Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 1.
Published in final edited form as: Neurosci Biobehav Rev. 2022 Apr 1;137:104651. doi: 10.1016/j.neubiorev.2022.104651

Control of complex behavior by astrocytes and microglia

PI Ortinski 1,*, KJ Reissner 2,*, J Turner 3,*, TA Anderson 1, A Scimemi 4,*
PMCID: PMC9119927  NIHMSID: NIHMS1799081  PMID: 35367512

Abstract

Evidence that glial cells influence behavior has been gaining a steady foothold in scientific literature. Out of the five main subtypes of glial cells in the brain, astrocytes and microglia have received an outsized share of attention with regard to shaping a wide spectrum of behavioral phenomena and there is growing appreciation that the signals intrinsic to these cells as well as their interactions with surrounding neurons reflect behavioral history in a brain region-specific manner. Considerable regional diversity of glial cell phenotypes is beginning to be recognized and may contribute to behavioral outcomes arising from circuit-specific computations within and across discrete brain nuclei. Here, we summarize current knowledge on the impact of astrocyte and microglia activity on behavioral outcomes, with a specific focus on brain areas relevant to higher cognitive control, reward-seeking, and circadian regulation.

Glial cells: a brief context on cell diversity and behavioral relevance

The term ‘glia’ refers to a heterogeneous population of cell types. These include: oligodendrocytes, that form axon-insulating myelin sheaths, oligodendrocyte progenitors (also called NG2-cells or polydendrocytes), ventricle-lining ependymal glia, astrocytes, and microglial cells1. Glia is a contraction of the word ‘neuroglia’ (Nervenkitt) or ‘nerve-glue’, first coined by Rudolf Virchow in 1856, who referred to these cells collectively as the substance “which lies between the proper nervous parts…and gives the whole its form in a greater or less degree”2,3. A few years after this statement, the close relationship between glial cells and blood vessels was acknowledged by anatomists, but it was not until 1891 that the term ‘astrocyte’ emerged as a distinct cell type mediating this relationship. It took another 30 years before ‘oligodendrocytes’ and ‘microglia’ were recognized as additional glial cell subtypes4. Within the broad scientific opinion, the brain glial cells have remained relegated to inert, neuron-supporting roles until about the 1990’s when research into active contributions of glial cells to neuronal activity started to gain ground.

According to recent historical perspectives, oligodendrocytes constitute 45–75% of all glial cells in the human brain, followed by astrocytes (19–40%) and microglial cells (5–10%)5,6. The frequently cited neuron-to-glia ratios of 1:107 appears to be a mistaken notion that is curiously perpetuated by many textbooks. In the 60–80’s, estimates of the number of neurons and glial cells in the human brain indicated that there are 70–85 billion neurons and 40–130 billion glial cells8,9, suggesting something close to a 1:1 ratio between neurons and glial cell numbers5. Some studies in the vestibular nuclei of the brainstem did find there were ten times more glial cells than neurons, but these estimates were not backed up by experimental evidence of brain-wide counts5,1012. Modern techniques, such as isotropic fractionation, flow fractionation and stereological optical fractionation, confirm the estimate of ~1:0.7 ratio between neurons and glial cells that is surprisingly close to the one obtained a long time ago using histological/stereological approaches9,1315. Overall, the emerging picture is that the glial cell density is relatively uniform across species and brain regions, and that the glia-to-neuron ratio is simply higher whenever the size of neurons and their processes, which do vary substantially, is higher and neuronal density is correspondingly lower1618.

In the evolutionary context, number of cells and, perhaps more importantly, number of cell types, have been proposed to vary linearly with organismal complexity. Indeed, astrocytes in the human brain are substantially more varied than rodent astrocytes19,20 and some have used this evidence to bolster the argument for glial cell role in cognitive processes4. The anatomical and functional heterogeneity of glia is now beyond doubt and mounting evidence suggest substantial transcriptome diversity2123. The multiple mechanisms by which glial cells may interact with neurons remain a subject of investigation, and it is not currently known whether unique mechanisms map onto specific computations performed by neuronal circuits mediating discrete behaviors. The impact of oligodendrocytes, oligodendrocyte progenitors, and ependymal glia on complex behavior has so far attracted a relatively small share of investigative attention. In the meantime, the role of astrocytes in behavior has been actively pursued and backed by substantial mechanistic insight 24,25. Microglia contributions to behavior have also been widely recognized as part of a complex interface between neuronal activity and immune system status26. Given such skewed accumulation of evidence in support of behavioral impact, this review focuses on astrocytes and microglia only. Specifically, here we examine contributions of astrocytes and microglia signals to behaviors thought to arise from activity within three relatively well-established circuits: frontocortical circuits underlying executive function, mesocorticolimbic circuits mediating reward-seeking behaviors, and suprachiasmatic nucleus (SCN) circuits driving circadian homeostasis (Fig.1).

Figure 1.

Figure 1

An outline of the discussed mechanisms underlying glial interactions with neuronal circuitry. Astrocytes (left) and microglia (right) display cytoskeletal changes in response to their microenvironment. These changes are linked to molecular signals underlying structural remodeling and a variety of associated functional changes. Note that binary representation of astrocyte and microglia morphologies is overly simplistic and intermediate phenotypes may be possible as discussed in the text for resting and active microglia. Structural and functional adaptations (dashed arrows) influence glial interactions with neuronal networks and impact diverse neuron types (schematized as purple and green cells). Central origin of the dashed arrows indicates that transition between morphologies is not a pre-requisite for functional changes. Discrete brain regions (top) at the focus of this review, may experience distinct changes in neuroglial dynamics arising from unique combinations of underlying adaptations. FC, frontal cortex; HPC, hippocampus; dSTR, dorsal striatum (including dorsomedial and dorsolateral striatum), NAc, nucleus accumbens (ventral striatum); AMG, amygdala; VTA, ventral tegmental area; SCN, suprachiasmatic nucleus.

Regulation of activity in frontocortical circuits underlying executive function

Many studies support glial cell contributions to neuropathology of cognitive decline, brain trauma, and neurodegeneration as well as regulation of cognitive function in the healthy brain. In this section, we review the literature that examines astrocytes and microglia in the context of their impact on executive control of behavior by the frontocortical circuits, including cortico-striatal, cortico-hippocampal, and cortico-amygdalar connections (Table 1). We begin with astrocytes and highlight converging evidence that regulation of frontocortical function by these cells involves glutamate recycling, release of transmitter molecules, secretion of inflammatory mediators and other signaling factors in addition to cytoskeletal adaptations that likely impose spatial constraints on neuroglial interactions. We recognize that these aspects of astrocyte activity do not represent an exhaustive list of mechanisms by which astrocytes modulate neuronal transmission and that other functions attributed to astrocytes (e.g., water balance, K+ buffering, glucose metabolism, etc.) are likely to impact frontocortical-dependent behaviors. Astrocytes represent a regionally heterogeneous population of cells2123,27 and it is not yet clear whether any combination of factors (e.g., gene expression, cell morphology, excitability profiles, etc.) can be used to define conserved astrocyte categories regardless of their regional placement in the brain. However, current evidence does indicate unique molecular phenotypes of astrocytes between brain regions and deeper understanding of the importance and role of such phenotypes is beginning to emerge28,29.

Table 1.

The table focuses on those studies where cell-specific manipulations occurred prior to the observed behavioral effects. Studies in which cellular changes were measured after a behavioral manipulation are excluded with a few exceptions. For constitutive gene knock-out models, brain area specificity column indicates brain areas in which cellular changes were measured. Bi-directional effect column lists evidence, if any, that measured behaviors could be either suppressed or enhanced with opposite cellular manipulations.

Experimental manipulationREF# Brain area specificity Behavioral impact Cellular mechanism Bi-directional effect
Astrocytes
Cognitive flexibility L-AAA injection (astrocyte toxicity)30,31 mPFC 30 ↓cognitive flexibility(set-shifting)
31↓texture discrimination
↓reversal learning and working memory
30 ↓gamma oscillations
31 Not investigated
Not investigated
hM3 DREADD activation30 mPFC ↑cognitive flexibility (set shifting) ↑Gq signaling in astrocytes Not investigated
S100b infusion30 mPFC ↑cognitive flexibility (set shifting) Not investigated Not investigated
GFAP & vimentin knockout mice41 Whole brain ↑memory extinction Not investigated Not investigated
Nestin knockout mice43 Whole brain ↑memory extinction Not investigated Not investigated
EAAT1 knockout mice54 Whole brain ↓ visual discrimination learning ↓ astrocyte glutamate reuptake transport Not investigated
Blockade of EAAT2 by dihydrokainate55 mPFC ↑ drinking latency & ↓ responding to PFC electrical self-stimulation55 ↓ astrocyte glutamate reuptake transport55 Not investigated
Mice IP injections of sub-chronic ketamine56 Hippocampus ↓ extinction of operant sucrose self-administration ↓ EAAT2 expression Not investigated
dnSNARE knockout mice65 PFC, hippocampus ↓ working and spatial memory ↓ theta-cycle synchronization between hippocampus and PFC Theta coherence and cognitive deficits rescued by d-serine supplementation
Deletion of astrocyte GABAB receptors in mice74 PFC Aberrant cortical synchronization and ↓ T-maze alternation performance astrocytic GABAB signaling activates mGluRs and recruits PV interneurons Not investigated
Urokinase plasminogen activator receptor knockout mice81 orbitofrontal cortex, dorsal striatum ↓ reversal learning ↓ PV interneurons in orbitofrontal cortex and dorsal striatum Reversal learning deficit rescued by up-regulation of hepatocyte growth factor in astrocytes
Human Chrdl1 mutations83 Retina, PFC ↑ executive function Corneal abnormalities and myelination deficits Not investigated
Overexpression of MHC1 in mice mPFC astrocytes88 mPFC ↓ reward-based visual discrimination ↓ neuronal spine density in dorsal striatum Not investigated
Chronic expression of IL-6 in transgenic mice89 PFC, amygdala ↓ avoidance learning Loss of synapses and calbindin-containing neurons due to chronic neuroinflammation Not investigated
Reward seeking Stimulation of hM3D receptors on astrocytes146 PFC ↑ ethanol drinking Activation of Gq-coupled signaling on astrocytes Not investigated
Stimulation of hM3D receptors on astrocytes147,148 NAc ↓ cocaine reinstatement and motivation to self-administer ethanol147,148 Stimulation of presynaptic mGluRs before hM3D stimulation147 Not investigated
Optogenetic stimulation of VTA astrocytes149 VTA ↑ avoidance behavior, overriding CPP for cocaine Astrocytes stimulation VTA GABA interneurons, inhibiting dopamine neurons Glutamate transporter (GLT-1) from VTA astrocytes blocks avoidance behavior and maintains CPP for cocaine
Optogenetic activation of hippocampal astrocytes151 Hippocampus ↓ consolidation of contextual fear memory Release of ATP and adenosine Pharmacological stimulation of A1 receptors elicited similar effects
hM3D activation of astrocytes167 Dorsal medial striatum Shifted behavior from habitual to goal-oriented hM3D activation reduced sEPSC frequency in D1 MSNs, but increased sEPSC amplitude in D2 MSNs The effect was not observed in adenosine transporter (ENT1) knockout mice
Training-induced upregulation of EAAT2/GLT-1171 Dorsal lateral striatum ↑ habit behavior in operant task for chocolate reward Astrocytes reinforce relative contributions of the DMS and DLS directed behaviors Inhibition of EAAT2/GLT-1 upregulation promoted goal-directed behavior
Overexpression of the plasma membrane Ca2+ pump in astrocytes173 Dorsal lateral striatum ↑ excessive and compulsive-like self-grooming behavior Removal of astrocyte Ca2+ signaling Not investigated
Circadian rhythms and sleep/wake cycle Ca2+ imaging of astrocytes in naïve animals270,271,272 Frontal cortex270,272, hippocampus, hypothalamus, pons, cerebellum, barrel cortex270,271 Ca2+ levels increase during wakefulness and decrease during REM sleep in several brain areas270,271,272 Not investigated Not investigated
Genetic reprogramming of SCN astrocyte clock genes220 SCN Genetic reprogramming of astrocyte clock genes reshapes circadian behavior Not investigated Not investigated
Genetic reprogramming of SCN neurons to have incompetent clock gene expression221 SCN Astrocytes reinstate clock gene expression and circadian function of SCN neurons autonomously Astrocyte glutamatergic signaling in the SCN Not investigated
Experimental manipulationREF# Brain area specificity Behavioral impact Cellular mechanism Bi-directional effect
Microglia
Cognitive flexibility Chronic unpredictable stressors in mice117,120 mPFC ↓ PFC-dependent temporal object recognition117 ↑ CSF1 receptor and somplement component mRNA117,120 Both mRNA and behavior rescued by treatment with RU486 (glucocorticoid antagonist). 117
7-day restraint stress119 dmPFC ↓ reversal learning in 4-choice odor discrimination ↑ dmPFC dendritic spine elimination Not investigated
Blockade of adenosine2A receptor in prenatal dexamethasone model for anxiety123,124 Hippocampus, PFC ↑ cognitive performance in recognition memory task124 Normalized frontocortical123 and hippocampal124 microglia morphology Not investigated
Adolescent social stress model in mice124 PFC ↓ cognitive flexibility performance Activated microglia release TNFα Cognitive flexibility deficits were rescued by increased TNFα release by ranylcypromine
Radioligand TSPO distribution PET scan125,126,127,128 PFC ↓ attention125
↓ memory and executive function126
↑ TSPO radioligand binding due to increased microglia activation125,126,127,128 Not investigated
Cognitive decline127,128
Reward seeking Inhibiting microglia with minocycline in alcohol-dependent mice188,189 Not investigated (IP injection) ↓ withdrawal-induced anxiety188,189 Not investigated Not investigated
Depletion of microglia with PLX-5622190 Whole brain Does not change escalation of voluntary alcohol consumption Not investigated PLX-5622 does block escalation under conditions of repeated immune activation
Depletion of microglia with PLX-5622191 Whole brain ↓ anxiety during alcohol withdrawal ↓ escalation of alcohol intake Normalized excitatory and inhibitory synaptic plasticity in central nucleus of the amygdala Not investigated
Depletion of microglia with PLX-5622192 Whole brain Normalized performance on marble burying and open field tests after nicotine withdrawal Elevation of NOX2 release from microglia Not investigated
Blockade of TLR4 signaling with LPS-RS or TLR4 knockout mice193 Whole brain ↓ CPP and self-administration of cocaine Inhibiting TLR4 ↓ extracellular dopamine from cocaine in the NAc by preventing downstream release of IL-1β Not investigated
Minocycline treatment194,195 Whole brain ↓ both maintenance and reinstatement of methamphetamine-induced CPP Minocycline ↓ extracellular dopamine from methamphetamine in the NAc Not investigated
Circadian rhythms and sleep/wake cycle Chronic sleep fragmentation model in wild-type mice281 PFC, hippocampus ↓ spatial learning and memory
↑ aggression and anxiety
↑ intracellular amyloid-β, dysfunction of endosome-autophagosome-lysosome pathway Not investigated
Depleting microglia with Cx3cr1 transgenic mice282 SCN, hippocampus Abnormal circadian body temperature and diurnal rhythms Disrupted expression of clock genes Per1, Per2, and Bmal1 Not investigated
Ablation of microglia following PLX-5622 treatment in mice283 Whole brain, Hippocampus ↑ sleep duration ↓ in light phase-dependent excitatory synaptic transmission in CA1 pyramidal neurons Not investigated
Sleep deprivation in rats288 Whole brain, hippocampus ↑ memory impairments
↓ performance in Morris water maze
Sleep deprivation increases activated microglia activity and minocycline blocks activation of resting microglia Memory and Morris water maze impairments were rescued by minocycline
Minocycline treatment in human290 and rodents291,292 Whole brain Both ↑290 and ↓291,292 sleep quality and episodic memory minocycline blocks activation of resting microglia288,290,291,292 Not investigated

Astrocyte lesions alter cognitive flexibility –

Several studies have investigated the impact of lesioning astrocytes in the prefrontal cortex (PFC) on relevant behavioral outcomes. In recent work, bilateral administration of the astrocyte-specific toxin, L-AAA, into the medial PFC (mPFC) impaired cognitive flexibility in an attentional set-shifting task and reduced the power of gamma oscillations in the mPFC without any effects on neuron morphology30. In contrast, stimulation of astrocyte activity via Gq receptor coupled DREADDs as well as via mPFC infusions of the astrocyte-secreted calcium-binding protein S100β, in the non-L-AAA lesioned groups, improved cognitive performance30. These observations are consistent with previous work that identified deficits in the ability to discriminate between texture-based, but not odor-based, stimuli, as well as impairments in odor-based reversal learning and working memory after infusions of L-AAA into the mPFC31. The L-AAA lesions of the PFC have also been reported to impair sucrose preference and novelty-suppressed feeding whereas excitotoxic lesioning of neurons by ibotenate did not impact performance on these tasks32,33. Consistent with the postulate of a close-knit relationship between local astrocyte function and neuron structure, substantial reductions in dendritic complexity of mPFC neurons have been observed after L-AAA administration30. In the pre-limbic cortex, L-AAA triggered swelling of neuronal cell bodies, but did not result in measurable neuronal cell death32. The ability of astrocytes to facilitate behavioral dysfunction without an outright neuronal loss has particular relevance to pathophysiology of drug seeking behaviors and depression, conditions characterized more by synaptic reorganization and plasticity rather than extensive neuronal degeneration34,35. However, results of L-AAA lesioning studies should be interpreted with caution given evidence that L-AAA inhibits glutamate uptake and glutamate metabolism leading to elevated extracellular glutamate levels36. An additional interpretational caveat of L-AAA studies has to do with findings that L-AAA increases microglial infiltration into the lesioned area37 which has clear implications for attribution of the behavioral effects of L-AAA exclusively to astrocytes.

The role of astrocyte cytoskeleton –

Several provocative reports have been published to examine the impact of astrocyte cytoskeletal proteins on behavior. GFAP and vimentin are astrocyte intermediate filament proteins often used for immunohistochemical identification of astrocytes, but also for examination of gross changes in astrocyte morphology. These molecules have been extensively used as markers of reactive gliosis, a phenomenon in which up-regulation of GFAP and vimentin is accompanied by morphological and a wide array of molecular changes38. Nestin is another intermediate filament protein that is normally expressed in progenitor cells during development, but is also re-expressed in mature reactive astrocytes, following brain trauma39,40. With regard to frontocortical function, one investigation found that transgenic mice null for GFAP and vimentin displayed improved extinction of memories linked to location of a food container in a reversal task41. While acquisition of place memories relies heavily on the hippocampus, spatial reversal learning implies suppression of existing information and requires additional processing in prefrontal areas42. GFAP/vimentin null mice maintained surprisingly normal performance on initial acquisition of hippocampus-dependent place memories, highlighting the specific impact on frontocortical processing41. Interestingly, mice with constitutive deletion of nestin showed a similar pattern of results: normal memory acquisition, but better extinction of acquired place memories43. Although behavioral specificity of GFAP deletion to reversal learning is intriguing, its absence since embryonic stage may have also impacted adult neuron populations or neural circuit interactions since GFAP is expressed in neural progenitor cells. Indeed, there are suggestions that altered cognitive function may be associated with highly local, cell-layer specific regulation of astrocyte number and morphology as found in a post-mortem analysis of GFAP expression in brains of schizophrenia patients44. Pre-clinical studies also support the idea that astrocyte interactions with neurons may be targeted to specific neuronal populations45. Overall, a vast number of reports outside the scope of this review has relied on intermediate filament expression, particularly GFAP, to evaluate astrocyte numbers and morphology throughout the brain. The most parsimonious conclusion from this literature is that astrocyte number is bidirectionally sensitive to and can be correlated with many behavioral antecedents, sequelae, or disease pathologies. However, since GFAP staining does not label fine astrocytic processes that are most likely to contact synapses, research using other markers to evaluate astrocyte plasticity is sorely needede.g..46 as neither astrocyte numbers on their own nor GFAP-based gross morphology reports are likely to provide mechanistic insight into behavioral relevance of these cells.

Astrocytic control of glutamate recycling and GABA synthesis –

Astrocytes recycle over 90% of extracellular glutamate47, with an impact not only on neuronal supply of glutamate, but also on availability of glutamate for GABA synthesis48. Among the four major subtypes of glutamate reuptake transporters, two – EAAT1 (GLAST in rodent) and EAAT2 (GLT-1 in rodent) – are abundantly expressed in astrocytes of the CNS. Expression of both EAAT1 and EAAT2 is strongly decreased in schizophrenia and substance use49,50, disorders known to prominently involve frontocortical activity. A decrease in astrocyte number and EAAT2 expression is also observed in Alzheimer’s disease (AD), in humans and in mouse models, where this deficit has been associated with slower clearance of extracellular glutamate51,52. Interestingly, overexpression of astrocytic glutamate transport may be beneficial for cognitive function, as suggested, for example, by a post-mortem analysis of entorhinal cortex of AD patients, where overexpression of EAAT2 was found to correlate with the absence of dementia or mild cognitive impairment53. A deficit in visual discrimination learning was reported in mice with constitutive deletion of EAAT154, suggesting impairment of executive function, although the learning deficit prevented a more direct evaluation of frontocortical control in a reversal task54. Reminiscent of the effects of astrocyte lesioning in L-AAA studies, blockade of EAAT2-dependent glutamate uptake by dihydrokainate increased drinking latency in a sucrose preference task and led to near-complete cessation of responding for electrical stimulation of the PFC in an intracranial self-stimulation paradigm55. In another study, reduced expression of EAAT2 in the mouse hippocampus was observed after treatment with sub-chronic ketamine which was associated with impaired extinction of an operant sucrose self-administration task56. These findings were speculated to reflect cognitive deficits characteristic of substance use and resemble data from the PFC of schizophrenic patients49. However, extinction of self-administration is not a widely used behavioral indicator of cognitive impairment, and future studies will be needed to more closely examine the impact of astrocytic EAAT2 on frontocortical control of behavior. In the meantime, repeated observations that disrupted glutamate uptake strongly impacts activity of NMDA receptors throughout the brain5759, is likely to have implications for astrocyte control of neuronal output as discussed in the next section.

Transmitter release by astrocytes –

A series of seminal studies used a mouse model with astrocyte-specific deletion of the SNARE protein (dnSNARE mice) that was argued to limit transmitter release60. Although the specificity of GFAP-driven dnSNARE transgene to astrocytes has been challenged61, this work formed the foundation for the view that astrocytes may promote synchronous neuronal activity and are instrumental for forms of long-term neuronal plasticity62. Later studies provided support for this hypothesis by demonstrating that astrocytes regulate sleep patterns and cortical oscillations with implications for cognitive performance63,64. The dnSNARE mice have also been recently used to demonstrate impaired theta-cycle synchronization between the hippocampus and the PFC accompanied by deficits in working and spatial memory, but intact neuronal morphology in both the PFC and the hippocampus65. Both the theta coherence and the cognitive deficits could be rescued by supplementation with d-serine65. D-serine acts as a co-agonist of NMDA receptors at the glycine site and d-serine release from astrocytes has been argued to impact synaptic long-term potentiation in the hippocampus66. Additionally, astrocyte-released ATP and its metabolic product, adenosine, may also influence NMDA receptor expression and signaling via activation of purinergic receptors67. The interaction between ATP/adenosine receptors and neuronal signals in the PFC is supported by experiments in slices indicating that astrocytic P2Y4 receptors are linked to vesicular release of glutamate and activation of neuronal metabotropic glutamate receptors (mGluRs) and NMDA receptors68. Altogether, there is substantial support for the view that NMDA receptors represent a critical component of astrocyte interaction with neurons. Particular attention must be paid to a potentially unique contribution of NMDA receptors located at extrasynaptic sites which have been proposed as primary mediators of neuronal response to astrocyte-released glutamate6972.

In addition to glutamate-mediated signaling, astrocytes also release GABA with an impact on inhibitory neurotransmission73. The specific effects of astrocyte-released GABA on behaviorally relevant frontocortical activity remain to be determined. However, a recent publication demonstrated that selective deletion of GABAB receptors in astrocytes of the mouse PFC resulted in aberrant cortical synchronization and deficits in a T-maze alternation task74. The study proposed that astrocytic GABAB signaling facilitates activation of group 1 mGluRs and recruitment of parvalbumin-expressing interneurons, suggesting a link between GABAB receptor activation and glutamate release from astrocytes. An interaction between astrocytes and parvalbumin positive interneurons has also been reported in the visual cortex, with an effect on orientation to visual stimuli75.

We would like to conclude this section by acknowledging an on-going debate over astrocytes ability to release transmitter molecules through vesicular (i.e. SNARE-mediated) or non-vesicular release mechanisms all grouped under the general definition of “gliotransmission”. The two most prominent aspects of this argument are an unresolved question over the role of astrocyte Ca2+ as mediator of gliotransmitter release and the degree to which ex vivo experimental conditions demonstrating astrocyte-mediated release reflect in vivo physiology of these cells76. The contentious nature of the debate might arise from the fact that mechanisms of transmitter release from astrocytes are potentially more diverse and certainly much less understood than in neurons76,77 and evaluations of astrocyte Ca2+ may require methods distinct from those applied to neuronal Ca2+ transients7880. As a consequence, trying to ‘squeeze’ gliotransmission into the framework of extensively characterized neuronal release mechanisms may not provide an accurate depiction of transmitter release from astrocytes. Similarly, any answers as to whether astrocyte contributions to neuronal activity align with intact physiology must await further experimentation, given that the overwhelming majority of current knowledge has been generated using astrocyte cultures or slice preparations.

Other astrocyte-secreted factors –

Astrocytes release a range of neuromodulators that interact with the surrounding neurons and impact behavioral measures of cognition. For example, Plaur transgenic mice, that lack urokinase plasminogen activator (uPA) receptors and show a specific loss of parvalbumin positive interneurons in the orbitofrontal cortex and dorsal striatum, display a pronounced deficit in performance on a reversal learning task81. This deficit, which does not generalize to a broad learning impairment, can be rescued by up-regulation of the hepatocyte growth factor in GFAP-positive astrocytes81. Loss of astrocyte reactivity could play a role in these behavioral outcomes since up-regulation of astrocyte uPA receptors in response to uPA secretion from neurons has been reported to mediate astrocyte activation, as measured by increased GFAP expression, after ischemic injury82. Another interesting secreted protein is chordin-like 1 (Chrdl1). Chrdl1 acts an antagonist of bone morphogenetic protein, which is critical in CNS development. A human study linked mutations in the Chrdl1 gene to development of corneal abnormalities and myelination deficits, but strikingly, superior performance on tests of executive function83. In mice, secretion of Chrdl1 from astrocytes is essential for increased developmental expression of Ca2+-impermeable, GluA2 subunit-containing AMPA receptors that impede synaptic plasticity84. Regulated expression of Ca2+-permeable versus Ca2+-impermeable AMPA receptors has been proposed as one of key characteristics of neuroplasticity within the mesolimbic dopamine reward system associated with cocaine use85. Related to this, up-regulation of Chrdl1 RNA was demonstrated in cultured astrocytes treated with dopamine86.

A vast literature supports the notion that astrocytic release of pro- and anti-inflammatory molecules as well as astrocytic response to these molecules has consequences for frontocortical signaling87. For example, astrocytic overexpression in the mPFC of major histocompatibility complex 1 (MHC1), which is involved in adaptive immune responses, led to decreased neuronal spine density in the dorsal striatum and deficient performance in a reward-based visual discrimination task, despite normal performance in a reversal learning task88. In another study, mice that constitutively expressed IL-6 in astrocytes showed deficits in avoidance learning that are thought to rely on PFC interactions with the amygdala89. The inflammatory role of astrocytes has been a prominent area of research on neurodegenerative disease with pathologies that span the gamut of behavioral impairments from memory deficits to dysregulation of executive function to depression, anxiety, etc. Numerous astrocyte-specific targets have been identified across neurodegenerative conditions including Parkinson’s disease90, Huntington’s disease91, AD92, multiple sclerosis93, stroke94, traumatic brain injury (TBI)95 as well as normal aging96,97. Since it appears unlikely that any one of the behavioral measures in these conditions can be attributed to a single astrocyte-specific factor, a deeper understanding of how combinations of secreted factors impact astrocyte interactions with local neurons and long-range signals between brain regions would be extremely valuable.

Microglial contributions to executive function –

Microglia-mediated inflammation has been observed to contribute to frontal cortex dependent behaviors across a range of pathologies, including AD, schizophrenia, TBI, aging, PTSD, depression, neurodegeneration as well as diet-induced inflammation98100. Similar to astrocytes, microglial morphology is highly sensitive to a wide range of physiological and pathological stimuli and many studies rely on morphology-based classification of activated microglia as evidence of altered microglial signaling. Indeed, physical appearance of the microglia can be indicative of the neuro-environment101. For example, resting microglia have long, thin, highly branched processes with a small cell body, and they are primarily in charge of querying the neuronal environment for perturbations to ensure effective communication between neurons. In line with their ‘surveillance’ role, these cells are regularly tiled throughout the brain to ensure local oversight by at least one microglial cell. However, when microglia detect a potential neuro-insult, these cells are galvanized into an active state, characterized by retraction of their processes and acquisition of a more amoeboid phenotype. Because these cells share a common lineage to macrophages in the peripheral immune system, microglial activation has been similarly described as falling under M1 (pro-inflammatory and expressing markers such as CD86, iNOS, CD16/32, and MHCII) or M2 (anti-inflammatory and expressing markers such as CD206, FIZZ1, and Arginase I) phenotypes. More recently, however application of M1 and M2 descriptors to microglia has been called into question based on growing support for existence of multiple intermediate and combinative phenotypes102.

There is substantial evidence that administration of minocycline, a tetracycline antibiotic that disrupts protein synthesis and prevents microglial activation elicits mild-to-substantial cognitive improvement across multiple domains including cognitive symptoms of bipolar disorder103, HIV104, addiction105, depression106 and psychotic disorders 107,108. Pro-cognitive effects of microglia depletion have also been observed after cranial irradiation, often used as a cancer therapy109. Reducing microglial population in mice by treatment with the colony stimulating factor (CSF)-1 receptor inhibitor, PLX-5622, has been reported to rescue irradiation-induced behavioral deficits on an ‘object in place’ and contextual fear-conditioning tasks both of which involve frontocortical signaling109. Interestingly, a recent study reported no effect of minocycline on cognitive decline associated with mild AD in human subjects110, although cognitive benefits have been reported in animal models111. To reconcile such disparate outcomes, some researchers advocate for use of minocycline in combination with other drugs as a strategy to combat cognitive impairment in AD patients112,113. It must be noted, however, that similarly to astrocyte depletion with L-AAA, microglia depletion with PLX-5622 or other CSF-1 inhibitors may have off-target effects that involve other cell types114.

Consistent with the body of knowledge that stress is both associated with cognitive deficits and promotes systemic inflammation, stress-activated microglia have also been found to facilitate cognitive impairment. Dysregulated stress responsivity within the PFC is thought to be one of core factors leading to emergence of depression and associated cognitive deficits115,116. For example, a regime of chronic unpredictable stressors led to elevation of CSF1 receptor and complement components C1q and C3 mRNA in the PFC and behavioral deficits in a PFC-dependent temporal object recognition task117. In this study, both the mRNA and the behavioral changes could be rescued by treatment with the glucocorticoid receptor antagonist, RU486. Glucocorticoid receptors are known to be expressed by a variety of cell types in the brain including neurons and RU486 treatment was noted to reduce stress-associated morphological remodeling of the PFC microglia as well as to prevent reduction of dendritic spine density in PFC layer V pyramidal cells117. These results complement other findings that stress-induced cognitive deficits trigger ramification of microglial processes and increase microglia-neuron contacts in the PFC leading to dendritic spine elimination118120. One molecular mechanism that links stress-induced PFC dysfunction with microglial activation has been proposed to involve ATP-gated P2X7 receptor signaling and the downstream release of PGE2 and IL-1β121. Extracellular ATP is generally subject to quick hydrolysis to adenosine, predicting a role for adenosine receptors in microglial response to stress. Indeed, blockade of adenosine A2A receptors has been shown to normalize frontocortical microglia morphology in the prenatal dexamethasone model of anxiety in male, but not female rats122. Using the same model, normalization of microglial morphology by A2A antagonism was observed in the hippocampus of female rats which was further linked to increased synchronization of hippocampal-PFC network and improved cognitive performance123.

Some evidence suggests that release of TNFα from activated microglia improves cognitive flexibility in an adolescent social stress model and that administration of the monoamine oxidase inhibitor, antidepressant, ranylcypromine, increases the number of microglia and TNFα release in the PFC124, a finding that suggests microglial activation may rescue, rather than promote, PFC-dependent cognitive impairment in some models and behavioral assays. Notwithstanding such evidence, human studies support deleterious effects of activated microglia on cognitive performance. This work has been greatly facilitated by the availability of radioligands to translocator protein (TSPO), a marker of activated microglia utilized in MRI and PET studies. For example, elevated TSPO distribution in the frontal cortex has been associated with lower attention scores in non-medicated patients with a major depressive disorder125. Similarly, in HIV patients, increased TSPO radioligand binding in the frontal cortex was linked to lower performance on tests of memory and executive function126. Increased microglial activation reported by TSPO binding has also been reported to predict cognitive decline in a model of experimentally-induced meningitis in rats127 and in a population of pre-manifest Huntington’s disease patients128. Although a specific role of PFC microglia was not investigated in these studies, they suggest that microglial activation can serve as a useful predictor of clinically manifesting symptoms of cognitive decline.

Modulation of reward-seeking behaviors

Regional heterogeneity of astrocyte responses to drugs of abuse –

The role of astrocytes may vary within distinct nodes of the reward circuit, including the PFC, caudate/putamen, nucleus accumbens (NAc), hippocampus, amygdala, and ventral tegmental area (VTA) as most prominent components. Indeed, while our understanding of astrocytes is still in its infancy, accumulating evidence indicates variability exists across brain regions with respect to the basal functional properties and engagement in pathology of these cells28,129. While a direct comparison has not been made between cortical and striatal astrocytes, several lines of evidence indicate differential features and expression profiles between striatal and hippocampal astrocytes, supporting other evidence for regional variability of both structure and function21,28,130133. Accordingly, it is not unreasonable to speculate that effects of drug exposure may have dissociable effects within the reward circuitry, and that astrocytes within these regions may differentially influence behavioral responses to drugs.

Early reports reflecting effects of drug self-administration on astrocytes indicated that astrocyte-enriched mediators of glutamate homeostasis, in particular system xC- and EAAT2/GLT-1, are chronically downregulated in the NAc134137. It was subsequently revealed that self-administration and extinction of cocaine-seeking led to downregulation of GFAP expression, as well as astrocyte surface area, volume, and colocalization of astrocyte membrane with synapses in the NAc138,139. This decrease in synaptic colocalization has since also been observed following self-administration and extinction from methamphetamine and heroin140,141. The effect of cocaine on astrocytes in the NAc was, in contrast, not observed to extend to the prelimbic PFC or the basolateral nucleus of the amygdala, suggesting regional susceptibility of astrocytes in this model139. However, decreased expression of EAAT1/GLAST and S100β-positive astrocytes has been observed in the SN/VTA following chronic non-contingent delivery of cocaine or methamphetamine142. Ethanol consumption in rats exhibits a time-dependent morphological effect on astrocytes in the mPFC, characterized by initial upregulation of GFAP followed by a decrease at 3 weeks of abstinence143,144. Likewise, adolescent intermittent ethanol administration results in decreased synaptic colocalization of dorsal hippocampal astrocytes in adulthood145. While many reports have found overall increases in GFAP expression and structural features of astrocytes following ethanol exposure, the general theme of reduced gene expression and structural features following post-administration abstinence has been observed across operant drug paradigms for review, see: 144.

How then do astrocytes affect drug-related behaviors? One prominent line of evidence comes from studies utilizing astrocyte-specific expression of Gq-coupled DREADD (hM3D) constructs. Stimulation of hM3D receptors within the murine PFC promotes ethanol drinking in ethanol-naïve mice in an intermittent access paradigm with free availability of ethanol every other day146. However, different results are observed in other brain regions. When expressed in the NAc astrocytes, hM3D stimulation opposes cocaine reinstatement and motivation to self-administer ethanol, respectively147,148. In the case of cocaine reinstatement, stimulation of presynaptic inhibitory mGluRs subsequent to hM3D stimulation was shown to be responsible for the observed behavioral effect147. It is unclear whether these divergent results of DREADDs in the mPFC and NAc reflect heterogeneity of astrocyte signaling, or are a function of the stimulation in naïve versus withdrawn, abstinent animals. The parallel between distinct effects on astrocyte morphology and behavioral drug-related output, however, is noteworthy. Moreover, results from the VTA indicate that astrocyte regulation of avoidance behavior is dependent on local circuitry and stimulation of GABAergic neurons149. Specifically, optogenetic stimulation of VTA astrocytes promotes avoidance behavior which overrides conditioned place preference for cocaine149. Ventral midbrain astrocytes exhibit distinct gene expression and physiological properties which may contribute to the microcircuit-level outcomes27.

Regional heterogeneity of astrocyte modulation of drug-related behaviors –

What is the mechanism by which striatal astrocytes could oppose drug-related, motivated behaviors? As addressed above, one possibility is stimulation of presynaptic, Gα and Gi/o-coupled metabotropic glutamate receptors147. In addition, considerable evidence indicates that astrocytes can oppose glutamatergic synaptic transmission via release of ATP/adenosine, acting also on presynaptic inhibitory adenosine A1 receptors150. For example, optogenetic activation of astrocytes in the hippocampus inhibited consolidation of contextual fear memory via release of ATP and adenosine and pharmacological stimulation of A1 receptors elicited similar effects151. In a similar fashion, glutamatergic synapses in the central nucleus of the amygdala can be inhibited by astrocytes via adenosine A1 receptor stimulation, while inhibitory synapses are activated via stimulation of adenosine A2A receptors152. Perhaps most germane, Corkrum et al.153 recently showed that Ca2+ responses are generated in NAc astrocytes subsequent to dopamine release and amphetamine exposure, resulting in ATP/adenosine release and reduced AMPA receptor-mediated synaptic activity. This effect may be facilitated by increased glutamate-mediated neuroglial coupling in the NAc observed after cocaine self-administration72, although multiple other regulators of AMPA signalinge.g.154156 could certainly be involved. Independent of drug exposure, Down Syndrome patient-derived induced pluripotent stem cells exhibit increased frequency of spontaneous calcium fluctuations, which inhibit co-cultured neurons via an adenosine-dependent mechanism157. This is in keeping with a rich literature on varied effects of astrocytes on potentiation of inhibition of synaptic transmission dependent on subtype of adenosine or glutamate receptors60,158160. Thus, while significant evidence indicates that astrocytes can promote and support synaptic transmission158,161 there is also considerable support for the hypothesis that astrocytes may oppose excitatory drive within the reward circuitry, and relatedly oppose behavioral output associated with drug craving and seeking162.

Striatal astrocytes in habit and compulsive behaviors –

Striatal astrocytes influence behaviors engaged by drugs beyond seeking. For example, both habit-directed and compulsive behaviors are associated with protracted drug seeking163. The transition from recreational drug use to more regulated use and relapse is associated with a shift in cellular activity from the ventral to dorsal striatum164. Relatedly, addiction is also associated with a shift from goal-directed, to more habit-based behaviors that are proposed to rely on recruitment of dorsomedial (DMS) and dorsolateral striatum (DLS), respectively165,166.

Recent studies have indicated supportive roles for astrocytes in these processes. For example, chemogenetic hM3D activation of astrocytes in the DMS reduced the frequency of spontaneous neuronal excitatory post-synaptic currents (EPSCs) in D1-receptor expressing medium spiny neurons (MSNs), but increased EPSC amplitude in D2-receptor positive MSNs and shifted behavior from habitual to goal-directed167. The effect of astrocyte Gq stimulation on promoting goal-directed behaviors was dependent on adenosine signaling, as the effect was not observed in adenosine transporter (ENT1) deficient mice. Considerable research has indicated both structural and functional interactions between A1-D1 and A2A-D2 receptors168170, underscoring the critical nature of adenosine in the regulation of synaptic and neuronal activity by astrocytes, and providing a mechanism whereby astrocytes may differentially affect different neuronal subtypes within the striatum. Precedent for specific interactions between astrocytes and neuronal subpopulations in the striatum has previously been reported45. In contrast to these findings subsequent to astrocyte hM3D stimulation in the DMS, training-induced upregulation of EAAT2/GLT-1 in the DLS resulted in strengthened habit behavior in an operant task for chocolate reinforcers, and inhibition of EAAT2/GLT-1 upregulation promoted goal-directed behavior171. Collectively, these studies in the dorsal striatum suggest that astrocytes may reinforce the relative contributions of the DMS and DLS to habit and goal-directed behaviors, respectively.

Substance use disorders are characterized by three stages: 1) binge and intoxication, 2) withdrawal and negative affect, and 3) preoccupation and anticipation172. The third stage is associated with impaired PFC-dependent regulation of behavior and compulsive drug seeking. Accordingly, it is of merit to understand how astrocytes may contribute to compulsive-like behaviors. Removal of astrocyte Ca2+ signaling in the DLS via expression of the plasma membrane Ca2+ pump resulted in excessive and compulsive-like self-grooming behavior associated with increased GABA transporter, GAT-3, expression in astrocytes and impaired tonic inhibition of medium spiny neurons173. These findings suggest that astrocytes may suppress dopamine release from axon terminals since compulsions manifesting as motor stereotypies are often observed in conditions characterized by excessive dopamine. Importantly, these results also highlight the ability of striatal astrocytes to modulate inhibitory as well as excitatory neuronal signaling echoing the findings in the frontal cortex.

Microglia and reward-seeking –

Microglia interact extensively with the dopamine system174 and show sensitivity to drugs across a spectrum of substance use disorder models. In alcohol use disorder, ethanol has been found to increase the number of microglia in the hippocampus and the PFC in the absence of astrocyte activation175. However, another study has found that although ethanol does induce microglial activation, microglial numbers in the hippocampus decrease after a “binge” pattern of exposure, finding that was supported by evidence of microglial dystrophy176. Microglial activation has also been shown after exposure to psychostimulant drugs, including amphetamine177, methamphetamine178 and cocaine179183. Interestingly, methamphetamine-induced activation of microglia could be prevented by maintaining animals at reduced temperature (10–12°C), whereas elevated ambient temperature alone (38–40°C) did not result in microglial activation178. Additionally, a PET study of human cocaine users using TSPO radioligands found no evidence of activated microglia184. These somewhat discrepant findings could be reconciled by acknowledging that there is not a binary distinction between activated and resting microglia. For example, while there is converging support for the idea of microglial activation by ethanol using morphological analyses185, binge ethanol exposure may result in increase of some, but not other markers of immunoreactive microglia186. Such observations of partial microglial activation call for a more nuanced classification scheme. Indeed, the need for discriminating between multiple microglia phenotypes has been increasingly recognized and particularly aided by the emergence of rapid and cost-effective gene sequencing techniques187.

In addition to morphological changes and expression of inflammatory markers following drug use, disrupting microglia has also been shown to have an effect on subsequent behavioral read-outs of drug exposure. Alcohol consumption and withdrawal-induced anxiety are both attenuated by inhibiting microglial activation with minocycline188,189. Depletion of microglia with PLX-5622 does not change escalation of voluntary alcohol consumption, but does block escalation of intake under conditions of repeated immune activation190. Another study has found that PLX-5622 normalized both excitatory and inhibitory synapse plasticity as well as prevented anxiety during alcohol withdrawal and reduced escalation of alcohol intake191. Consistent with this, microglial depletion normalized behavioral performance on marble burying and open field tests (measures of anxiety) after withdrawal from nicotine, an effect associated with elevation of NOX2, a source of reactive oxygen species expressed predominantly in the microglia192. Inhibition of microglia has also been generally reported to attenuate behavioral effects of psychostimulants. Thus, preventing microglia activation by blockade of toll like receptor 4 (TLR4) signaling and the downstream release of IL-1β attenuated conditioned place preference and self-administration of cocaine193. Minocycline treatment has also been reported to attenuate both maintenance and reinstatement of methamphetamine-induced conditioned place preference194,195 as well as subjective rewarding effects of dextroamphetamine in human subjects196. Clinical studies generally support the idea that microglial inhibition suppresses behavioral effects of commonly abused drugs197, although some results argue against this198. In preclinical work, locomotor sensitization to cocaine appears to break the pattern with findings that sensitization is suppressed by activated rather than inhibited microglia via release of TNFα183 and findings that microglial depletion has no effect on cocaine sensitization199. It is possible that microglial control of substance-use related behaviors is proportional to the level of stress response that accompanies experimental manipulations. This has been suggested in a recent review200 and is supported by findings that immune activation is required for microglial control of drug-related behavior190.

Astrocyte control of circadian rhythmicity

In mammals, a specific nucleus of the hypothalamus, the SCN, acts as the master circadian pacemaker. Its function is required to drive daily rhythms in behavior, and lesions of the SCN abolish rhythms in mammals. Accordingly, in hamsters, transplanting an explant of the SCN into an SCN-lesioned animal restores circadian rhythms201. Of note, these rhythms have the same period as in the donor, indicating that they arise from cell-autonomous mechanisms present within the SCN201. This form of cell-autonomous rhythmicity is generated by two interlocking transcription/translation feedback loops driven by four clock proteins: the activators CLOCK and BMAL1 and the repressors PER and CRY. CLOCK and BMAL1 activate the transcription of the Per and Cry genes. The proteins PER and CRY inhibit the transcriptional activation of CLOCK and BMAL1. As the proteins PER and CRY are degraded through ubiquitination, the repression of CLOCK and BMAL1 is relieved and the cycle begins again202,203. The period and phase with which these reactions take place can be modified and reset or entrained by cycles of light and darkness in the environment. Light contributes to reset circadian clocks through the activation of retino-hypothalamic (RHT) glutamatergic inputs to the SCN that, according to studies of changes in the expression of the immediate early gene c-Fos, can activate both neurons and astrocytes204,205.

The components of the molecular machinery regulating clock gene expression in mammals were first characterized in hamster neurons. Multiple groups hypothesized that astrocytes may also contribute and perhaps drive circadian rhythmicity in the SCN and experimental evidence showing that astrocytes express clock genes and function as competent circadian oscillators arrived with the use of reporter gene constructs206,207. The identification of clock genes in astrocytes puts these cells under the spotlight and suggests that these cells are not mere modulators or targets of neuronal pacemakers, but they can act in conjunction with neurons to establish circadian rhythms206. One should note, however, that the level of expression of clock genes in astrocytes is substantially lower or more diffuse than in neurons, making it perhaps more difficult to detect206. This might have delayed the due recognition of astrocytes in the circadian pacemaker process, despite the early work demonstrating an essential role of astrocytes for the proper function of the circadian clock208211. There is now a growing awareness that just like the number of a given cell type does not speak to the relevance of its function, the lower expression of clock genes in astrocytes is not a measure of their physiological relevance in the generation and maintenance of circadian rhythms.

How do astrocytes contribute to the pacemaker activity of the SCN? In 1993, Lavialle and Serviere first reported that, in hamsters, there are light-independent (circadian) changes in the spatial distribution of the astrocyte protein GFAP and in astrocyte morphology, suggesting that these cells undergo circadian structural remodeling212. Since astrocyte remodeling is associated with changes in glucose consumption212,213, and astrocytes are main sources of glycogen, these findings have been thought to reflect the participation of astrocytes in the regulation of energy metabolism212,214. However, due to their abundant expression of glutamate transporters215, a retraction of astrocyte processes could also imply a longer time for glutamate clearance from the extracellular space216. This, in turn, could alter the time course of glutamatergic transmission at RHT synapses onto the SCN without the need for altering glutamate transporter expression in these cells217. The structural remodeling of astrocytes has been shown to account for circadian changes in the glial coverage of the soma and dendrites of VIP and AVP neurons in the SCN implicated with clock entrainment and pacemaker resetting, respectively218,219. Despite these findings, there are still uncertainties about the implications that the remodeling of astrocytes in the SCN has for glutamate clearance, synaptic plasticity, and metabolic exchange208. What has emerged in recent years, however, is that SCN astrocytes are not mere followers but rather partners in crime with the neuronal circadian pacemaker machinery220. Neurons are metabolically active during circadian daytime. Astrocytes, instead, are active during circadian nighttime and suppress neuronal activity by regulating extracellular glutamate concentration and activation of GluN2C-containing NMDA receptors. More importantly, astrocytes in the SCN can instruct neurons without a competent molecular clock to initiate and sustain circadian patterns of activity221.

Astrocytes and circadian clocks outside the SCN –

The first demonstration that astrocytes are competent circadian oscillators came from luciferase assay studies in rat cultures206. This work was important for several reasons that we would like to reiterate. First, it showed that the circadian period of astrocytes is genetically determined and differs between mice and rats, in parallel with period differences in the locomotor behavior of these species, potentially reconciling experimental differences detected when working with experimental preparations from different rodent species. Second, it showed that the expression of clock genes like Per1 is substantially lower or more diffuse in astrocytes than in neurons, potentially explaining initial difficulties in detectability of clock genes in astrocytes. Third, it showed that, in vitro, the circadian rhythms of astrocytes are not detected before the first week of culture. This is in stark contrast with the in vitro developmental timeline of cultured neurons, which display circadian rhythmicity earlier. This finding may provide insights into the different developmental profiles of circadian clocks in neurons and astrocytes in vivo222. Last, and arguably most importantly, the cultured astrocytes used in this work did not derive from the SCN but from the cortex, suggesting that regions of the brain outside the SCN can act as auxiliary, ancillary or accessory circadian oscillators.

Accordingly, the core circadian molecular machinery of astrocytes has been characterized in several regions of the nervous system (e.g., retina, olfactory bulb and hippocampus) and even outside the brain (e.g., cultured fibroblasts, hepatocytes, leukocytes, adipocytes, and muscle cells)223233. Although cells in these tissues can display intrinsic circadian rhythms, each of them would run at its own pace, desynchronize, and dampen population rhythms without SCN activity. This contrasts with what happens in the SCN, where neurons and astrocytes synchronize to each other234241. Several studies have attempted to reveal the identity of paracrine signals capable of synchronizing neuronal and astrocytes rhythms in auxiliary circadian oscillators (i.e., regions that express clock genes, but follow the timekeeping system set by the SCN). One of the first ones to be identified was the peptide hormone VIP, which coordinates circadian rhythms among astrocytes where VIP is released and where its receptor VPAC2R is expressed, like the olfactory bulb, retina, and the neocortex242245. Although the VIP/VPAC2R signaling pathway dominates circadian rhythmicity, other signaling pathways like the AVP/V1a/b and the GRP/ BB2 can also sustain daily cycling246.

Astrocytes and circadian clocks in the hippocampus –

Clock genes are expressed in the hippocampus, a brain region involved in encoding spatial and episodic memories247. Recent work has shown that not only clock genes, but also a larger set of hippocampal genes (10%) and proteins (11%) are expressed with circadian rhythmicity (cf. 19% in the SCN)248. These oscillations in the molecular landscape of the hippocampus are altered in experimental temporal lobe epilepsy248, which is of particular interest given that seizure onsets within individuals display strong circadian rhythmicity249.

In vivo behavioral studies indicate that lesioning the SCN eliminates circadian rhythmicity in a passive avoidance task250, and desynchronization of the circadian system impairs recall of a spatial task251. More specific tests on hippocampal-dependent learning and memory show that these vary between the circadian daytime and nighttime252257. The work of Chaudhury et al. is particularly important in this context as it shows that there is a circadian regulation of memory acquisition, recall and extinction in mice tested using a fear conditioning protocol258.

These periodical variations in hippocampal function can also be detected using in vivo electrophysiology recordings, although there are some differences in the results reported in the literature. Early studies in the dentate gyrus showed that the field EPSP and population spike amplitude are larger during the active phase (nighttime for rodents)259. Potentiation of the field EPSP amplitude, but not of the population spike amplitude, was confirmed in a later study260. Others also showed that the dentate gyrus is more excitable during the inactive phase (daytime for rodents)261. When describing these findings, it is important to keep in mind that the results of these works are sometimes described by comparing two clock times which can be misleading, since circadian rhythmicity is a phenomenon with a sinusoidal profile. The use of two points forces linearization which makes the comparison across datasets collected from different laboratories difficult, if not inaccurate, especially when the two time points are not twelve hours apart from each other (i.e., half the period of a circadian cycle) or when different labs use different reference times. This is important because it can lead to opposite conclusions about whether the described phenomenon increases or decreases during daytime/nighttime.

Perhaps surprisingly, there are circadian changes in hippocampal function that are retained in reduced slice preparations. These show that the incidence and magnitude of long-term potentiation (LTP) varies with opposite phase in the dentate gyrus versus hippocampal area CA1, and may therefore shape in different ways the activity of synapses in different domains of the same brain region262. Until recently, the synaptic mechanisms accounting for this effect remained unknown. What has now emerged is that these circadian changes in synaptic plasticity are differentially regulated by neurons and astrocytes263. During the nighttime, when LTP is reduced in hippocampal area CA1, pyramidal cells reduce their surface pool of functional NMDA receptors263. At the same time, glutamate clearance from astrocytes becomes slower, due to a retraction of astrocytic processes enriched with glutamate transporters from synapses, similar to what has also been observed in the SCN212. Glutamate inactivates AMPA receptors, and the longer glutamate remains in the extracellular space, the longer it takes for these receptors to recover from inactivation263. This has implications for synaptic integration of EPSPs, which is impaired during nighttime in CA1 pyramidal cells. The diffusible agent mediating these effects is not d-serine, because there are no detectable circadian changes in the occupancy of the NMDA receptor glycine binding site263,264. It is likely corticosterone, whose peak production time is during the active phase (i.e., nighttime for mice)263,265. Accordingly, one can rescue the loss of NMDA receptors and synaptic integration in slices prepared during the nighttime by treating them with antagonists of NR3C1/2 mineralocorticoid and glucocorticoid receptors263. What is also notable from this work is the fact that the magnitude of the circadian changes in synaptic integration are frequency dependent and are most pronounced for stimulation frequencies that can be detected in vivo during exploratory behaviors266,267. In other words, circadian rhythms do not alter hippocampal activity as a whole: rather, they modulate hippocampal-dependent learning while preserving memory recall.

Together, these findings suggest that in a subordinate oscillator like the hippocampus, the circadian changes in gene and protein expression may change the rules of synaptic plasticity, but the direction and extent of these changes vary not only across but also within brain regions, in cell-specific ways. On the other hand, different sets of cells can be active to mediate the same behavior at different times268. The ultimate effect of circadian clocks on more complex phenomena and behaviors may be activity-dependent, adding additional levels of complexity. Perhaps, our attempts to draw general rules on the functional properties of the brain, has led us to underestimate how dynamic this organ is and how profoundly our circadian clock can shape what we think we can always do well.

Astrocytes and sleep/wake cycles –

The earliest evidence that astrocyte activity impacts sleep homeostasis came from a 2009 publication showing that mice with astrocytic dnSNARE expression show decreased extracellular accumulation of adenosine, a molecule that in wild type mice promotes sleep and increases sleep pressure, also observed with sleep deprivation63. These findings implicated astrocyte-derived adenosine and A1 adenosine receptor activation as the factors underlying these behavioral effects, because pharmacological, in vivo blockade of A1 receptors attenuated sleep pressure accumulation in these mice63. The majority of extracellular adenosine is derived from metabolism of ATP and astrocytes have been recognized as a major source of extracellular ATP269. In addition to these findings, Papouin et al (2017) showed that d-serine levels oscillate in the hippocampus as a function of wakefulness, not of circadian rhythms264. This causes saturation of the NMDA receptor glycine binding site during wakefulness, that wanes down during sleep. Activation of Ca2+-permeable α7-nicotinic acetylcholine receptors via septal cholinergic afferents to the hippocampus is thought to be the trigger for d-serine release from astrocytes during wakefulness264. Consistent with these findings, recent Ca2+ fiber photometry data show that Ca2+ levels in astrocytes increase during wakefulness and decrease during REM sleep in different brain regions including the cortex, hippocampus, hypothalamus, pons and cerebellum270272. However, the magnitude of these oscillations and the detailed Ca2+ dynamics change between astrocytes in the: (i) cortex and hippocampus; (ii) hypothalamus and pons; (iii) cerebellum270. These findings suggest that astrocytes may contribute differently to the regulation of sleep and wakefulness, depending on the brain region270. The emerging evidence in support of brain region-specific patterns of astrocyte gene expression21,132,273,274 may help in identification of molecular drivers underlying such divergent functional and behavioral outcomes.

Microglia involvement in circadian rhythms –

Isolated microglia display intrinsic daily fluctuations of circadian clock genes, Bmal1, Rev-erb, Per1, and Per2275 with inflammatory factors, TNFα, IL-1β, and IL6 oscillating in phase with Per1 and Per2 and in anti-phase with Rev-erb276. Dysregulated Per1 and Per2 expression in aged animals has been associated with elevated TNFα and IL-1β mRNA expression275 linking circadian rhythms with evidence of microglial contribution to age-related neurodegeneration277. Moreover, daily microglial cytokine fluctuations may underlie increased sensitivity of neuroinflammatory response to stimulation by lipopolysaccharide during the resting (sleep) phase276 with the implication that therapeutic efficacy of inflammation-focused treatments may be improved by alignment to daily sleep/wake cycles. The link between microglial immunoreactivity and circadian rhythms is further borne out in studies that reduce or eliminate clock gene expression, although the direction of gene regulation differs between reports. For example, elevated levels of IL-1β and TNFα have been reported in striatal microglia of Bmal1 knock-out mice with further elevation after challenge with LPS or treatment with MPTH, a model of Parkinsonian neurodegeneration278. Elevated levels of IL-1β, IL-6 and TNFα have also been reported in the Rev-erba knock-out model279. However, in homogenized brain tissue, Bmal1 deletion was seen to decrease expression of IL-1β and inflammation-related gene, Nox2280, highlighting potential influence of region-specific differences in microglial response to Bmal1 deletion.

Microglia involvement in sleep/wake cycles –

It is well-documented and, indeed, accepted as common knowledge that disrupted sleep leads to a range of adverse psychological and physiological consequences. Evidence of microglial activation caused by sleep fragmentation281 is consistent with this line of reasoning although it is possible that fragmented sleep may arise secondary to other conditions (e.g., depression, drug use) associated with microglial pathologies. Supporting the former contention, depleting microglia by activating the diphtheria toxin receptor under the Cx3cr1 promoter in a transgenic rat model, resulted in disrupted expression of clock genes Per1, Per2, and Bmal1 in the SCN and the hippocampus, abnormal circadian body temperature, and diurnal rhythm disruptions282. Systemic ablation of microglia following PLX-5622 treatment was reported to increase sleep duration and eliminate light phase-dependent difference in synaptic transmission in mouse hippocampal CA1 pyramidal neurons283. In the healthy brain, microglial morphology fluctuates in rhythm with circadian cycles with longer processes and branching points during wakefulness, phenomena dependent on microglial expression of lysosomal cysteine protease cathepsin S and the adenosine diphosphate, P2Y12 receptors284,285. Microglial morphology may also be generally sensitive to arousal states with increased arborization of processes and increased process motility observed following anesthesia relative to wakeful conditions286. Partial activation of microglia during sleep may contribute to synapse elimination, suggesting an intriguing mechanism for the role of sleep in memory consolidation287. Indeed, sleep deprivation-induced memory impairments on a Morris water maze could be rescued in rats treated with minocycline288. Minocycline treatment has also been reported to increase slow-wave activity during sleep and improve episodic memory in human subjects289, although others have found sleep disruption by minocycline in both human290 and rodent291,292 studies. Notably, minocycline effects on sleep are unlikely due to its antibiotic activity as ampicillin treatment was not seen to impact sleep homeostasis290.

Concluding remarks

The emerging picture of structural and functional diversity of glial cells across distinct brain regions suggests co-existence of multiple mechanisms by which glial cells regulate neuronal activity. The diversity of mechanisms likely reflects unique physiological and computational demands placed upon circuits underlying specific behavioral outputs (Table 1). Within brain regions, dynamic glial cell responses to external perturbations, circadian rhythms or other internal changes are echoed by the flexible, adaptive nature of many behaviors. Understanding the functional roles of a molecularly diverse populations of glial cells will advance not only our knowledge of the mechanisms by which these cells control executive function, reward seeking, and circadian function, but will also shed light on phenomenology of a variety of neurodegenerative, substance use, and other disorders. The unique molecular signatures of glial cells make them attractive candidates for development of new therapeutics. Future studies utilizing single cell transcriptomics may shed light on region- and cell-type selective targets for use in both psychiatric and neurological disorders.

Highlights.

  • Astrocytes and microglia display diverse properties across brain regions

  • Synaptic and neuronal circuit activity are highly sensitive to astrocyte and microglial signals

  • Higher cognitive function, reward-seeking, and circadian behaviors are associated with structurally and functionally dynamic responses in glial cells

Acknowledgements:

A.S. NSF, IOS1655365, IOS2011998

P.I.O. NIH, DA04041513, DA053070

J.T. NIH, DA044311, DA053070

K.J.R. NIH, DA041455, DA052447

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Kandel ER, Principles of neural science. 5th ed. 2013, New York: McGraw-Hill. l, 1709 p. [Google Scholar]
  • 2.Virchow RRLK, Gesammelte Abhandlungen zur Wissenschaftlichen Medicin / von Rudolf Virchow. 1856, University of Glasgow: University of Glasgow. [Google Scholar]
  • 3.Parpura V and Verkhratsky A, Neuroglia at the crossroads of homoeostasis, metabolism and signalling: evolution of the concept. ASN Neuro, 2012. 4(4): p. 201–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Verkhratsky A, Parpura V, and Rodriguez JJ, Where the thoughts dwell: the physiology of neuronal-glial “diffuse neural net”. Brain Res Rev, 2011. 66(1–2): p. 133–51. [DOI] [PubMed] [Google Scholar]
  • 5.von Bartheld CS, Bahney J, and Herculano-Houzel S, The search for true numbers of neurons and glial cells in the human brain: A review of 150 years of cell counting. J Comp Neurol, 2016. 524(18): p. 3865–3895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pelvig DP, et al. , Neocortical glial cell numbers in human brains. Neurobiol Aging, 2008. 29(11): p. 1754–62. [DOI] [PubMed] [Google Scholar]
  • 7.Hyden H and Pigon A, A cytophysiological study of the functional relationship between oliodendroglial cells and nerve cells of Deiters’ nucleus. J Neurochem, 1960. 6: p. 57–72. [DOI] [PubMed] [Google Scholar]
  • 8.Blinkov S.M.l. and Glezer I.i.a.I., The human brain in figures and tables; a quantitative handbook. 1968, New York: Basic Books. xxxii, 482 p. [Google Scholar]
  • 9.Haug H, History of neuromorphometry. J Neurosci Methods, 1986. 18(1–2): p. 1–17. [DOI] [PubMed] [Google Scholar]
  • 10.Brachet J and Mirsky AE, The cell: biochemistry, physiology, morphology. 1959, New York,: Academic Press. [Google Scholar]
  • 11.Hyden H, Dynamic aspects of the neuron-glia relationship—A study with microchemical methods., in The Neuron, Hyden H, Editor. 1967, Elsevier: Netherlands. p. 179–217. [Google Scholar]
  • 12.von Bartheld CS, Myths and truths about the cellular composition of the human brain: A review of influential concepts. J Chem Neuroanat, 2018. 93: p. 2–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Azevedo FA, et al. , Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J Comp Neurol, 2009. 513(5): p. 532–41. [DOI] [PubMed] [Google Scholar]
  • 14.Herculano-Houzel S and Lent R, Isotropic fractionator: a simple, rapid method for the quantification of total cell and neuron numbers in the brain. J Neurosci, 2005. 25(10): p. 2518–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Miller DJ, et al. , Three counting methods agree on cell and neuron number in chimpanzee primary visual cortex. Front Neuroanat, 2014. 8: p. 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hawkins A and Olszewski J, Glia/nerve cell index for cortex of the whale. Science, 1957. 126(3263): p. 76–7. [DOI] [PubMed] [Google Scholar]
  • 17.Herculano-Houzel S, The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia, 2014. 62(9): p. 1377–91. [DOI] [PubMed] [Google Scholar]
  • 18.Keller D, Ero C, and Markram H, Cell Densities in the Mouse Brain: A Systematic Review. Front Neuroanat, 2018. 12: p. 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Oberheim NA, et al. , Astrocytic complexity distinguishes the human brain. Trends Neurosci, 2006. 29(10): p. 547–53. [DOI] [PubMed] [Google Scholar]
  • 20.Oberheim NA, et al. , Uniquely hominid features of adult human astrocytes. J Neurosci, 2009. 29(10): p. 3276–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chai H, et al. , Neural Circuit-Specialized Astrocytes: Transcriptomic, Proteomic, Morphological, and Functional Evidence. Neuron, 2017. 95(3): p. 531–549 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Morel L, et al. , Molecular and Functional Properties of Regional Astrocytes in the Adult Brain. J Neurosci, 2017. 37(36): p. 8706–8717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Boisvert MM, et al. , The Aging Astrocyte Transcriptome from Multiple Regions of the Mouse Brain. Cell Rep, 2018. 22(1): p. 269–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lalo U, et al. , The tripartite glutamatergic synapse. Neuropharmacology, 2021. 199: p. 108758. [DOI] [PubMed] [Google Scholar]
  • 25.Semyanov A and Verkhratsky A, Astrocytic processes: from tripartite synapses to the active milieu. Trends Neurosci, 2021. 44(10): p. 781–792. [DOI] [PubMed] [Google Scholar]
  • 26.Ferro A, Auguste YSS, and Cheadle L, Microglia, Cytokines, and Neural Activity: Unexpected Interactions in Brain Development and Function. Front Immunol, 2021. 12: p. 703527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Xin W, et al. , Ventral midbrain astrocytes display unique physiological features and sensitivity to dopamine D2 receptor signaling. Neuropsychopharmacology, 2019. 44(2): p. 344–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Khakh BS and Deneen B, The Emerging Nature of Astrocyte Diversity. Annu Rev Neurosci, 2019. 42: p. 187–207. [DOI] [PubMed] [Google Scholar]
  • 29.Yu X, et al. , Context-Specific Striatal Astrocyte Molecular Responses Are Phenotypically Exploitable. Neuron, 2020. 108(6): p. 1146–1162 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Brockett AT, et al. , Evidence supporting a role for astrocytes in the regulation of cognitive flexibility and neuronal oscillations through the Ca2+ binding protein S100beta. PLoS One, 2018. 13(4): p. e0195726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lima A, et al. , Astrocyte pathology in the prefrontal cortex impairs the cognitive function of rats. Mol Psychiatry, 2014. 19(7): p. 834–41. [DOI] [PubMed] [Google Scholar]
  • 32.Banasr M and Duman RS, Glial loss in the prefrontal cortex is sufficient to induce depressive-like behaviors. Biol Psychiatry, 2008. 64(10): p. 863–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee Y, et al. , Glutamine deficiency in the prefrontal cortex increases depressive-like behaviours in male mice. J Psychiatry Neurosci, 2013. 38(3): p. 183–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Duman RS, Neuronal damage and protection in the pathophysiology and treatment of psychiatric illness: stress and depression. Dialogues Clin Neurosci, 2009. 11(3): p. 239–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Luscher C and Malenka RC, Drug-evoked synaptic plasticity in addiction: from molecular changes to circuit remodeling. Neuron, 2011. 69(4): p. 650–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McBean GJ, Inhibition of the glutamate transporter and glial enzymes in rat striatum by the gliotoxin, alpha aminoadipate. Br J Pharmacol, 1994. 113(2): p. 536–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Khurgel M, Koo AC, and Ivy GO, Selective ablation of astrocytes by intracerebral injections of alpha-aminoadipate. Glia, 1996. 16(4): p. 351–8. [DOI] [PubMed] [Google Scholar]
  • 38.Zamanian JL, et al. , Genomic analysis of reactive astrogliosis. J Neurosci, 2012. 32(18): p. 6391–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lin RC, et al. , Re-expression of the intermediate filament nestin in reactive astrocytes. Neurobiol Dis, 1995. 2(2): p. 79–85. [DOI] [PubMed] [Google Scholar]
  • 40.Krum JM and Rosenstein JM, Transient coexpression of nestin, GFAP, and vascular endothelial growth factor in mature reactive astroglia following neural grafting or brain wounds. Exp Neurol, 1999. 160(2): p. 348–60. [DOI] [PubMed] [Google Scholar]
  • 41.Wilhelmsson U, et al. , The role of GFAP and vimentin in learning and memory. Biol Chem, 2019. 400(9): p. 1147–1156. [DOI] [PubMed] [Google Scholar]
  • 42.Avigan PD, Cammack K, and Shapiro ML, Flexible spatial learning requires both the dorsal and ventral hippocampus and their functional interactions with the prefrontal cortex. Hippocampus, 2020. 30(7): p. 733–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wilhelmsson U, et al. , Nestin Null Mice Show Improved Reversal Place Learning. Neurochem Res, 2020. 45(1): p. 215–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rajkowska G, et al. , Layer-specific reductions in GFAP-reactive astroglia in the dorsolateral prefrontal cortex in schizophrenia. Schizophr Res, 2002. 57(2–3): p. 127–38. [DOI] [PubMed] [Google Scholar]
  • 45.Martin R, et al. , Circuit-specific signaling in astrocyte-neuron networks in basal ganglia pathways. Science, 2015. 349(6249): p. 730–4. [DOI] [PubMed] [Google Scholar]
  • 46.Lavialle M, et al. , Structural plasticity of perisynaptic astrocyte processes involves ezrin and metabotropic glutamate receptors. Proc Natl Acad Sci U S A, 2011. 108(31): p. 12915–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhou Y, et al. , EAAT2 (GLT-1; slc1a2) glutamate transporters reconstituted in liposomes argues against heteroexchange being substantially faster than net uptake. J Neurosci, 2014. 34(40): p. 13472–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ortinski PI, et al. , Selective induction of astrocytic gliosis generates deficits in neuronal inhibition. Nat Neurosci, 2010. 13(5): p. 584–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Spangaro M, et al. , Cognitive dysfunction and glutamate reuptake: effect of EAAT2 polymorphism in schizophrenia. Neurosci Lett, 2012. 522(2): p. 151–5. [DOI] [PubMed] [Google Scholar]
  • 50.Roberts-Wolfe DJ and Kalivas PW, Glutamate Transporter GLT-1 as a Therapeutic Target for Substance Use Disorders. CNS Neurol Disord Drug Targets, 2015. 14(6): p. 745–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Meeker KD, Meabon JS, and Cook DG, Partial Loss of the Glutamate Transporter GLT-1 Alters Brain Akt and Insulin Signaling in a Mouse Model of Alzheimer’s Disease. J Alzheimers Dis, 2015. 45(2): p. 509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Scimemi A, et al. , Amyloid-beta1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1. J Neurosci, 2013. 33(12): p. 5312–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kobayashi E, et al. , Activated forms of astrocytes with higher GLT-1 expression are associated with cognitive normal subjects with Alzheimer pathology in human brain. Sci Rep, 2018. 8(1): p. 1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Karlsson RM, et al. , Assessment of glutamate transporter GLAST (EAAT1)-deficient mice for phenotypes relevant to the negative and executive/cognitive symptoms of schizophrenia. Neuropsychopharmacology, 2009. 34(6): p. 1578–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.John CS, et al. , Blockade of astrocytic glutamate uptake in the prefrontal cortex induces anhedonia. Neuropsychopharmacology, 2012. 37(11): p. 2467–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Featherstone RE, et al. , Subchronic ketamine treatment leads to permanent changes in EEG, cognition and the astrocytic glutamate transporter EAAT2 in mice. Neurobiol Dis, 2012. 47(3): p. 338–46. [DOI] [PubMed] [Google Scholar]
  • 57.Scimemi A, et al. , NR2B-containing receptors mediate cross talk among hippocampal synapses. J Neurosci, 2004. 24(20): p. 4767–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nie H and Weng HR, Glutamate transporters prevent excessive activation of NMDA receptors and extrasynaptic glutamate spillover in the spinal dorsal horn. J Neurophysiol, 2009. 101(4): p. 2041–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ortinski PI, Turner JR, and Pierce RC, Extrasynaptic targeting of NMDA receptors following D1 dopamine receptor activation and cocaine self-administration. J Neurosci, 2013. 33(22): p. 9451–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pascual O, et al. , Astrocytic purinergic signaling coordinates synaptic networks. Science, 2005. 310(5745): p. 113–6. [DOI] [PubMed] [Google Scholar]
  • 61.Fujita T, et al. , Neuronal transgene expression in dominant-negative SNARE mice. J Neurosci, 2014. 34(50): p. 16594–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Durkee C, et al. , Astrocyte and neuron cooperation in long-term depression. Trends Neurosci, 2021. 44(10): p. 837–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Halassa MM, et al. , Astrocytic modulation of sleep homeostasis and cognitive consequences of sleep loss. Neuron, 2009. 61(2): p. 213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Poskanzer KE and Yuste R, Astrocytes regulate cortical state switching in vivo. Proc Natl Acad Sci U S A, 2016. 113(19): p. E2675–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sardinha VM, et al. , Astrocytic signaling supports hippocampal-prefrontal theta synchronization and cognitive function. Glia, 2017. 65(12): p. 1944–1960. [DOI] [PubMed] [Google Scholar]
  • 66.Henneberger C, et al. , Long-term potentiation depends on release of D-serine from astrocytes. Nature, 2010. 463(7278): p. 232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Deng Q, et al. , Astrocytic activation of A1 receptors regulates the surface expression of NMDA receptors through a Src kinase dependent pathway. Glia, 2011. 59(7): p. 1084–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wirkner K, et al. , Modulation of NMDA receptor current in layer V pyramidal neurons of the rat prefrontal cortex by P2Y receptor activation. Cereb Cortex, 2007. 17(3): p. 621–31. [DOI] [PubMed] [Google Scholar]
  • 69.Fellin T, et al. , Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron, 2004. 43(5): p. 729–43. [DOI] [PubMed] [Google Scholar]
  • 70.Henneberger C, et al. , LTP Induction Boosts Glutamate Spillover by Driving Withdrawal of Perisynaptic Astroglia. Neuron, 2020. 108(5): p. 919–936 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ortinski PI, Cocaine-induced changes in NMDA receptor signaling. Mol Neurobiol, 2014. 50(2): p. 494–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.O’Donovan B, et al. , Cocaine experience induces functional adaptations in astrocytes: Implications for synaptic plasticity in the nucleus accumbens shell. Addict Biol, 2021: p. e13042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Yoon BE and Lee CJ, GABA as a rising gliotransmitter. Front Neural Circuits, 2014. 8: p. 141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mederos S, et al. , GABAergic signaling to astrocytes in the prefrontal cortex sustains goal-directed behaviors. Nat Neurosci, 2021. 24(1): p. 82–92. [DOI] [PubMed] [Google Scholar]
  • 75.Perea G, et al. , Optogenetic astrocyte activation modulates response selectivity of visual cortex neurons in vivo. Nat Commun, 2014. 5: p. 3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fiacco TA and McCarthy KD, Multiple Lines of Evidence Indicate That Gliotransmission Does Not Occur under Physiological Conditions. J Neurosci, 2018. 38(1): p. 3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Savtchouk I and Volterra A, Gliotransmission: Beyond Black-and-White. J Neurosci, 2018. 38(1): p. 14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Khakh BS and McCarthy KD, Astrocyte calcium signaling: from observations to functions and the challenges therein. Cold Spring Harb Perspect Biol, 2015. 7(4): p. a020404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Semyanov A, Henneberger C, and Agarwal A, Making sense of astrocytic calcium signals - from acquisition to interpretation. Nat Rev Neurosci, 2020. 21(10): p. 551–564. [DOI] [PubMed] [Google Scholar]
  • 80.Neugornet A, O’Donovan B, and Ortinski PI, Comparative Effects of Event Detection Methods on the Analysis and Interpretation of Ca(2+) Imaging Data. Front Neurosci, 2021. 15: p. 620869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bissonette GB, et al. , Astrocyte-mediated hepatocyte growth factor/scatter factor supplementation restores GABAergic interneurons and corrects reversal learning deficits in mice. J Neurosci, 2010. 30(8): p. 2918–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Diaz A, et al. , A Cross Talk between Neuronal Urokinase-type Plasminogen Activator (uPA) and Astrocytic uPA Receptor (uPAR) Promotes Astrocytic Activation and Synaptic Recovery in the Ischemic Brain. J Neurosci, 2017. 37(43): p. 10310–10322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Webb TR, et al. , X-linked megalocornea caused by mutations in CHRDL1 identifies an essential role for ventroptin in anterior segment development. Am J Hum Genet, 2012. 90(2): p. 247–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Blanco-Suarez E, et al. , Astrocyte-Secreted Chordin-like 1 Drives Synapse Maturation and Limits Plasticity by Increasing Synaptic GluA2 AMPA Receptors. Neuron, 2018. 100(5): p. 1116–1132 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wolf ME and Tseng KY, Calcium-permeable AMPA receptors in the VTA and nucleus accumbens after cocaine exposure: when, how, and why? Front Mol Neurosci, 2012. 5: p. 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Galloway A, et al. , Dopamine Triggers CTCF-Dependent Morphological and Genomic Remodeling of Astrocytes. J Neurosci, 2018. 38(21): p. 4846–4858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Sofroniew MV, Multiple roles for astrocytes as effectors of cytokines and inflammatory mediators. Neuroscientist, 2014. 20(2): p. 160–72. [DOI] [PubMed] [Google Scholar]
  • 88.Caudal LC, et al. , The Paradox of Astroglial Ca(2 +) Signals at the Interface of Excitation and Inhibition. Front Cell Neurosci, 2020. 14: p. 609947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Heyser CJ, et al. , Progressive decline in avoidance learning paralleled by inflammatory neurodegeneration in transgenic mice expressing interleukin 6 in the brain. Proc Natl Acad Sci U S A, 1997. 94(4): p. 1500–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Booth HDE, Hirst WD, and Wade-Martins R, The Role of Astrocyte Dysfunction in Parkinson’s Disease Pathogenesis. Trends Neurosci, 2017. 40(6): p. 358–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Khakh BS, et al. , Unravelling and Exploiting Astrocyte Dysfunction in Huntington’s Disease. Trends Neurosci, 2017. 40(7): p. 422–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Carter SF, et al. , Astrocyte Biomarkers in Alzheimer’s Disease. Trends Mol Med, 2019. 25(2): p. 77–95. [DOI] [PubMed] [Google Scholar]
  • 93.Brambilla R, The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol, 2019. 137(5): p. 757–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pekny M, et al. , Astrocyte activation and reactive gliosis-A new target in stroke? Neurosci Lett, 2019. 689: p. 45–55. [DOI] [PubMed] [Google Scholar]
  • 95.Burda JE, Bernstein AM, and Sofroniew MV, Astrocyte roles in traumatic brain injury. Exp Neurol, 2016. 275 Pt 3: p. 305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Rodriguez-Arellano JJ, et al. , Astrocytes in physiological aging and Alzheimer’s disease. Neuroscience, 2016. 323: p. 170–82. [DOI] [PubMed] [Google Scholar]
  • 97.Cohen J and Torres C, Astrocyte senescence: Evidence and significance. Aging Cell, 2019. 18(3): p. e12937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Veniaminova E, et al. , Prefrontal cortex inflammation and liver pathologies accompany cognitive and motor deficits following Western diet consumption in non-obese female mice. Life Sci, 2020. 241: p. 117163. [DOI] [PubMed] [Google Scholar]
  • 99.Val-Laillet D, et al. , A maternal Western diet during gestation and lactation modifies offspring’s microglial cell density and morphology in the hippocampus and prefrontal cortex in Yucatan minipigs. Neurosci Lett, 2020. 739: p. 135395. [DOI] [PubMed] [Google Scholar]
  • 100.Bocarsly ME, et al. , Obesity diminishes synaptic markers, alters microglial morphology, and impairs cognitive function. Proc Natl Acad Sci U S A, 2015. 112(51): p. 15731–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.De Biase LM, et al. , Local Cues Establish and Maintain Region-Specific Phenotypes of Basal Ganglia Microglia. Neuron, 2017. 95(2): p. 341–356 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Butovsky O and Weiner HL, Microglial signatures and their role in health and disease. Nat Rev Neurosci, 2018. 19(10): p. 622–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rosenblat JD and McIntyre RS, Are medical comorbid conditions of bipolar disorder due to immune dysfunction? Acta Psychiatr Scand, 2015. 132(3): p. 180–91. [DOI] [PubMed] [Google Scholar]
  • 104.Lin SP, et al. , Clinical Treatment Options and Randomized Clinical Trials for Neurocognitive Complications of HIV Infection: Combination Antiretroviral Therapy, Central Nervous System Penetration Effectiveness, and Adjuvants. Curr Top Behav Neurosci, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kohno M, et al. , Neuroinflammation in addiction: A review of neuroimaging studies and potential immunotherapies. Pharmacol Biochem Behav, 2019. 179: p. 34–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bortolato B, et al. , Cognitive remission: a novel objective for the treatment of major depression? BMC Med, 2016. 14: p. 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jeppesen R, et al. , Efficacy and safety of anti-inflammatory agents in treatment of psychotic disorders - A comprehensive systematic review and meta-analysis. Brain Behav Immun, 2020. 90: p. 364–380. [DOI] [PubMed] [Google Scholar]
  • 108.Solmi M, et al. , Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr, 2017. 22(5): p. 415–426. [DOI] [PubMed] [Google Scholar]
  • 109.Acharya MM, et al. , Elimination of microglia improves cognitive function following cranial irradiation. Sci Rep, 2016. 6: p. 31545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Howard R, et al. , in Minocycline 200 mg or 400 mg versus placebo for mild Alzheimer’s disease: the MADE Phase II, three-arm RCT. 2020: Southampton (UK). [PubMed] [Google Scholar]
  • 111.Choi Y, et al. , Minocycline attenuates neuronal cell death and improves cognitive impairment in Alzheimer’s disease models. Neuropsychopharmacology, 2007. 32(11): p. 2393–404. [DOI] [PubMed] [Google Scholar]
  • 112.Daulatzai MA, Pharmacotherpy and Alzheimer’s Disease: The M-Drugs (Melatonin, Minocycline, Modafinil, and Memantine) Approach. Curr Pharm Des, 2016. 22(16): p. 2411–30. [DOI] [PubMed] [Google Scholar]
  • 113.Fessel J, Prevention of Alzheimer’s disease by treating mild cognitive impairment with combinations chosen from eight available drugs. Alzheimers Dement (N Y), 2019. 5: p. 780–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Green KN, Crapser JD, and Hohsfield LA, To Kill a Microglia: A Case for CSF1R Inhibitors. Trends Immunol, 2020. 41(9): p. 771–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.McEwen BS and Morrison JH, The brain on stress: vulnerability and plasticity of the prefrontal cortex over the life course. Neuron, 2013. 79(1): p. 16–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Fogaca MV and Duman RS, Cortical GABAergic Dysfunction in Stress and Depression: New Insights for Therapeutic Interventions. Front Cell Neurosci, 2019. 13: p. 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Horchar MJ and Wohleb ES, Glucocorticoid receptor antagonism prevents microglia-mediated neuronal remodeling and behavioral despair following chronic unpredictable stress. Brain Behav Immun, 2019. 81: p. 329–340. [DOI] [PubMed] [Google Scholar]
  • 118.Hinwood M, et al. , Chronic stress induced remodeling of the prefrontal cortex: structural reorganization of microglia and the inhibitory effect of minocycline. Cereb Cortex, 2013. 23(8): p. 178–497. [DOI] [PubMed] [Google Scholar]
  • 119.Liu T, et al. , Stress induces microglia-associated synaptic circuit alterations in the dorsomedial prefrontal cortex. Neurobiol Stress, 2021. 15: p. 100342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wohleb ES, et al. , Stress-Induced Neuronal Colony Stimulating Factor 1 Provokes Microglia-Mediated Neuronal Remodeling and Depressive-like Behavior. Biol Psychiatry, 2018. 83(1): p. 38–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Furuyashiki T, Roles of dopamine and inflammation-related molecules in behavioral alterations caused by repeated stress. J Pharmacol Sci, 2012. 120(2): p. 63–9. [DOI] [PubMed] [Google Scholar]
  • 122.Caetano L, et al. , Adenosine A2A receptor regulation of microglia morphological remodeling-gender bias in physiology and in a model of chronic anxiety. Mol Psychiatry, 2017. 22(7): p. 1035–1043. [DOI] [PubMed] [Google Scholar]
  • 123.Duarte JM, et al. , Region-specific control of microglia by adenosine A2A receptors: uncoupling anxiety and associated cognitive deficits in female rats. Glia, 2019. 67(1): p. 182–192. [DOI] [PubMed] [Google Scholar]
  • 124.Zhang Y, et al. , Deficiencies of microglia and TNFalpha in the mPFC-mediated cognitive inflexibility induced by social stress during adolescence. Brain Behav Immun, 2019. 79: p. 256–266. [DOI] [PubMed] [Google Scholar]
  • 125.Li H, Sagar AP, and Keri S, Microglial markers in the frontal cortex are related to cognitive dysfunctions in major depressive disorder. J Affect Disord, 2018. 241: p. 305–310. [DOI] [PubMed] [Google Scholar]
  • 126.Rubin LH, et al. , Microglial activation is inversely associated with cognition in individuals living with HIV on effective antiretroviral therapy. AIDS, 2018. 32(12): p. 1661–1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Giridharan VV, et al. , Neuroinflammation trajectories precede cognitive impairment after experimental meningitis-evidence from an in vivo PET study. J Neuroinflammation, 2020. 17(1): p. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Politis M, et al. , Microglial activation in regions related to cognitive function predicts disease onset in Huntington’s disease: a multimodal imaging study. Hum Brain Mapp, 2011. 32(2): p. 258–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Polyzos AA, et al. , Metabolic Reprogramming in Astrocytes Distinguishes Region-Specific Neuronal Susceptibility in Huntington Mice. Cell Metab, 2019. 29(6): p. 1258–1273 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Khakh BS, Astrocyte-Neuron Interactions in the Striatum: Insights on Identity, Form, and Function. Trends Neurosci, 2019. 42(9): p. 617–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Xin W and Bonci A, Functional Astrocyte Heterogeneity and Implications for Their Role in Shaping Neurotransmission. Front Cell Neurosci, 2018. 12: p. 141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Batiuk MY, et al. , Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun, 2020. 11(1): p. 1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Clarke BE, et al. , Regionally encoded functional heterogeneity of astrocytes in health and disease: A perspective. Glia, 2021. 69(1): p. 20–27. [DOI] [PubMed] [Google Scholar]
  • 134.Baker DA, et al. , Neuroadaptations in cystine-glutamate exchange underlie cocaine relapse. Nat Neurosci, 2003. 6(7): p. 743–9. [DOI] [PubMed] [Google Scholar]
  • 135.Knackstedt LA, Melendez RI, and Kalivas PW, Ceftriaxone restores glutamate homeostasis and prevents relapse to cocaine seeking. Biol Psychiatry, 2010. 67(1): p. 81–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Scofield MD, et al. , The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev, 2016. 68(3): p. 816–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Spencer S and Kalivas PW, Glutamate Transport: A New Bench to Bedside Mechanism for Treating Drug Abuse. Int J Neuropsychopharmacol, 2017. 20(10): p. 797–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Scofield MD, et al. , Cocaine Self-Administration and Extinction Leads to Reduced Glial Fibrillary Acidic Protein Expression and Morphometric Features of Astrocytes in the Nucleus Accumbens Core. Biol Psychiatry, 2016. 80(3): p. 207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Testen A, et al. , Region-Specific Reductions in Morphometric Properties and Synaptic Colocalization of Astrocytes Following Cocaine Self-Administration and Extinction. Front Cell Neurosci, 2018. 12: p. 246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kruyer A, et al. , Heroin Cue-Evoked Astrocytic Structural Plasticity at Nucleus Accumbens Synapses Inhibits Heroin Seeking. Biol Psychiatry, 2019. 86(11): p. 811–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Siemsen BM, et al. , Effects of Methamphetamine Self-Administration and Extinction on Astrocyte Structure and Function in the Nucleus Accumbens Core. Neuroscience, 2019. 406: p. 528–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sharpe A, Trzeciak M, Douglas P, Beckstead MJ., Repeated treatment with cocaine or methamphetamine increases CRF2 and decreases astrocytic markers in the ventral tegmental area and substantia nigra, in bioRxiv. 2019. [Google Scholar]
  • 143.Bull C, et al. , Differential response of glial fibrillary acidic protein-positive astrocytes in the rat prefrontal cortex following ethanol self-administration. Alcohol Clin Exp Res, 2015. 39(4): p. 650–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kim R, et al. , Astroglial correlates of neuropsychiatric disease: From astrocytopathy to astrogliosis. Prog Neuropsychopharmacol Biol Psychiatry, 2018. 87(Pt A): p. 126–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Healey KL, et al. , Enduring alterations in hippocampal astrocytesynaptic proximity following adolescent alcohol exposure: reversal by gabapentin. Neural Regen Res, 2020. 15(8): p. 1496–1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Erickson EK, et al. , Cortical astrocytes regulate ethanol consumption and intoxication in mice. Neuropsychopharmacology, 2021. 46(3): p. 500–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Scofield MD, et al. , Gq-DREADD Selectively Initiates Glial Glutamate Release and Inhibits Cue-induced Cocaine Seeking. Biol Psychiatry, 2015. 78(7): p. 441–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bull C, et al. , Rat nucleus accumbens core astrocytes modulate reward and the motivation to self-administer ethanol after abstinence. Neuropsychopharmacology, 2014. 39(12): p. 2835–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Gomez JA, et al. , Ventral tegmental area astrocytes orchestrate avoidance and approach behavior. Nat Commun, 2019. 10(1): p. 1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kofuji P and Araque A, G-Protein-Coupled Receptors in Astrocyte-Neuron Communication. Neuroscience, 2021. 456: p. 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Li Y, et al. , Activation of astrocytes in hippocampus decreases fear memory through adenosine A1 receptors. Elife, 2020. 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Martin-Fernandez M, et al. , Synapse-specific astrocyte gating of amygdala-related behavior. Nat Neurosci, 2017. 20(11): p. 1540–1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Corkrum M, et al. , Dopamine-Evoked Synaptic Regulation in the Nucleus Accumbens Requires Astrocyte Activity. Neuron, 2020. 105(6): p. 1036–1047 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ortinski PI, et al. , Cocaine-seeking is associated with PKC-dependent reduction of excitatory signaling in accumbens shell D2 dopamine receptor-expressing neurons. Neuropharmacology, 2015. 92: p. 80–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Briand LA, et al. , Disruption of glutamate receptor-interacting protein in nucleus accumbens enhances vulnerability to cocaine relapse. Neuropsychopharmacology, 2014. 39(3): p. 759–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.White SL, et al. , A Critical Role for the GluA1 Accessory Protein, SAP97, in Cocaine Seeking. Neuropsychopharmacology, 2016. 41(3): p. 736–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Mizuno GO, et al. , Aberrant Calcium Signaling in Astrocytes Inhibits Neuronal Excitability in a Human Down Syndrome Stem Cell Model. Cell Rep, 2018. 24(2): p. 355–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Papouin T, et al. , Astrocytic control of synaptic function. Philos Trans R Soc Lond B Biol Sci, 2017. 372(1715). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Newman EA, Glial cell inhibition of neurons by release of ATP. J Neurosci, 2003. 23(5): p. 1659–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Koizumi S, et al. , Dynamic inhibition of excitatory synaptic transmission by astrocyte-derived ATP in hippocampal cultures. Proc Natl Acad Sci U S A, 2003. 100(19): p. 11023–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Allen NJ and Eroglu C, Cell Biology of Astrocyte-Synapse Interactions. Neuron, 2017. 96(3): p. 697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Corkrum M and Araque A, Astrocyte-neuron signaling in the mesolimbic dopamine system: the hidden stars of dopamine signaling. Neuropsychopharmacology, 2021. 46(11): p. 1864–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Lipton DM, Gonzales BJ, and Citri A, Dorsal Striatal Circuits for Habits, Compulsions and Addictions. Front Syst Neurosci, 2019. 13: p. 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Fouyssac M and Belin D, Beyond drug-induced alteration of glutamate homeostasis, astrocytes may contribute to dopamine-dependent intrastriatal functional shifts that underlie the development of drug addiction: A working hypothesis. Eur J Neurosci, 2019. 50(6): p. 3014–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Vandaele Y and Ahmed SH, Habit, choice, and addiction. Neuropsychopharmacology, 2021. 46(4): p. 689–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Everitt BJ and Robbins TW, Drug Addiction: Updating Actions to Habits to Compulsions Ten Years On. Annu Rev Psychol, 2016. 67: p. 23–50. [DOI] [PubMed] [Google Scholar]
  • 167.Kang S, et al. , Activation of Astrocytes in the Dorsomedial Striatum Facilitates Transition From Habitual to Goal-Directed Reward-Seeking Behavior. Biol Psychiatry, 2020. 88(10): p. 797–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Fuxe K, et al. , Adenosine-dopamine interactions in the pathophysiology and treatment of CNS disorders. CNS Neurosci Ther, 2010. 16(3): p. e18–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Ferre S, et al. , Adenosine-dopamine receptor-receptor interactions as an integrative mechanism in the basal ganglia. Trends Neurosci, 1997. 20(10): p. 482–7. [DOI] [PubMed] [Google Scholar]
  • 170.Ferre S, et al. , Adenosine-dopamine interactions in the brain. Neuroscience, 1992. 51(3): p. 501–12. [DOI] [PubMed] [Google Scholar]
  • 171.Boender AJ, et al. , Striatal Astrocytes Shape Behavioral Flexibility via Regulation of the Glutamate Transporter EAAT2. Biol Psychiatry, 2021. 89(11): p. 1045–1057. [DOI] [PubMed] [Google Scholar]
  • 172.Volkow ND, Koob GF, and McLellan AT, Neurobiologic Advances from the Brain Disease Model of Addiction. N Engl J Med, 2016. 374(4): p. 363–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Yu X, et al. , Reducing Astrocyte Calcium Signaling In Vivo Alters Striatal Microcircuits and Causes Repetitive Behavior. Neuron, 2018. 99(6): p. 1170–1187 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Thomas Broome S, et al. , Dopamine: an immune transmitter. Neural Regen Res, 2020. 15(12): p. 2173–2185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.West RK, Rodgers SP, and Leasure JL, Neural Perturbations Associated With Recurrent Binge Alcohol in Male and Female Rats. Alcohol Clin Exp Res, 2021. 45(2): p. 365–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Marshall SA, et al. , Microglia Dystrophy Following Binge-Like Alcohol Exposure in Adolescent and Adult Male Rats. Front Neuroanat, 2020. 14: p. 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Marchese NA, et al. , Angiotensin II modulates amphetamine-induced glial and brain vascular responses, and attention deficit via angiotensin type 1 receptor: Evidence from brain regional sensitivity to amphetamine. Eur J Neurosci, 2020. 51(4): p. 1026–1041. [DOI] [PubMed] [Google Scholar]
  • 178.Thomas DM, et al. , Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglial activation. J Pharmacol Exp Ther, 2004. 311(1): p. 1–7. [DOI] [PubMed] [Google Scholar]
  • 179.Cotto B, et al. , Cocaine-mediated activation of microglia and microglial MeCP2 and BDNF production. Neurobiol Dis, 2018. 117: p. 28–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wang ZJ, et al. , Activin A is increased in the nucleus accumbens following a cocaine binge. Sci Rep, 2017. 7: p. 43658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Jarvis R, et al. , Cocaine Self-administration and Extinction Inversely Alter Neuron to Glia Exosomal Dynamics in the Nucleus Accumbens. Front Cell Neurosci, 2019. 13: p. 581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Burkovetskaya ME, et al. , Cocaine self-administration differentially activates microglia in the mouse brain. Neurosci Lett, 2020. 728: p. 134951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Lewitus GM, et al. , Microglial TNF-alpha Suppresses Cocaine-Induced Plasticity and Behavioral Sensitization. Neuron, 2016. 90(3): p. 483–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Narendran R, et al. , Cocaine abuse in humans is not associated with increased microglial activation: an 18-kDa translocator protein positron emission tomography imaging study with [11C]PBR28. J Neurosci, 2014. 34(30): p. 9945–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Melbourne JK, et al. , Its complicated: The relationship between alcohol and microglia in the search for novel pharmacotherapeutic targets for alcohol use disorders. Prog Mol Biol Transl Sci, 2019. 167: p. 179–221. [DOI] [PubMed] [Google Scholar]
  • 186.Marshall SA, et al. , Microglial activation is not equivalent to neuroinflammation in alcohol-induced neurodegeneration: The importance of microglia phenotype. Neurobiol Dis, 2013. 54: p. 239–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Boche D and Gordon MN, Diversity of transcriptomic microglial phenotypes in aging and Alzheimer’s disease. Alzheimers Dement, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Agrawal RG, et al. , Minocycline reduces ethanol drinking. Brain Behav Immun, 2011. 25 Suppl 1: p. S165–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Gajbhiye SV, et al. , Minocycline in Alcohol Withdrawal Induced Anxiety and Alcohol Relapse in Rats. Curr Clin Pharmacol, 2018. 13(1): p. 65–72. [DOI] [PubMed] [Google Scholar]
  • 190.Warden AS, et al. , Microglia depletion and alcohol: Transcriptome and behavioral profiles. Addict Biol, 2021. 26(2): p. e12889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Warden AS, et al. , Microglia Control Escalation of Drinking in Alcohol-Dependent Mice: Genomic and Synaptic Drivers. Biol Psychiatry, 2020. 88(12): p. 910–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Adeluyi A, et al. , Microglia morphology and proinflammatory signaling in the nucleus accumbens during nicotine withdrawal. Sci Adv, 2019. 5(10): p. eaax7031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Northcutt AL, et al. , DAT isn’t all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry, 2015. 20(12): p. 1525–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Fujita Y, et al. , The antibiotic minocycline prevents methamphetamine-induced rewarding effects in mice. Pharmacol Biochem Behav, 2012. 101(2): p. 303–6. [DOI] [PubMed] [Google Scholar]
  • 195.Attarzadeh-Yazdi G, Arezoomandan R, and Haghparast A, Minocycline, an antibiotic with inhibitory effect on microglial activation, attenuates the maintenance and reinstatement of methamphetamine-seeking behavior in rat. Prog Neuropsychopharmacol Biol Psychiatry, 2014. 53: p. 142–8. [DOI] [PubMed] [Google Scholar]
  • 196.Sofuoglu M, et al. , Minocycline attenuates subjective rewarding effects of dextroamphetamine in humans. Psychopharmacology (Berl), 2011. 213(1): p. 61–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Bachtell RK, et al. , Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend, 2017. 180: p. 156–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Arout CA, et al. , Minocycline does not affect experimental pain or addiction-related outcomes in opioid maintained patients. Psychopharmacology (Berl), 2019. 236(10): p. 2857–2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Wu CM and Lai TW, Microglia depletion by PLX3397 has no effect on cocaine-induced behavioral sensitization in male mice. Brain Res, 2021. 1761: p. 147391. [DOI] [PubMed] [Google Scholar]
  • 200.McGrath AG and Briand LA, A potential role for microglia in stress- and drug-induced plasticity in the nucleus accumbens: A mechanism for stress-induced vulnerability to substance use disorder. Neurosci Biobehav Rev, 2019. 107: p. 360–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Ralph MR, et al. , Transplanted suprachiasmatic nucleus determines circadian period. Science, 1990. 247(4945): p. 975–8. [DOI] [PubMed] [Google Scholar]
  • 202.Buhr ED and Takahashi JS, Molecular components of the Mammalian circadian clock. Handb Exp Pharmacol, 2013(217): p. 3–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Partch CL, Green CB, and Takahashi JS, Molecular architecture of the mammalian circadian clock. Trends Cell Biol, 2014. 24(2): p. 90–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Rea MA, Light increases Fos-related protein immunoreactivity in the rat suprachiasmatic nuclei. Brain Res Bull, 1989. 23(6): p. 577–81. [DOI] [PubMed] [Google Scholar]
  • 205.Bennett MR and Schwartz WJ, Are glia among the cells that express immunoreactive c-Fos in the suprachiasmatic nucleus? Neuroreport, 1994. 5(14): p. 1737–40. [DOI] [PubMed] [Google Scholar]
  • 206.Prolo LM, Takahashi JS, and Herzog ED, Circadian rhythm generation and entrainment in astrocytes. J Neurosci, 2005. 25(2): p. 404–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yagita K, et al. , Real-time monitoring of circadian clock oscillations in primary cultures of mammalian cells using Tol2 transposon-mediated gene transfer strategy. BMC Biotechnol, 2010. 10: p. 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Serviere J and Lavialle M, Astrocytes in the mammalian circadian clock: putative roles. Prog Brain Res, 1996. 111: p. 57–73. [DOI] [PubMed] [Google Scholar]
  • 209.van den Pol AN, Finkbeiner SM, and Cornell-Bell AH, Calcium excitability and oscillations in suprachiasmatic nucleus neurons and glia in vitro. J Neurosci, 1992. 12(7): p. 2648–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Shinohara K, et al. , Circadian release of excitatory amino acids in the suprachiasmatic nucleus culture is Ca(2+)-independent. Neurosci Res, 2000. 36(3): p. 245–50. [DOI] [PubMed] [Google Scholar]
  • 211.Prosser RA, et al. , A possible glial role in the mammalian circadian clock. Brain Res, 1994. 643(1–2): p. 296–301. [DOI] [PubMed] [Google Scholar]
  • 212.Lavialle M and Serviere J, Circadian fluctuations in GFAP distribution in the Syrian hamster suprachiasmatic nucleus. Neuroreport, 1993. 4(11): p. 1243–6. [DOI] [PubMed] [Google Scholar]
  • 213.Schwartz WJ and Gainer H, Suprachiasmatic nucleus: use of 14C-labeled deoxyglucose uptake as a functional marker. Science, 1977. 197(4308): p. 1089–91. [DOI] [PubMed] [Google Scholar]
  • 214.Brown AM and Ransom BR, Astrocyte glycogen and brain energy metabolism. Glia, 2007. 55(12): p. 1263–71. [DOI] [PubMed] [Google Scholar]
  • 215.Lehre KP and Danbolt NC, The number of glutamate transporter subtype molecules at glutamatergic synapses: chemical and stereological quantification in young adult rat brain. J Neurosci, 1998. 18(21): p. 8751–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Sweeney AM, et al. , PAR1 activation induces rapid changes in glutamate uptake and astrocyte morphology. Sci Rep, 2017. 7: p. 43606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Beaule C, et al. , Circadian modulation of gene expression, but not glutamate uptake, in mouse and rat cortical astrocytes. PLoS One, 2009. 4(10): p. e7476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Becquet D, et al. , Ultrastructural plasticity in the rat suprachiasmatic nucleus. Possible involvement in clock entrainment. Glia, 2008. 56(3): p. 294–305. [DOI] [PubMed] [Google Scholar]
  • 219.Herzog ED, et al. , Regulating the Suprachiasmatic Nucleus (SCN) Circadian Clockwork: Interplay between Cell-Autonomous and Circuit-Level Mechanisms. Cold Spring Harb Perspect Biol, 2017. 9(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Brancaccio M, et al. , Astrocytes Control Circadian Timekeeping in the Suprachiasmatic Nucleus via Glutamatergic Signaling. Neuron, 2017. 93(6): p. 1420–1435 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Brancaccio M, et al. , Cell-autonomous clock of astrocytes drives circadian behavior in mammals. Science, 2019. 363(6423): p. 187–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Welsh DK, et al. , Individual neurons dissociated from rat suprachiasmatic nucleus express independently phased circadian firing rhythms. Neuron, 1995. 14(4): p. 697–706. [DOI] [PubMed] [Google Scholar]
  • 223.Tosini G and Menaker M, Circadian rhythms in cultured mammalian retina. Science, 1996. 272(5260): p. 419–21. [DOI] [PubMed] [Google Scholar]
  • 224.Ruby NF, Burns DE, and Heller HC, Circadian rhythms in the suprachiasmatic nucleus are temperature-compensated and phase-shifted by heat pulses in vitro. J Neurosci, 1999. 19(19): p. 8630–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Herzog ED and Huckfeldt RM, Circadian entrainment to temperature, but not light, in the isolated suprachiasmatic nucleus. J Neurophysiol, 2003. 90(2): p. 763–70. [DOI] [PubMed] [Google Scholar]
  • 226.Izumo M, Johnson CH, and Yamazaki S, Circadian gene expression in mammalian fibroblasts revealed by real-time luminescence reporting: temperature compensation and damping. Proc Natl Acad Sci U S A, 2003. 100(26): p. 16089–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Granados-Fuentes D, et al. , The suprachiasmatic nucleus entrains, but does not sustain, circadian rhythmicity in the olfactory bulb. J Neurosci, 2004. 24(3): p. 615–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Granados-Fuentes D, et al. , Olfactory bulb neurons express functional, entrainable circadian rhythms. Eur J Neurosci, 2004. 19(4): p. 898–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Lavery DJ, et al. , Circadian expression of the steroid 15 alpha-hydroxylase (Cyp2a4) and coumarin 7-hydroxylase (Cyp2a5) genes in mouse liver is regulated by the PAR leucine zipper transcription factor DBP. Mol Cell Biol, 1999. 19(10): p. 6488–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Arjona A and Sarkar DK, Circadian oscillations of clock genes, cytolytic factors, and cytokines in rat NK cells. J Immunol, 2005. 174(12): p. 7618–24. [DOI] [PubMed] [Google Scholar]
  • 231.Du YZ, et al. , Circadian expression of clock and screening of clock-controlled genes in peripheral lymphocytes of rat. Biochem Biophys Res Commun, 2005. 336(4): p. 1069–73. [DOI] [PubMed] [Google Scholar]
  • 232.Durgan DJ, et al. , The intrinsic circadian clock within the cardiomyocyte. Am J Physiol Heart Circ Physiol, 2005. 289(4): p. H1530–41. [DOI] [PubMed] [Google Scholar]
  • 233.Chalmers JA, et al. , Vascular circadian rhythms in a mouse vascular smooth muscle cell line (Movas-1). Am J Physiol Regul Integr Comp Physiol, 2008. 295(5): p. R1529–38. [DOI] [PubMed] [Google Scholar]
  • 234.Pando MP, et al. , Phenotypic rescue of a peripheral clock genetic defect via SCN hierarchical dominance. Cell, 2002. 110(1): p. 107–17. [DOI] [PubMed] [Google Scholar]
  • 235.Balsalobre A, Marcacci L, and Schibler U, Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Curr Biol, 2000. 10(20): p. 1291–4. [DOI] [PubMed] [Google Scholar]
  • 236.Yamazaki S, et al. , Resetting central and peripheral circadian oscillators in transgenic rats. Science, 2000. 288(5466): p. 682–5. [DOI] [PubMed] [Google Scholar]
  • 237.Silver R, et al. , A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms. Nature, 1996. 382(6594): p. 810–3. [DOI] [PubMed] [Google Scholar]
  • 238.Aton SJ and Herzog ED, Come together, right…now: synchronization of rhythms in a mammalian circadian clock. Neuron, 2005. 48(4): p. 531–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Nagoshi E, et al. , Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell, 2004. 119(5): p. 693–705. [DOI] [PubMed] [Google Scholar]
  • 240.Welsh DK, et al. , Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression. Curr Biol, 2004. 14(24): p. 2289–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Carr AJ and Whitmore D, Imaging of single light-responsive clock cells reveals fluctuating free-running periods. Nat Cell Biol, 2005. 7(3): p. 319–21. [DOI] [PubMed] [Google Scholar]
  • 242.Marpegan L, Krall TJ, and Herzog ED, Vasoactive intestinal polypeptide entrains circadian rhythms in astrocytes. J Biol Rhythms, 2009. 24(2): p. 135–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Gall C, Seroogy KB, and Brecha N, Distribution of VIP- and NPY-like immunoreactivities in rat main olfactory bulb. Brain Res, 1986. 374(2): p. 389–94. [DOI] [PubMed] [Google Scholar]
  • 244.Sims KB, et al. , Vasoactive intestinal polypeptide (VIP) in mouse and rat brain: an immunocytochemical study. Brain Res, 1980. 186(1): p. 165–83. [DOI] [PubMed] [Google Scholar]
  • 245.Okamoto S, et al. , Localization of vasoactive intestinal peptide (VIP) messenger RNA (mRNA) in amacrine cells of rat retina. Curr Eye Res, 1992. 11(7): p. 711–5. [DOI] [PubMed] [Google Scholar]
  • 246.Maywood ES, et al. , A diversity of paracrine signals sustains molecular circadian cycling in suprachiasmatic nucleus circuits. Proc Natl Acad Sci U S A, 2011. 108(34): p. 14306–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Chun LE, et al. , Variations in Phase and Amplitude of Rhythmic Clock Gene Expression across Prefrontal Cortex, Hippocampus, Amygdala, and Hypothalamic Paraventricular and Suprachiasmatic Nuclei of Male and Female Rats. J Biol Rhythms, 2015. 30(5): p. 417–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Debski KJ, et al. , The circadian dynamics of the hippocampal transcriptome and proteome is altered in experimental temporal lobe epilepsy. Sci Adv, 2020. 6(41). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Quigg M, et al. , Temporal distribution of partial seizures: comparison of an animal model with human partial epilepsy. Ann Neurol, 1998. 43(6): p. 748–55. [DOI] [PubMed] [Google Scholar]
  • 250.Stephan FK and Kovacevic NS, Multiple retention deficit in passive avoidance in rats is eliminated by suprachiasmatic lesions. Behav Biol, 1978. 22(4): p. 456–62. [DOI] [PubMed] [Google Scholar]
  • 251.Devan BD, et al. , Circadian phase-shifted rats show normal acquisition but impaired long-term retention of place information in the water task. Neurobiol Learn Mem, 2001. 75(1): p. 51–62. [DOI] [PubMed] [Google Scholar]
  • 252.Gerstner JR, et al. , Cycling behavior and memory formation. J Neurosci, 2009. 29(41): p. 12824–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Ruby NF, et al. , Hippocampal-dependent learning requires a functional circadian system. Proc Natl Acad Sci U S A, 2008. 105(40): p. 15593–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Shimizu K, et al. , SCOP/PHLPP1beta mediates circadian regulation of long-term recognition memory. Nat Commun, 2016. 7: p. 12926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Smarr BL, et al. , A time to remember: the role of circadian clocks in learning and memory. Behav Neurosci, 2014. 128(3): p. 283–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Snider KH, et al. , Modulation of learning and memory by the targeted deletion of the circadian clock gene Bmal1 in forebrain circuits. Behav Brain Res, 2016. 308: p. 222–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Rawashdeh O, Parsons R, and Maronde E, Clocking In Time to Gate Memory Processes: The Circadian Clock Is Part of the Ins and Outs of Memory. Neural Plast, 2018. 2018: p. 6238989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Chaudhury D and Colwell CS, Circadian modulation of learning and memory in fear-conditioned mice. Behav Brain Res, 2002. 133(1): p. 95–108. [DOI] [PubMed] [Google Scholar]
  • 259.Barnes CA, et al. , Circadian rhythm of synaptic excitability in rat and monkey central nervous system. Science, 1977. 197(4298): p. 91–2. [DOI] [PubMed] [Google Scholar]
  • 260.Cauller LJ, Boulos Z, and Goddard GV, Circadian rhythms in hippocampal responsiveness to perforant path stimulation and their relation to behavioral state. Brain Res, 1985. 329(1–2): p. 117–30. [DOI] [PubMed] [Google Scholar]
  • 261.West MO and Deadwyler SA, Circadian modulation of granule cell response to perforant path synaptic input in the rat. Neuroscience, 1980. 5(9): p. 1597–602. [DOI] [PubMed] [Google Scholar]
  • 262.Harris KM and Teyler TJ, Age differences in a circadian influence on hippocampal LTP. Brain Res, 1983. 261(1): p. 69–73. [DOI] [PubMed] [Google Scholar]
  • 263.McCauley JP, et al. , Circadian Modulation of Neurons and Astrocytes Controls Synaptic Plasticity in Hippocampal Area CA1. Cell Rep, 2020. 33(2): p. 108255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Papouin T, et al. , Septal Cholinergic Neuromodulation Tunes the Astrocyte-Dependent Gating of Hippocampal NMDA Receptors to Wakefulness. Neuron, 2017. 94(4): p. 840–854 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Diotel N, et al. , Steroid Transport, Local Synthesis, and Signaling within the Brain: Roles in Neurogenesis, Neuroprotection, and Sexual Behaviors. Front Neurosci, 2018. 12: p. 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Colgin LL, et al. , Frequency of gamma oscillations routes flow of information in the hippocampus. Nature, 2009. 462(7271): p. 353–7. [DOI] [PubMed] [Google Scholar]
  • 267.Zheng C, et al. , Fast Gamma Rhythms in the Hippocampus Promote Encoding of Novel Object-Place Pairings. eNeuro, 2016. 3(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Lamothe-Molina P, Franzelin A, Auksutat L, Laprell L, Alhbeck J, Kneussel M, Engel AK., Morellini F, Oertner TG, cFos ensembles in the dentate gyrus rapidly segregate over time and do not form a stable map of space, in bioRxiv. 2020. [Google Scholar]
  • 269.Boison D, Chen JF, and Fredholm BB, Adenosine signaling and function in glial cells. Cell Death Differ, 2010. 17(7): p. 1071–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Tsunematsu T, et al. , Region-Specific and State-Dependent Astrocyte Ca(2+) Dynamics during the Sleep-Wake Cycle in Mice. J Neurosci, 2021. 41(25): p. 5440–5452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Bojarskaite L, et al. , Astrocytic Ca(2+) signaling is reduced during sleep and is involved in the regulation of slow wave sleep. Nat Commun, 2020. 11(1): p. 3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Ingiosi AM, et al. , A Role for Astroglial Calcium in Mammalian Sleep and Sleep Regulation. Curr Biol, 2020. 30(22): p. 4373–4383 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Zeisel A, et al. , Molecular Architecture of the Mouse Nervous System. Cell, 2018. 174(4): p. 999–1014 e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Bayraktar OA, et al. , Astrocyte layers in the mammalian cerebral cortex revealed by a single-cell in situ transcriptomic map. Nat Neurosci, 2020. 23(4): p. 500–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Fonken LK, et al. , Diminished circadian rhythms in hippocampal microglia may contribute to age-related neuroinflammatory sensitization. Neurobiol Aging, 2016. 47: p. 102–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Fonken LK, et al. , Microglia inflammatory responses are controlled by an intrinsic circadian clock. Brain Behav Immun, 2015. 45: p. 171–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Graykowski D and Cudaback E, Don’t know what you got till it’s gone: microglial depletion and neurodegeneration. Neural Regen Res, 2021. 16(10): p. 1921–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Liu WW, et al. , BMAL1 regulation of microglia-mediated neuroinflammation in MPTP-induced Parkinson’s disease mouse model. FASEB J, 2020. 34(5): p. 6570–6581. [DOI] [PubMed] [Google Scholar]
  • 279.Griffin P, et al. , Circadian clock protein Rev-erbalpha regulates neuroinflammation. Proc Natl Acad Sci U S A, 2019. 116(11): p. 5102–5107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Wang XL, et al. , Deficiency of the Circadian Clock Gene Bmal1 Reduces Microglial Immunometabolism. Front Immunol, 2020. 11: p. 586399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Xie Y, et al. , Chronic sleep fragmentation shares similar pathogenesis with neurodegenerative diseases: Endosome-autophagosome-lysosome pathway dysfunction and microglia-mediated neuroinflammation. CNS Neurosci Ther, 2020. 26(2): p. 215–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Sominsky L, et al. , Microglial ablation in rats disrupts the circadian system. FASEB J, 2021. 35(2): p. e21195. [DOI] [PubMed] [Google Scholar]
  • 283.Corsi G, et al. , Microglia modulate hippocampal synaptic transmission and sleep duration along the light/dark cycle. Glia, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Nakanishi H, et al. , Microglial circadian clock regulation of microglial structural complexity, dendritic spine density and inflammatory response. Neurochem Int, 2021. 142: p. 104905. [DOI] [PubMed] [Google Scholar]
  • 285.Hayashi Y, et al. , The intrinsic microglial molecular clock controls synaptic strength via the circadian expression of cathepsin S. Sci Rep, 2013. 3: p. 2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Stowell RD, et al. , Noradrenergic signaling in the wakeful state inhibits microglial surveillance and synaptic plasticity in the mouse visual cortex. Nat Neurosci, 2019. 22(11): p. 1782–1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Choudhury ME, et al. , Phagocytic elimination of synapses by microglia during sleep. Glia, 2020. 68(1): p. 44–59. [DOI] [PubMed] [Google Scholar]
  • 288.Wadhwa M, et al. , Inhibiting the microglia activation improves the spatial memory and adult neurogenesis in rat hippocampus during 48 h of sleep deprivation. J Neuroinflammation, 2017. 14(1): p. 222. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 289.Besedovsky L, et al. , Signs of enhanced sleep and sleep-associated memory processing following the anti-inflammatory antibiotic minocycline in men. J Psychopharmacol, 2017. 31(2): p. 204–210. [DOI] [PubMed] [Google Scholar]
  • 290.Nonaka K, Nakazawa Y, and Kotorii T, Effects of antibiotics, minocycline and ampicillin, on human sleep. Brain Res, 1983. 288(1–2): p. 253–9. [DOI] [PubMed] [Google Scholar]
  • 291.Wisor JP and Clegern WC, Quantification of short-term slow wave sleep homeostasis and its disruption by minocycline in the laboratory mouse. Neurosci Lett, 2011. 490(3): p. 165–9. [DOI] [PubMed] [Google Scholar]
  • 292.Wisor JP, Schmidt MA, and Clegern WC, Evidence for neuroinflammatory and microglial changes in the cerebral response to sleep loss. Sleep, 2011. 34(3): p. 261–72. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES