Previous Article in Journal
Distribution and Levels of Insulin-like Growth Factor 2 Receptor Across Mouse Brain Cell Types
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Astrocytic Receptor Systems of the Basal Ganglia

1
Institute of Human Anatomy, Department of Neurosciences, University of Padova, 35121 Padua, Italy
2
Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35131 Padua, Italy
3
Movement Disorders Unit, Neurology Clinic, University Hospital of Padova, 35128 Padua, Italy
*
Author to whom correspondence should be addressed.
Receptors 2026, 5(1), 2; https://doi.org/10.3390/receptors5010002
Submission received: 10 June 2025 / Revised: 2 September 2025 / Accepted: 4 December 2025 / Published: 23 December 2025

Abstract

Astrocytes are increasingly recognized as active participants of synaptic communication, yet their role in the basal ganglia circuitry remains poorly defined. Emerging evidence indicates that astrocytes in this region express a diverse array of neurotransmitter receptors thought to regulate intracellular calcium signaling, gliotransmitter release, synaptic plasticity, and neuroimmune responses. However, the literature is limited by methodological variability and a pronounced focus on the striatum, with comparatively little data on other basal ganglia nuclei. This review aims to organize the current literature on astrocytic receptor systems within the basal ganglia, including dopaminergic (D1–D5), glutamatergic (AMPA, NMDA, mGluRs), GABAergic (GABA-A, GABA-B), purinergic (P1, P2), and adrenergic (α, β) receptors. By organizing receptor-specific findings across basal ganglia structures, this review provides a foundation for future investigations into astrocytic function in this complex neural network.

1. Introduction

The basal ganglia are a group of deep cerebral nuclei implicated in the pathophysiology of various neurodegenerative and neuropsychiatric conditions [1,2]. In health, functional subdivisions and parallel processing loops allow the basal ganglia to support sensorimotor, associative, and limbic processes [3,4,5,6,7,8,9,10,11]. Accordingly, pathological changes within the basal ganglia give rise to a broad spectrum of brain-based disorders. While neurodegeneration in the various nuclei of the region is thought to trigger conditions such as Parkinson’s Disease (PD) [12], Huntington’s disease (HD) [13], multiple systems atrophy (MSA) [14], and progressive supranuclear palsy (PSP) [15], disrupted basal ganglia function has also been linked to several psychiatric conditions. Obsessive–compulsive disorder (OCD) involves altered corticostriatal connectivity within limbic circuits [16,17], while substance use disorders show both hypo- and hyperactivity in the ventral striatum [18,19,20]. Moreover, structural changes are observed in schizophrenia and bipolar disorder [21,22], and dopaminergic dysregulation within the basal ganglia is implicated in major depressive disorder (MDD) and generalized anxiety disorder (GAD) [23,24,25].
Despite extensive research across anatomical, clinical, and functional domains, our understanding of the basal ganglia circuitry remains incomplete. Key questions remain regarding their functional organization, cellular regulation, and the mechanisms driving dysfunction in neurological and psychiatric conditions. One possible bottleneck in answering these remaining questions is the lack of a clear understanding of astrocytic involvements in the basal ganglia circuitry.
While astrocytes were initially viewed as mere supportive cells, researchers have begun to appreciate their active participation in the central nervous system (CNS)’s processing [26]. The tripartite synapse model is illustrated in Figure 1 and describes CNS synaptic transmission in the context of pre- and postsynaptic neurons as well as perisynaptic astrocytic processes. Araque et al. [27] specify that astrocytes actively detect synaptic activity, release gliotransmitters and regulate the extracellular environment. Astrocytes express a diverse array of neurotransmitter receptors, including those for glutamate, GABA, dopamine, noradrenaline and purines such as ATP and adenosine. These include both fast-acting ionotropic receptors and slower-acting metabotropic G protein-coupled receptors (GPCRs) [28,29]. The effects of these receptors are multifaceted, ranging from inducing morphological changes to triggering intracellular excitability through calcium signaling [30,31,32]. Although astrocytes do not depolarize like neurons, they respond to synaptic activity with transient elevations in intracellular Ca2+ concentration [33]. While minimal input typically induces rapid localized Ca2+ responses confined to astrocytic microdomains, more intense neuronal activity can evoke slower Ca2+ events in the soma [34,35,36]. It has been proposed that somatic Ca2+ signals reflect the integration of activity from multiple synaptic inputs [34,37]. Astrocytes form functionally interconnected networks with individual astrocytes contacting as many as 100,000 synapses [38,39,40]. These synapses can be independently regulated though the synapse-specific release of gliotransmitters but also allow for the integrated coordination of neurons and astrocytes [38,41,42,43,44]. Ultimately, astrocytes influence synapse turnover, neuroinflammatory responses and the neural microenvironment [45,46,47,48,49].
Astrocytes also seem to be involved in the pathogenesis and pathophysiology of various CNS disorders [50,51]. Indeed, the loss of normal astrocyte functioning may be a primary driver of neurodegeneration [52,53]. Animal models of HD suggest a causal role of enhanced astrocytic calcium release [54], while a loss of astrocytic GABA release might be responsible for the tonic inhibition of striatal neurons in HD [55]. In Parkinson’s disease, α-synuclein has been linked to astrocyte dysfunction, triggering the release of proinflammatory cytokines and chemokines [56]. Moreover, mutations or deletions in the DJ-1 gene, which are known to cause a rare form of autosomal recessive parkinsonism [57], have been shown to exert neuroprotective effects through an astrocyte-mediated mechanism [58]. Studies on MDD consistently report a reduction in astroglial density [59], and emerging evidence suggests that the therapeutic effects of antidepressants may depend on modifying of astrocytic function, potentially reversing the glial deficits commonly observed in MDD patients [60,61]. Molecular and functional abnormalities of astrocytes have also been reported in schizophrenia and seem to critically affect the pathogenesis through neurodevelopmental and homeostatic processes [50]. Recent evidence points to a central role for astrocytes in OCD, implicating dysfunctions in their regulation of glutamate and GABA homeostasis [62,63,64,65].
Given the emerging role of astrocytes in pathophysiology and healthy brain function, the cells involvements in the basal ganglia circuitry should be thoroughly investigated. The region’s intricate connectivity indicates that astrocytes may contribute substantially to circuit homeostasis, with their mechanisms potentially contributing to complex basal ganglia motor and non-motor pathologies that are not yet fully understood. Although individual studies have examined the functional integration and receptor expression of astrocytes within this circuitry, the field still lacks a cohesive framework of astrocytic receptor systems to guide future research. To address this gap, this review aims to synthesize an overview of receptor-specific findings across the basal ganglia structures. Advancing our understanding of bidirectional interactions between neurons and astrocytes may offer key insights into the mechanisms underlying neuroinflammation, neuromodulation and synaptic transmission in health and disease. By providing a microanatomical background, this review sets the stage for future breakthroughs in theoretical understanding of the basal ganglia circuitry as well as translational applications. Astrocytic receptors might reveal novel mechanisms of intercellular communication and serve as therapeutic targets for neurological as well as neuropsychiatric disorders. The following sections will explore the basal ganglia’s most relevant astrocytic receptor systems based on the currently available literature.

2. Dopaminergic System

Dopamine receptors are GPCRs with diverse downstream effects. They can be split up into two broad categories D1-like (D1, D5) receptors, coupled to Gs/Golf proteins and D2-like (D2, D3, D4) receptors, coupled to Gi/o proteins. Functionally, D1-like receptors are known to stimulate adenylyl cyclase, leading to increased cyclic adenosine monophosphate (cAMP) levels, the activation of protein kinase A and CREB phosphorylation [66]. D2-like receptors, on the other hand, exert opposite effects [67]. Thus, the effects of D1-like receptor stimulation are generally excitatory and those of D2-like receptor stimulation inhibitory. A wide body of literature explores the expression of dopamine receptors within the basal ganglia. While most studies are focused on the striatum, there are also reports of astrocytic dopamine receptors in the GP [68], STh [69] and SN [29,70]. Studies exploring the expression of dopaminergic receptors on basal ganglia astrocytes are listed in Table 1.
Reuss et al. [71] found no constitutive expression of D1 receptors (D1Rs) in striatal and mesencephalic astrocytes, however, observed an upregulation upon fibroblast growth factor 2 administration. This finding stands in contrast to other studies showing a clear expression of D1Rs in the striatum [29,71,72] and mesencephalon [29,70]. Striatal astrocytes seem to follow the same reactivity pattern of their neuronal counterparts [70], suggesting that astrocytes might engage in the same circuit-specific signaling observed in basal ganglia neurons [83]. Interestingly, striatal D1Rs seem to be of a different size than their neuronal counterparts [29]. In the substantia nigra reticulata (SNr), a particularly strong expression of astrocytic D1Rs in combination with relatively weaker neuronal expression might suggest that the astrocytic receptors are the target of dendritically released dopamine [70]. In the nucleus accumbens (NAcc) of freely behaving mice and in slices, astrocytic D1R stimulation increased intracellular Ca2+ concentrations, stimulated ATP/adenosine release, and depressed excitatory synaptic transmission [84].
While there was early evidence for D2 receptor (D2R) mRNA in the rat striatum, initial studies failed to find proof for the expression or function of astrocytic D2R proteins [29,72]. Later, studies of synaptic suppression through astrocytes delivered functional evidence for the expression of astrocytic D2Rs in the striatum. The preadministration of a selective D2R agonist could prevent metabolically or chemogenetically induced depressions of striatal neurotransmission [85]. By now there is ample evidence of the presence of astrocytic D2Rs in the human and rodent basal ganglia [29,68,69,71,73,85]. Astrocytic D2R expression in the basal ganglia appears to be partially regulated by Dysbindin-1 (Dys1A), as Dys1A knockout mice show increased D2R levels in the globus pallidus externus (GPe) and striatum, but not in the prefrontal cortex [68]. Shao et al. [73] observed that mice lacking astrocytic D2Rs showed hyperreactivity to immune stimuli and a significantly increased vulnerability of nigral dopaminergic neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced neurotoxicity. The study suggests that astrocytic D2Rs suppress neuroinflammation through an αB-crystallin-dependent mechanism. Additionally, a wide body of evidence suggests that astrocytic D2Rs are involved in the regulation of synaptic transmission [82,85].
Similarly, Astrocytic D3 receptors (D3Rs) seem to play a crucial role in neuroinflammation. While the receptor is not expressed in microglia, a clustered pattern of expression was observed in astroglia [74]. Clustered patterns of expression promote the interaction of various cell-surface proteins and facilitate intracellular signaling [86]. Similarly to D2Rs, MPTP models of Parkinson’s disease suggest that astrocytic D3R deficiencies exacerbate inflammatory responses and neuronal loss [74,75]. Specifically, it seems likely that D3R signaling is involved in the activation of microglia and astrocytes as well as in the regulation of the functional phenotype of microglial cells. The mechanistic and temporal details of this effect are still a topic of discussion [74,75,87,88]. While some observed a protective role of D3R agonists in parkinsonian animals [74,88], others observed therapeutic effects of D3R antagonists [75]. However, what does seem certain is that astrocytic D3R dysregulation exacerbates the loss of dopaminergic neurons in the nigrostriatal pathway.
Few studies have investigated the expression of D4 and D5 receptors (D4R; D5R) in the basal ganglia. Evidence suggests that both receptors are expressed in the striatum as well as the mesencephalon [29,76,77]. Interestingly, examining all five dopamine receptor subtypes in the basal ganglia, Miyazaki et al. [29] observed the strongest immunoreactivity for D4Rs. While investigating the developmental interaction of brain-derived neurotrophic factor (BDNF) and dopaminergic input in the neostriatum, Brito et al. [77] observed that BDNF selectively regulates astrocytic D5Rs. This effect was not observed for D1Rs or neuronal receptors.
Astrocytic dopamine receptors in the basal ganglia seem to engage in heteromer formations. Cervetto et al. [78,79] were the first to demonstrate receptor-receptor interactions and heteromerization of astrocytic A2A and D2Rs in the rat striatum. Later, these findings were confirmed by in situ proximity ligation assays (PLA). Interestingly, while almost all D2Rs immunoprecipitated together with A2ARs, this was only true for a fraction of A2ARs [80]. These findings seem contradictory to Cervetto et al. ‘s [78,79] functional observation that astrocytic A2AR activation was only consequential under D2R coactivation. The complex down-stream interactions and functional outcomes of the receptor heteromer are excellently reviewed elsewhere [89,90], but in short, the A2A-D2R heteromers control the release of glutamate from striatal astrocytes. Analyses of post-mortem human tissue showed compelling early evidence for the existence of A2A-D2R heteromers also on astrocytes in the human STh [69]. Co-immunoprecipitation and PLA evidence suggests the heteromerization of D2Rs also with oxytocin (OT) receptors in the rat striatum [81]. Moreover, Amato et al. [82] confirmed the existence of A2A-D2-OT receptor complex with ternary membrane structure. While all three receptors interact to regulate the astrocytic release of glutamate, the D2R appears to be the main controlling factor. OT receptors seem D2-facilitating and A2ARs exert inhibitory control on both D2Rs and OT receptors [78,79,81,82].

3. Glutamatergic System

Astrocytic glutamate receptors are broadly classified into two main subtypes, the ionotropic (iGluRs) and the metabotropic (mGluRs) receptors, each of which is characterized by distinct structure and signaling mechanisms that lead to different functional outcomes. Studies exploring the expression of glutamatergic receptors on basal ganglia astrocytes are listed in Table 2.

3.1. Ionotropic Glutamate Receptors

The subreceptors of Ionotropic Glutamate Receptors are ligand-gated channels where Glutamate binding directly opens the channel. The astrocytic iGluRs of the basal ganglia express NMDA receptors (NMDARs), AMPA receptors (AMPARs), and Kainate receptors (KARs), nomenclature that reflects the types of synthetic agonists that activate them. The NMDA receptors are highly permeable receptors to Ca2+, and their activation is dependent on glycine/D-serine, which acts as a co-agonist, allowing astrocytes to influence synaptic plasticity and excitotoxicity [109]. Studies on astrocytic NMDARs have shown that, compared to neuronal NMDARs, they rarely contain an Mg2+ block, which has been argued to contribute towards activation at resting membrane potentials [110]. Palygin et al. [91] have shown that astrocytic NMDARs predominantly express the subunits GluN1/GluN2B/GluN3A, which confer the reduced Mg2+ sensitivity and the increased Ca2+ permeability.
Furthermore, astrocytic processes ensheath synapses, positioning NMDARs to detect spillover glutamate that escapes synaptic reuptake by EAAT1/2 transporters [111,112]. When high-frequency neuronal activity occurs, synaptic glutamate spills over to perisynaptic zones, leading to activation of the astrocytic NMDARs and hence triggering Ca2+ influx that results in release of gliotransmitters (e.g., ATP, D-serine) that regulate synaptic plasticity [41,92]. Studies have shown that the release of D-serine potentiates neuronal NMDARs and enhances LTP in the striatal MSNs [92], while depending on the receptor subtypes, ATP/glutamate release from the astrocytes can presynaptically inhibit or facilitate neurotransmitter release [113]. Studies of the cortico-striatal synapses show that by controlling extracellular glutamate levels, astrocytic NMDARs regulate STDP [28].
On the other hand, AMPA receptors have been found to be present in abundance in astrocytes, where unlike neurons, they express calcium-permeable AMPARs lacking the GluA2 subunit, which ensures high Ca2+ permeability [114,115]. As shown by Höft et al. [93], rapid transient Ca2+ influx in response to glutamate over-spill in the synapses is mediated by GluA1/GluA4 homomers, while Sun et al. [94] showed that striatal astrocytes present GluA2-lacking receptors, which enable Ca2-dependent gliotransmitter release. This wave-like propagation of Ca2+ signals through astrocytic networks facilitates long-range modulation of synaptic activity in the striatum and nucleus accumbens [93]. The regulation of AMPARs in astrocytes seems to have an activity-dependent feature, where high-frequency stimulation increases their expression on the astrocytic surface and leads to enhanced glutamate buffering in the basal ganglia system [95,96,116]. The role of the astrocytic AMPARs has been implicated further in glutamate homeostasis in the basal ganglia by expressing high-affinity excitatory amino acid transporters (EAAT1/2; GLAST/GLT-1), crucial in maintaining extracellular glutamate concentrations within physiological ranges. This feature is achieved through AMPAR-mediated depolarization of astrocytes, which increases glutamate transport via EAATs [97]. Impaired astrocytic glutamate uptake can lead to elevated extracellular glutamate levels and increased neuronal excitability, as demonstrated in studies focusing on hippocampal astrocytes [94,117]. Once glutamate is taken up by astrocytes, it is converted to glutamine and shuttled back to neurons for GABA and glutamate synthesis, completing the metabolic glutamate-glutamine cycle that maintains the excitatory/inhibitory balance in the brain [118]. Furthermore, studies show that AMPARs activation stimulates glycolysis in astrocytes and increases lactate production, serving as an energy substrate for neurons during high synaptic activity [119]. Astrocytic AMPA receptor activation can lead to ATP release, which is subsequently converted to adenosine by ectonucleotidases like CD73. This adenosine can then modulate synaptic plasticity in the striatum by acting on neuronal A2A receptors, influencing processes such as LTD [89,120,121].
Unlike AMPAR, most Kainate receptors (KARs), except GluK1 homomers, show limited Ca2+ permeability [122], exhibiting rapid desensitization kinetics, with time constants in the range of a few milliseconds, which can prevent sustained depolarization during prolonged glutamate exposure [123,124]. Astrocytic KARs in the basal ganglia predominantly consist of subunits that either mediate ionotropic effects like the influx of Ca2+ and Na+ (GluK1-GluK3 *GluR5-7* subunits) or modulate receptor kinetics when coassembled (GluK4-GluK5 *KA1-2* subunits) [125]. Astrocytic glutamate transporters, particularly EAAT2, play a crucial role in maintaining extracellular glutamate homeostasis. EAAT2 is responsible for approximately 90% of glutamate uptake, effectively preventing excitotoxicity by clearing excess glutamate from the synaptic cleft. The expression of EAAT2 in astrocytes is dynamically regulated and can be upregulated in response to increased synaptic activity, thereby enhancing glutamate clearance capacity [126,127]. Furthermore, glutamate uptake by astrocytes is closely linked to their metabolic activity. The process of glutamate transport is coupled with sodium influx, which stimulates the Na+/K+-ATPase pump, leading to increased glycolytic activity within astrocytes. This metabolic shift results in the production and release of lactate, which serves as an energy substrate for neurons [128]. Although the role of KARs in astrocytic signaling within the basal ganglia remains less well defined than that of AMPARs or NMDARs, evidence suggests that KARs contribute to synaptic modulation in this region. Jin and Smith [98] demonstrated that presynaptic KAR activation in the globus pallidus reduces GABA release, pointing to a regulatory role in inhibitory transmission. While direct evidence of astrocytic KAR involvement in striatal LTD is limited, the presence of KARs in glial cells and their known ability to modulate glutamate uptake and cellular metabolism suggest a potential, yet underexplored, contribution to synaptic plasticity. Cavaccini et al. [99] demonstrated that high-frequency stimulation (HFS) leads to the activation of astrocytic metabotropic glutamate receptor 5 (mGluR5), resulting in increased intracellular calcium levels in striatal astrocytes. This calcium signaling is essential for the induction of LTD in direct-pathway MSNs. The study further revealed that this form of LTD is dependent on the activation of adenosine A1 receptors on presynaptic terminals, suggesting that astrocyte-derived adenosine modulates glutamatergic transmission to facilitate LTD. These findings highlight a mechanism wherein astrocytic activity influences synaptic plasticity through purinergic signaling pathways [121].

3.2. Metabotropic Glutamate Receptors

The metabotropic glutamate receptors are G protein-coupled subreceptors that indirectly gate channels through the production of secondary messengers [129]. In the astrocytes of the basal ganglia, the mGluR are mostly of the subcategory of Gorup I and Group II, while Group III mGluRs are mostly found presynaptically on neurons, with some evidence for mGluR4/8 localized to perisynaptic astrocytic processes in the subthalamic nucleus playing a role in glutamate clearance [106,107,108]. These regional patterns of astrocytic mGluR expression in the basal ganglia is characteristic also for Group I mGluRs, usually found in striatal astrocytes [100], and Group II mGluRs predominantly expressed in the substantia nigra and globus pallidus [102]. During early postnatal development, astrocytic expression of group I metabotropic glutamate receptors, particularly mGluR5, is transiently elevated and plays a crucial role in synaptic maturation and glutamate homeostasis. In mice, mGluR5 expression in astrocytes peaks around postnatal day 7 and declines markedly after the second postnatal week, becoming nearly undetectable in adulthood [130]. This temporal expression pattern supports a role for astrocytic mGluR5 in the developmental tuning of corticostriatal circuits.
Functionally, activation of mGluR5 in astrocytes during this developmental period is linked to Gq protein-coupled signaling cascades, leading to increased intracellular Ca2+ levels [131]. These Ca2+ transients regulate the release of gliotransmitters and contribute to the modulation of synaptic strength and plasticity.
Moreover, mGluR5 signaling has been shown to regulate the expression and function of the glutamate transporter GLT-1 (EAAT2), enhancing glutamate clearance from the synaptic cleft and thereby maintaining extracellular glutamate concentrations within physiological limits [132]. These mechanisms are especially relevant in the basal ganglia, where astrocytic glutamate handling is critical for preventing excitotoxicity and refining synaptic signaling during early development.
Activation of astrocytic Group I mGlu5 triggers Ca2+ oscillations and gliotransmitter release, as shown by Cavaccini et al. [99], by application of mGluR agonist DHPG or cortical stimulation. This Ca2+ burst results in ATP release and conversion to adenosine, which mediates presynaptic A1R LTD in the direct pathway synapses. On the other hand, astrocytic mGlu5 in the basal ganglia circuits have been implicated to play a role in LTP via astrocyte-mediated glutamate release acting on neuronal mGluRs [83]. This receptor also plays a role in dopamine release via P2Y1 receptors through mGluR5-driven ATP [84]. In a study by Spampinato et al. [101], the authors found that blocking astrocytic mGlu5 reduces pro-inflammatory cytokine release, such as IL-6 and IL-8, and protects neurons from excitotoxicity. A similar function has been established for Group II mGluRs, particularly mGluR3, which, by modulating and reducing cAMP levels, promotes the release of pro-inflammatory cytokines such as TNF-α in Parkinson’s disease models. This signaling pathway is implicated in neuroinflammation, a key feature in neurodegenerative diseases [102]. The neuroprotective role of mGlu3 has been further established in studies that show that mGlu3 signaling induces anti-oxidant and anti-inflammatory gene expression in striatal glia [103], while genetic loss of the receptor worsens nigrostriatal damage and microglial activation in PD models [104]. A study by Matute et al. [105] demonstrates that activation of Group II mGluRs enhances glutamate uptake by astrocytes, contributing to the prevention of excitotoxicity. This process plays a neuroprotective role by regulating glutamate levels in the striatum and other regions of the central nervous system [133].

4. GABAergic System

Early studies on GABAergic receptors done by Fraser et al. [134] showed functional ionotropic GABA-A receptors in striatal astrocytes, characterized by chloride currents. Building on this work, Lee et al. [135] confirmed the presence of subunit compositions such as α1, β2/3, in rodent models of basal ganglia astrocytes. High GABA-A receptor density, linked to tonic inhibition regulation, has been found in astrocytes in the striatum [136]. Aside from the ionotropic GABARs, studies also report the expression of metabotropic GABA-B receptors across the astrocytes of the basal ganglia, especially in the circuits involving substantia nigra and globus pallidus, where they modulate cAMP levels via Gi/o coupling and influence the dopaminergic neuronal activity [137,138]. Studies focusing on astrocyte cultures show that GABA-A receptor activation results in rapid currents and evocation of astrocytic Ca2+ signals by activation of voltage-gated Ca2+ channels, type L and T, leading to Ca2+ entry [139]. Such findings imply that astrocytic GABA-A receptors can utilize intracellular Ca and signaling pathways through depolarization-activated channels or indirect metabotropic cascade [140]. As shown by Liu et al. [139] GABA-A receptors are localized on astrocyte membranes, including the soma, processes, and endfeet, near synapses, suggesting their critical role in maintaining homeostasis in the nervous system. Similarly, astrocytic GABA-B receptors are mostly expressed on the somata and processes of the astrocytes [138,141]. Striatal astrocytes abundantly express GABA-B1 and GABA-B2 transcripts, where GABA-B1 protein has been detected on Aldh1L1+ striatal astrocytes [142]. Activation of these receptors through GABA and GABA-B agonist baclofen produces slow intracellular Ca2+ elevations through the IP3, evoking Ca signals [142]. Furthermore, astrocytic GABA transporters, mainly GAT-3, within BG regulate extracellular GABA and prevent over-inhibition [143,144]. As shown by Jin et al. [145] blocking astrocytic GATs leads to increase in extracellular GABA, tonically activating neuronal GABA-A and GABA-B receptors. Striatal astrocytes’ GABA uptake has been shown to significantly limit tonic GABA inhibition of dopamine release [146,147]. For example, in Parkinson’s models, dysregulated GABA uptake by striatal astrocytes through downregulation of GAT-3 is associated with reduced dopamine release and motor deficits [146]. On the other hand, astrocytic GABA-B receptors in the BG suppress glutamate release, leading to LTD in corticostriatal synapses [148]. Table 3 presents an overview of studies that have examined the GABAergic system on basal ganglia astrocytes.

5. Purinergic System

Purinergic receptors are broadly classified into two main families based on the type of ligand they bind: P1 and P2 receptors. P1 receptors, which include A1, A2A, A2B, and A3, are GPCRs that respond to the nucleoside adenosine. A1 and A3 receptors (A1R; A3R) are primarily coupled to Gi proteins, resulting in the inhibition of adenylyl cyclase. In contrast, A2A and A2B receptors (A2AR; A2BR) couple to Gs or Golf proteins, which activate adenylyl cyclase and promote the production of cAMP. A2BRs require significantly higher concentrations of adenosine to influence cAMP signaling but can also couple with Gq and G12 proteins [149,150]. The P2 receptor family binds primarily to adenosine triphosphate (ATP) and adenosine diphosphate (ADP) and is divided into two subtypes based on their mechanism of action [151,152]. P2X receptors (P2X1R to P2X7R) function as ATP-sensitive ion channels [153], whereas P2Y receptors (P2Y1R, P2Y2R, P2Y4R, P2Y6R, and P2Y11R to P2Y14R) are GPCRs that respond to various nucleotides including ATP and ADP [154]. Evidence for the expression of P1 and P2 receptors is listed in Table 4 and Table 5, respectively.

5.1. P1 Receptor System

Astrocytic A1Rs have been described in mouse embryonic primary astrocyte cultures and the adult brain of mice and rats [49,155,156,167]. In striatal astrocytes, they appear to modulate phospholipase C (PLC) activation. El-Etr et al. [156,167] reported that A1Rs do not directly regulate PLC, but instead enhance noradrenaline- or carbamylcholine-induced PLC signaling by increasing extracellular glutamate and subsequently activating mGluRs. In contrast, Biber et al. [155] observed direct PLC potentiation via the Gβγ subunit of Gi/o proteins. However, this effect was not observed in striatal astrocytes where the study reports only low levels A1R mRNA and protein expression. The discrepancy may be due to differences in the experimental models employed. Biber et al. [155] used adult rat tissue, while El-Etr et al. [167] also studied embryonic mouse striatal cultures. More recently, striatal astrocytic A1Rs have been shown to regulate neuroinflammatory factors in response to endotoxins. While a specific ablation of astrocytic A1Rs prevented early glial reactivity and reduced neuroinflammation, the stimulation of Gi signaling in A1R-deficient astrocytes could restore inflammation [49].
Studies have shown the expression of A2ARs on astrocytes in the striatum, and subthalamic nucleus and in the rodents as well as human brain [69,80,149,157,158,159,160,168,169]. In vitro studies showed that selective A2A antagonists are able to abolish astrocytic reactivity induced by basic fibroblast growth factor (bFGF). However, they also show that A2A agonists by itself are not sufficient to trigger astrogliosis, suggesting that A2ARs act in concert with other bFGF-related mechanisms [157]. Basal ganglia activity is modulated by A2ARs via glutamate-dependent mechanisms. In the short-term, astrocytic A2ARs regulate glutamate reuptake through the modulation of Na+/K+-ATPases (NKAs) [158]. Long-term, the A2AR triggers a cAMP pathway resulting in the decrease in both glutamate transporter 1 (GLT-I) and glutamate-aspartate transporter (GLAST) expression [168]. Moreover, the absence of astrocytic A2ARs results in the upregulation of the NMDA-R 2B subunit and the downregulation AMPA receptors in the NAcc [169]. A2AR antagonists prevent the effects of various glutamate uptake inhibitors that otherwise drastically increase extracellular glutamate levels [159].
Evidence on the astrocytic expression of A2BRs in the basal ganglia is very limited but suggests that A2BRs stimulate cAMP accumulation in primary rat astrocytes [161]. Additionally, there is evidence that A2BRs modulate the release of interleukin-6 (IL-6) in the mouse striatum in vitro and in vivo [162]. While the increments in IL-6 concentration are likely caused by astrocytic release, the study could not exclude a neuronal origin of the cytokine [162,170].
Astrocytic A3Rs exert dual actions on rat striatal astrocytes in culture [163,171]. At sub-threshold concentrations, selective agonists induce morphological changes characterized by stress fibres and numerous cell protrusions. While these changes proved cell-protective, the observed reduction in spontaneous apoptosis was completely counteracted by a selective A3R antagonist. Strikingly, higher concentrations of the same agonists induced cell death in both rat astrocytes and human astrocytoma cells.

5.2. P2 Receptor Systems

Centemeri et al. [30] were the first to characterize the astrocytic Ca2+ response to purinergic nucleotides within the rodent striatum. Through fluorescence spectroscopy the group found that ATP induces fast and transient elevations of cytosolic Ca2+ levels by triggering the discharge of intracellularly stored Ca2+ from striatal astrocytes. The response was mediated by receptors belonging to the P2Y-receptor family and independent of ionotropic P2X-channels. In separate studies, the group around Franke et al. [164,172] found genetic evidence for the expression of all P2X receptor subtypes P2X1-7 as well as P2Y receptor subtypes P2Y1,2,4,6,12. However, the localization of these receptors via double immunofluorescence with receptor-specific and GFAP antibodies proved more complex. Non-injured astrocytes express P2X2-4Rs on GFAP positive processes, with only P2X4Rs appearing also on cell bodies. Interestingly, upon mechanical injury, P2X receptors significantly upscale their expression with P2X2-4 subtypes expressed on astrocytic fibres and cell bodies. Moreover, a previously non-existent expression of P2X1,5,7Rs is now evident. While P2X1Rs and P2X7Rs are confined to fibres and cell bodies, respectively, P2X5Rs are expressed on both [165,172].
A similar pattern was observed for P2Y receptors. Astrocytes in the untreated rat striatum and NAcc displayed a pronounced expression of P2Y1Rs on fibres and cell bodies [164,166]. However, showed no immunoreactivity for receptor subtypes P2Y2,6,12, and only a weak reactivity for P2Y4Rs confined to astrocytic fibres [164]. In contrast, four days after a mechanical injury, almost all the examined (P2Y1,2,4,6) receptor subtypes were clearly expressed on GFAP-positive astrocytes. While P2Y1Rs remained the dominant phenotype, a significant general upregulation was evident where only P2Y12Rs remained absent. Lastly, triple-labeling for P2Y1 and P2X3 receptors as well as GFAP revealed the receptors’ co-expression on the same GFAP-positive astrocyte in the injured rat NAcc [164].
P2 receptors seem to regulate the astroglial response to injury in striatal tissue. While ATP analogous substances up-regulate astroglial responses to injury, P2 antagonists reduce astrocytic proliferation and furthermore, counteract the upregulation caused by ATP analogues [31]. Specifically, the mitogenic effects include the appearance of hypertrophic astrocytes, elongated and thickened astrocytic processes as well as an increase in astrocytic GFAP expression. This reaction is evident for both P2X and P2Y receptor agonists but seems stronger for P2Y agonists than P2X agonists [32,173,174]. However, reasonable uncertainties remain around the roles of specific receptor subtypes within these processes. While the involvement of P2Y1Rs is well established [32,164], the participation of P2Y2, P2Y4, and P2Y6 subtypes appears less likely [172]. The roles of P2Y11, P2Y12, P2Y13, and P2Y14 remain unclear. Although there is limited in vivo evidence of P2Y12R mediated astrogliosis [32], later studies failed to detect this receptor on striatal astrocytes [164]. The astrocytic expression of P2Y11, P2Y13, and P2Y14 receptors has not yet been studied in the basal ganglia. In terms of P2X receptors, in vivo evidence suggests the involvement of homomeric P2X1,3Rs as well as the heteromeric P2X4/6R in mediating astrogliosis within the NAcc [32,172]. Beyond their role in regulating astroglial responses to brain injury, P2 receptors in the basal ganglia may also influence neural adaptations associated with drug sensitization. Repeated administration of d-amphetamine (AMPH) in rats induces both behavioral sensitization and astrogliosis, along with increased expression of P2Y1Rs on astrocytes in the striatum and nucleus accumbens. Notably, Franke et al. [166] reported that pre-treatment with a P2 receptor antagonist inhibited the development of behavioral sensitization, AMPH-induced astrogliosis, and the associated upregulation of P2Y1R expression.

6. Adrenergic System

Similarly to the expression of adrenergic receptors on astrocytes in the cerebral cortex, studies have reported their expression in the basal ganglia [175,176]. In a study on cultured striatal astrocytes by Giaume et al. [177], the authors found expression of both α1- and β-AR, which respond to α1 agonists with transient Ca2+ elevations [178]. On the other hand, astrocytic heterogeneity is evident when compared to recent in situ RNA-seq studies that found lack of astrocytic α1A expression in the ventral midbrain, specifically SN and the ventral tegmental area, with almost no Ca2+ responses to norepinephrine [179]. However, β- and α2-ARs may still be expressed by astrocytes of the SN, but their functional role in this region is unclear.
Furthermore, autoradiographic studies show that striatal astrocytes carry α1- and β-adrenoceptors [180], which in cultures exhibit norepinephrine-stimulated phosphoinositide turnover [181], while transient Ca2+ spikes in striatal astrocytes caused by α1-agonists have been confirmed in functional assays [178]. Additionally, more than 85% of the astrocytes in the striatum express β-adrenoceptors, most of which are β2 subtypes over β1 [180]. Stimulation of astrocytic α1-Ars in the striatum has been shown to result in enhanced glutamate uptake, indicative of their presynaptic localization [176].
However, contradictions in the literature can be found regarding β2-AR expression on astrocytes, while some studies emphasize their role in glial energetics [182], Hertz et al. [176] argue β2-AR expression on astrocytes to be uncertain. When it comes to α1-AR, studies find that activation of this subreceptor reliably triggers transient intracellular Ca2+ elevations in astrocytes [183], increasing glycogen synthesis and mitochondrial oxidative metabolism [176], supporting energy storage during low activity. Interestingly, NE signaling can also prime astrocytes to release gliotransmitters (e.g., ATP, D-serine) that influence local circuits. For example, astrocytic NE action has been shown to modulate inhibitory networks; α1-driven astrocyte Ca2+ waves can facilitate GABAergic inhibition in nearby neurons [183].
On another note, chronic exposures to drugs, such as cocaine, have been linked to disrupting astrocytic noradrenergic signaling and changes in morphology, which influence basal ganglia circuits involved in reward and addictive behaviour [175]. However, concrete links between astrocytic ARs and basal ganglia disorders await further study. An overview of the available literature on the astrocytic adrenergic receptor expression in the basal ganglia is presented in Table 6.

7. Discussion

The neurophysiology of the basal ganglia is regulated by a network of astrocytic receptors, which play an active role in this region’s complex functions. The localization of these receptors varies across subregions, and emerging studies are increasingly exploring their precise roles. Most of the studies address the striatum where astrocyte processes envelop synapses and express multiple dopamine receptors (D1, D3–D5) [29], as well as glutamate receptors and transporters, GABA_A and GABA_B receptors [139], adenosine A_1/A_2A receptors, and adrenergic receptors (α_1A, α_2A, β_1) [176]. On the other hand, studies on the substantia nigra show exceptionally strong immunoreactivity for the D1R [70]. High expression of GABA transporter GAT-3 to clear GABA has been found in the GPe [184], participating in the circuit control. Here, the astrocytic GAT3 expression dynamically changes with behavior: increased GAT3 during habitual learning was necessary for astrocyte-driven shifts toward goal-directed behavior. On the other hand, the subthalamic nucleus is gaining more interest in research, where high-frequency stimulation in PD models modulates astrocytic NF-κB signaling and cytokine release in GP, linking astrocyte activation to improved motor outcomes [185]. Furthermore, astrocytic receptors often interact with each other and with neuronal signaling. For example, A2A–D2 heteromer in striatal astrocyte processes co-localizes and physically interact [80]. In addition, astrocytic D1R engagement may couple to cAMP or Ca2+, but its impact on astrocyte physiology is unknown. Similarly, metabotropic GABA_B activation clearly raises astrocytic Ca2+, but how this shapes BG network activity is yet unclear. Another evident gap that remains in the literature is considering the subpopulations of astrocytes. Satellite astrocytes near cholinergic interneurons of the BG may have distinct receptors influencing network activity [186]. Also underexplored remain the dynamics of receptor interactions, in particular heteromer formation and anatomical distribution. Finally, the physiology of downstream pathways such as IP_3/Ca2+, cAMP/PKA, NF-κB, glycogen metabolism, linking receptor activity to astrocytic output, remains to be detailed.
While we have focused on the most extensively studied astrocytic receptors within the BG-dopaminergic, glutamatergic, GABAergic, purinergic, and adrenergic—this review acknowledges that numerous receptor families remain insufficiently explored. For example, notably absent from this review are astrocytic endocannabinoid receptors (CB), which play a critical role in BG physiology. Although neuronal CB1 receptors have been extensively investigated in modulating synaptic plasticity, astrocytic expression and function of cannabinoid receptors are less understood and often context-dependent [187,188,189]. Similarly, serotonin, acetylcholine, and histamine receptors expressed on astrocytes may influence BG function [190,191,192], but detailed studies in this area are sparse. These underexplored receptor systems offer opportunities for future studies, especially in understanding how diverse astrocytic receptors may coordinate to modulate BG output.
Furthermore, the study of astrocytic receptor function in the BG is met with several technical and conceptual challenges. One major limitation is the heterogeneity of astrocyte populations, both across and within basal ganglia nuclei [193]. Receptor expression can vary significantly depending on the developmental stage, disease state, and even local microenvironment [194]. Additionally, differentiating astrocyte-specific receptor function from neuronal counterparts remains difficult without highly selective tools [195]. Conventional pharmacological methods often lack the precision needed to isolate astrocytic effects. [196]. Recent technological advances such as the use of astrocyte-specific viral vectors, genetically encoded calcium indicators, and cell-type-specific optogenetic, offer some promising potential [197,198,199,200].
Looking forward, future studies should aim to clarify how astrocytic receptors shape local circuit computations and behavioral outputs, particularly in disease contexts such as Parkinson’s and Huntington’s disease. Investigating the dynamic regulation of astrocytic receptor expression and signaling in response to dopaminergic or glutamatergic dysfunctions may reveal new avenues for therapeutic targets. Furthermore, understanding how astrocytes coordinate with microglia and endothelial cells to maintain homeostasis in basal ganglia networks remains a key area of interest.

8. Conclusions

In conclusion, astrocytic receptors are indispensable components of basal ganglia circuitry. Despite significant advances, much remains to be understood about their cell-type specificity, functional roles, and contributions to disease. A more integrative approach, that builds upon incorporating of molecular, cellular, and network-level analyses, could be essential in unlocking the full potential of astrocytic signaling in the basal ganglia.

Author Contributions

Conceptualization, A.T., L.H., A.P. and A.E.; writing—original draft preparation, A.T., L.H.; writing—review and editing, A.T., L.H., A.P. and A.E.; visualization, A.T., L.H.; supervision, A.P., E.S., R.D.C., V.M., A.E.; project administration, A.T., L.H., A.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Figure 1 was created in https://BioRender.com (accessed on 12 June 2025).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAlzheimer’s disease
ADPAdenosine diphosphate
AMPAα-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
AMPHD-amphetamine
ANLSAstrocyte-neuron lactate shuttle
ATPAdenosine triphosphate
BDNFBrain-derived neurotrophic factor
bFGFBasic fibroblast growth factor
cAMPCyclic adenosine monophosphate
CNSCentral nervous system
DADopamine
EAAT2Excitatory amino acid transporter 2
ERKExtracellular signal-regulated kinase
GABAGamma-aminobutyric acid
GADGeneralized anxiety disorder
GATGABA transporter
GLASTGlutamate-aspartate transporter
GLT-1Glutamate transporter 1
GPGlobus pallidus
GPeGlobus pallidus externus
GPiGlobus pallidus internus
GPCRG protein-coupled receptor
HDHuntington’s disease
iGluRIonotropic glutamate receptor
IL-6Interleukin-6
iNOSInducible nitric oxide synthase
KARKainate receptor
LTDLong-term depression
LTPLong-term potentiation
MDDMajor depressive disorder
mGluRMetabotropic glutamate receptor
MPTP1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
MSAMultiple system atrophy
MSNMedium spiny neuron
NAccNucleus accumbens
NKANa+/K+-ATPase
NMDAN-methyl-D-aspartate
OCDObsessive-compulsive disorder
OTOxytocin
PDParkinson’s disease
PLAIn-situ proximity ligation assay
PLCPhospholipase C

References

  1. Macpherson, T.; Hikida, T. Role of Basal Ganglia Neurocircuitry in the Pathology of Psychiatric Disorders. Psychiatry Clin. Neurosci. 2019, 73, 289–301. [Google Scholar] [CrossRef]
  2. Vitanova, K.S.; Stringer, K.M.; Benitez, D.P.; Brenton, J.; Cummings, D.M. Dementia Associated with Disorders of the Basal Ganglia. J. Neurosci. Res. 2019, 97, 1728–1741. [Google Scholar] [CrossRef]
  3. Mink, J.W. The Basal Ganglia: Focused Selection and Inhibition of Competing Motor Programs. Prog. Neurobiol. 1996, 50, 381–425. [Google Scholar] [CrossRef] [PubMed]
  4. Frank, M.J.; Seeberger, L.C.; O’Reilly, R.C. By Carrot or by Stick: Cognitive Reinforcement Learning in Parkinsonism. Science 2004, 306, 1940–1943. [Google Scholar] [CrossRef] [PubMed]
  5. Nambu, A. A New Dynamic Model of the Cortico-Basal Ganglia Loop. In Progress in Brain Research; Elsevier: Amsterdam, The Netherlands, 2004; Volume 143, pp. 461–466. ISBN 978-0-444-51389-2. [Google Scholar]
  6. Ashby, F.G.; Ennis, J.M.; Spiering, B.J. A Neurobiological Theory of Automaticity in Perceptual Categorization. Psychol. Rev. 2007, 114, 632–656. [Google Scholar] [CrossRef]
  7. Schroll, H.; Vitay, J.; Hamker, F.H. Working Memory and Response Selection: A Computational Account of Interactions among Cortico-Basalganglio-Thalamic Loops. Neural Netw. 2012, 26, 59–74. [Google Scholar] [CrossRef] [PubMed]
  8. Trapp, S.; Schroll, H.; Hamker, F.H. Open and Closed Loops: A Computational Approach to Attention and Consciousness. Adv. Cogn. Psychol. 2012, 8, 1–8. [Google Scholar] [CrossRef]
  9. Packard, M.G.; Knowlton, B.J. Learning and Memory Functions of the Basal Ganglia. Annu. Rev. Neurosci. 2002, 25, 563–593. [Google Scholar] [CrossRef]
  10. Schroll, H.; Hamker, F.H. Computational Models of Basal-Ganglia Pathway Functions: Focus on Functional Neuroanatomy. Front. Syst. Neurosci. 2013, 7, 122. [Google Scholar] [CrossRef]
  11. Nambu, A.; Tokuno, H.; Hamada, I.; Kita, H.; Imanishi, M.; Akazawa, T.; Ikeuchi, Y.; Hasegawa, N. Excitatory Cortical Inputs to Pallidal Neurons Via the Subthalamic Nucleus in the Monkey. J. Neurophysiol. 2000, 84, 289–300. [Google Scholar] [CrossRef]
  12. Zhou, Z.D.; Yi, L.X.; Wang, D.Q.; Lim, T.M.; Tan, E.K. Role of Dopamine in the Pathophysiology of Parkinson’s Disease. Transl. Neurodegener. 2023, 12, 44. [Google Scholar] [CrossRef] [PubMed]
  13. Ehrlich, M.E. Huntington’s Disease and the Striatal Medium Spiny Neuron: Cell-Autonomous and Non-Cell-Autonomous Mechanisms of Disease. Neurotherapeutics 2012, 9, 270–284. [Google Scholar] [CrossRef] [PubMed]
  14. Ubhi, K.; Low, P.; Masliah, E. Multiple System Atrophy: A Clinical and Neuropathological Perspective. Trends Neurosci. 2011, 34, 581–590. [Google Scholar] [CrossRef] [PubMed]
  15. Dickson, D.W.; Rademakers, R.; Hutton, M.L. Progressive Supranuclear Palsy: Pathology and Genetics. Brain Pathol. 2007, 17, 74–82. [Google Scholar] [CrossRef]
  16. Whiteside, S.P.; Port, J.D.; Abramowitz, J.S. A Meta–Analysis of Functional Neuroimaging in Obsessive–Compulsive Disorder. Psychiatry Res. Neuroimaging 2004, 132, 69–79. [Google Scholar] [CrossRef]
  17. Maia, T.V.; Cooney, R.E.; Peterson, B.S. The Neural Bases of Obsessive–Compulsive Disorder in Children and Adults. Dev. Psychopathol. 2008, 20, 1251–1283. [Google Scholar] [CrossRef]
  18. Hommer, D.W.; Bjork, J.M.; Gilman, J.M. Imaging Brain Response to Reward in Addictive Disorders. Ann. N. Y. Acad. Sci. 2011, 1216, 50–61. [Google Scholar] [CrossRef]
  19. Diekhof, E.K.; Falkai, P.; Gruber, O. Functional Neuroimaging of Reward Processing and Decision-Making: A Review of Aberrant Motivational and Affective Processing in Addiction and Mood Disorders. Brain Res. Rev. 2008, 59, 164–184. [Google Scholar] [CrossRef]
  20. Leyton, M.; Vezina, P. Striatal Ups and Downs: Their Roles in Vulnerability to Addictions in Humans. Neurosci. Biobehav. Rev. 2013, 37, 1999–2014. [Google Scholar] [CrossRef]
  21. Hokama, H.; Shenton, M.E.; Nestor, P.G.; Kikinis, R.; Levitt, J.J.; Metcalf, D.; Wible, C.G.; O’Donnella, B.F.; Jolesz, F.A.; McCarley, R.W. Caudate, Putamen, and Globus Pallidus Volume in Schizophrenia: A Quantitative MRI Study. Psychiatry Res. Neuroimaging 1995, 61, 209–229. [Google Scholar] [CrossRef]
  22. Womer, F.Y.; Wang, L.; Alpert, K.I.; Smith, M.J.; Csernansky, J.G.; Barch, D.M.; Mamah, D. Basal Ganglia and Thalamic Morphology in Schizophrenia and Bipolar Disorder. Psychiatry Res. Neuroimaging 2014, 223, 75–83. [Google Scholar] [CrossRef] [PubMed]
  23. Cannon, D.M.; Klaver, J.M.; Peck, S.A.; Rallis-Voak, D.; Erickson, K.; Drevets, W.C. Dopamine Type-1 Receptor Binding in Major Depressive Disorder Assessed Using Positron Emission Tomography and [11C]NNC-112. Neuropsychopharmacology 2009, 34, 1277–1287. [Google Scholar] [CrossRef] [PubMed]
  24. Lee, L.-T.; Tsai, H.C.; Chi, M.H.; Chang, W.H.; Chen, K.C.; Lee, I.H.; Chen, P.S.; Yao, W.J.; Chiu, N.T.; Yang, Y.K. Lower Availability of Striatal Dopamine Transporter in Generalized Anxiety Disorder: A Preliminary Two-Ligand SPECT Study. Int. Clin. Psychopharmacol. 2015, 30, 175–178. [Google Scholar] [CrossRef]
  25. Meyer, J.H.; McNeely, H.E.; Sagrati, S.; Boovariwala, A.; Martin, K.; Verhoeff, N.P.L.G.; Wilson, A.A.; Houle, S. Elevated Putamen D2 Receptor Binding Potential in Major Depression with Motor Retardation: An [11C]Raclopride Positron Emission Tomography Study. Am. J. Psychiatry 2006, 163, 1594–1602. [Google Scholar] [CrossRef] [PubMed]
  26. Barres, B.A. The Mystery and Magic of Glia: A Perspective on Their Roles in Health and Disease. Neuron 2008, 60, 430–440. [Google Scholar] [CrossRef]
  27. Araque, A.; Parpura, V.; Sanzgiri, R.P.; Haydon, P.G. Tripartite Synapses: Glia, the Unacknowledged Partner. Trends Neurosci. 1999, 22, 208–215. [Google Scholar] [CrossRef]
  28. Min, R.; Nevian, T. Astrocyte Signaling Controls Spike Timing–Dependent Depression at Neocortical Synapses. Nat. Neurosci. 2012, 15, 746–753. [Google Scholar] [CrossRef]
  29. Miyazaki, I.; Asanuma, M.; Diaz-Corrales, F.J.; Miyoshi, K.; Ogawa, N. Direct Evidence for Expression of Dopamine Receptors in Astrocytes from Basal Ganglia. Brain Res. 2004, 1029, 120–123. [Google Scholar] [CrossRef]
  30. Centemeri, C.; Bolego, C.; Abbracchio, M.P.; Cattabeni, F.; Puglisi, L.; Burnstock, G.; Nicosia, S. Characterization of the Ca2+ Responses Evoked by ATP and Other Nucleotides in Mammalian Brain Astrocytes. Br. J. Pharmacol. 1997, 121, 1700–1706. [Google Scholar] [CrossRef]
  31. Franke, H.; Krügel, U.; Illes, P. P2 Receptor-Mediated Proliferative Effects on Astrocytes in Vivo. Glia 1999, 28, 190–200. [Google Scholar] [CrossRef]
  32. Franke, H.; Krügel, U.; Schmidt, R.; Grosche, J.; Reichenbach, A.; Illes, P. P2 Receptor-Types Involved in Astrogliosis in Vivo. Br. J. Pharmacol. 2001, 134, 1180–1189. [Google Scholar] [CrossRef] [PubMed]
  33. Zorec, R.; Araque, A.; Carmignoto, G.; Haydon, P.G.; Verkhratsky, A.; Parpura, V. Astroglial Excitability and Gliotransmission: An Appraisal of Ca2+ as a Signalling Route. ASN Neuro 2012, 4, AN20110061. [Google Scholar] [CrossRef] [PubMed]
  34. Araque, A.; Carmignoto, G.; Haydon, P.G.; Oliet, S.H.R.; Robitaille, R.; Volterra, A. Gliotransmitters Travel in Time and Space. Neuron 2014, 81, 728–739. [Google Scholar] [CrossRef] [PubMed]
  35. Di Castro, M.A.; Chuquet, J.; Liaudet, N.; Bhaukaurally, K.; Santello, M.; Bouvier, D.; Tiret, P.; Volterra, A. Local Ca2+ Detection and Modulation of Synaptic Release by Astrocytes. Nat. Neurosci. 2011, 14, 1276–1284. [Google Scholar] [CrossRef]
  36. Panatier, A.; Vallée, J.; Haber, M.; Murai, K.K.; Lacaille, J.-C.; Robitaille, R. Astrocytes Are Endogenous Regulators of Basal Transmission at Central Synapses. Cell 2011, 146, 785–798. [Google Scholar] [CrossRef]
  37. Beierlein, M.; Regehr, W.G. Brief Bursts of Parallel Fiber Activity Trigger Calcium Signals in Bergmann Glia. J. Neurosci. 2006, 26, 6958–6967. [Google Scholar] [CrossRef]
  38. Bushong, E.A.; Martone, M.E.; Jones, Y.Z.; Ellisman, M.H. Protoplasmic Astrocytes in CA1 Stratum Radiatum Occupy Separate Anatomical Domains. J. Neurosci. 2002, 22, 183–192. [Google Scholar] [CrossRef]
  39. Cornell-Bell, A.H.; Finkbeiner, S.M.; Cooper, M.S.; Smith, S.J. Glutamate Induces Calcium Waves in Cultured Astrocytes: Long-Range Glial Signaling. Science 1990, 247, 470–473. [Google Scholar] [CrossRef]
  40. Charles, A.C.; Merrill, J.E.; Dirksen, E.R.; Sandersont, M.J. Intercellular Signaling in Glial Cells: Calcium Waves and Oscillations in Response to Mechanical Stimulation and Glutamate. Neuron 1991, 6, 983–992. [Google Scholar] [CrossRef]
  41. Pascual, O.; Casper, K.B.; Kubera, C.; Zhang, J.; Revilla-Sanchez, R.; Sul, J.-Y.; Takano, H.; Moss, S.J.; McCarthy, K.; Haydon, P.G. Astrocytic Purinergic Signaling Coordinates Synaptic Networks. Science 2005, 310, 113–116. [Google Scholar] [CrossRef]
  42. Serrano, A.; Haddjeri, N.; Lacaille, J.-C.; Robitaille, R. GABAergic Network Activation of Glial Cells Underlies Hippocampal Heterosynaptic Depression. J. Neurosci. 2006, 26, 5370–5382. [Google Scholar] [CrossRef] [PubMed]
  43. Covelo, A.; Araque, A. Lateral Regulation of Synaptic Transmission by Astrocytes. Neuroscience 2016, 323, 62–66. [Google Scholar] [CrossRef] [PubMed]
  44. Covelo, A.; Araque, A. Neuronal Activity Determines Distinct Gliotransmitter Release from a Single Astrocyte. eLife 2018, 7, e32237. [Google Scholar] [CrossRef] [PubMed]
  45. Attwell, D.; Buchan, A.M.; Charpak, S.; Lauritzen, M.; MacVicar, B.A.; Newman, E.A. Glial and Neuronal Control of Brain Blood Flow. Nature 2010, 468, 232–243. [Google Scholar] [CrossRef]
  46. Brown, A.M.; Ransom, B.R. Astrocyte Glycogen and Brain Energy Metabolism. Glia 2007, 55, 1263–1271. [Google Scholar] [CrossRef]
  47. Deitmer, J.W. pH Regulation and Acid/Base-Mediated Transport in Glial Cells. In Glial ⇔ Neuronal Signaling; Hatton, G.I., Parpura, V., Eds.; Springer: Boston, MA, USA, 2004; pp. 263–277. ISBN 978-1-4757-1069-4. [Google Scholar]
  48. Simard, M.; Nedergaard, M. The Neurobiology of Glia in the Context of Water and Ion Homeostasis. Neuroscience 2004, 129, 877–896. [Google Scholar] [CrossRef]
  49. Guo, Q.; Gobbo, D.; Zhao, N.; Zhang, H.; Awuku, N.-O.; Liu, Q.; Fang, L.-P.; Gampfer, T.M.; Meyer, M.R.; Zhao, R.; et al. Adenosine Triggers Early Astrocyte Reactivity That Provokes Microglial Responses and Drives the Pathogenesis of Sepsis-Associated Encephalopathy in Mice. Nat. Commun. 2024, 15, 6340. [Google Scholar] [CrossRef]
  50. Notter, T. Astrocytes in Schizophrenia. Brain Neurosci. Adv. 2021, 5, 23982128211009148. [Google Scholar] [CrossRef]
  51. Ben Haim, L.; Carrillo-de Sauvage, M.-A.; Ceyzériat, K.; Escartin, C. Elusive Roles for Reactive Astrocytes in Neurodegenerative Diseases. Front. Cell. Neurosci. 2015, 9, 278. [Google Scholar] [CrossRef]
  52. Brenner, M.; Johnson, A.B.; Boespflug-Tanguy, O.; Rodriguez, D.; Goldman, J.E.; Messing, A. Mutations in GFAP, Encoding Glial Fibrillary Acidic Protein, Are Associated with Alexander Disease. Nat. Genet. 2001, 27, 117–120. [Google Scholar] [CrossRef]
  53. Li, R.; Johnson, A.B.; Salomons, G.; Goldman, J.E.; Naidu, S.; Quinlan, R.; Cree, B.; Ruyle, S.Z.; Banwell, B.; D’Hooghe, M.; et al. Glial Fibrillary Acidic Protein Mutations in Infantile, Juvenile, and Adult Forms of Alexander Disease. Ann. Neurol. 2005, 57, 310–326. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, W.; Reyes, R.C.; Gottipati, M.K.; Lewis, K.; Lesort, M.; Parpura, V.; Gray, M. Enhanced Ca2+-Dependent Glutamate Release from Astrocytes of the BACHD Huntington’s Disease Mouse Model. Neurobiol. Dis. 2013, 58, 192–199. [Google Scholar] [CrossRef] [PubMed]
  55. Wójtowicz, A.M.; Dvorzhak, A.; Semtner, M.; Grantyn, R. Reduced Tonic Inhibition in Striatal Output Neurons from Huntington Mice Due to Loss of Astrocytic GABA Release through GAT-3. Front. Neural Circuits 2013, 7, 188. [Google Scholar] [CrossRef]
  56. Lee, H.-J.; Suk, J.-E.; Patrick, C.; Bae, E.-J.; Cho, J.-H.; Rho, S.; Hwang, D.; Masliah, E.; Lee, S.-J. Direct Transfer of α-Synuclein from Neuron to Astroglia Causes Inflammatory Responses in Synucleinopathies. J. Biol. Chem. 2010, 285, 9262–9272. [Google Scholar] [CrossRef] [PubMed]
  57. Bonifati, V.; Rizzu, P.; Van Baren, M.J.; Schaap, O.; Breedveld, G.J.; Krieger, E.; Dekker, M.C.J.; Squitieri, F.; Ibanez, P.; Joosse, M.; et al. Mutations in the DJ-1 Gene Associated with Autosomal Recessive Early-Onset Parkinsonism. Science 2003, 299, 256–259. [Google Scholar] [CrossRef]
  58. Mullett, S.J.; Hinkle, D.A. DJ-1 Knock-down in Astrocytes Impairs Astrocyte-Mediated Neuroprotection against Rotenone. Neurobiol. Dis. 2009, 33, 28–36. [Google Scholar] [CrossRef]
  59. Wang, Q.; Jie, W.; Liu, J.; Yang, J.; Gao, T. An Astroglial Basis of Major Depressive Disorder? An Overview. Glia 2017, 65, 1227–1250. [Google Scholar] [CrossRef]
  60. Bowley, M.P.; Drevets, W.C.; Öngür, D.; Price, J.L. Low Glial Numbers in the Amygdala in Major Depressive Disorder. Biol. Psychiatry 2002, 52, 404–412. [Google Scholar] [CrossRef]
  61. Peng, L.; Huang, J. Astrocytic 5-HT2B Receptor as in Vitro and in Vivo Target of SSRIs. Recent Patents CNS Drug Discov. 2012, 7, 243–253. [Google Scholar] [CrossRef]
  62. Gonzalez, L.; Bezzi, P. Astrocyte Dysfunctions in Obsessive Compulsive Disorder: Rethinking Neurobiology and Therapeutic Targets. J. Neurochem. 2025, 169, e70092. [Google Scholar] [CrossRef]
  63. Nagai, J.; Yu, X.; Papouin, T.; Cheong, E.; Freeman, M.R.; Monk, K.R.; Hastings, M.H.; Haydon, P.G.; Rowitch, D.; Shaham, S.; et al. Behaviorally Consequential Astrocytic Regulation of Neural Circuits. Neuron 2021, 109, 576–596. [Google Scholar] [CrossRef]
  64. Mederos, S.; Perea, G. GABAergic-Astrocyte Signaling: A Refinement of Inhibitory Brain Networks. Glia 2019, 67, 1842–1851. [Google Scholar] [CrossRef] [PubMed]
  65. Soto, J.S.; Jami-Alahmadi, Y.; Chacon, J.; Moye, S.L.; Diaz-Castro, B.; Wohlschlegel, J.A.; Khakh, B.S. Astrocyte–Neuron Subproteomes and Obsessive–Compulsive Disorder Mechanisms. Nature 2023, 616, 764–773. [Google Scholar] [CrossRef]
  66. Moritz, A.E.; Madaras, N.S.; Rankin, M.L.; Inbody, L.R.; Sibley, D.R. Delineation of G Protein-Coupled Receptor Kinase Phosphorylation Sites within the D1 Dopamine Receptor and Their Roles in Modulating β-Arrestin Binding and Activation. Int. J. Mol. Sci. 2023, 24, 6599. [Google Scholar] [CrossRef]
  67. Beaulieu, J.; Espinoza, S.; Gainetdinov, R.R. Dopamine Receptors—IUPHAR R Eview 13. Br. J. Pharmacol. 2015, 172, 1–23. [Google Scholar] [CrossRef]
  68. Mastrogiacomo, R.; Trigilio, G.; Devroye, C.; Dautan, D.; Ferretti, V.; Losi, G.; Caffino, L.; Orso, G.; Marotta, R.; Maltese, F.; et al. Dysbindin-1A Modulation of Astrocytic Dopamine and Basal Ganglia Dependent Behaviors Relevant to Schizophrenia. Mol. Psychiatry 2022, 27, 4201–4217. [Google Scholar] [CrossRef] [PubMed]
  69. Emmi, A.; Antonini, A.; Sandre, M.; Baldo, A.; Contran, M.; Macchi, V.; Guidolin, D.; Porzionato, A.; De Caro, R. Topography and Distribution of Adenosine A2A and Dopamine D2 Receptors in the Human Subthalamic Nucleus. Front. Neurosci. 2022, 16, 945574. [Google Scholar] [CrossRef]
  70. Nagatomo, K.; Suga, S.; Saitoh, M.; Kogawa, M.; Kobayashi, K.; Yamamoto, Y.; Yamada, K. Dopamine D1 Receptor Immunoreactivity on Fine Processes of GFAP-Positive Astrocytes in the Substantia Nigra Pars Reticulata of Adult Mouse. Front. Neuroanat. 2017, 11, 3. [Google Scholar] [CrossRef]
  71. Reuss, B.; Leung, D.S.Y.; Ohlemeyer, C.; Kettenmann, H.; Unsicker, K. Regionally Distinct Regulation of Astroglial Neurotransmitter Receptors by Fibroblast Growth Factor-2. Mol. Cell. Neurosci. 2000, 16, 42–58. [Google Scholar] [CrossRef]
  72. Zanassi, P.; Paolillo, M.; Montecucco, A.; Avvedimento, E.V.; Schinelli, S. Pharmacological and Molecular Evidence for Dopamine D1 Receptor Expression by Striatal Astrocytes in Culture. J. Neurosci. Res. 1999, 58, 544–552. [Google Scholar] [CrossRef]
  73. Shao, W.; Zhang, S.; Tang, M.; Zhang, X.; Zhou, Z.; Yin, Y.; Zhou, Q.; Huang, Y.; Liu, Y.; Wawrousek, E.; et al. Suppression of Neuroinflammation by Astrocytic Dopamine D2 Receptors via αB-Crystallin. Nature 2013, 494, 90–94. [Google Scholar] [CrossRef]
  74. Montoya, A.; Elgueta, D.; Campos, J.; Chovar, O.; Falcón, P.; Matus, S.; Alfaro, I.; Bono, M.R.; Pacheco, R. Dopamine Receptor D3 Signalling in Astrocytes Promotes Neuroinflammation. J. Neuroinflammation 2019, 16, 258. [Google Scholar] [CrossRef] [PubMed]
  75. Elgueta, D.; Aymerich, M.S.; Contreras, F.; Montoya, A.; Celorrio, M.; Rojo-Bustamante, E.; Riquelme, E.; González, H.; Vásquez, M.; Franco, R.; et al. Pharmacologic Antagonism of Dopamine Receptor D3 Attenuates Neurodegeneration and Motor Impairment in a Mouse Model of Parkinson’s Disease. Neuropharmacology 2017, 113, 110–123. [Google Scholar] [CrossRef] [PubMed]
  76. Svingos, A.L.; Periasamy, S.; Pickel, V.M. Dopamine D4 Receptors Are Strategically Localized for Primary Involvement in the Presynaptic Effects of Dopamine in the Rat Nucleus Accumbens Shell. Ann. N. Y. Acad. Sci. 1999, 877, 679–683. [Google Scholar] [CrossRef] [PubMed]
  77. Brito, V.; Beyer, C.; Küppers, E. BDNF-dependent Stimulation of Dopamine D5 Receptor Expression in Developing Striatal Astrocytes Involves PI3-kinase Signaling. Glia 2004, 46, 284–295. [Google Scholar] [CrossRef]
  78. Cervetto, C.; Venturini, A.; Passalacqua, M.; Guidolin, D.; Genedani, S.; Fuxe, K.; Borroto-Esquela, D.O.; Cortelli, P.; Woods, A.; Maura, G.; et al. A2A-D2 Receptor–Receptor Interaction Modulates Gliotransmitter Release from Striatal Astrocyte Processes. J. Neurochem. 2017, 140, 268–279. [Google Scholar] [CrossRef]
  79. Cervetto, C.; Venturini, A.; Guidolin, D.; Maura, G.; Passalacqua, M.; Tacchetti, C.; Cortelli, P.; Genedani, S.; Candiani, S.; Ramoino, P.; et al. Homocysteine and A2A-D2 Receptor-Receptor Interaction at Striatal Astrocyte Processes. J. Mol. Neurosci. 2018, 65, 456–466. [Google Scholar] [CrossRef]
  80. Pelassa, S.; Guidolin, D.; Venturini, A.; Averna, M.; Frumento, G.; Campanini, L.; Bernardi, R.; Cortelli, P.; Calandra Buonaura, G.; Maura, G.; et al. A2A-D2 Heteromers on Striatal Astrocytes: Biochemical and Biophysical Evidence. Int. J. Mol. Sci. 2019, 20, 2457. [Google Scholar] [CrossRef]
  81. Amato, S.; Averna, M.; Guidolin, D.; Ceccoli, C.; Gatta, E.; Candiani, S.; Pedrazzi, M.; Capraro, M.; Maura, G.; Agnati, L.F.; et al. Heteromerization of Dopamine D2 and Oxytocin Receptor in Adult Striatal Astrocytes. Int. J. Mol. Sci. 2023, 24, 4677. [Google Scholar] [CrossRef]
  82. Amato, S.; Averna, M.; Farsetti, E.; Guidolin, D.; Pedrazzi, M.; Gatta, E.; Candiani, S.; Maura, G.; Agnati, L.F.; Cervetto, C.; et al. Control of Dopamine Signal in High-Order Receptor Complex on Striatal Astrocytes. Int. J. Mol. Sci. 2024, 25, 8610. [Google Scholar] [CrossRef]
  83. Martín, R.; Bajo-Grañeras, R.; Moratalla, R.; Perea, G.; Araque, A. Circuit-Specific Signaling in Astrocyte-Neuron Networks in Basal Ganglia Pathways. Science 2015, 349, 730–734. [Google Scholar] [CrossRef] [PubMed]
  84. Corkrum, M.; Covelo, A.; Lines, J.; Bellocchio, L.; Pisansky, M.; Loke, K.; Quintana, R.; Rothwell, P.E.; Lujan, R.; Marsicano, G.; et al. Dopamine-Evoked Synaptic Regulation in the Nucleus Accumbens Requires Astrocyte Activity. Neuron 2020, 105, 1036–1047.e5. [Google Scholar] [CrossRef] [PubMed]
  85. Adermark, L.; Lagström, O.; Loftén, A.; Licheri, V.; Havenäng, A.; Loi, E.A.; Stomberg, R.; Söderpalm, B.; Domi, A.; Ericson, M. Astrocytes Modulate Extracellular Neurotransmitter Levels and Excitatory Neurotransmission in Dorsolateral Striatum via Dopamine D2 Receptor Signaling. Neuropsychopharmacology 2022, 47, 1493–1502. [Google Scholar] [CrossRef] [PubMed]
  86. Ishii, T.; Ohnuma, K.; Murakami, A.; Takasawa, N.; Kobayashi, S.; Dang, N.H.; Schlossman, S.F.; Morimoto, C. CD26-Mediated Signaling for T Cell Activation Occurs in Lipid Rafts through Its Association with CD45RO. Proc. Natl. Acad. Sci. USA 2001, 98, 12138–12143. [Google Scholar] [CrossRef]
  87. Beier, E.E.; Neal, M.; Alam, G.; Edler, M.; Wu, L.-J.; Richardson, J.R. Alternative Microglial Activation Is Associated with Cessation of Progressive Dopamine Neuron Loss in Mice Systemically Administered Lipopolysaccharide. Neurobiol. Dis. 2017, 108, 115–127. [Google Scholar] [CrossRef]
  88. Ohta, K.; Kuno, S.; Inoue, S.; Ikeda, E.; Fujinami, A.; Ohta, M. The Effect of Dopamine Agonists: The Expression of GDNF, NGF, and BDNF in Cultured Mouse Astrocytes. J. Neurol. Sci. 2010, 291, 12–16. [Google Scholar] [CrossRef]
  89. Cervetto, C.; Maura, G.; Guidolin, D.; Amato, S.; Ceccoli, C.; Agnati, L.F.; Marcoli, M. Striatal Astrocytic A2A-D2 Receptor-Receptor Interactions and Their Role in Neuropsychiatric Disorders. Neuropharmacology 2023, 237, 109636. [Google Scholar] [CrossRef]
  90. Guidolin, D.; Marcoli, M.; Tortorella, C.; Maura, G.; Agnati, L.F. Adenosine A2A-Dopamine D2 Receptor-Receptor Interaction in Neurons and Astrocytes: Evidence and Perspectives. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2020; Volume 169, pp. 247–277. ISBN 978-0-12-817929-1. [Google Scholar]
  91. Palygin, O.; Lalo, U.; Pankratov, Y. Distinct Pharmacological and Functional Properties of NMDA Receptors in Mouse Cortical Astrocytes. Br. J. Pharmacol. 2011, 163, 1755–1766. [Google Scholar] [CrossRef]
  92. Henneberger, C.; Bard, L.; King, C.; Jennings, A.; Rusakov, D.A. NMDA Receptor Activation: Two Targets for Two Co-Agonists. Neurochem. Res. 2013, 38, 1156–1162. [Google Scholar] [CrossRef]
  93. Höft, S.; Griemsmann, S.; Seifert, G.; Steinhäuser, C. Heterogeneity in Expression of Functional Ionotropic Glutamate and GABA Receptors in Astrocytes across Brain Regions: Insights from the Thalamus. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130602. [Google Scholar] [CrossRef]
  94. Sun, D.; Tan, Z.-B.; Sun, X.-D.; Liu, Z.-P.; Chen, W.-B.; Milibari, L.; Ren, X.; Yao, L.-L.; Lee, D.; Shen, C.; et al. Hippocampal Astrocytic Neogenin Regulating Glutamate Uptake, a Critical Pathway for Preventing Epileptic Response. Proc. Natl. Acad. Sci. USA 2021, 118, e2022921118. [Google Scholar] [CrossRef] [PubMed]
  95. Devaraju, P.; Sun, M.-Y.; Myers, T.L.; Lauderdale, K.; Fiacco, T.A. Astrocytic Group I mGluR-Dependent Potentiation of Astrocytic Glutamate and Potassium Uptake. J. Neurophysiol. 2013, 109, 2404–2414. [Google Scholar] [CrossRef] [PubMed]
  96. Tawfik, V.L.; Chang, S.-Y.; Hitti, F.L.; Roberts, D.W.; Leiter, J.C.; Jovanovic, S.; Lee, K.H. Deep Brain Stimulation Results in Local Glutamate and Adenosine Release. Neurosurgery 2010, 67, 367–375. [Google Scholar] [CrossRef] [PubMed]
  97. Sheldon, A.L.; Robinson, M.B. The Role of Glutamate Transporters in Neurodegenerative Diseases and Potential Opportunities for Intervention. Neurochem. Int. 2007, 51, 333–355. [Google Scholar] [CrossRef]
  98. Jin, X.-T.; Smith, Y. Activation of Presynaptic Kainate Receptors Suppresses GABAergic Synaptic Transmission in the Rat Globus Pallidus. Neuroscience 2007, 149, 338–349. [Google Scholar] [CrossRef]
  99. Cavaccini, A.; Durkee, C.; Kofuji, P.; Tonini, R.; Araque, A. Astrocyte Signaling Gates Long-Term Depression at Corticostriatal Synapses of the Direct Pathway. J. Neurosci. 2020, 40, 5757–5768. [Google Scholar] [CrossRef]
  100. Testa, C.M.; Standaert, D.G.; Landwehrmeyer, G.B.; Penney, J.B.; Young, A.B. Differential Expression of mGluR5 Metabotropic Glutamate Receptor mRNA by Rat Striatal Neurons. J. Comp. Neurol. 1995, 354, 241–252. [Google Scholar] [CrossRef]
  101. Spampinato, S.F.; Copani, A.; Nicoletti, F.; Sortino, M.A.; Caraci, F. Metabotropic Glutamate Receptors in Glial Cells: A New Potential Target for Neuroprotection? Front. Mol. Neurosci. 2018, 11, 414. [Google Scholar] [CrossRef]
  102. D’Antoni, S.; Berretta, A.; Bonaccorso, C.M.; Bruno, V.; Aronica, E.; Nicoletti, F.; Catania, M.V. Metabotropic Glutamate Receptors in Glial Cells. Neurochem. Res. 2008, 33, 2436–2443. [Google Scholar] [CrossRef]
  103. Durand, D.; Carniglia, L.; Caruso, C.; Lasaga, M. mGlu3 Receptor and Astrocytes: Partners in Neuroprotection. Neuropharmacology 2013, 66, 1–11. [Google Scholar] [CrossRef]
  104. Di Menna, L.; Alborghetti, M.; De Bartolo, M.I.; Borro, M.; Gentile, G.; Zinni, M.; Bologna, M.; Cutrona, C.; D’Errico, G.; Imbriglio, T.; et al. Preclinical and Clinical Study on Type 3 Metabotropic Glutamate Receptors in Parkinson’s Disease. NPJ Park. Dis. 2025, 11, 9. [Google Scholar] [CrossRef]
  105. Matute, C.; Domercq, M.; Sánchez-Gómez, M. Glutamate-mediated Glial Injury: Mechanisms and Clinical Importance. Glia 2006, 53, 212–224. [Google Scholar] [CrossRef] [PubMed]
  106. Awad, H.; Hubert, G.W.; Smith, Y.; Levey, A.I.; Conn, P.J. Activation of Metabotropic Glutamate Receptor 5 Has Direct Excitatory Effects and Potentiates NMDA Receptor Currents in Neurons of the Subthalamic Nucleus. J. Neurosci. 2000, 20, 7871–7879. [Google Scholar] [CrossRef] [PubMed]
  107. Corti, C.; Aldegheri, L.; Somogyi, P.; Ferraguti, F. Distribution and Synaptic Localisation of the Metabotropic Glutamate Receptor 4 (mGluR4) in the Rodent CNS. Neuroscience 2002, 110, 403–420. [Google Scholar] [CrossRef] [PubMed]
  108. Shen, K.; Johnson, S.W. Group II Metabotropic Glutamate Receptor Modulation of Excitatory Transmission in Rat Subthalamic Nucleus. J. Physiol. 2003, 553, 489–496. [Google Scholar] [CrossRef]
  109. Verkhratsky, A.; Chvátal, A. NMDA Receptors in Astrocytes. Neurochem. Res. 2020, 45, 122–133. [Google Scholar] [CrossRef]
  110. Verkhratsky, A.; Kirchhoff, F. NMDA Receptors in Glia. Neuroscientist 2007, 13, 28–37. [Google Scholar] [CrossRef]
  111. Bergles, D.E.; Jahr, C.E. Synaptic Activation of Glutamate Transporters in Hippocampal Astrocytes. Neuron 1997, 19, 1297–1308. [Google Scholar] [CrossRef]
  112. Lehre, K.P.; Danbolt, N.C. The Number of Glutamate Transporter Subtype Molecules at Glutamatergic Synapses: Chemical and Stereological Quantification in Young Adult Rat Brain. J. Neurosci. 1998, 18, 8751–8757. [Google Scholar] [CrossRef]
  113. Pannasch, U.; Vargová, L.; Reingruber, J.; Ezan, P.; Holcman, D.; Giaume, C.; Syková, E.; Rouach, N. Astroglial Networks Scale Synaptic Activity and Plasticity. Proc. Natl. Acad. Sci. USA 2011, 108, 8467–8472. [Google Scholar] [CrossRef]
  114. Schiffmann, S.N.; Fisone, G.; Moresco, R.; Cunha, R.A.; Ferré, S. Adenosine A2A Receptors and Basal Ganglia Physiology. Prog. Neurobiol. 2007, 83, 277–292. [Google Scholar] [CrossRef] [PubMed]
  115. Seifert, G.; Zhou, M.; Steinhäuser, C. Analysis of AMPA Receptor Properties During Postnatal Development of Mouse Hippocampal Astrocytes. J. Neurophysiol. 1997, 78, 2916–2923. [Google Scholar] [CrossRef] [PubMed]
  116. Lalo, U.; Palygin, O.; Rasooli-Nejad, S.; Andrew, J.; Haydon, P.G.; Pankratov, Y. Exocytosis of ATP From Astrocytes Modulates Phasic and Tonic Inhibition in the Neocortex. PLoS Biol. 2014, 12, e1001747. [Google Scholar] [CrossRef] [PubMed]
  117. Pannasch, U.; Dossi, E.; Ezan, P.; Rouach, N. Astroglial Cx30 Sustains Neuronal Population Bursts Independently of Gap-junction Mediated Biochemical Coupling. Glia 2019, 67, 1104–1112. [Google Scholar] [CrossRef]
  118. Andersen, J.V. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J. Neurochem. 2025, 169, e70029. [Google Scholar] [CrossRef]
  119. Pellerin, L.; Magistretti, P.J. Glutamate Uptake into Astrocytes Stimulates Aerobic Glycolysis: A Mechanism Coupling Neuronal Activity to Glucose Utilization. Proc. Natl. Acad. Sci. USA 1994, 91, 10625–10629. [Google Scholar] [CrossRef]
  120. Lopes, C.R.; Cunha, R.A.; Agostinho, P. Astrocytes and Adenosine A2A Receptors: Active Players in Alzheimer’s Disease. Front. Neurosci. 2021, 15, 666710. [Google Scholar] [CrossRef]
  121. Zhai, S. Emerging Role of Astrocytes in Striatal Synaptic Plasticity. J. Neurosci. 2021, 41, 2088–2090. [Google Scholar] [CrossRef]
  122. LERMA, J. Kainate Receptor Physiology. Curr. Opin. Pharmacol. 2006, 6, 89–97. [Google Scholar] [CrossRef]
  123. Jonas, P. Differences in Ca2+ Permeability of AMPA-Type Glutamate Receptor Channels in Neocortical Neurons Caused by Differential GluR-B Subunit Expression. Neuron 1994, 12, 1281–1289. [Google Scholar] [CrossRef]
  124. Partin, K.M.; Patneau, D.K.; Winters, C.A.; Mayer, M.L.; Buonanno, A. Selective Modulation of Desensitization at AMPA versus Kainate Receptors by Cyclothiazide and Concanavalin A. Neuron 1993, 11, 1069–1082. [Google Scholar] [CrossRef] [PubMed]
  125. Pinheiro, P.; Mulle, C. Kainate Receptors. Cell Tissue Res. 2006, 326, 457–482. [Google Scholar] [CrossRef] [PubMed]
  126. Karki, P.; Smith, K.; Johnson, J.; Aschner, M.; Lee, E.Y. Genetic Dys-Regulation of Astrocytic Glutamate Transporter EAAT2 and Its Implications in Neurological Disorders and Manganese Toxicity. Neurochem. Res. 2015, 40, 380–388. [Google Scholar] [CrossRef] [PubMed]
  127. Lutgen, V.; Narasipura, S.D.; Sharma, A.; Min, S.; Al-Harthi, L. β-Catenin Signaling Positively Regulates Glutamate Uptake and Metabolism in Astrocytes. J. Neuroinflammation 2016, 13, 242. [Google Scholar] [CrossRef] [PubMed]
  128. Weber, B.; Barros, L.F. The Astrocyte: Powerhouse and Recycling Center. Cold Spring Harb. Perspect. Biol. 2015, 7, a020396. [Google Scholar] [CrossRef]
  129. Niswender, C.M.; Conn, P.J. Metabotropic Glutamate Receptors: Physiology, Pharmacology, and Disease. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 295–322. [Google Scholar] [CrossRef]
  130. Sun, W.; McConnell, E.; Pare, J.-F.; Xu, Q.; Chen, M.; Peng, W.; Lovatt, D.; Han, X.; Smith, Y.; Nedergaard, M. Glutamate-Dependent Neuroglial Calcium Signaling Differs Between Young and Adult Brain. Science 2013, 339, 197–200. [Google Scholar] [CrossRef]
  131. Balázs, R.; Miller, S.; Romano, C.; De Vries, A.; Chun, Y.; Cotman, C.W. Metabotropic Glutamate Receptor mGluR5 in Astrocytes: Pharmacological Properties and Agonist Regulation. J. Neurochem. 1997, 69, 151–163. [Google Scholar] [CrossRef]
  132. Higashimori, H.; Morel, L.; Huth, J.; Lindemann, L.; Dulla, C.; Taylor, A.; Freeman, M.; Yang, Y. Astroglial FMRP-Dependent Translational down-Regulation of mGluR5 Underlies Glutamate Transporter GLT1 Dysregulation in the Fragile X Mouse. Hum. Mol. Genet. 2013, 22, 2041–2054. [Google Scholar] [CrossRef]
  133. Yao, H.; Ding, J.; Zhou, F.; Wang, F.; Hu, L.; Sun, T.; Hu, G. Enhancement of Glutamate Uptake Mediates the Neuroprotection Exerted by Activating Group II or III Metabotropic Glutamate Receptors on Astrocytes. J. Neurochem. 2005, 92, 948–961. [Google Scholar] [CrossRef]
  134. Fraser, D.D.; Mudrick-Donnon, L.A.; Macvicar, B.A. Astrocytic GABA Receptors. Glia 1994, 11, 83–93. [Google Scholar] [CrossRef] [PubMed]
  135. Lee, M.; McGeer, E.G.; McGeer, P.L. Mechanisms of GABA Release from Human Astrocytes. Glia 2011, 59, 1600–1611. [Google Scholar] [CrossRef] [PubMed]
  136. Yoon, B.-E.; Jo, S.; Woo, J.; Lee, J.-H.; Kim, T.; Kim, D.; Lee, C.J. The Amount of Astrocytic GABA Positively Correlates with the Degree of Tonic Inhibition in Hippocampal CA1 and Cerebellum. Mol. Brain 2011, 4, 42. [Google Scholar] [CrossRef] [PubMed]
  137. Brickley, S.G.; Mody, I. Extrasynaptic GABAA Receptors: Their Function in the CNS and Implications for Disease. Neuron 2012, 73, 23–34. [Google Scholar] [CrossRef]
  138. Charles, K.J.; Deuchars, J.; Davies, C.H.; Pangalos, M.N. GABAB Receptor Subunit Expression in Glia. Mol. Cell. Neurosci. 2003, 24, 214–223. [Google Scholar] [CrossRef]
  139. Liu, J.; Feng, X.; Wang, Y.; Xia, X.; Zheng, J.C. Astrocytes: GABAceptive and GABAergic Cells in the Brain. Front. Cell. Neurosci. 2022, 16, 892497. [Google Scholar] [CrossRef]
  140. Nilsson, M.; Eriksson, P.S.; Rönnbäck, L.; Hansson, E. GABA Induces Ca2+ Transients in Astrocytes. Neuroscience 1993, 54, 605–614. [Google Scholar] [CrossRef]
  141. Charles, K.J.; Calver, A.R.; Jourdain, S.; Pangalos, M.N. Distribution of a GABAB-like Receptor Protein in the Rat Central Nervous System. Brain Res. 2003, 989, 135–146. [Google Scholar] [CrossRef]
  142. Nagai, J.; Rajbhandari, A.K.; Gangwani, M.R.; Hachisuka, A.; Coppola, G.; Masmanidis, S.C.; Fanselow, M.S.; Khakh, B.S. Hyperactivity with Disrupted Attention by Activation of an Astrocyte Synaptogenic Cue. Cell 2019, 177, 1280–1292.e20. [Google Scholar] [CrossRef]
  143. Beenhakker, M.P.; Huguenard, J.R. Astrocytes as Gatekeepers of GABA B Receptor Function. J. Neurosci. 2010, 30, 15262–15276. [Google Scholar] [CrossRef]
  144. Goubard, V.; Fino, E.; Venance, L. Contribution of Astrocytic Glutamate and GABA Uptake to Corticostriatal Information Processing. J. Physiol. 2011, 589, 2301–2319. [Google Scholar] [CrossRef] [PubMed]
  145. Jin, X.-T.; Galvan, A.; Wichmann, T.; Smith, Y. Localization and Function of GABA Transporters GAT-1 and GAT-3 in the Basal Ganglia. Front. Syst. Neurosci. 2011, 5, 63. [Google Scholar] [CrossRef] [PubMed]
  146. Roberts, B.M.; Lopes, E.F.; Cragg, S.J. Axonal Modulation of Striatal Dopamine Release by Local γ-Aminobutyric Acid (GABA) Signalling. Cells 2021, 10, 709. [Google Scholar] [CrossRef] [PubMed]
  147. Yoon, B.-E.; Woo, J.; Justin Lee , C. Astrocytes as GABA-Ergic and GABA-Ceptive Cells. Neurochem. Res. 2012, 37, 2474–2479. [Google Scholar] [CrossRef]
  148. Galvan, A.; Kuwajima, M.; Smith, Y. Glutamate and GABA Receptors and Transporters in the Basal Ganglia: What Does Their Subsynaptic Localization Reveal about Their Function? Neuroscience 2006, 143, 351–375. [Google Scholar] [CrossRef]
  149. Boison, D.; Chen, J.-F.; Fredholm, B.B. Adenosine Signaling and Function in Glial Cells. Cell Death Differ. 2010, 17, 1071–1082. [Google Scholar] [CrossRef]
  150. Fredholm, B.B.; Arslan, G.; Halldner, L.; Kull, B.; Schulte, G.; Ådén, U.; Svenningsson, P. Adenosine Receptor Signaling in Vitro and in Vivo. Drug Dev. Res. 2001, 52, 274–282. [Google Scholar] [CrossRef]
  151. Burnstock, G.; Kennedy, C. Is There a Basis for Distinguishing Two Types of P2-Purinoceptor? Gen. Pharmacol. Vasc. Syst. 1985, 16, 433–440. [Google Scholar] [CrossRef]
  152. Köles, L.; Leichsenring, A.; Rubini, P.; Illes, P. P2 Receptor Signaling in Neurons and Glial Cells of the Central Nervous System. In Advances in Pharmacology; Elsevier: Amsterdam, The Netherlands, 2011; Volume 61, pp. 441–493. ISBN 978-0-12-385526-8. [Google Scholar]
  153. North, R.A. Molecular Physiology of P2X Receptors. Physiol. Rev. 2002, 82, 1013–1067. [Google Scholar] [CrossRef]
  154. Abbracchio, M.P.; Burnstock, G.; Boeynaems, J.-M.; Barnard, E.A.; Boyer, J.L.; Kennedy, C.; Knight, G.E.; Fumagalli, M.; Gachet, C.; Jacobson, K.A.; et al. International Union of Pharmacology LVIII: Update on the P2Y G Protein-Coupled Nucleotide Receptors: From Molecular Mechanisms and Pathophysiology to Therapy. Pharmacol. Rev. 2006, 58, 281–341. [Google Scholar] [CrossRef]
  155. Biber, K.; Klotz, K.-N.; Berger, M.; Gebicke-Härter, P.J.; Van Calker, D. Adenosine A1 Receptor-Mediated Activation of Phospholipase C in Cultured Astrocytes Depends on the Level of Receptor Expression. J. Neurosci. 1997, 17, 4956–4964. [Google Scholar] [CrossRef] [PubMed]
  156. El-Etr, M.; Marin, P.; Tencé, M.; Delumeau, J.C.; Cordier, J.; Glowinski, J.; Premont, J. 24-Chloroadenosine Potentiates the A1-Adrenergic Activation of Phospholipase C through a Mechanism Involving Arachidonic Acid and Glutamate in Striatal Astrocytes. J. Neurosci. 1992, 12, 1363–1369. [Google Scholar] [CrossRef] [PubMed]
  157. Brambilla, R.; Cottini, L.; Fumagalli, M.; Ceruti, S.; Abbracchio, M.P. Blockade of A2A Adenosine Receptors Prevents Basic Fibroblast Growth Factor-induced Reactive Astrogliosis in Rat Striatal Primary Astrocytes. Glia 2003, 43, 190–194. [Google Scholar] [CrossRef] [PubMed]
  158. Matos, M.; Augusto, E.; Agostinho, P.; Cunha, R.A.; Chen, J.-F. Antagonistic Interaction between Adenosine A2A Receptors and Na+ /K+ -ATPase-α2 Controlling Glutamate Uptake in Astrocytes. J. Neurosci. 2013, 33, 18492–18502. [Google Scholar] [CrossRef]
  159. Pintor, A.; Galluzzo, M.; Grieco, R.; Pèzzola, A.; Reggio, R.; Popoli, P. Adenosine A2A Receptor Antagonists Prevent the Increase in Striatal Glutamate Levels Induced by Glutamate Uptake Inhibitors. J. Neurochem. 2004, 89, 152–156. [Google Scholar] [CrossRef]
  160. Hettinger, B.D.; Lee, A.; Linden, J.; Rosin, D.L. Ultrastructural Localization of Adenosine A2A Receptors Suggests Multiple Cellular Sites for Modulation of GABAergic Neurons in Rat Striatum. J. Comp. Neurol. 2001, 431, 331–346. [Google Scholar] [CrossRef]
  161. Peakman, M.-C.; Hill, S.J. Adenosine A2B-Receptor-Mediated Cyclic AMP Accumulation in Primary Rat Astrocytes. Br. J. Pharmacol. 1994, 111, 191–198. [Google Scholar] [CrossRef]
  162. Vazquez, J.F.; Clement, H.; Sommer, O.; Schulz, E.; Van Calker, D. Local Stimulation of the Adenosine A2B Receptors Induces an Increased Release of IL-6 in Mouse Striatum: An in Vivo Microdialysis Study. J. Neurochem. 2008, 105, 904–909. [Google Scholar] [CrossRef]
  163. Abbracchio, M.P.; Ceruti, S.; Brambilla, R.; Barbieri, D.; Camurri, A.; Franceschi, C.; Giammarioli, A.M.; Jacobson, K.A.; Cattabeni, F.; Malorni, W. Adenosine A3 Receptors and Viability of Astrocytes. Drug Dev. Res. 1998, 45, 379–386. [Google Scholar] [CrossRef]
  164. Franke, H.; Krügel, U.; Grosche, J.; Heine, C.; Härtig, W.; Allgaier, C.; Illes, P. P2Y Receptor Expression on Astrocytes in the Nucleus Accumbens of Rats. Neuroscience 2004, 127, 431–441. [Google Scholar] [CrossRef]
  165. Franke, H.; Krügel, U.; Grosche, J.; Illes, P. Immunoreactivity for Glial Fibrillary Acidic Protein and P2 Receptor Expression on Astrocytes in Vivo. Drug Dev. Res. 2003, 59, 175–189. [Google Scholar] [CrossRef]
  166. Franke, H.; Kittner, H.; Grosche, J.; Illes, P. Enhanced P2Y1 Receptor Expression in the Brain after Sensitisation with D-Amphetamine. Psychopharmacology 2003, 167, 187–194. [Google Scholar] [CrossRef]
  167. El-Etr, M.; Cordier, J.; Glowinski, J.; Premont, J. A Neuroglial Cooperativity Is Required for the Potentiation by 2Ghloroadenosine of the Muscarinic-Sensitive Phospholipase C in the Striatum. J. Neurosci. 1989, 9, 1473–1480. [Google Scholar] [CrossRef]
  168. Matos, M.; Augusto, E.; Santos-Rodrigues, A.D.; Schwarzschild, M.A.; Chen, J.; Cunha, R.A.; Agostinho, P. Adenosine A2A Receptors Modulate Glutamate Uptake in Cultured Astrocytes and Gliosomes. Glia 2012, 60, 702–716. [Google Scholar] [CrossRef]
  169. Matos, M.; Shen, H.-Y.; Augusto, E.; Wang, Y.; Wei, C.J.; Wang, Y.T.; Agostinho, P.; Boison, D.; Cunha, R.A.; Chen, J.-F. Deletion of Adenosine A2A Receptors From Astrocytes Disrupts Glutamate Homeostasis Leading to Psychomotor and Cognitive Impairment: Relevance to Schizophrenia. Biol. Psychiatry 2015, 78, 763–774. [Google Scholar] [CrossRef] [PubMed]
  170. Van Wagoner, N.J.; Benveniste, E.N. Interleukin-6 Expression and Regulation in Astrocytes. J. Neuroimmunol. 1999, 100, 124–139. [Google Scholar] [CrossRef]
  171. Abbracchio, M.P.; Saffrey, M.J.; Ho¨pker, V.; Burnstock, G. Modulation of Astroglial Cell Proliferation by Analogues of Adenosine and ATP in Primary Cultures of Rat Striatum. Neuroscience 1994, 59, 67–76. [Google Scholar] [CrossRef] [PubMed]
  172. Franke, H.; Grosche, J.; Schädlich, H.; Krügel, U.; Allgaier, C.; Illes, P. P2X Receptor Expression on Astrocytes in the Nucleus Accumbens of Rats. Neuroscience 2001, 108, 421–429. [Google Scholar] [CrossRef] [PubMed]
  173. Franke, H.; Krügel, U.; Illes, P. P2Y and P2X Receptor-Mediated Mitogenic Effcts on Astrocytes in Vivo. Br. J. Pharmacol. 1999, 128, 153. [Google Scholar]
  174. Franke, H.; Krügel, U.; Schmidt, R.; Illes, P. P2Y-receptor mediated astrogliosis in vivo. Drug Dev. Res. 2000, 50, 91. [Google Scholar]
  175. Hertz, L.; Chen, Y.; Gibbs, M.; Zang, P.; Peng, L. Astrocytic Adrenoceptors: A Major Drug Target in Neurological and Psychiatric Disorders? Curr. Drug Target-CNS Neurol. Disord. 2004, 3, 239–268. [Google Scholar] [CrossRef] [PubMed]
  176. Hertz, L.; Lovatt, D.; Goldman, S.A.; Nedergaard, M. Adrenoceptors in Brain: Cellular Gene Expression and Effects on Astrocytic Metabolism and [Ca2+]i. Neurochem. Int. 2010, 57, 411–420. [Google Scholar] [CrossRef] [PubMed]
  177. Giaume, C.; Marin, P.; Cordier, J.; Glowinski, J.; Premont, J. Adrenergic Regulation of Intercellular Communications between Cultured Striatal Astrocytes from the Mouse. Proc. Natl. Acad. Sci. USA 1991, 88, 5577–5581. [Google Scholar] [CrossRef]
  178. Delumeau, J.C.; Tencé, M.; Marin, P.; Cordier, J.; Glowinski, J.; Prémont, J. Synergistic Regulation of Cytosolic Ca2+ Concentration by Adenosine and α1-Adrenergic Agonists in Mouse Striatal Astrocytes. Eur. J. Neurosci. 1991, 3, 539–550. [Google Scholar] [CrossRef] [PubMed]
  179. Xin, W.; Schuebel, K.E.; Jair, K.; Cimbro, R.; De Biase, L.M.; Goldman, D.; Bonci, A. Ventral Midbrain Astrocytes Display Unique Physiological Features and Sensitivity to Dopamine D2 Receptor Signaling. Neuropsychopharmacology 2019, 44, 344–355. [Google Scholar] [CrossRef]
  180. Shao, Y.; Sutin, J. Expression of Adrenergic Receptors in Individual Astrocytes and Motor Neurons Isolated from the Adult Rat Brain. Glia 1992, 6, 108–117. [Google Scholar] [CrossRef]
  181. Puig, J.F.; Pacitti, A.J.; Guzman, N.J.; Crews, F.T.; Sumners, C.; Raizada, M.K. A1-Adrenergic Receptors in the Brain: Characterization in Astrocytic Glial Cultures and Comparison with Neuronal Cultures. Brain Res. 1990, 527, 318–325. [Google Scholar] [CrossRef]
  182. Jensen, C.J.; Demol, F.; Bauwens, R.; Kooijman, R.; Massie, A.; Villers, A.; Ris, L.; De Keyser, J. Astrocytic B2 Adrenergic Receptor Gene Deletion Affects Memory in Aged Mice. PLoS ONE 2016, 11, e0164721. [Google Scholar] [CrossRef]
  183. Wahis, J.; Holt, M.G. Astrocytes, Noradrenaline, A1-Adrenoreceptors, and Neuromodulation: Evidence and Unanswered Questions. Front. Cell. Neurosci. 2021, 15, 645691. [Google Scholar] [CrossRef]
  184. Kang, S.; Hong, S.-I.; Kang, S.; Song, M.; Yang, M.A.; Essa, H.; Baker, M.; Lee, J.; Bruce, R.A.; Lee, S.W.; et al. Astrocyte Activities in the External Globus Pallidus Regulate Action-Selection Strategies in Reward-Seeking Behaviors. Sci. Adv. 2023, 9, eadh9239. [Google Scholar] [CrossRef]
  185. Campos, A.C.P.; Kikuchi, D.S.; Paschoa, A.F.N.; Kuroki, M.A.; Fonoff, E.T.; Hamani, C.; Pagano, R.L.; Hernandes, M.S. Unraveling the Role of Astrocytes in Subthalamic Nucleus Deep Brain Stimulation in a Parkinson’s Disease Rat Model. Cell. Mol. Neurobiol. 2020, 40, 939–954. [Google Scholar] [CrossRef] [PubMed]
  186. Stedehouder, J.; Roberts, B.M.; Raina, S.; Bossi, S.; Liu, A.K.L.; Doig, N.M.; McGerty, K.; Magill, P.J.; Parkkinen, L.; Cragg, S.J. Rapid Modulation of Striatal Cholinergic Interneurons and Dopamine Release by Satellite Astrocytes. Nat. Commun. 2024, 15, 10017. [Google Scholar] [CrossRef] [PubMed]
  187. Njoo, C.; Agarwal, N.; Lutz, B.; Kuner, R. The Cannabinoid Receptor CB1 Interacts with the WAVE1 Complex and Plays a Role in Actin Dynamics and Structural Plasticity in Neurons. PLoS Biol. 2015, 13, e1002286. [Google Scholar] [CrossRef] [PubMed]
  188. Tsou, K.; Brown, S.; Sañudo-Peña, M.C.; Mackie, K.; Walker, J.M. Immunohistochemical Distribution of Cannabinoid CB1 Receptors in the Rat Central Nervous System. Neuroscience 1998, 83, 393–411. [Google Scholar] [CrossRef]
  189. Bénard, G.; Massa, F.; Puente, N.; Lourenço, J.; Bellocchio, L.; Soria-Gómez, E.; Matias, I.; Delamarre, A.; Metna-Laurent, M.; Cannich, A.; et al. Mitochondrial CB1 Receptors Regulate Neuronal Energy Metabolism. Nat. Neurosci. 2012, 15, 558–564. [Google Scholar] [CrossRef]
  190. Hösli, L.; Hösli, E.; Schneider, U.; Wiget, W. Evidence for the Existence of Histamine H1- and H2-Receptors on Astrocytes of Cultured Rat Central Nervous System. Neurosci. Lett. 1984, 48, 287–291. [Google Scholar] [CrossRef]
  191. Miyazaki, I.; Asanuma, M. Serotonin 1A Receptors on Astrocytes as a Potential Target for the Treatment of Parkinson’s Disease. Curr. Med. Chem. 2016, 23, 686–700. [Google Scholar] [CrossRef]
  192. Liu, Y.; Zeng, X.; Hui, Y.; Zhu, C.; Wu, J.; Taylor, D.H.; Ji, J.; Fan, W.; Huang, Z.; Hu, J. Activation of A7 Nicotinic Acetylcholine Receptors Protects Astrocytes against Oxidative Stress-Induced Apoptosis: Implications for Parkinson’s Disease. Neuropharmacology 2015, 91, 87–96. [Google Scholar] [CrossRef]
  193. Holt, M.G. Astrocyte Heterogeneity and Interactions with Local Neural Circuits. Essays Biochem. 2023, 67, 93–106. [Google Scholar] [CrossRef]
  194. Rusnakova, V.; Honsa, P.; Dzamba, D.; Ståhlberg, A.; Kubista, M.; Anderova, M. Heterogeneity of Astrocytes: From Development to Injury—Single Cell Gene Expression. PLoS ONE 2013, 8, e69734. [Google Scholar] [CrossRef]
  195. Kruyer, A. Astrocyte Heterogeneity in Regulation of Synaptic Activity. Cells 2022, 11, 3135. [Google Scholar] [CrossRef]
  196. Nimmerjahn, A. Astrocytes Going Live: Advances and Challenges. J. Physiol. 2009, 587, 1639–1647. [Google Scholar] [CrossRef]
  197. Kanemaru, K.; Sekiya, H.; Xu, M.; Satoh, K.; Kitajima, N.; Yoshida, K.; Okubo, Y.; Sasaki, T.; Moritoh, S.; Hasuwa, H.; et al. In Vivo Visualization of Subtle, Transient, and Local Activity of Astrocytes Using an Ultrasensitive Ca2+ Indicator. Cell Rep. 2014, 8, 311–318. [Google Scholar] [CrossRef]
  198. Qin, H.; He, W.; Yang, C.; Li, J.; Jian, T.; Liang, S.; Chen, T.; Feng, H.; Chen, X.; Liao, X.; et al. Monitoring Astrocytic Ca2+ Activity in Freely Behaving Mice. Front. Cell. Neurosci. 2020, 14, 603095. [Google Scholar] [CrossRef]
  199. Losi, G.; Mariotti, L.; Sessolo, M.; Carmignoto, G. New Tools to Study Astrocyte Ca2+ Signal Dynamics in Brain Networks In Vivo. Front. Cell. Neurosci. 2017, 11, 134. [Google Scholar] [CrossRef] [PubMed]
  200. Tong, X.; Shigetomi, E.; Looger, L.L.; Khakh, B.S. Genetically Encoded Calcium Indicators and Astrocyte Calcium Microdomains. Neuroscientist 2013, 19, 274–291. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tripartite synapse between (a) astrocyte, (b) presynaptic neuron and (c) postsynaptic neuron. Neuroactive molecules are released into the synaptic cleft and may bind to receptor proteins expressed on the membrane of the perisynaptic astrocyte [27].
Figure 1. Tripartite synapse between (a) astrocyte, (b) presynaptic neuron and (c) postsynaptic neuron. Neuroactive molecules are released into the synaptic cleft and may bind to receptor proteins expressed on the membrane of the perisynaptic astrocyte [27].
Receptors 05 00002 g001
Table 1. Expression of dopaminergic receptors on basal ganglia astrocytes.
Table 1. Expression of dopaminergic receptors on basal ganglia astrocytes.
ReceptorReferencesBrain Region(s)Species
D1RMiyazaki et al., 2004 [29]Striatum, mesencephalonRat
Mastrogiacomo et al., 2022 [68]Striatum, GPeMouse
Nagatomo et al., 2017 [70]SNrMouse
Reuss et al., 2000 [71]Striatum, GPRat
Zanassi et al., 1999 [72]StriatumRat
D2RMiyazaki et al., 2004 [29]Striatum, mesencephalonRat
Mastrogiacomo et al., 2022 [68]Striatum, GPeMouse
Emmi et al., 2022 [69]SThHuman
Reuss et al., 2000 [71]Striatum, GPRat
Shao et al., 2013 [73]StriatumMouse
D3RMiyazaki et al., 2004 [29]Striatum, mesencephalonRat
Mastrogiacomo et al., 2022 [68]Striatum, GPeMouse
Montoya et al. 2019 [74]Striatum, mesencephalonMouse
Elgueta et al., 2017 [75]Striatum, mesencephalon Mouse
D4RMiyazaki et al., 2004 [29]Striatum, mesencephalonRat
Svingos et al., 1999 [76]NAccRat
D5RMiyazaki et al., 2004 [29]Striatum, mesencephalonRat
Brito et al., 2004 [77]StriatumMouse
A2A-D2RCervetto et al., 2017 [78]StriatumRat
Cervetto et al., 2018 [79]StriatumRat
Pelassa et al., 2019 [80]StriatumRat
D2-OTRAmato et al., 2023 [81]StriatumRat
A2A-D2-OTRAmato et al., 2024 [82]StriatumRat
Table 2. Expression of glutamatergic receptors on basal ganglia astrocytes.
Table 2. Expression of glutamatergic receptors on basal ganglia astrocytes.
ReceptorReferencesBrain Region(s)Species
NMDAMin & Nevian, 2012 [28]Cortico-striatal synapsesMouse
Pascual et al., 2005 [41]Basal ganglia (general)Rat
Palygin et al., 2011 [91]StriatumMouse
Henneberger et al., 2013 [92]StriatumRat
Höft et al., 2014 [93]Striatum, NAccMouse
Sun et al., 2021 [94]StriatumMouse
Devaraju et al., 2013 [95]Basal ganglia (general)Mouse
Tawfik et al., 2010 [96]Basal ganglia (general)Rat
Sheldon & Robinson, 2007 [97]Basal ganglia (general)Rat
KainateJin & Smith, 2007 [98]Globus PallidusRat
Cavaccini et al., 2020 [99]StriatumMouse
mGluR5 (Group I)Martín et al., 2015 [83]Basal ganglia (general)Mouse
Corkrum et al., 2020 [84]NAccMouse
Cavaccini et al., 2020 [99]StriatumMouse
Testa et al., 1995 [100]StriatumRat
Spampinato et al., 2018 [101]Basal ganglia (general)Mouse
mGluR3 (Group II)D’Antoni et al., 2008 [102]SN, GP, StriatumRat
Durand et al., 2013 [103]StriatumMouse
Di Menna et al., 2025 [104]Striatum, SNMouse
Matute et al., 2006 [105]StriatumRat
mGluR4/8 (Group III)Awad et al., 2000 [106]SThRat
Corti et al., 2002 [107]
Shen & Johnson, 2003 [108]
Table 3. Expression of GABAergic receptors on basal ganglia astrocytes.
Table 3. Expression of GABAergic receptors on basal ganglia astrocytes.
ReceptorReferencesBrain Region(s)Species
GABA-αFraser et al., 1994 [134]StriatumRat
Lee et al., 2011 [135]Basal ganglia (general)Rodent
Yoon et al., 2011 [136]StriatumRat
Liu et al., 2022 [139]Astrocyte membranes (soma, processes, endfeet)Rat
GABA-βCharles et al., 2003 [138]Substantia nigra, Globus pallidusRat
Charles et al., 2003 [141]Somata and processes of astrocytesRat
Nagai et al., 2019 [142]StriatumMouse
Jin et al., 2011 [145]Basal ganglia (general)Rat
Roberts et al., 2021 [146]StriatumRat/Mouse
Galvan et al., 2006 [148]Corticostriatal synapsesRat
Table 4. Expression of purinergic P1 receptors on basal ganglia astrocytes.
Table 4. Expression of purinergic P1 receptors on basal ganglia astrocytes.
ReceptorReferencesBrain Region(s)Species
A1RGuo et al., 2024 [49]StriatumMouse
Biber et al., 1997 [155]Striatum, tegmentumRat
El-Etr et al., 1989 [156]StriatumMouse, Rat
A2AREmmi et al., 2022 [69]SThHuman
Pelassa et al., 2019 [80]StriatumRat
Brambilla et al., 2003 [157]StriatumRat
Matos et al., 2013 [158]StriatumMouse
Pintor et al., 2004 [159]Striatum Rat
Hettinger et al., 2001 [160]Dorsolateral striatumRat
A2BRPeakman et al., 1994 [161]Telencephalon, diencephalonRat
Vazquez et al., 2008 [162]StriatumMouse
A3RAbbracchio et al., 1998 [163]StriatumRat
Table 5. Expression of purinergic P2 receptors on basal ganglia astrocytes.
Table 5. Expression of purinergic P2 receptors on basal ganglia astrocytes.
ReceptorReferencesBrain RegionsSpecies
P2XFranke et al., 2001 [32]NAccRat
Franke et al., 2001 [164]NAccRat
Franke et al., 2003 [165]NAccRat
P2YCentemeri et al., 1997 [30]StriatumRat
Franke et al., 2001 [32]NAccRat
Franke et al., 2004 [164]NAccRat
Franke et al., 2003 [165]NAccRat
Franke et al., 2003 [166]StriatumRat
Table 6. Expression of adrenergic receptors on basal ganglia astrocytes.
Table 6. Expression of adrenergic receptors on basal ganglia astrocytes.
ReceptorReferencesBrain Region(s)Species
α1-ARHertz et al., 2004 [175]GeneralReview
Giaume et al., 1991 [177]StriatumRat
Delumeau et al., 1991 [178]StriatumRat
Xin et al., 2019 [179]Ventral midbrain (SN, VTA)Mouse
Shao & Sutin, 1992 [180]StriatumRat
Wahis & Holt, 2021 [183]GeneralReview
Hertz et al., 2004 [175]GeneralReview
β-ARGiaume et al., 1991 [177]StriatumRat
Shao & Sutin, 1992 [180]StriatumRat
Jensen et al., 2016 [182]GeneralMouse
α2-ARXin et al., 2019 [179]SNMouse
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tushevski, A.; Happe, L.; Stocco, E.; De Caro, R.; Macchi, V.; Porzionato, A.; Emmi, A. Astrocytic Receptor Systems of the Basal Ganglia. Receptors 2026, 5, 2. https://doi.org/10.3390/receptors5010002

AMA Style

Tushevski A, Happe L, Stocco E, De Caro R, Macchi V, Porzionato A, Emmi A. Astrocytic Receptor Systems of the Basal Ganglia. Receptors. 2026; 5(1):2. https://doi.org/10.3390/receptors5010002

Chicago/Turabian Style

Tushevski, Aleksandar, Linus Happe, Elena Stocco, Raffaele De Caro, Veronica Macchi, Andrea Porzionato, and Aron Emmi. 2026. "Astrocytic Receptor Systems of the Basal Ganglia" Receptors 5, no. 1: 2. https://doi.org/10.3390/receptors5010002

APA Style

Tushevski, A., Happe, L., Stocco, E., De Caro, R., Macchi, V., Porzionato, A., & Emmi, A. (2026). Astrocytic Receptor Systems of the Basal Ganglia. Receptors, 5(1), 2. https://doi.org/10.3390/receptors5010002

Article Metrics

Back to TopTop