Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 177(5): 1091-1093, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31100265

ABSTRACT

Are neurons solely responsible for determining behavioral output, or can other brain cells modulate behavior? In this issue of Cell, Nagai et al. demonstrate that striatal astrocytes, through GABAB receptor signaling, regulate behaviors including hyperactivity and attention by inducing new synapse formation between neurons.


Subject(s)
Astrocytes , Cues , Attention , Neurons , Receptors, GABA-B
2.
Nat Rev Neurosci ; 24(1): 23-39, 2023 01.
Article in English | MEDLINE | ID: mdl-36316501

ABSTRACT

There is increasing appreciation that non-neuronal cells contribute to the initiation, progression and pathology of diverse neurodegenerative disorders. This Review focuses on the role of astrocytes in disorders including Alzheimer disease, Parkinson disease, Huntington disease and amyotrophic lateral sclerosis. The important roles astrocytes have in supporting neuronal function in the healthy brain are considered, along with studies that have demonstrated how the physiological properties of astrocytes are altered in neurodegenerative disorders and may explain their contribution to neurodegeneration. Further, the question of whether in neurodegenerative disorders with specific genetic mutations these mutations directly impact on astrocyte function, and may suggest a driving role for astrocytes in disease initiation, is discussed. A summary of how astrocyte transcriptomic and proteomic signatures are altered during the progression of neurodegenerative disorders and may relate to functional changes is provided. Given the central role of astrocytes in neurodegenerative disorders, potential strategies to target these cells for future therapeutic avenues are discussed.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Astrocytes/physiology , Proteomics , Neurodegenerative Diseases/pathology , Amyotrophic Lateral Sclerosis/pathology , Neurons/pathology
3.
Annu Rev Cell Dev Biol ; 30: 439-63, 2014.
Article in English | MEDLINE | ID: mdl-25288116

ABSTRACT

Astrocytes regulate multiple aspects of neuronal and synaptic function from development through to adulthood. Instead of addressing each function independently, this review provides a comprehensive overview of the different ways astrocytes modulate neuronal synaptic function throughout life, with a particular focus on recent findings in each area. It includes the emerging functions of astrocytes, such as a role in synapse formation, as well as more established roles, including the uptake and recycling of neurotransmitters. This broad approach covers the many ways astrocytes and neurons constantly interact to maintain the correct functioning of the brain. It is important to consider all of these diverse functions of astrocytes when investigating how astrocyte-neuron interactions regulate synaptic behavior to appreciate the complexity of these ongoing interactions.


Subject(s)
Astrocytes/physiology , Nerve Tissue Proteins/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Brain/cytology , Brain/growth & development , Calcium Signaling , Cell Communication , Glutamic Acid/physiology , Humans , Ion Transport , Lipids/biosynthesis , Neurons/physiology , Neurotransmitter Agents/physiology , Neurotransmitter Transport Proteins/physiology , Potassium/metabolism , Receptors, Neurotransmitter/physiology
4.
Nat Chem Biol ; 19(2): 187-197, 2023 02.
Article in English | MEDLINE | ID: mdl-36266352

ABSTRACT

Lipids contribute to the structure, development, and function of healthy brains. Dysregulated lipid metabolism is linked to aging and diseased brains. However, our understanding of lipid metabolism in aging brains remains limited. Here we examined the brain lipidome of mice across their lifespan using untargeted lipidomics. Co-expression network analysis highlighted a progressive decrease in 3-sulfogalactosyl diacylglycerols (SGDGs) and SGDG pathway members, including the potential degradation products lyso-SGDGs. SGDGs show an age-related decline specifically in the central nervous system and are associated with myelination. We also found that an SGDG dramatically suppresses LPS-induced gene expression and release of pro-inflammatory cytokines from macrophages and microglia by acting on the NF-κB pathway. The detection of SGDGs in human and macaque brains establishes their evolutionary conservation. This work enhances interest in SGDGs regarding their roles in aging and inflammatory diseases and highlights the complexity of the brain lipidome and potential biological functions in aging.


Subject(s)
Aging , Lipids , Animals , Humans , Mice , Aging/genetics , Anti-Inflammatory Agents , Brain/metabolism , Microglia/metabolism , NF-kappa B/metabolism
5.
Cell ; 139(2): 380-92, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818485

ABSTRACT

Synapses are asymmetric cellular adhesions that are critical for nervous system development and function, but the mechanisms that induce their formation are not well understood. We have previously identified thrombospondin as an astrocyte-secreted protein that promotes central nervous system (CNS) synaptogenesis. Here, we identify the neuronal thrombospondin receptor involved in CNS synapse formation as alpha2delta-1, the receptor for the anti-epileptic and analgesic drug gabapentin. We show that the VWF-A domain of alpha2delta-1 interacts with the epidermal growth factor-like repeats common to all thrombospondins. alpha2delta-1 overexpression increases synaptogenesis in vitro and in vivo and is required postsynaptically for thrombospondin- and astrocyte-induced synapse formation in vitro. Gabapentin antagonizes thrombospondin binding to alpha2delta-1 and powerfully inhibits excitatory synapse formation in vitro and in vivo. These findings identify alpha2delta-1 as a receptor involved in excitatory synapse formation and suggest that gabapentin may function therapeutically by blocking new synapse formation.


Subject(s)
CD36 Antigens/metabolism , Calcium Channels/metabolism , Neurogenesis , Synapses , Amines/pharmacology , Animals , Calcium Channels, L-Type , Cyclohexanecarboxylic Acids/pharmacology , Gabapentin , Mice , Neuronal Plasticity , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , gamma-Aminobutyric Acid/pharmacology
6.
Am J Hum Genet ; 104(5): 914-924, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982611

ABSTRACT

Glypicans are a family of cell-surface heparan sulfate proteoglycans that regulate growth-factor signaling during development and are thought to play a role in the regulation of morphogenesis. Whole-exome sequencing of the Australian family that defined Keipert syndrome (nasodigitoacoustic syndrome) identified a hemizygous truncating variant in the gene encoding glypican 4 (GPC4). This variant, located in the final exon of GPC4, results in premature termination of the protein 51 amino acid residues prior to the stop codon, and in concomitant loss of functionally important N-linked glycosylation (Asn514) and glycosylphosphatidylinositol (GPI) anchor (Ser529) sites. We subsequently identified seven affected males from five additional kindreds with novel and predicted pathogenic variants in GPC4. Segregation analysis and X-inactivation studies in carrier females provided supportive evidence that the GPC4 variants caused the condition. Furthermore, functional studies of recombinant protein suggested that the truncated proteins p.Gln506∗ and p.Glu496∗ were less stable than the wild type. Clinical features of Keipert syndrome included a prominent forehead, a flat midface, hypertelorism, a broad nose, downturned corners of mouth, and digital abnormalities, whereas cognitive impairment and deafness were variable features. Studies of Gpc4 knockout mice showed evidence of the two primary features of Keipert syndrome: craniofacial abnormalities and digital abnormalities. Phylogenetic analysis demonstrated that GPC4 is most closely related to GPC6, which is associated with a bone dysplasia that has a phenotypic overlap with Keipert syndrome. Overall, we have shown that pathogenic variants in GPC4 cause a loss of function that results in Keipert syndrome, making GPC4 the third human glypican to be linked to a genetic syndrome.


Subject(s)
Deafness/congenital , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genetic Variation , Glypicans/genetics , Lower Extremity Deformities, Congenital/genetics , Lower Extremity Deformities, Congenital/pathology , Adult , Child , Child, Preschool , Deafness/genetics , Deafness/pathology , Female , Humans , Infant , Male , Pedigree , Phenotype , Young Adult
7.
Nature ; 592(7854): 360-361, 2021 04.
Article in English | MEDLINE | ID: mdl-33828277
8.
Nature ; 578(7793): 46-47, 2020 02.
Article in English | MEDLINE | ID: mdl-32020106
9.
Neurobiol Dis ; 143: 105008, 2020 09.
Article in English | MEDLINE | ID: mdl-32622920

ABSTRACT

Aging is the strongest risk factor for metabolic, vascular and neurodegenerative diseases. Aging alone is associated with a gradual decline of cognitive and motor functions. Considering an increasing elderly population in the last century, understanding the cellular and molecular mechanisms contributing to brain aging is of vital importance. Recent genetic and transcriptomic findings strongly suggest that glia are the first cells changing with aging. Glial cells constitute around 50% of the total cells in the brain and play key roles regulating brain homeostasis in health and disease. Their essential functions include providing nutritional support to neurons, activation of immune responses, and regulation of synaptic transmission and plasticity. In this review we discuss how glia are altered in the aging brain and whether these alterations are protective or contribute to the age-related pathological cascade. We focus on the major morphological, transcriptional and functional changes affecting glia in a range of systems, including human, non-human primates, and rodents. We also highlight future directions for investigating the roles of glia in brain aging.


Subject(s)
Aging/pathology , Brain/pathology , Neuroglia/pathology , Aging/metabolism , Animals , Brain/metabolism , Humans , Neuroglia/metabolism
10.
Nature ; 486(7403): 410-4, 2012 May 27.
Article in English | MEDLINE | ID: mdl-22722203

ABSTRACT

In the developing central nervous system (CNS), the control of synapse number and function is critical to the formation of neural circuits. We previously demonstrated that astrocyte-secreted factors powerfully induce the formation of functional excitatory synapses between CNS neurons. Astrocyte-secreted thrombospondins induce the formation of structural synapses, but these synapses are postsynaptically silent. Here we use biochemical fractionation of astrocyte-conditioned medium to identify glypican 4 (Gpc4) and glypican 6 (Gpc6) as astrocyte-secreted signals sufficient to induce functional synapses between purified retinal ganglion cell neurons, and show that depletion of these molecules from astrocyte-conditioned medium significantly reduces its ability to induce postsynaptic activity. Application of Gpc4 to purified neurons is sufficient to increase the frequency and amplitude of glutamatergic synaptic events. This is achieved by increasing the surface level and clustering, but not overall cellular protein level, of the GluA1 subunit of the AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) glutamate receptor (AMPAR). Gpc4 and Gpc6 are expressed by astrocytes in vivo in the developing CNS, with Gpc4 expression enriched in the hippocampus and Gpc6 enriched in the cerebellum. Finally, we demonstrate that Gpc4-deficient mice have defective synapse formation, with decreased amplitude of excitatory synaptic currents in the developing hippocampus and reduced recruitment of AMPARs to synapses. These data identify glypicans as a family of novel astrocyte-derived molecules that are necessary and sufficient to promote glutamate receptor clustering and receptivity and to induce the formation of postsynaptically functioning CNS synapses.


Subject(s)
Astrocytes/metabolism , Excitatory Postsynaptic Potentials/physiology , Glypicans/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Astrocytes/cytology , Cerebellum/cytology , Cerebellum/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Female , Glypicans/deficiency , Glypicans/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Synapses/drug effects , Synapses/pathology
11.
J Physiol ; 595(6): 1903-1916, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27381164

ABSTRACT

Astrocytes comprise half of the cells in the brain. Although astrocytes have traditionally been described as playing a supportive role for neurons, they have recently been recognized as active participants in the development and plasticity of dendritic spines and synapses. Astrocytes can eliminate dendritic spines, induce synapse formation, and regulate neurotransmission and plasticity. Dendritic spine and synapse impairments are features of many neurological disorders, including autism spectrum disorder, schizophrenia, and Alzheimer's disease. In this review we will present evidence from multiple neurological disorders demonstrating that changes in astrocyte-synapse interaction contribute to the pathologies. Genomic analysis has connected altered astrocytic gene expression with synaptic deficits in a number of neurological disorders. Alterations in astrocyte-secreted factors have been implicated in the neuronal morphology and synaptic changes seen in neurodevelopmental disorders, while alteration in astrocytic glutamate uptake is a core feature of multiple neurodegenerative disorders. This evidence clearly demonstrates that maintaining astrocyte-synapse interaction is crucial for normal central nervous system functioning. Obtaining a better understanding of the role of astrocytes at synapses in health and disease will provide a new avenue for future therapeutic targeting.


Subject(s)
Astrocytes/physiology , Nervous System Diseases/physiopathology , Neurodevelopmental Disorders/physiopathology , Synapses/physiology , Animals , Dendritic Spines/physiology , Humans
12.
Biochem Soc Trans ; 42(5): 1263-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25233401

ABSTRACT

Astrocytes modulate many aspects of neuronal function, including synapse formation and the response to injury. Heparan sulfate proteoglycans (HSPGs) mediate some of the effects of astrocytes on synaptic function, and participate in the astrocyte-mediated brain injury response. HSPGs are a highly conserved class of proteoglycans, with variable heparan sulfate (HS) chains that play a major role in determining the function of these proteins, such as binding to growth factors and receptors. Expression of both the core proteins and their HS chains can vary depending on cellular origin, thus the functional impact of HSPGs may be determined by the cell type in which they are expressed. In the brain, HSPGs are expressed by both neurons and astrocytes; however, the specific contribution of neuronal HSPGs compared with astrocyte-derived HSPGs to development and the injury response is largely unknown. The present review examines the current evidence regarding the roles of HSPGs in the brain, describes the cellular origins of HSPGs, and interrogates the roles of HSPGs from astrocytes and neurons in synaptogenesis and injury. The importance of considering cell-type-specific expression of HSPGs when studying brain function is discussed.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Heparan Sulfate Proteoglycans/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Animals , Astrocytes/cytology , Astrocytes/pathology , Brain Injuries/pathology , Humans , Neuronal Plasticity , Neurons/cytology , Neurons/pathology , Synapses/metabolism , Synapses/pathology
13.
Proc Natl Acad Sci U S A ; 108(32): E440-9, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788491

ABSTRACT

Astrocytes regulate synaptic connectivity in the CNS through secreted signals. Here we identified two astrocyte-secreted proteins, hevin and SPARC, as regulators of excitatory synaptogenesis in vitro and in vivo. Hevin induces the formation of synapses between cultured rat retinal ganglion cells. SPARC is not synaptogenic, but specifically antagonizes synaptogenic function of hevin. Hevin and SPARC are expressed by astrocytes in the superior colliculus, the synaptic target of retinal ganglion cells, concurrent with the excitatory synaptogenesis. Hevin-null mice had fewer excitatory synapses; conversely, SPARC-null mice had increased synaptic connections in the superior colliculus. Furthermore, we found that hevin is required for the structural maturation of the retinocollicular synapses. These results identify hevin as a positive and SPARC as a negative regulator of synapse formation and signify that, through regulation of relative levels of hevin and SPARC, astrocytes might control the formation, maturation, and plasticity of synapses in vivo.


Subject(s)
Astrocytes/metabolism , Calcium-Binding Proteins/metabolism , Central Nervous System/metabolism , Extracellular Matrix Proteins/metabolism , Neurogenesis , Osteonectin/metabolism , Synapses/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/ultrastructure , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/deficiency , Central Nervous System/cytology , Central Nervous System/ultrastructure , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/deficiency , HEK293 Cells , Humans , Mice , Neurogenesis/drug effects , Osteonectin/chemistry , Osteonectin/deficiency , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/ultrastructure , Superior Colliculi/cytology , Superior Colliculi/drug effects , Superior Colliculi/metabolism , Superior Colliculi/ultrastructure , Synapses/drug effects , Synapses/ultrastructure
14.
Article in English | MEDLINE | ID: mdl-38346858

ABSTRACT

Astrocytes play an integral role in the development, maturation, and refinement of neuronal circuits. Astrocytes secrete proteins and lipids that instruct the formation of new synapses and induce the maturation of existing synapses. Through contact-mediated signaling, astrocytes can regulate the formation and state of synapses within their domain. Through phagocytosis, astrocytes participate in the elimination of excess synaptic connections. In this work, we will review key findings on the molecular mechanisms of astrocyte-synapse interaction with a focus on astrocyte-secreted factors, contact-mediated mechanisms, and synapse elimination. We will discuss this in the context of typical brain development and maintenance, as well as consider the consequences of dysfunction in these pathways in neurological disorders, highlighting a role for astrocytes in health and disease.

15.
bioRxiv ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38559139

ABSTRACT

Neural circuits in many brain regions are refined by experience. Sensory circuits support higher plasticity at younger ages during critical periods - times of circuit refinement and maturation - and limit plasticity in adulthood for circuit stability. The mechanisms underlying these differing plasticity levels and how they serve to maintain and stabilize the properties of sensory circuits remain largely unclear. By combining a transcriptomic approach with ex vivo electrophysiology and in vivo imaging techniques, we identify that astrocytes release cellular communication network factor 1 (CCN1) to maintain synapse and circuit stability in the visual cortex. By overexpressing CCN1 in critical period astrocytes, we find that it promotes the maturation of inhibitory circuits and limits ocular dominance plasticity. Conversely, by knocking out astrocyte CCN1 in adults, binocular circuits are destabilized. These studies establish CCN1 as a novel astrocyte-secreted factor that stabilizes neuronal circuits. Moreover, they demonstrate that the composition and properties of sensory circuits require ongoing maintenance in adulthood, and that these maintenance cues are provided by astrocytes.

16.
STAR Protoc ; 4(4): 102599, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37742178

ABSTRACT

Astrocytes are glial cells of the central nervous system that modulate neuronal function. Here, we present glyoxal-fixed astrocyte nuclei transcriptomics (GFAT), a protocol for the purification and transcriptomic analysis of astrocyte nuclei from the cortex and cerebellum of adult and aged fresh mouse brain. We describe steps for tissue dissection, glyoxal fixation, homogenization, nuclei isolation, antibody staining, fluorescence-activated cell sorting, and RT-qPCR or bulk RNA sequencing. GFAT does not require transgenic lines or viral injection and allows parallel astrocyte and neuron profiling.


Subject(s)
Astrocytes , Cell Nucleus , Mice , Animals , Astrocytes/metabolism , Cell Nucleus/metabolism , Neuroglia , Gene Expression Profiling/methods , Glyoxal/metabolism
17.
bioRxiv ; 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37808668

ABSTRACT

Neuronal dendrite patterning and synapse formation are tightly regulated during development to promote proper connectivity. Astrocyte-secreted proteins act as guidance and pro-synaptogenic factors during development, but little is known about how astrocytes may contribute to neurodevelopmental disorders. Here we identify down-regulation of the astrocyte-secreted molecule pleiotrophin as a major contributor to neuronal morphological alterations in the Ts65Dn mouse model of Down Syndrome. We find overlapping deficits in neuronal dendrites, spines and intracortical synapses in Ts65Dn mutant and pleiotrophin knockout mice. By targeting pleiotrophin overexpression to astrocytes in adult Ts65Dn mutant mice in vivo , we show that pleiotrophin can rescue dendrite morphology and spine density and increase excitatory synapse number. We further demonstrate functional improvements in behavior. Our findings identify pleiotrophin as a molecule that can be used in Down Syndrome to promote proper circuit connectivity, importantly at later stages of development after typical periods of circuit refinement have completed.

18.
Sci Rep ; 12(1): 4176, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264691

ABSTRACT

Ischemic injury occurs when the brain is deprived of blood flow, preventing cells from receiving essential nutrients. The injury core is the brain region directly deprived and is surrounded by the peri-infarct area, the region with recovery potential. In the peri-infarct area neurons undergo acute loss of dendritic spines, which modifies synaptic plasticity and determines neuronal survival. Astrocytes can be protective or detrimental to the ischemic injury response depending on the specific stage, yet we lack clear understanding of the underlying mechanisms. Chordin-like 1 (Chrdl1) is an astrocyte-secreted protein that promotes synaptic maturation and limits experience-dependent plasticity in the mouse visual cortex. Given this plasticity-limiting function we asked if Chrdl1 regulates the response to ischemic injury, modelled using photothrombosis (PT). We find that Chrdl1 mRNA is upregulated in astrocytes in the peri-infarct area in both acute and sub-acute phases post-PT. To determine the impact of increased Chrdl1 on the response to PT we analyzed Chrdl1 knock-out mice. We find that absence of Chrdl1 prevents ischemia-induced spine loss in the peri-infarct area and reduces cell death in the core, without impacting gliosis. These findings highlight the important role of astrocyte-secreted proteins in regulating structural plasticity in response to brain ischemic injuries.


Subject(s)
Brain Injuries , Brain Ischemia , Animals , Astrocytes/metabolism , Brain Injuries/metabolism , Brain Ischemia/metabolism , Eye Proteins/metabolism , Glycoproteins , Infarction , Intercellular Signaling Peptides and Proteins , Ischemia/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
19.
Nat Neurosci ; 25(9): 1163-1178, 2022 09.
Article in English | MEDLINE | ID: mdl-36042312

ABSTRACT

Astrocytes negatively impact neuronal development in many models of neurodevelopmental disorders (NDs); however, how they do this, and if mechanisms are shared across disorders, is not known. In this study, we developed a cell culture system to ask how astrocyte protein secretion and gene expression change in three mouse models of genetic NDs (Rett, Fragile X and Down syndromes). ND astrocytes increase release of Igfbp2, a secreted inhibitor of insulin-like growth factor (IGF). IGF rescues neuronal deficits in many NDs, and we found that blocking Igfbp2 partially rescues inhibitory effects of Rett syndrome astrocytes, suggesting that increased astrocyte Igfbp2 contributes to decreased IGF signaling in NDs. We identified that increased BMP signaling is upstream of protein secretion changes, including Igfbp2, and blocking BMP signaling in Fragile X and Rett syndrome astrocytes reverses inhibitory effects on neurite outgrowth. This work provides a resource of astrocyte-secreted proteins in health and ND models and identifies novel targets for intervention in diverse NDs.


Subject(s)
Neurodevelopmental Disorders , Rett Syndrome , Animals , Astrocytes/metabolism , Mice , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurogenesis , Neurons/metabolism , Rett Syndrome/metabolism
20.
Neurosci Res ; 167: 17-29, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33316304

ABSTRACT

Glial cells are non-neuronal cells in the nervous system that are crucial for proper brain development and function. Three major classes of glia in the central nervous system (CNS) include astrocytes, microglia and oligodendrocytes. These cells have dynamic morphological and functional properties and constantly surveil neural activity throughout life, sculpting synaptic plasticity. Astrocytes form part of the tripartite synapse with neurons and perform many homeostatic functions essential to proper synaptic function including clearing neurotransmitter and regulating ion balance; they can modify these properties, in addition to additional mechanisms such as gliotransmitter release, to influence short- and long-term plasticity. Microglia, the resident macrophage of the CNS, monitor synaptic activity and can eliminate synapses by phagocytosis or modify synapses by release of cytokines or neurotrophic factors. Oligodendrocytes regulate speed of action potential conduction and efficiency of information exchange through the formation of myelin, having important consequences for the plasticity of neural circuits. A deeper understanding of how glia modulate synaptic and circuit plasticity will further our understanding of the ongoing changes that take place throughout life in the dynamic environment of the CNS.


Subject(s)
Neuroglia , Neuronal Plasticity , Astrocytes , Neurons , Synapses , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL