, computational modeling, perturbation studies, and multiple sclerosis (MS) patient samples to evaluate the effects of environmental exposure on CNS inflammation. We found that the herbicide linuron amplifies astrocyte pro-inflammatory activities by activating signaling via sigma receptor 1, inositol-requiring enzyme-1α (IRE1α), and X-box binding protein 1 (XBP1). Indeed, astrocyte-specific shRNA- and CRISPR/Cas9-driven gene inactivation combined with RNA-seq, ATAC-seq, ChIP-seq, and study of patient samples suggest that IRE1α-XBP1 signaling promotes CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, MS. In summary, these studies define environmental mechanisms that control astrocyte pathogenic activities and establish a multidisciplinary approach for the systematic investigation of the effects of environmental exposure in neurologic disorders.


Subject(s)
Astrocytes/metabolism , Central Nervous System/metabolism , Animals , Central Nervous System/immunology , Computational Biology/methods , Encephalomyelitis, Autoimmune, Experimental/immunology , Endoribonucleases/metabolism , Environment , Environmental Exposure/adverse effects , Genome , Genomics , Humans , Inflammation/metabolism , Linuron/adverse effects , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Protein Serine-Threonine Kinases/metabolism , Receptors, sigma/drug effects , Receptors, sigma/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , Zebrafish
11.
Cell ; 177(6): 1522-1535.e14, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31130380

ABSTRACT

Metabolic coordination between neurons and astrocytes is critical for the health of the brain. However, neuron-astrocyte coupling of lipid metabolism, particularly in response to neural activity, remains largely uncharacterized. Here, we demonstrate that toxic fatty acids (FAs) produced in hyperactive neurons are transferred to astrocytic lipid droplets by ApoE-positive lipid particles. Astrocytes consume the FAs stored in lipid droplets via mitochondrial ß-oxidation in response to neuronal activity and turn on a detoxification gene expression program. Our findings reveal that FA metabolism is coupled in neurons and astrocytes to protect neurons from FA toxicity during periods of enhanced activity. This coordinated mechanism for metabolizing FAs could underlie both homeostasis and a variety of disease states of the brain.


Subject(s)
Astrocytes/metabolism , Fatty Acids/metabolism , Neurons/metabolism , Animals , Apolipoproteins E/metabolism , Apolipoproteins E/physiology , Astrocytes/physiology , Brain/metabolism , Fatty Acids/toxicity , Homeostasis , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
12.
Immunity ; 56(7): 1502-1514.e8, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37160117

ABSTRACT

Glial cells and central nervous system (CNS)-infiltrating leukocytes contribute to multiple sclerosis (MS). However, the networks that govern crosstalk among these ontologically distinct populations remain unclear. Here, we show that, in mice and humans, CNS-resident astrocytes and infiltrating CD44hiCD4+ T cells generated interleukin-3 (IL-3), while microglia and recruited myeloid cells expressed interleukin-3 receptor-ɑ (IL-3Rɑ). Astrocytic and T cell IL-3 elicited an immune migratory and chemotactic program by IL-3Rɑ+ myeloid cells that enhanced CNS immune cell infiltration, exacerbating MS and its preclinical model. Multiregional snRNA-seq of human CNS tissue revealed the appearance of IL3RA-expressing myeloid cells with chemotactic programming in MS plaques. IL3RA expression by plaque myeloid cells and IL-3 amount in the cerebrospinal fluid predicted myeloid and T cell abundance in the CNS and correlated with MS severity. Our findings establish IL-3:IL-3RA as a glial-peripheral immune network that prompts immune cell recruitment to the CNS and worsens MS.


Subject(s)
Multiple Sclerosis , Animals , Humans , Mice , Central Nervous System , Interleukin-3 , Microglia , Neuroglia/metabolism
13.
Annu Rev Neurosci ; 46: 101-121, 2023 07 10.
Article in English | MEDLINE | ID: mdl-36854317

ABSTRACT

Astrocyte endfeet enwrap the entire vascular tree within the central nervous system, where they perform important functions in regulating the blood-brain barrier (BBB), cerebral blood flow, nutrient uptake, and waste clearance. Accordingly, astrocyte endfeet contain specialized organelles and proteins, including local protein translation machinery and highly organized scaffold proteins, which anchor channels, transporters, receptors, and enzymes critical for astrocyte-vascular interactions. Many neurological diseases are characterized by the loss of polarization of specific endfoot proteins, vascular dysregulation, BBB disruption, altered waste clearance, or, in extreme cases, loss of endfoot coverage. A role for astrocyte endfeet has been demonstrated or postulated in many of these conditions. This review provides an overview of the development, composition, function, and pathological changes of astrocyte endfeet and highlights the gaps in our knowledge that future research should address.


Subject(s)
Astrocytes , Blood-Brain Barrier , Astrocytes/physiology , Blood-Brain Barrier/metabolism , Central Nervous System , Protein Biosynthesis , Brain/pathology
14.
Genes Dev ; 35(21-22): 1431-1444, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34675062

ABSTRACT

During neocortical development, tight regulation of neurogenesis-to-astrogenesis switching of neural precursor cells (NPCs) is critical to generate a balanced number of each neural cell type for proper brain functions. Accumulating evidence indicates that a complex array of epigenetic modifications and the availability of extracellular factors control the timing of neuronal and astrocytic differentiation. However, our understanding of NPC fate regulation is still far from complete. Bone morphogenetic proteins (BMPs) are renowned as cytokines that induce astrogenesis of gliogenic late-gestational NPCs. They also promote neurogenesis of mid-gestational NPCs, although the underlying mechanisms remain elusive. By performing multiple genome-wide analyses, we demonstrate that Smads, transcription factors that act downstream from BMP signaling, target dramatically different genomic regions in neurogenic and gliogenic NPCs. We found that histone H3K27 trimethylation and DNA methylation around Smad-binding sites change rapidly as gestation proceeds, strongly associated with the alteration of accessibility of Smads to their target binding sites. Furthermore, we identified two lineage-specific Smad-interacting partners-Sox11 for neurogenic and Sox8 for astrocytic differentiation-that further ensure Smad-regulated fate-specific gene induction. Our findings illuminate an exquisite regulation of NPC property change mediated by the interplay between cell-extrinsic cues and -intrinsic epigenetic programs during cortical development.


Subject(s)
Neural Stem Cells , Brain , Cell Differentiation/genetics , Epigenesis, Genetic , Female , Genome-Wide Association Study , Humans , Neurogenesis/genetics , Pregnancy , SOXE Transcription Factors/genetics
15.
EMBO J ; 42(15): e111247, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37357972

ABSTRACT

Social behavior is essential for health, survival, and reproduction of animals; however, the role of astrocytes in social behavior remains largely unknown. The transmembrane protein CD38, which acts both as a receptor and ADP-ribosyl cyclase to produce cyclic ADP-ribose (cADPR) regulates social behaviors by promoting oxytocin release from hypothalamic neurons. CD38 is also abundantly expressed in astrocytes in the postnatal brain and is important for astroglial development. Here, we demonstrate that the astroglial-expressed CD38 plays an important role in social behavior during development. Selective deletion of CD38 in postnatal astrocytes, but not in adult astrocytes, impairs social memory without any other behavioral abnormalities. Morphological analysis shows that depletion of astroglial CD38 in the postnatal brain interferes with synapse formation in the medial prefrontal cortex (mPFC) and hippocampus. Moreover, astroglial CD38 expression promotes synaptogenesis of excitatory neurons by increasing the level of extracellular SPARCL1 (also known as Hevin), a synaptogenic protein. The release of SPARCL1 from astrocytes is regulated by CD38/cADPR/calcium signaling. These data demonstrate a novel developmental role of astrocytes in neural circuit formation and regulation of social behavior in adults.


Subject(s)
Antigens, CD , Cyclic ADP-Ribose , Animals , ADP-ribosyl Cyclase 1/genetics , Antigens, CD/metabolism , Cyclic ADP-Ribose/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Astrocytes/metabolism , Synapses/metabolism
16.
Annu Rev Neurosci ; 42: 187-207, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31283899

ABSTRACT

Astrocytes are morphologically complex, ubiquitous cells that are viewed as a homogeneous population tiling the entire central nervous system (CNS). However, this view has been challenged in the last few years with the availability of RNA sequencing, immunohistochemistry, electron microscopy, morphological reconstruction, and imaging data. These studies suggest that astrocytes represent a diverse population of cells and that they display brain area- and disease-specific properties and functions. In this review, we summarize these observations, emphasize areas where clear conclusions can be made, and discuss potential unifying themes. We also identify knowledge gaps that need to be addressed in order to exploit astrocyte diversity as a biological phenomenon of physiological relevance in the CNS. We thus provide a summary and a perspective on astrocyte diversity in the vertebrate CNS.


Subject(s)
Astrocytes/classification , Animals , Astrocytes/physiology , Astrocytes/ultrastructure , Biomarkers , Calcium Signaling , Cell Compartmentation , Cell Lineage , Cell Shape , Cell Size , Electrophysiology , Forecasting , Mice , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/physiology , Neurogenesis , Vertebrates/anatomy & histology , Vertebrates/physiology
17.
Annu Rev Neurosci ; 42: 107-127, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31283900

ABSTRACT

Maturation of neuronal circuits requires selective elimination of synaptic connections. Although neuron-intrinsic mechanisms are important in this process, it is increasingly recognized that glial cells also play a critical role. Without proper functioning of these cells, the number, morphology, and function of synaptic contacts are profoundly altered, resulting in abnormal connectivity and behavioral abnormalities. In addition to their role in synaptic refinement, glial cells have also been implicated in pathological synapse loss and dysfunction following injury or nervous system degeneration in adults. Although mechanisms regulating glia-mediated synaptic elimination are still being uncovered, it is clear this complex process involves many cues that promote and inhibit the removal of specific synaptic connections. Gaining a greater understanding of these signals and the contribution of different cell types will not only provide insight into this critical biological event but also be instrumental in advancing knowledge of brain development and neural disease.


Subject(s)
Central Nervous System/embryology , Nerve Degeneration/physiopathology , Nervous System Diseases/physiopathology , Neuroglia/physiology , Neurons/physiology , Peripheral Nervous System/embryology , Synapses/physiology , Animals , Astrocytes/physiology , Biological Evolution , Central Nervous System/growth & development , Cues , Exosomes/physiology , Humans , Invertebrates/embryology , Microglia/physiology , Morphogenesis , Myelin Sheath/physiology , Neuromuscular Junction/embryology , Peripheral Nervous System/growth & development , Synapses/pathology
18.
Semin Immunol ; 67: 101751, 2023 05.
Article in English | MEDLINE | ID: mdl-36989541

ABSTRACT

Immunity to fungal infections of the central nervous system (CNS) is one of the most poorly understood subjects within the field of medical mycology. Yet, the majority of deaths from invasive fungal infections are caused by brain-tropic fungi. In recent years, there have been several significant discoveries in the regulation of neuroinflammation and the role of the immune system in tissue homeostasis within the CNS. In this review, I highlight five important advances in the neuroimmunology field over the last decade and discuss how we should capitalise on these discoveries to better understand the pathogenesis of fungal CNS infections. In addition, the latest insights into fungal invasion tactics, microglia-astrocyte crosstalk and regulation of antifungal adaptive immune responses are summarised in the context of our contemporary understanding of CNS-specific immunity.


Subject(s)
Central Nervous System Infections , Mycoses , Humans , Central Nervous System , Microglia , Immunity
19.
Proc Natl Acad Sci U S A ; 121(22): e2315690121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781206

ABSTRACT

The prion-like spread of protein aggregates is a leading hypothesis for the propagation of neurofibrillary lesions in the brain, including the spread of tau inclusions associated with Alzheimer's disease. The mechanisms of cellular uptake of tau seeds and subsequent nucleated polymerization of cytosolic tau are major questions in the field, and the potential for coupling between the entry and nucleation mechanisms has been little explored. We found that in primary astrocytes and neurons, endocytosis of tau seeds leads to their accumulation in lysosomes. This in turn leads to lysosomal swelling, deacidification, and recruitment of ESCRT proteins, but not Galectin-3, to the lysosomal membrane. These observations are consistent with nanoscale damage of the lysosomal membrane. Live cell imaging and STORM superresolution microscopy further show that the nucleation of cytosolic tau occurs primarily at the lysosome membrane under these conditions. These data suggest that tau seeds escape from lysosomes via nanoscale damage rather than wholesale rupture and that nucleation of cytosolic tau commences as soon as tau fibril ends emerge from the lysosomal membrane.


Subject(s)
Cytosol , Lysosomes , tau Proteins , tau Proteins/metabolism , Lysosomes/metabolism , Cytosol/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Neurons/metabolism , Neurons/pathology , Humans , Intracellular Membranes/metabolism , Endocytosis , Mice , Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 121(22): e2400648121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781210

ABSTRACT

After central nervous system injury, a rapid cellular and molecular response is induced. This response can be both beneficial and detrimental to neuronal survival in the first few days and increases the risk for neurodegeneration if persistent. Semaphorin4B (Sema4B), a transmembrane protein primarily expressed by cortical astrocytes, has been shown to play a role in neuronal cell death following injury. Our study shows that after cortical stab wound injury, cytokine expression is attenuated in Sema4B-/- mice, and microglia/macrophage reactivity is altered. In vitro, Sema4B enhances the reactivity of microglia following injury, suggesting astrocytic Sema4B functions as a ligand. Moreover, injury-induced microglia reactivity is attenuated in the presence of Sema4B-/- astrocytes compared to Sema4B+/- astrocytes. In vitro experiments indicate that Plexin-B2 is the Sema4B receptor on microglia. Consistent with this, in microglia/macrophage-specific Plexin-B2-/- mice, similar to Sema4B-/- mice, microglial/macrophage reactivity and neuronal cell death are attenuated after cortical injury. Finally, in Sema4B/Plexin-B2 double heterozygous mice, microglial/macrophage reactivity is also reduced after injury, supporting the idea that both Sema4B and Plexin-B2 are part of the same signaling pathway. Taken together, we propose a model in which following injury, astrocytic Sema4B enhances the response of microglia/macrophages via Plexin-B2, leading to increased reactivity.


Subject(s)
Astrocytes , Mice, Knockout , Microglia , Nerve Tissue Proteins , Semaphorins , Animals , Mice , Astrocytes/metabolism , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/genetics , Cell Communication , Macrophages/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Semaphorins/metabolism , Semaphorins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL