Abstract
In recent years it has becoming clear that glial cells of the central and peripheral nervous system play a crucial role from the earliest stages of development throughout adult life. Glial cells are important for neuronal plasticity, axonal conduction and synaptic transmission. In this respect, glial cells are able to produce, uptake and metabolize many factors that are essential for neuronal physiology, including classic neurotransmitters and neuroactive steroids. In particular, neuroactive steroids, which are mainly synthesized by glial cells, are able to modulate some neurotransmitter receptors affecting both glia and neurons. Among the signaling systems that are specialized for neuron-glial communication, we can include neurotransmitter GABA.
The main focus of this review is to illustrate the cross-talk between neurons and glial cells in terms of GABA neurotransmission and actions of neuroactive steroids. To this purpose, we will review the presence of the different GABA receptors in the glial cells of the central and peripheral nervous system. Then, we will discuss their modulation by some neuroactive steroids.
Key Words: GABA-A receptor, GABA-B receptor, neurosteroids, microglia, macroglia
INTRODUCTION
For a long time neurons have been considered the principal cells in the nervous system while glial cells, originally described by Virchow early in the1846 [201], were believed to be only a mechanical support for them. It is now evident that glial cells exert an essential role in many processes and functions of the nervous system, such as differentiation, development and metabolism [45,61,81,227,232]. Indeed, glial cells control the microenvironment of the brain [85,210], regulate neuronal activity [61,200] and participate in the regulation of synaptic transmission [91,183,249], stabilizing and refining axonal branches within the target area. Nevertheless, glial cells have been recognized to have a crucial role in several pathological states, such as injury, inflammation and pain [4,254].
In the nervous system, glial cells are classified in two main groups: microglia and macroglia. The microglial cells represent a minor, but important subpopulation of cells possessing phagocytic functions [60]. Namely, they are the major immunocompetent cells in the nervous system, since they express many features of monocytes, including signaling cascades that involve chemokines, cytokines and their receptors. Moreover, in the central nervous system (CNS) these cells are also able to respond to specific neurotransmitters. Indeed, these cells express GluR5 glutamate receptors, P2Y and P2X purinergic receptors, γ-aminobutyric acid (GABA) type B (GABA-B) receptors (see below), functional β1, β2 adrenergic receptors and amino-5-methyl-4-isoxazole-propionic acid (AMPA) receptors [58].
Macroglia are composed of two classes of cells, namely astrocytes and myelin-forming cells. It is possible to distinguish two populations of astrocytes, the so-called type 1 and type 2 astrocytes [160,234]. The type 1 astrocytes seem to be the prevalent form in the gray matter [208,258], while the type 2 astrocytes, first described by Raff and colleagues [209], have a stellate appearance and are located only in the white matter. Astrocytes contribute to brain homeostasis, regulating the local concentration of ions and neuroactive substances [21,134,174]. In the central and peripheral nervous system (PNS) the myelin-forming cells are respectively oligodendrocytes and Schwann cells [227]. These cells are deputed to ensheath the axons forming the myelin and allowing the faster propagation of action potentials.
Finally another kind of glial cells are the satellite glial cells of sensory ganglia. These cells wrap completely the sensory neurons [84,195]. Usually, they are structurally identified like a type of astrocytes or considered as a specialization of Schwann cells. However, because the satellite cells are similar neither to astrocytes nor to myelin forming cells, they should be classified as a distinct type of glial cells [84,195].
In the last few years, great attention has been devoted to the study of progenitor stem cells. During forebrain development, most neurons and glial cells arise from neural progenitors located mainly in two specialized germinative zones, the ventricular and subventricular zone [5,136]. The earliest neonatal neural progenitors consist of cells that express the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). Among PSA-NCAM positive neural progenitors, a subpopulation isolated from postnatal day 1 has been named early PSA-NCAM-positive progenitors [69,70]. Depending on the site of transplantation in the brain of newborn mice, these early progenitors differentiate into glial cells or neurons [250]. In particular, these cells differentiate first into oligodendrocyte precursor cells (OPC) then in turn into oligodendrocytes or astrocytes [50]. Among glial progenitor cells, the O-2A is another strain of glial cellsantigenically and physiologically similar to OPC. These progenitors are classified as self-renewing glial progenitors located in both grey and white matter [42,137]. O-2A cells in vitro develop into oligodendrocytes and type 2 astrocytes [209]. Moreover, another subpopulation of glial cells in the CNS which are NG2 positive expresses both the chondroitin sulphate proteoglycan NG2 and the receptor for platelet-derived growth factor alpha (PDGF- R) [135,190]. These glial cell precursors are stellate-shaped and possess a small cell body with thin radiating processes, which distinguish them from astrocytes, oligodendrocytes and microglia [140, 141]. Therefore, NG2 should be considered as a distinct macroglial cell population [189], which is ubiquitously found in both gray and white matter of the developing and adult CNS [49].
The emerging role of the GABAergic system [via the GABA type A (GABA-A) and GABA-B receptors] in the glial cells of the CNS and PNS will be described in this review. However, the mechanisms by which these receptors become activated in the different classes of glia and, consequently, the responses that these receptors evoke in vivo remain poorly understood. Therefore, in light of the involvement of the neuroactive steroids in some of these actions, we will discuss the possible interactions between the GABAergic system and the neuroactive steroids, which are of relevance in the neuron-glia signaling.
NEURON-GLIAL SIGNALING
The role exerted by glial cells in nervous system physiology may be ascribed to a bidirectional neuron-glial communication. Glial cells respond to the same signals acting on neurons and they modulate the neuronal response. These signaling pathways include ion flux, neurotransmitter release, cell adhesion molecules and specialized signaling molecules released from synaptic and/or extrasynaptic regions of the neurons [61]. In the hippocampus, the axons that are stimulated to fire action potentials, produce Ca++ responses that propagate in surrounding astrocytic networks [48,204]. Moreover, glial cells communicate with each other over long distances throughout intracellular Ca++ waves [185], as well as via diffusion of chemical messengers [61]. Notably, astrocytes express a wide variety of neurotransmitter receptors, including acetylcholine, adenosine, histamine, glutamate, GABA (see below), norepinephrine [205], that after activation can raise intracellular Ca++ [6,23,56,116,127, 204,213,228,247]. Similarly, in the PNS the stimulation of motor axons induces Ca++ waves in the terminal presynaptic Schwann cells [105,213,214]. Likely, these observations suggest that molecules released during synaptic transmission bind to the receptors on glial cells, which in turn determine the intracellular Ca++ increase. On the other hand, studies performed in CNS and PNS indicate that neural impulse activity during fetal and early post-natal life may influence the development of myelinating glia, mainly through the release of non-synaptic neurotransmitters [12,236]. Experiments performed in co-cultures of mouse dorsal root ganglion (DRG) neurons and Schwann cell or OPC, revealed a Ca++ signal increase in glia following electrical stimulation [236,237]. This effect was entirely mediated by the rise in extracellular ATP [236]. Adenosine was able to cause a similar large Ca++response in OPC, although no response was detected in Schwann cells [237].
The glial cells also possess transport mechanisms that represent a characteristic clearance function for neurotransmitters, which are presynaptically released by neurons. Examples are the uptake and glial accumulation of neurotransmitters, like glutamate and GABA [93].
However, glial cells not only represent a target for neurotransmitters, but they are also able to synthesize and to secrete them [100,142,252]. About ten years ago a study by Pow and Robinson [206], performed in the retina, indicated the glial cells as active in leading to de novo synthesis of glutamate. In fact, the enzymes involved in the formation of glutamate have been preferentially localized in glial cells [198]. Consequently, these findings argued that much of the de novo synthesis of glutamate, aspartate and GABA occurs in glia [23,106,198]. Therefore, since GABA is relevant among the signaling systems that are specialized for bidirectional neuron-glial communication, it will be further considered in detail in this review.
GABA AND ITS RECEPTORS
GABA is the major inhibitory neurotransmitter in the mammalian nervous system [253]. In the CNS, GABA is primarily produced by inhibitory neurons and released during the firing of action potentials [129], in a process known as phasic inhibition [59]. Tonic inhibition, resulting from continuous activation of extrasynaptic receptors, however is also present in some neurons [17,59,226]. Moreover, like other classic neurotransmitters, GABA is produced by glial cells (e.g. astrocytes). GABA is particularly important in the developing CNS, since different mechanisms involving the activation of GABA receptors influence some processes, such as neuronal or glial precursor proliferation, differentiation and migration [3,11,18,161,194,211]. Nevertheless, it has been recently shown that non-synaptic release of GABA from neuroblasts is fundamental to provide negative feedback to neural stem cells, inhibiting the production of new neuroblasts throughout the block of cell-cycle re-entry [144]. Very recent observations attempt to extend this GABA capability also to the control of glial progenitor development [10,90,145].
A precursor of GABA is glutamate that in the brain is synthesized from glucose. The first step in GABA de novo synthesis is the one-way conversion of glutamate to GABA, by a decarboxylation reaction catalyzed by the enzyme glutamate decarboxylase (GAD). GAD, which is the rate-limiting enzyme in GABA synthesis, requires pyridoxal phosphate as a cofactor [212]. Mammalian species express two isoforms of GAD that differ in their subcellular distribution: the membrane anchored form of 65 kDa (GAD65) and the cytoplasmic form of 67 kDa (GAD67). GAD was also demonstrated in PNS; for instance, GAD-containing neurons have been identified in the enteric sympathetic and parasympathetic nervous system [107,117], as well as in DRG and trigeminal ganglion [89,215]. The enzyme GABA transaminase (GABA-T) is responsible for GABA catabolism leading to the synthesis of succinic semialdehyde, successively oxidized by the enzyme succinic semialdehydedehydrogenase (SSADH) to succinate [244]. GABA-T in turn acts to diminish the quantity of neurotransmitter available to signal at the synaptic cleft.
The rapid termination of GABA transmission is achieved throughout a high affinity GABA uptake by GABAergic neurons and glial cells [223]. There are at least three different GABA transporters (GAT). The one targeted by tiagabine is neuronal (GAT-1), while the others are on glia and other non-neural cells [158].
Pharmacological and electrophysiological studies, have profiled three types of GABA receptors which have been identified in the nervous system: the ionotropic GABA-A and GABA type C (GABA-C) receptors and the metabotropic GABA-B receptor [22,29].
The GABA-A receptor is a member of the ligand-gated ion channel family. GABA-A is composed of five subunits drawn from a repertoire of 1-6, β1-3, γ1-3, δ, ε and π, ρ 1-3, θ [131,256,257]. The assembly of five of these subunits forms a Cl − channel. The GABA-A receptor is blocked by bicuculline and picrotoxin, whereas currents are enhanced by benzodiazepines, barbiturates, and a variety of general anesthetics as well as by the neuroactive steroids [17,196]. Examples of neuroactive steroids, which act as potent allosteric modulators of the GABA-A receptor, are tetrahydroprogesterone (THP, also called allopregnanolone) and tetrahy drodeoxicorticosterone (THDOC) [17]. Downstream of Cl − channel activation, multiple mechanisms of action may apply although a common regulation of the GABA-A receptor function remains phosphorylation [240]. The GABA-A receptor is widely distributed in adult mammalian brain and localized both in neurons [66,231] and in glial cells (see below). Furthermore, functionally active GABA-A receptors have been localized also in DRG of cat, rat, frog and in humans [52,72,89,101,245].
The designation GABA-B was given to a distinct, baclofen-sensitive, metabotropic receptor, in order to distinguish it from the bicuculline-sensitive, ionotropic GABA-A receptor [30,31]. GABA-B receptors are members of the seven transmembrane G-protein-coupled receptor superfamily [30] that may influence presynaptic neurotransmitter release and cause postsynaptic “silencing” of excitatory neurotransmission. These actions occur via the activation of second messenger systems, mainly by modulating the adenylate cyclase or the calcium and potassium channels [29,157]. The cDNAs encoding two GABA-B receptor proteins, initially named GABA-B-1a and GABA-B-1b were identified in 1997 [118]. Subsequently, other GABA-B receptor isoforms were cloned [102,202]. Of particular interest, a number of independent laboratories identified the cDNA encoding for the GABA-B receptor isoform 2 (GABA-B-2) [109,119,128,255]. The GABA-B-1 is retained in the endoplasmic reticulum and is transported to the cell surface only in presence of the GABA-B-2. Therefore, the formation of a functional heterodimeric complex depends on the presence of both −1 and −2 subunits [38,109,119,128,186,255]. A detailed analysis has indicated that GABA-B heterodimer component proteins (i.e., GABA-B-1 and GABA-B-2) are widely expressed throughout the neuronal compartment of the white matter in the brain and spinal cord [44,156].
Moreover, the presence of the GABA-B receptor has also been demonstrated in the rat DRG [242].
NEUROACTIVE STEROIDS
The studies performed in the early ‘80s by Baulieu and colleagues introduced the concept that steroids could be synthesized de novo in the brain [14]. These steroids were named “neurosteroids” to refer to their atypical origin and to differentiate them from the steroids derived from classic steroidogenic tissues (i.e. gonads, placenta and adrenal glands). Subsequently the term “neuroactive steroids” was used to include both the neurosteroids and the classic hormonal steroids from the periphery that are metabolized to neuroactive compounds in the nervous system. The nervous system, thus, is now considered a steroidogenic organ.
In particular, the formation of neurosteroids and neuroactive steroids takes place mainly in the glial compartment of the CNS and PNS (i.e. astrocytes, oligodendrocytes and Schwann cells), which possess the enzymatic pathways capable of synthesizing neurosteroids or convert hormonal steroids into neuroactive metabolites [41,83,124,165,166,167, 224]. In particular, it should be mentioned that progesterone (P) and testosterone (T) may be converted by the 5reductase (5-R) respectively in dihydroprogesterone (DHP) and dihydrotestosterone (DHT) and successively by the 3hydroxysteroid-dehydrogenase (3-HSD) respectively in THP and 3-diol (androstanediol) [41,171]. Additionally, also some glial progenitor cells, such as the early PSANCAM progenitors, can synthesize neuroactive steroids, in a process that is developmentally regulated [69].
The central and peripheral glial cells not only possess the capability to form neuroactive steroids, but are also a possible target for some of them. In fact, neuroactive steroids may exert their actions via an interaction with classical or nonclassical steroid receptors, which are localized both in the neuronal and in the glial compartments [171]. Classic receptors for glucocorticoids (GR) and mineralocorticoids (MR) are present in astrocytes, oligodendrocytes and microglial cells [25,112,248]. The estrogen receptor type (ER) and β (ER β), the androgen receptor (AR) and the P receptor (PR) are also expressed in vivo and in vitro in astrocytes, oligodendrocytes and microglial cells [7,36,40,62,132,178,220]. The presence of steroid receptors has been analyzed also in peripheral nerves, as demonstrated by in vivo and in vitro experiments [110,151,152]. Schwann cells express GR and MR [184] as well as ER and PR [114,151,152,241]. Particular emphasis has been placed on the analysis of the AR, since in vivo this receptor seems to be present in the sciatic nerve, but not in the Schwann cells [152].
Generally, the faster non-genomic actions of neuroactive steroids are mediated through the non-classic steroid receptors. Neuroactive steroids, thus, may act via the interaction with membrane-bound steroid receptors, or through some neurotransmitter receptors, like GABA-A, the N-methyl-Daspartate (NMDA) glutamate receptor, AMPA, 5HT3 and receptors [41,131,165,218]. In particular neuroactive steroids, like dehydroepiandrosterone (DHEA), pregnenolone (PREG) and their sulphates, respectively DHEA-S and PREG-S, as well as the 3-HSD derivatives THP, THDOC and 3-diol are able to interact with the GABA-A receptor [17,68,88,235], which will be further discussed below. Similarly, also the NMDA receptor is modulated by neuroactive steroids. DHEA, DHEA-S and PREG are able to activate allosterically this receptor, while PREG-S acts as a negative modulator [197,260]. Although NMDA receptors are widely distributed in the CNS, their functional expression in glial cells is still a matter of debate [225]. Finally, very recent observations indicate that neuroactive steroids may also modulate voltage-dependent ion channels, a phenomenon which has been characterized mainly for Ca++ channels and to a lesser extent for K+ channels [221].
NEUROACTIVE STEROIDS INTERACTS WITH GABA RECEPTORS
The positive allosteric modulation of GABA-A receptors exerted by THP and THDOC, which accounts for the proposed use of neuroactive steroids as general sedatives, represents the most known non-genomic action of neuroactive steroids [131,154,218]. This kind of modulation has been widely demonstrated in astrocytes [27] and in Schwann cells [169]. Moreover, the GABA modulatory effects exerted by neuroactive steroids are also evident in the spinal cord [121]. In the dorsal horn neurons of the spinal cord the strength of GABA-A mediated synaptic inhibition, during development and under physio-pathological conditions, can be locally modulated by controlling the synthesis of the 5-reduced neuroactive steroid metabolites [121]. However, as previously mentioned, GABA-A receptors can be inhibited by steroids. Sulphated neurosteroids such as DHEA-S or PREGS in fact are negative modulators of GABA-A receptors, whereas un-sulphated steroids can exert both the stimulatory or inhibitory action [196]. From a mechanistic point of view, the neuroactive steroids act allosterically on GABA-A receptors in the nanomolar range, enhancing the action of GABA [87,130,203,218]. Conversely, at higher (micromolar range) concentrations they directly gate the GABA-A receptor channel [37,207]. Androsterone, as well as T or P, however, are only active at high micromolar concentrations (EC50> 20 μM) [196], which are relatively unlikely to be reached in vivo. DHEA has a slight but non-significant effect at a concentration of 100 μM, but DHEA-S dose-dependently inhibits GABA-A receptor mediated currents (EC50 10 μM) in a manner comparable to that of PREG-S (EC507 μM) [196]. Recent observations performed on hippocampal neurons grown in synaptic isolation, demonstrated that neuroactive steroids directly gate GABA-A receptors also at lower (about 100 nM) concentrations [230]. The kinetics of this receptor activation are relatively slow, but this effect may underpin some important cellular and behavioral effects of neuroactive steroids that are difficult to explain [230]. However, after relatively short exposure periods, neuroactive steroids may be retained in the plasma membrane and/or in the intracellular compartments, constituting a reserve able to modulate the GABA-A system for long time after the initial exposure [16]. This process is achieved by the limited access of the neuroactive steroids to the enzyme responsible to terminate their action, representing a kind of autocrine loop of neuroactive steroids [16]. Very recently, the report by Akk et al. [1] strengthened this slow effect of neuroactive steroids on GABA-A receptor, since confirmed that the membrane and the intracellular interactions (depth of embedding or rate of transport through the membrane) may affect the time course of neuroactive steroid action at the GABA-A receptor [1]. In fact, once the neuroactive steroids reach the site of action they diffuse through the plasma membrane in order to bind directly to the GABA-A receptor, rather than to interact to the external part of the receptor. Secondly, neuroactive steroids may be accumulated in the intracellular compartment and then re-supply the plasma membrane with steroids capable to modulate the GABA-A receptor, so that the rate of this transport can affect the kinetics of neuroactive steroid actions [1].
The regulation of GABA-A receptor function by neuroactive steroids involves different mechanisms, even though this modulation is enantioselective and is partially dependent upon the receptor subunit composition [131]. There is no special requirement concerning the nature of the and β subunits able to confer sensitivity at GABA-A receptor to neuroactive steroids. However, γ2 subunits confer more sensitivity than γ1 or γ3 [15] while the δ subunit potentiates the action of the 3α,5 α -reduced neurosteroids [15,176]. Phosphorylation of GABA-A receptors by protein kinase C (PKC) also influences the sensitivity to neuroactive steroids [35].
These steroids also possess the capability to modulate the expression of the GABA-A receptor subunits [63,229]. For instance, fluctuations in the concentration of THP regulate both the expression and function of GABA-A receptor subunits in different brain regions [63]. In hippocampal CA1 of female rats, a 2-days exposure to pregnenolone or estradiol (E2) plus P increased the expression of the GABA-A δ subunit [229], which specifically confers an extrasynaptic localization to the GABA-A receptor [191]. Similarly, short-term exposure of P19-derived neuron (from murine multipotent embryonic carcinoma cell line) to THP and DHEA significantly increased the levels of the subunits α1 and β2 [261]. Less clear are the mechanisms downstream to the GABA-A receptor activation, which account for the effect exerted by the neuroactive steroids. For instance, THP exerted a neuroprotective mechanism on P19-derived neurons, decreasing the apoptotic cell death as well as cytochrome C and Bax translocations [1].
It has been suggested that some effects of neuroactive steroids might be also mediated via other neurotransmitter receptors, like for instance the metabotropic GABA-B receptor. Indeed, the variations of P occurring during the estrous cycle seem to be partially responsible for the regulation of GABA-B receptors in the rat neocortex [3].
Examples of the interaction between neuroactive steroids and GABA receptors are also represented by the GABA-induced regulation of the endogenous neuroactive steroid synthesis. In the frog hypothalamus, GABA inhibits in a dose-dependent manner the activity of several key steroidogenic enzymes, including 3 α-HSD and cytochrome P450C17 [54,175]. This effect was mimicked by the synthetic GABA-A agonist muscimol and blocked by the specific GABA-A antagonist bicuculline [54,175]. However, in the rat retinal ganglion cells, muscimol stimulates the biosynthesis of PREG, an effect reversed by the antagonists bicuculline and picrotoxin [82]. The discrepancies in these studies could be due to the different species utilized and/or to the different pharmacological properties of GABA respect to those of synthetic ligands utilized. Moreover, in rat cortex a GABA-B mediated mechanism is responsible of the increase in THP and THDOC synthesis induced by γ-hydroxybutyric acid (GHB), a metabolite of GABA [17,219]. The neuroactive steroids so formed, in turn, acting as amplifiers of the GABA neurotransmission play a role in the GABA-A mediated action of GHB [9]. Although, these mechanisms occur mainly in neurons rather than in glial cells, they may give evidence on the bidirectional cross-talk between the GABA ergic system and neuroactive steroids. Indeed, neuroactive steroids may regulate GABA receptors as well as GABA, via GABA-A or GABA-B receptors, may control neuroactive steroid synthesis.
Finally, it has been reported that neuroactive steroids not only interact with the GABA receptors, but also may regulate GAD expression, thus, the synthesis of GABA [86]. For instance, P regulates GAD expression in the rat brain during a physiological status, like the surge of gonadotropin hormone releasing hormone (GnRH) [32]. Namely, GAD67 mRNA levels were suppressed by P treatment in the medial preoptic area of the hypothalamus in E2-primed ovariectomized rats [32], suggesting a role for P in the GABA synthesis [32]. In addition, cortisol treatment resulted in an increase of the number of GAD positive cells in the superior cervical ganglia of early postnatal rat [86].
GABA AND NEUROACTIVE STEROIDS: INTERACTIONS IN GLIAL CELLS
Based on the observations so far reported, it might be assumed that GABA possesses the capability to regulate neuron-glial cross-talk in the nervous system [3,161]. In particular, these effects occur because glia expresses some neurotransmitter receptors, including those for GABA. Different electrophysiological and immunohistochemical approaches, in fact, confirmed that GABA-A receptors are widely distributed in adult mammalian brain [231] and particularly in astrocytes [20,95,103,116] and in some OPC [80,122,123]. Consequently, these glial cells might be potentially considered as a target for GABA-A receptor ligands and for neuroactive steroids. Moreover, recent observations suggest that certain types of CNS glial cells (i.e. astrocytes and activated microglia) exhibit GABA-B receptor immunoreactivity [43, 94] and might be considered also a target for the action of GABA-B receptor modulators [47,116,126]. Furthermore, since functionally active GABA-A and GABA-B receptors are present in the Schwann cells of the PNS [149,151,169], it indicates that also these peripheral glial cells are a target for GABA ligands and neuroactive steroids. Additionally, also the glial precursor O-2A cells express GABA-A receptors [251,259].
Finally, it should be further underlined that glial cells are not only a putative target of GABA, but may release this neurotransmitter that in turn might act in a paracrine or autocrine way. Several evidences for the release of GABA come from early experiments in which glial cells, in vitro or in situ, release preloaded neurotransmitter (e.g. 3 H-GABA), in response to depolarizing stimuli [73,177]. Nonetheless, glial cells normally synthesize and secrete GABA or, instead, take them up from the extracellular fluid. As well as the capacity for GABA synthesis, glial and endothelial cells have also been to found the catabolic enzyme GABA-T [64].
In general, the GABAergic system via its main GABA-A and GABA-B receptors assumes a fundamental role in the biology of glial cells and might be important in the cross-talk with neurons. Neuroactive steroids may be involved in this signaling pathway. Therefore, the importance of GABA signaling in the biology of the different glia cell types (e.g. astrocytes, oligodendrocytes, Schwann cells, etc.) will be discussed in the following sections. In addition, we will review the available information on the interaction of the GABAergic system and neuroactive steroids in glial cells.
ASTROCYTES
Many in vitro and in vivo observations demonstrated the presence of functionally active GABA-A and GABA-B receptors in astroglia [20,26,43,94,95,103,116,147] (Table 1). For instance, functional GABA-A receptors are expressed by protoplasmatic and fibrous astrocytes and by specialized astroglia (i.e. Bergmann glia and pituicytes) from many different CNS regions, like cerebellum, hippocampus, optic nerve and spinal cord [65,147,180,181,199] (Table 1). In particular the GABA-A subunits expressed by Bergmann glia have been examined. Immunoreactivity for α2, α3, and δ GABA-A subunits were detected on the cell bodies, as well as on the large radial and the small lateral processes of Bergmann glia [181]. The presence of these subunits appears to be developmentally regulated [181].
Table 1.
In Vivo and In Vitro Localization of GABA-A and GABA-B Receptors in Glial Cells of CNS and PNS
In Vivo or Ex Vivo Localization | In Vitro Localization | ||
---|---|---|---|
ASTROCYTES | GABA-A | • Mouse corpus callosum [20] | • Culture from whole rat brain [26] |
• Bergmann glial cells of mouse cerebellum; in particular α 2, α3 δ subunits [181] | • Culture from rat cerebellum, hippocampus and spinal cord [95] | ||
• Rat hypothalamus [103] | |||
• Stellate cells of the rat pituitary pars intermedia [180] | |||
• Rat hippocampus [65,147] | |||
• Rat hippocampus [65,147] | • Rat spinal cord [199] | ||
GABA-B | • Rat hippocampus [43,116] | • Culture from rat brain [2] | |
• Rat developing cerebellum; in particular -R1 subunit [146] | • Culture from rat hippocampus [43] | ||
• Culture from rat cerebellum, brain stem, and spinal cord [94] | |||
Oligodendrocytes | GABA-A | • Mouse corpus callosum [20] | • Culture from mouse spinal cord [80] |
GABA-B | • Absent?? [43,44] | • n.i. | |
Microglia | GABA-A | • n.i. | • n.i. |
GABA-B | • Rat Brain and facial nucleus [126] | • Culture from rat hippocampus and cerebellar cortex [43,126] | |
Schwann cells | GABA-A | • Rat Sciatic nerve, dorsal roots [24] | • Culture from rat sciatic nerve; in particular α 2, α 3, β1-3 subunits [149,151,169] |
• Rat sciatic nerve; in particular α 2, α 3, β1-3 subunits [149,151,169] | |||
GABA-B | • Rat sciatic nerve; in particular –1a, -1b, -2 subunits[148,149] | • Culture from rat sciatic nerve; in particular -1, -2 subunits [148,149] | |
Satellite cells of DRG | GABA-A | • n.i. | • Culture from rat DRG [96] |
GABA-B | • Mouse DRG, in particular –1, -2 subunits [see detail in the text] | • n.i. | |
Glial Precursor Cells | GABA-A | • In NG2 cells in mouse hippocampus [139] | • In O-2A cells from mouse culture [251,259] |
• In PSA-NCAM from rat brain, in particular α 1-5, β 2, β 3, γ2 [71] and rat striatum [187] | |||
GABA-B | • n.i. | • n.i. |
The first analysis of the presence of GABA-B receptors in astrocytes was performed on pure glial cultures from cerebellum, cortex, brain stem and spinal cord, indicating that GABA-B receptor is present in all these structures [2,94] (Table 1). Further analysis revealed that GABA-B receptors subunits are also expressed on astrocytic processes surrounding both symmetrical and asymmetrical synapses in the CA1 sub-region of the hippocampus [43]. Very recently, immunohistochemical evidence demonstrated that GABA-B-1 is transiently expressed in Bergmann glia and cerebellar astrocytes during the first two weeks of postnatal development [146].
In the adult rat brain, GABAergic signaling mediates the morphological organization of immature astrocytes and the expression of an important and specific marker of astrocytes, the glial fibrillary acidic protein (GFAP) [217], which is involved in modulating astrocyte shape and motility [133]. GABA-A receptor activation, in fact, increases the proliferation of cultured astrocytes [74]. The presence of GABA-A receptors in astrocytes, therefore, suggests that these cells may respond to GABA released from synaptic terminals.
Concerning the mechanism of action, early observations in culture of cat cortex indicated that astrocytes may respond to neurotransmitter by a change in membrane potential [125]. However, the depolarization of astrocytes in culture, that follows the GABA stimulation, was supposed to be indirectly due to a K+ release from adjacent stimulated neurons [233], rather than to a direct stimulation of GABA receptors on glial cells [94,98]. Conversely, analysis of astrocytes from neonatal rat optic nerve (8 to 12 days old), revealed that ABA exerts a depolarization due to Cl- efflux. This is likely a GABA-A mediated mechanism, because muscimol mimics such effect, while bicuculline act as antagonist [180,199]. In the hippocampal astrocytes, the depolarization produced by GABA-receptor stimulation was sufficient to activate voltage-dependent Ca++ channels, inducing an influx of Ca++ [65]. Culture of type 1 astrocytes from rat cortex revealed that the stimulation of both GABA-A and GABA-B receptors transiently evokes Ca++ increase [188]. More recently, observations by Kang et al. [116] have shown that the GABA-mediated astrocytic Ca++ increase in hippocampal slices is prevented by treatment with GABA-B receptor antagonists [116]. However, other authors have proposed different roles for GABA-B receptors in CNS astrocytes. For instance, in culture of rat cortical astrocytes, the block of the GABA-A receptor with the antagonist bicuculline involves a reduction in Ca++ efflux in response to GABA or baclofen [2]. It has been suggested that GABA-B receptor expression is independent of astrocyte activation status and therefore GABA-B receptor likely plays a role in the tonic modulation of astrocytic function [43]. Altogether, these observations indicate that the different mechanisms of action proposed likely depend upon the firing pattern of GABAergic cells involved [116].
As mentioned above, astrocytes are considered as a target for the action of neuroactive steroids. For instance, many observations have indicated that neuroactive steroids are able to influence the expression of GFAP. In particular, the finding that in androgen-insensitive testicular feminized mice and in the hypogonadal mice (hpg), hypothalamic astrocytes show an increased GFAP immunoreactivity [162,163], accounts for a relationship between androgens and astrocytic functions. Moreover, after a penetrating brain injury, neuroactive steroids, like E2, P and T, were able to decrease the process of gliosis and of astrocytic proliferation, resulting in a decrease in the number of GFAP-positive astrocytes in the cerebral cortex and in the hippocampus [75]. However, both the GFAP expression and immunoreactivity are sexually dimorphic in the rat arcuate nucleus, with lower levels in female than in male [46]. Moreover, in the arcuate nucleus and in the hilus of the dentate gyrus of adult female rats, the surface density of GFAP-immunoreactive cells fluctuates throughout the estrous cycle [76,77]. In addition, Stone et al. [238] have demonstrated that estrogens may act directly on astrocytes, although the direction of the transcriptional response is influenced by neuron-astrocyte interactions. A systematic in vitro study on astrocyte cultures was performed in order to answer the question whether neuroactive steroids, such as P or T, act directly on the astrocytes, or need to be converted into their active metabolites (i.e. respectively, DHP and THP in case of P, DHT and 3 α-diol in case of T). The exposure to DHP or THP produced a significant modulation of the GFAP mRNA levels [173], with a stimulatory effect by DHP and an inhibition by THP. It has been proposed that the effect exerted by THP is mediated by the activation of the GABA-A receptors. Conversely, in the case of androgens, DHT induced a significant decrease of GFAP mRNA levels, while T and 3 α-diol were ineffective [173].
Altogether, these observations indicate that in the CNS some of the modulatory actions exerted by neuroactive steroids may be ascribed to an interaction or modulation of the GABA-A receptors. Recently Maguire and colleagues [153], have found that changes in the levels of steroid hormones during the estrous cycle in mice are associated with modifications in the hippocampal GABA-A receptor composition (i.e. that are higher during the late diestrus) [153]. Moreover, more evidence is provided by the finding that pulsatile infusion of the GABA-A blocker bicuculline, in female rhesus monkey, is able to control the release of luteinizing hormone releasing hormone (LHRH) from hypothalamic neurons and the onset of puberty [120]. In rat, LHRH neurons express ER α immunoreactivity [99], whereas the ER β has not been identified in the same cells [92], suggesting the involvement of other cells, likely astrocytes, in the estrogen control of LHRH. However, the absence of ER α in LHRH neurons [92] and the co-localization of ER with the hypothalamic GABA neurons [104] might also support the hypothesis that GABA is implicated in the control of estrogen-mediated gonadotropin release and further indicates an interaction between neuroactive steroids and GABA. It might be argued that astrocytes in the hypothalamus play a central role in the synaptic plasticity across the estrous cycle, a function relevant to the LH surge on proestrus [161]. In fact, E2 increases the synthesis of neuronal GABA by up-regulating the expression of GAD, in turn GABA diffuses from neurons and binds the GABA-A receptors on nearby astrocytes. This binding activates a transient influx of Ca++ in astrocytes that triggers a series of linked phenomena, including the polymerization of GFAP and cellular differentiation [161].
Also corticosteroids, like corticosterone, dihydrocorticosterone (DHC), deoxicorticosterone (DOC), dihydrodeoxicorticosterone (DHDOC) and THDOC act as neuroactive steroids influencing GFAP gene expression [168]. This effect is dependent on the corticosteroid considered. Namely, an exposure of cultured astrocytes to corticosterone induces an increase in GFAP mRNA and protein [168,216], while DHDOC strongly inhibits GFAP gene expression [168]. THDOC (i.e. a neuroactive steroid able to interact with GABA-A receptor) was ineffective in such an effect excluding a possible role for the GABA-A mediated effects by corticosteroids on astrocytes. The effects of neuroactive steroids have been also evaluated on ammonia-induced astrocyte swelling in culture. Bender and Norenberg [19] reported that THP and PREG-S (at nanomolar concentration) diminished the induced swelling, likely via a GABA-A mechanism. These observations might be considered very useful for new therapeutic approaches to the acute hyperammonemic syndromes and other associated pathological conditions [19].
On the other hand, despite the demonstration of the presence of the GABA-B receptors on astrocytes [2,44,96,146], their physiological significance and their correlation with the neuroactive steroid action, has been poorly investigated.
It is important to highlight that astrocytes may also synthesize and release the neurotransmitter GABA [143]. The GABA synthesis in rat cortical brain slice depends on the glutamine produced in astrocytes, which is a quantitatively important precursor of GABA [13]. In the CNS, GABA astrocytic release exerts several actions on the neuronal compartment. In the hippocampus, for instance, GABA via persistent activation of GABA-A receptor, regulates neuronal activity and tonic inhibition [111,143]. Cocultures of neurons and astrocytes from the neonatal rat olfactory bulb, reveal that when astrocytes are pretreated with muscimol before the addition of neurons they support neuronal branching better [159]. These results suggest that various aspects of the GABA-promoted dendritic branching of neurons are mediated by astrocytes[159].
OLIGODENDROCYTE
The early observation by Gilbert and colleagues [80] evidenced that oligodendrocytes from mouse spinal cord are depolarized in a dose-dependent manner by GABA. Because this response is not due to K+ released from active neurons [80], it was suggested that GABA directly influences the physiology of the oligodendrocytes. This effect is mimicked by muscimol and counteracted by bicuculline or picrotoxin, indicating an involvement of the GABA-A receptor in such effect [80]. Notably, in order to strengthen this picture, it should be highlighted that further observations have indicated that oligodendrocytes express the GABA-A receptors [20] 1). The stimulation of thin brain slices at the corpus callosum, in fact, showed several GABA responses that may be ascribed to GABA-A activation in the oligodendrocytes [20]. However, GABA depolarization of these cells is a heterogeneous phenomenon, since only one third of oligodendrocytes are responsive [80]. To date, although only the oligodendrocytes obtained from the white matter of the spinal cord have been investigated, the GABA-B receptor expression has not been found in the myelin forming cells, identified by the presence of the most important protein of central myelin [i.e. myelin basic protein (MBP)] [43,44].
Indirect proof of the association of the GABAergic function within the CNS myelin arises from the recent observations performed on SSADH deficient mice [79]. These mice display different features associated with CNS hypomyelination, like an increase in GHB and GABA contents. In the hippocampus and cortex of these mice the levels of two myelin proteins [i.e. MBP and myelin associated glycoprotein (MAG)] were decreased, while corresponding spinal cord levels were normal or up-regulated [79].
Some observations have indicated direct effects of neuroactive steroids on the oligodendrocytes. In primary cultures of this kind of glial cells, P and E2 are able to increase the MBP expression [112,113]. P and its metabolite DHP also stimulate myelination, by increasing MBP expression, in organotypic slice cultures of 7-day-old rat and mouse cerebellum [78]. This effect likely involves classic PR, since is mimicked with the selective PR agonist R5020 and blocked with the PR antagonist mifepristone [78]. THP significantly stimulated the MBP expression in 7-day-old rat and mouse cerebellum cultures [78]. These effects seem to be mediated by an interaction with the GABA-A receptor, because the antagonist bicuculline proved to counteract the THP stimulatory effect [78]. Also T produces an increase in the degree of myelination, as observed in the forebrain and in the cerebellum of juvenile male zebra finches [115]. However the mechanism involved in this effect is not so far investigated. Corticosteroids also affect MBP expression in the oligodendrocytes. It has been demonstrated that a 7 day treatment of neonatal rats with a synthetic glucocorticoid dexamethasone, is able to significantly decrease in the cerebrum (at 20th and 30th days of life), and in the cerebellum (at 10th days of life) the relative abundance of MBP mRNA levels [243]. Moreover, experiments in vitro, performed in cultures of rat oligodendrocytes demonstrated that among the neuroactive steroids considered (i.e. DHC, DOC, DHDOC and THDOC), only DHDOC was effective in diminishing the mRNA levels of MBP, suggesting an involvement of the MR receptor [168].
MICROGLIA
In the CNS, activated microglial cells exhibit GABA-B receptor immunoreactivity [43] (Table 1). In particular, the number of GABA-B receptor expressing microglial cells, present in the nucleus of the facial nerve, increased in response to nerve axotomy [126].
A functional analysis revealed that microglial cells in culture activated with lipopolysaccharide (LPS) increase the release of interleukin-6 (IL6) and interleukin-12 (IL12). This release activity was attenuated by the simultaneous activation of the GABA-B receptors, indicating that GABA can modulate the microglial immune response [126]. Moreover, agonists of the GABA-B receptor trigger the induction of outwardly rectifying K+ conductance in microglial cells [126].
Furthermore, to the author’s knowledge, the functional presence of the GABA-A receptor has never been considered in the microglial cells.
Neuroactive steroids exert an anti-inflammatory action in the CNS acting mainly on the microglia. For instance, Drew and Chavis [55] have proposed that the P-mediated inhibition of microglial cell activation may contribute to the decreased severity of multiple sclerosis symptoms, usually observed with pregnancy. Furthermore, Vegeto et al. [246] have found that E2 also reduces LPS induced production of inflammatory mediators, such as PGE2 and metalloproteinase-9, in cultured microglia. E2 is also able to enhance uptake of amyloid beta-protein by microglia derived from the human cortex [138]. This finding may be relevant for the possible protective effect exerted by E2 in Alzheimer’s disease. However, all these effects seem to be due to an interaction with the classic steroid receptors.
SCHWANN CELLS
GABA is a neurotransmitter also in the PNS, as suggested by the early study of Jessen and collaborators on the vertebrate peripheral autonomic nervous system [108]. Similarly, it has been shown that also the myelinated and unmyelinated fibers possess GABA receptors and GABA carriers [33,34,179,193]. In fact, early in the 70’s, the accumulation of 3H-GABA was shown in Schwann cells of the taste buds of the amphibian Necturus maculosus [182]. GABA-A receptors are present on normal mammalian sensory axons and are re-established on regenerated sensory axons [24], although the presence of these receptors on Schwann cells has not been further investigated in detail. Recent studies performed in our laboratory using RT-PCR and immunohistochemistry, demonstrated that Schwann cells in culture express several subunits of the GABA-A receptor, such as the α2, α 3, β1-3 subunits [149,151,169] (Table 1). The presence of GABA-B receptor in the PNS, has been previously demonstrated in the rat DRG, in peripheral axons, in autonomic nerve terminals and in pig nodose ganglion cells [28,51, 142,239,242,262], although its cellular localization has never been considered. Very recently, by RT-PCR, western blot analysis and immunohistochemistry we have observed that also Schwann cells express the different isoforms of GABAB receptors, i.e., -1a, -1b, -1c, and -2 [148,149] (Table 1).
In order to investigate the physiological meaning of the expression of GABA-A and GABA-B receptors in Schwann cells, the effects of specific receptor ligands on important features and properties of Schwann cells have been taken in consideration. A 24-hour exposure of Schwann cells to a relatively low concentration of muscimol (1 μM) exerted a clear stimulatory effect on the level of one of the most important protein of the peripheral myelin, i.e. the peripheral myelin protein of 22 kDa (PMP22). This suggests that PMP22 might be under the control of the GABA-A receptor [151, 165]. In addition, the specific GABA-B agonist baclofen influences the proliferation of Schwann cells. Namely, baclofen decreases the forskolin-induced proliferation after 4 days in vitro, and this effect became more evident at later times of exposure (i.e. 5 and 6 day in vitro) [148]. Additionally, baclofen reduced the percentage of Schwann-BrdUrd immunopositive cells and decreased the levels of PMP22 and glycoprotein zero (P0), another important protein of the peripheral myelin [148]. Collectively, these results indicate that Schwann cells are a potential target for GABA actions. Therefore, at least in the case of PMP22, depending on the GABA receptor involved, the neurotransmitter GABA may increase or decrease the synthesis of this myelin protein.
The neuroactive steroids P, DHP and THP, in vivo and in vitro revealed to be potent modulators of several biochemical and morphological parameters of the PNS. In particular, all these neuroactive steroids stimulate the expression of P0 and PMP22 [151,165]. Concerning THP, even though it is also able to increase the levels of P0 mRNA, this neuroactive steroid seems to be the main steroid that specifically stimulates the PMP22 mRNA and protein levels [164,169,170, 172]. Moreover, also 3α-diol, another positive allosteric modulator of the GABA-A receptor [67,68] significantly increases the gene expression and the protein levels of PMP22 [150,151].
Altogether, these data suggest that, in the Schwann cells, the expression of PMP22 is preferentially under the control of the GABA-A receptors [149]. Interestingly, in the rat Schwann cell culture the neuroactive steroids, P, DHP, and THP are also able to control the expression of the GABA-B receptor subunits. Indeed, the mRNA levels of the different GABA-B subunits (i.e. 1a, 1b and 2) are increased mainly by THP, and in a lesser extent also by its precursors P and DHP [149]. In this light, it has been hypothesized that, at least in the PNS, the neuroactive steroid THP exerts a GABA-A mediated regulation of the GABA-B receptor expression. To strengthen this hypothesis it should be recalled that the effect exerted by THP on the expression of GABA-B receptor was mimicked by muscimol and GABA [149]. To the author’s knowledge this is the first demonstration that in the nervous system neuroactive steroids are able to modulate the GABAB receptor via a modulation of the GABA-A receptor (discussed in Fig. 1). The intracellular mechanism underpins this control is presumably complex and to date has not been so far identified. However, some possible mechanisms that tempt to explain how a GABA-A receptor activation may control transcriptional activity have been suggested [155, 192]. In the developing rat cortex, for instance, the neuronal nitric oxide synthase and brain derived neurotrophic factor levels are controlled via GABA-A receptor [155]. In this case it has been proposed, that following GABA-A receptor activation the depolarization leads to opening of L-type voltage-gated Ca++ channels with an increase of Ca++ influx, that in turn leads to phosphorylation and activation of the transcription factor cAMP response element-binding protein(CREB)[155]
Fig. (1).
The GABA-A receptor, the GABA-B receptor and the neuroactive steroids cross-interact in the PNS. These interactions are particularly relevant for the basic bidirectional cross-talk between neurons and Schwann cells. It is possible to hypothesize that GABA neurotransmitter, coming from the neuronal compartment or produced by the Schwann cells, may affect the paracrine cross-talk between these cells. The data obtained with the specific agonist baclofen, in fact, suggests that the extracellular GABA might interact with the GABA-B receptor on the Schwann cells (1), decreasing their proliferation. This challenge prompts the Schwann cells to start differentiation. The neuroactive steroids, such as THP (which is produced by Schwann cells), modulate the expression and the responsivity of the GABA-B receptor, and in turn its desensitization. Successively, THP further decrease Schwann cell proliferation and simultaneously stimulate their differentiation. By a direct interaction with the GABA-A receptor (2), in fact, THP increases some myelin protein expressions. Similarly, muscimol, a specific GABA-A agonist induces an identical effect on the Schwann cells, supporting the hypothesis that not only THP but also GABA might participate in such a control of the Schwann cell differentiation (2). Taken together, these results suggest that the GABA-mediated control of the Schwann cell proliferation/differentiation is particularly relevant to explain the mechanisms affecting the Schwann cell physiology. GABA is a classic neurotransmitter released by neurons, nonetheless, an involvement of the neuronal compartment in such a control is hypothesized.
SATELLITE CELLS OF THE DRG
The first in vitro studies performed on DRG showed that exogenously administered GABA is selectively accumulated by satellite cells [222]. Moreover, in cultured rat DRG, GABA at 100 μM first depolarizes all the neurons, then, with a delay depolarizes also the satellite cells [96]. Because this effect appeared to be bicuculline-sensitive, it suggested an involvement of the GABA-A receptor (Table 1). On the other hand, using specific K+ channel blockers such as 4aminopyridine, this effect appeared to be indirect (i.e., via a GABA-induced release of K+ from the neurons into the narrow cleft between them and the satellite cells) [97]. Furthermore, the early study of Villegas and colleagues in squid axons and satellite cells produced the first demonstration of non-synaptic GABA release [249]. However, neither the presence of functional GABA receptors nor the capability to synthesize GABA was specifically investigated in these satellite glial cells. Very recently, Hayasaki et al. [89] found that the majority of satellite glial cells in the rat trigeminal ganglion are immunopositive for GABA, whereas GAD65 and GAD67 immunoreactivity were not detected in the same cells. These findings suggest that satellite cells in the rat trigeminal ganglion take up GABA from the extracellular space [89].
Concerning the GABA-B receptors some electrophysiological evidences clearly revealed their presence in DRG neuron cultures [39,53]. Subsequently, GABA-B receptors have been generally reported to be present in the DRG [44,242], whereas their cellular localization has not been deeply analyzed. Very recently, high levels of -1a subunit and low levels of the -1b protein were demonstrated in the rat DRG [57]. On the contrary, the -2 subunit was practically undetectable [57]. This is consistent with the proposed existence of an atypical receptor composed of GABA-B-1 homodimers in the lumbar DRG [57]. However, also in this study the satellite cells were not investigated. To this end, an immunocytochemical study was recently performed in order to investigate the possible cellular localization of the GABAB subunits in the DRG. The observations obtained indicated that both neurons and satellite cells of the mouse lumbar DRG are immunopositive for the GABA-B-1 (Fig. 2a). Conversely, the GABA-B-2 appeared to be present mainly in the satellite cells, although its labeling was generally weaker than that found for subunit -1 (Fig. 2b).
Fig. (2).
Localization of GABA -B receptor subunits 1 and 2 in satellite cells of the mouse dorsal root ganglion (DRG).The mouse DRGs were fixed in PBS-PFA 4%, then coronal sections were incubated overnight at 4°C with one of the primary antibodies against GABA-B subunits. We have used the guinea pig pan-anti-GABA-B-1 (1:300), that recognizes both 1a and 1b subunits, and the anti-GABA-B-2 (1:250), both by Chemicon (USA) as described in Magnaghi et al. [46]. The immunoreactivities were revealed with Alexa-488anti-guinea pig secondary antibody (1:600) and the samples mounted with PermaFluorTM. Controls for antibody specificity included a lack of a primary antibody. Confocal laser microscopy was performed using a Biorad Radiance 2100 Confocal System (Biorad, Italy) and a Nikon TE2000-S Eclipse microscope, utilizing the 488 nm laser. (a) Immunopositivity for the GABA-B-1 subunit is evident both in the neuron soma (arrows) and in some satellite cells of the mouse lumbar DRG (arrowheads). In particular, the neuronal soma shows a typical patchy distribution of the GABA-B-1 subunit. (b) Satellite cells are immunopositive also for the -2 subunit (arrowheads), while the signal for the GABA-B-2 seems to be absent in the neuron soma. Scale bar 20¼m.
To date, the possible effects exerted by the neuroactive steroids on the satellite cells have never been considered.
GLIAL PRECURSOR CELLS
In the OPC cells of the developing and adult rat hippocampus the quantal release of GABA from interneurons elicits GABA-A receptor currents, with a rapid rise times. These currents did not exhibit features of spillover transmission, suggesting that interneuronal terminals are in direct contact with OPC. Moreover, in OPC the GABA responses regulate the efficacy of AMPA receptor currents and glutamatergic signaling, influencing the OPC maturation into O-2A and then into oligodendrocytes [141]. On the other hand, glutamine synthase, an enzyme that subserves important function in regulating GABA metabolism, is expressed in O-2A progenitors and is stimulated during oligodendrocyte developmental maturation [8]. This suggests that in O-2A precursors the possible synthesis of GABA and subsequently its release might be important in addressing the maturation of these precursor cells. This may be supported by the finding that GABA can be detected in O-2A cells cultured in medium lacking any source of this neurotransmitter [11]. Moreover, it should be underlined that O-2A cells in vitro express GABA-A receptors [251,259] (Table 1).
In parallel, it has been revealed that also the NG2, which are a distinct macroglial cell population [189], may communicate throughout GABAergic pathways. In particular, it has been found that focal application of THIP (i.e. a selective GABA-A receptor agonist) to voltage-clamped NG2 cells, in hippocampal slices, elicited an inward current that was blocked by the GABA-A receptor antagonist SR-95531 (i.e. gabazine) [139]. This confirms that NG2 cells possess functional GABA-A receptor in situ (Table 1), and it suggests that this rapid form of glial communication may regulate the proliferation rate of NG2 cells or their development into mature oligodendrocytes [139].
Gago and colleagues [71] have recently observed that neuroactive steroids and GABA signaling are involved in autocrine/paracrine loops, which control PSA-NCAM progenitor proliferation and differentiation [71]. The PSANCAM positive cells possess many GABA-A receptor subunits [71,187], like for instance the α1-5, β2, β3 and γ2 [71] (Table 1). GABA increased in a dose dependent manner the proliferation of these cells and this effect was bicuculline sensitive, revealing that it was actually mediated by GABAA receptor. Notably, this effect on the PSA-NCAM proliferation was mimicked by THP in a nanomolar range of concentration. Moreover, P via its conversion in THP, is able to stimulate the early PSA-NCAM progenitor proliferation [71]. Collectively, these data suggest a key role for GABA and THP in the development of the nervous system, and specifically into its glial components (e.g. astrocytes, oligodendrocytes,etc.)[18,71]
CONCLUSIONS
In this review we report the functional presence of GABA receptors and we describe some of the physiological effects consequent to their activation, in the different glial cells of the CNS and PNS. These observations have been further re-evaluated, in the light of the involvement of the neuroactive steroids in some of the effects exerted by GABA. For instance, it has been reported that the neuroactive steroid THP is the major regulator of the expression of the GABA-B receptor subunits in the Schwann cells. Since THP is known to be a potent allosteric modulator of the GABA-A receptor, therefore, it has been hypothesized that the THP-induced regulation of GABA-B receptors represents a new type of GABA-A mediated mechanism. Moreover, GABA itself may be present in some glial cells, where it also affects the synthesis of neuroactive steroids.
Altogether, these observations suggest that the close relationship between the GABAergic system and neuroactive steroids may be fundamental for the neuron-glial cross-talk in the central and peripheral nervous system. In the field of neurobiology, for instance, the study of these signaling pathways might be promising to investigate the possible factors involved in the development of the glial progenitor cells.
ACKNOWLEDGEMENTS
The author is grateful to Prof. Patrizia Procacci, University of Milan, for assistance with confocal microscopy. The author would like to thank Prof. R.C. Melcangi, University of Milan, and Prof. L.R. Garcia-Segura, Instituto Cajal, Spain for helpful discussions.
Abbreviations
- 3α-HSD
3α-hydroxysteroid-dehydrogenase
- 5α-R
5α-reductase
- AMPA
Amino-5-methyl-4-isoxazole-propionic acid
- AR
Androgen receptor
- CNS
Central nervous system
- CREB
cAMP response element-binding protein
- DHEA
Dehydroepiandrosterone
- DHC
Dihydrocorticosterone
- DHDOC
Dihydrodeoxicorticosterone
- DHP
Dihydroprogesterone
- DHT
Dihydrotestosterone
- DOC
Deoxicorticosterone
- DRG
Dorsal root ganglion
- ER α
Estrogen receptor type α
- ER β
Estrogen receptor type β
- GABA
γ-aminobutyric acid
- GABA-A
GABA type A receptor
- GABA-B
GABA type B receptor
- GABA-C
GABA type C receptor
- GABA-T
GABA transaminase
- GAD
Glutamate decarboxylase
- GAD65
Glutamate decarboxylase of 65 kDa
- GAD67
Glutamate decarboxylase of 67 kDa
- GAT
GABA transporters
- GFAP
Glial fibrillary acidic protein
- GHB
γ-hydroxybutyric acid
- GnRH
Gonadotropin hormone releasing hormone
- GR
Glucocorticoid receptor
- IL6
Interleukin-6
- IL12
Interleukin-12
- LPS
Lipopolysaccharide
- MAG
Myelin associated glycoprotein
- MBP
Myelin basic protein
- MR
Mineralocorticoid receptor
- NMDA
N-methyl-D-aspartate
- OPC
Oligodendrocyte precursor cells
- P0
Glycoprotein zero
- PMP22
Peripheral myelin protein of 22 kDa
- P
Progesterone
- PNS
Peripheral nervous system
- PSA-NCAM
Polysialylated form of the neural cell adhesion molecule
- PR
Progesterone receptor
- PREG
Pregnenolone
- SSADH
Succinic semialdehyde dehydrogenase
- T
Testosterone
- THDOC
Tetrahydrodeoxicorticosterone
- THP
Tetrahydroprogesterone
REFERENCES
- 1.Akk G, Shu HJ, Wang C, Steinbach JH, Zorumski CF, Covey DF, Mennerick S. Neurosteroid access to the GABA-A receptor. J Neurosci. 2005;25:11605–11613. doi: 10.1523/JNEUROSCI.4173-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Albrecht J, Pearce B, Murphy S. Evidence for an interaction between GABA-B and glutamate receptors in astrocytes as revealed by changes in Ca2+ flux. Eur J Pharmacol. 1986;125:463–464. doi: 10.1016/0014-2999(86)90805-8. [DOI] [PubMed] [Google Scholar]
- 3.Al-Dahan MI, Thalmann RH. Progesterone regulates gamma-aminobutyric acid B (GABA-B) receptors in the neocortex of female rats. Brain Res. 1996;727:40–48. [PubMed] [Google Scholar]
- 4.Aldskogius H, Kozlova EN. Central neuron-glial and glial-glial interactions following axon injury. Prog Neurobiol. 1998;55:1–26. doi: 10.1016/s0301-0082(97)00093-2. [DOI] [PubMed] [Google Scholar]
- 5.Altman J. Autoradiographic and histological studies of postnatal neurogenesis IV Cell proliferation and migration in the anterior forebrain, with special reference to persisting neurogenesis in the olfactory bulb. J Comp Neurol. 1969;137:433–458. doi: 10.1002/cne.901370404. [DOI] [PubMed] [Google Scholar]
- 6.Araque A, Martin ED, Perea G, Arellano JI, Buño W. Synaptically-released acetylcholine evokes Ca2+ elevations in astrocytes in hippocampal slices. J Neurosci. 2002;22:2443–2450. doi: 10.1523/JNEUROSCI.22-07-02443.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Azcoitia I, Sierra A, Garcia-Segura LM. Localization of estrogen receptor beta-immunoreactivity in astrocytes of the adult rat brain. Glia. 1999;26:260–267. [PubMed] [Google Scholar]
- 8.Baas D, Dalencon D, Fressinaud C, Vitkovic L, Sarlieve LL. Oligodendrocyte-typt-2 astrocyte (O-2A) progenitor cells express glutamine synthetase: developmental and cell type-specific regulation. Mol Psychiatry. 1998;3:356–361. doi: 10.1038/sj.mp.4000379. [DOI] [PubMed] [Google Scholar]
- 9.Barbaccia ML, Colombo G, Affricano D, Carai MA, Vacca G, Melis S, Purdy RH, Gessa GL. GABA(B) receptor-mediated increase of neurosteroids by gamma-hydroxybutyric acid. Neuropharmacology. 2002;42:782–791. doi: 10.1016/s0028-3908(02)00026-6. [DOI] [PubMed] [Google Scholar]
- 10.Barker JL, Behar T, Li Y-X, Liu Q-Y, Ma W, Maric D, Maric I, Schaffner AE, Serafini R, Smith SV, Somogyi R, Vautrin JY, Wen X-L, Xian H. GABAergic cells and signals in CNS development. Perspect DevNeurobiol. 1998;5:305–322. [PubMed] [Google Scholar]
- 11.Barres BA, Koroshetz WJ, Swartz KJ, Chun LL, Corey DP. Ion channel expression by white matter glia: the O-2A glial progenitor cell. Neuron. 1990;4:507–524. doi: 10.1016/0896-6273(90)90109-s. [DOI] [PubMed] [Google Scholar]
- 12.Barres BA, Raff M. Proliferation of oligodendrocyte precursor cells depends on electrical activity in axons. Nature. 1993;361:258–260. doi: 10.1038/361258a0. [DOI] [PubMed] [Google Scholar]
- 13.Battaglioli G, Martin DL. GABA synthesis in brain slices is dependent on glutamine produced in astrocytes. Neurochem Res. 1991;16:151–156. doi: 10.1007/BF00965703. [DOI] [PubMed] [Google Scholar]
- 14.Baulieu EE. Neurosteroids: of the nervous system, by the nervous system, for the nervous system. Rec Prog Horm Res. 1997;52:1–32. [PubMed] [Google Scholar]
- 15.Belelli D, Casula A, Ling A, Lambert J J. The influence of subunit composition on the interaction of neurosteroids with GABA(A) receptors. Neuropharmacology. 2002;43:651–661. doi: 10.1016/s0028-3908(02)00172-7. [DOI] [PubMed] [Google Scholar]
- 16.Belelli D, Herd M B. The contraceptive agent Provera enhances GABAA receptor-mediated inhibitory neurotransmission in the rat hippocampus: evidence for endogenous neurosteroids? J Neurosci. 2003;23:10013–10020. doi: 10.1523/JNEUROSCI.23-31-10013.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Belelli D, Lambert JJ. Neurosteroids: endogenous regulators of the GABA-A receptor. Nat Rev Neurosci. 2005;6:565–575. doi: 10.1038/nrn1703. [DOI] [PubMed] [Google Scholar]
- 18.Ben-Ari Y. Excitatory actions of GABA during development the nature of the nurture. Nat Rev Neurosci. 2002;3:728–739. doi: 10.1038/nrn920. [DOI] [PubMed] [Google Scholar]
- 19.Bender AS, Norenberg MD. Effect of benzodiazepines and neurosteroids on ammonia-induced swelling in cultured astro-cytes. J NeurosciRes. 1988;54:673–680. doi: 10.1002/(SICI)1097-4547(19981201)54:5<673::AID-JNR12>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 20.Berger T, Walz W, Schnitzer J, Kettenmann H. GABA- and glutamate-activated currents in glial cells of the mouse corpus callosum slice. J Neurosci Res. 1992;31:21–7. doi: 10.1002/jnr.490310104. [DOI] [PubMed] [Google Scholar]
- 21.Bergles DE, Jahr CE. Synaptic activation of glutamate transporters in hippocampal astrocytes. Neuron. 1997;19:1297–1308. doi: 10.1016/s0896-6273(00)80420-1. [DOI] [PubMed] [Google Scholar]
- 22.Bettler B, Kaupmann K, Mosbacher J, Gassmann M. Molecular structure and physiological functions of GABA(B) receptors. Physiol Rev. 2004;84:835–867. doi: 10.1152/physrev.00036.2003. [DOI] [PubMed] [Google Scholar]
- 23.Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Rizzini B, Pozzan T, Volterra A. Prostaglandins stimulate Ca2+-dependent glutamate release in astrocytes. Nature. 1998;391:281–285. doi: 10.1038/34651. [DOI] [PubMed] [Google Scholar]
- 24.Bhisitkul RB, Villa JE, Kocsis JD. Axonal GABA receptors are selectively present on normal and regenerated sensory fibers in rat peripheral nerve. Exp Brain Res. 1987;66:659–63. doi: 10.1007/BF00270698. [DOI] [PubMed] [Google Scholar]
- 25.Bohn MC, Howard E, Vielkind U, Krozowski Z. Glial cells express both mineralocorticoid and glucocorticoid receptors. J Steroid Biochem Mol Biol. 1991;40:105–111. doi: 10.1016/0960-0760(91)90173-3. [DOI] [PubMed] [Google Scholar]
- 26.Bormann J, Kettenmann H. Patch-clamp study of GABA receptor Cl channels in cultured astrocytes. Proc Nat1 Acad Sci USA. 1988;85:9336–9340. doi: 10.1073/pnas.85.23.9336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bovolin P, Santi MR, Puia G, Costa E, Grayson D. Expression patterns of gamma-aminobutyric acid type A receptor subunit mRNAs in primary cultures of granule neurons and astro-cytes from neonatal rat cerebella. Proc Natl Acad Sci USA. 1992;89:9344–9348. doi: 10.1073/pnas.89.19.9344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bowery NG, Doble A, Hill DR, Hudson AL, Shaw JS, Turnbull MJ, Warrington R. Bicuculline-insensitive GABA receptors on peripheral autonomic nerve terminals. Eur J Pharmacol. 1981;71:53–70. doi: 10.1016/0014-2999(81)90386-1. [DOI] [PubMed] [Google Scholar]
- 29.Bowery NG, Enna SJ. γ-Aminobutyric acidB receptors: first of the functional metabotropic heterodimers. J Pharmacol Exp Ther. 2000;292:2–7. [PubMed] [Google Scholar]
- 30.Bowery NG, Enna SJ, Olsen RW. Six decades of GABA. Biochem Pharmacol. 2004;68:1477–1478. doi: 10.1016/j.bcp.2004.07.033. [DOI] [PubMed] [Google Scholar]
- 31.Bowery NG, Hill DR, Hudson AL, Doble A, Middlemiss DN, Shaw J, Turnbull M. (-)Baclofen decreases neuro-transmitter release in the mammalian CNS by an action at a novel GABA receptor. Nature. 1980;283:92–94. doi: 10.1038/283092a0. [DOI] [PubMed] [Google Scholar]
- 32.Brann DW, Unda R, Mahesh VB. Role of progesterone receptor in restrained glutamic acid decaboxylase gene expression in the hypothalamus during the preovulatory luteinising hormone surge. Neuroendocrinology. 2002;76:283–289. doi: 10.1159/000066628. [DOI] [PubMed] [Google Scholar]
- 33.Brown DA, Adams PR, Higgins AJ, Marsh S. Distribution of gaba-receptors and gaba-carriers in the mammalian nervous system. J Physiol Paris. 1979;75:667–671. [PubMed] [Google Scholar]
- 34.Brown DA, Marsh S. Axonal GABA-receptors in mammalian peripheral nerve trunks. Brain Res. 1978;156:187–191. doi: 10.1016/0006-8993(78)90098-7. [DOI] [PubMed] [Google Scholar]
- 35.Brussaard AB, Koksma JJ. Conditional regulation of neurosteroid sensitivity of GABAA receptors. Ann N Y Acad Sci. 2003;1007:29–36. doi: 10.1196/annals.1286.003. [DOI] [PubMed] [Google Scholar]
- 36.Buchanan CD, Mahesh VB, Brann DW. Estrogen-astrocyte-luteinizing hormone-releasing hormone signaling: a role for transforming growth factor-beta(1) Biol Reprod. 2000;62:1710–1721. doi: 10.1095/biolreprod62.6.1710. [DOI] [PubMed] [Google Scholar]
- 37.Callachan H, Cottrell GA, Hather NY, Lambert JJ, Nooney JM, Peters JA. Modulation of the GABA-A receptor by progesterone metabolites. Proc R Soc Lond B Biol Sci. 1987;231:359–369. doi: 10.1098/rspb.1987.0049. [DOI] [PubMed] [Google Scholar]
- 38.Calver AR, Robbins MJ, Cosio G, Rice SQ, Babbs AJ, Hirst WD, Boyfield I, Wood MD, Russell RB, Price GW, Couve A, Moss SJ, Pangalos MN. The C-terminal domains of the GABA-B receptor subunits mediate intracellular trafficking but are not required for receptor signalling. J Neurosci. 2001;21:1203–1210. doi: 10.1523/JNEUROSCI.21-04-01203.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Campbell V, Berrow N, Dolphin AC. GABA-B receptor modulation of Ca2+ currents in rat sensory neurones by the G protein G(0): antisense oligonucleotide studies. J Physiol. 1993;470:1–11. doi: 10.1113/jphysiol.1993.sp019842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cardona-Gomez GP, Don Carlos L, Garcia-Segura LM. Insulin-like growth factor I receptors and estrogen receptors colo-calize in female rat brain. Neuroscience. 2000;99:751–760. doi: 10.1016/s0306-4522(00)00228-1. [DOI] [PubMed] [Google Scholar]
- 41.Celotti F, Melcangi RC, Martini L. The 5α-reductase in the brain: molecular aspects and relation to brain function. Front Neuroendocrinol. 1992;13:163–215. [PubMed] [Google Scholar]
- 42.Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J Neurosci. 2000;20:6404–6412. doi: 10.1523/JNEUROSCI.20-17-06404.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Charles KJ, Deuchars J, Davies CH, Pangalos MN. GABAB receptor subunit expression in glia. Mol Cell Neurosci. 2003;24:214–223. doi: 10.1016/s1044-7431(03)00162-3. [DOI] [PubMed] [Google Scholar]
- 44.Charles KJ, Evans ML, Robbins MJ, Calver AR, Leslie RA, Pangalos MN. Comparative immunohistochemical localisation of GABAB1a, GABAB1b, and GABAB2 subunits in rat brain, spinal cord and dorsal root ganglion. Neuroscience. 2001;106:447–467. doi: 10.1016/s0306-4522(01)00296-2. [DOI] [PubMed] [Google Scholar]
- 45.Chotard C, Salecker I. Neurons and glia: team players in axon guidance. Trends Neurosci. 2004;27:655–661. doi: 10.1016/j.tins.2004.09.001. [DOI] [PubMed] [Google Scholar]
- 46.Chowen JA, Busiguina S, Garcia-Segura LM. Sexual dimorphism and sex steroid modulation of glial fibrillary acidic protein messenger RNA and immunoreactivity levels in the rat hypothalamus. Neuroscience. 1995;69:519–532. doi: 10.1016/0306-4522(95)00250-m. [DOI] [PubMed] [Google Scholar]
- 47.Clark JA, Mezey E, Lam AS, Bonner TI. Distribution of the GABAB receptor gb2 in rat CNS. Brain Res. 2000;860:41–52. doi: 10.1016/s0006-8993(00)01958-2. [DOI] [PubMed] [Google Scholar]
- 48.Dani JW, Chernjavsky A, Smith SJ. Neuronal activity triggers calcium waves in hippocampal astrocyte networks. Neuron. 1992;8:429–440. doi: 10.1016/0896-6273(92)90271-e. [DOI] [PubMed] [Google Scholar]
- 49.Dawson MR, Polito A, Levine JM, Reynolds R. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neuro-sci. 2003;24:476–488. doi: 10.1016/s1044-7431(03)00210-0. [DOI] [PubMed] [Google Scholar]
- 50.Decker L, Avellana-Adalid V, Nait-Oumesmar B, Durbec P, Baron-Van Evercooren A. Oligodendrocyte precursor migration and differentiation: combined effects of PSA residues, growth factors, and substrates. Mol Cell Neurosci. 2000;16:422–439. doi: 10.1006/mcne.2000.0885. [DOI] [PubMed] [Google Scholar]
- 51.Desarmenien M, Feltz P, Occhipinti G, Santangelo F, Schlichter R. Coexistence of GABA-A and GABA-B receptors on A delta and C primary afferents. Br J Pharmacol. 1984;81:327–333. doi: 10.1111/j.1476-5381.1984.tb10082.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Deschennes M, Feltz P, Lamour Y. A model for an estimate in vivo of the ionic basis of presynaptic inhibition: an intra-cellular analysis of the GABA-induced depolarization in rat dorsal root ganglia. Brain Res. 1976;118:486–493. doi: 10.1016/0006-8993(76)90318-8. [DOI] [PubMed] [Google Scholar]
- 53.Dolphin AC, Scott RH. Inhibition of calcium currents in cultured rat dorsal ganglion neurones by (-)-baclofen. Br J Phar-macol. 1986;88:213–220. doi: 10.1111/j.1476-5381.1986.tb09489.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Do-Rego JL, Mensah-Nyagan GA, Beaujean D, Vaudry D, Sieghart W, Luu-The V, Pelletier G, Vaudry H. γ-Aminobutyric acid, acting through γ-aminobutyric acid type A receptors, inhibits the biosynthesis of neurosteroids in the frog hypothalamus. Proc Natl Acad Sci USA. 2000;97:13925–13930. doi: 10.1073/pnas.240269897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Drew PD, Chavis JA. Female sex steroids: effects upon microglial cell activation. J Neuroimmunol. 2000;111:77–85. doi: 10.1016/s0165-5728(00)00386-6. [DOI] [PubMed] [Google Scholar]
- 56.Duffy S, MacVicar BA. Adrenergic calcium signaling in astrocyte networks within the hippocampal slice. J Neurosci. 1995;15:5535–5550. doi: 10.1523/JNEUROSCI.15-08-05535.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Engle MP, Gassman M, Sykes KT, Bettler B, Hammond DL. Spinal nerve ligation does not alter the expression or function of GABA(B) receptors in spinal cord and dorsal root ganglia of the rat. Neuroscience. 2006;138:1277–1287. doi: 10.1016/j.neuroscience.2005.11.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Farber K, Kettenmann H. Physiology of microglial cells. Brain Res Rev. 2005;48:133–143. doi: 10.1016/j.brainresrev.2004.12.003. [DOI] [PubMed] [Google Scholar]
- 59.Farrant M, Nusser Z. Variation of an inhibitory theme: phasic and tonic activation of GABA(A) receptors. Nat Rev Neu-rosci. 2005;6:215–229. doi: 10.1038/nrn1625. [DOI] [PubMed] [Google Scholar]
- 60.Fedoroff S. Development in microglia. In: Kettenmann H, Ransom BR, editors. New York: Oxford Univ. Press.; 1995. pp. 162–181. [Google Scholar]
- 61.Fields RD, Stevens-Graham B. New insights into neuron-glia communication. Science. 2002;298:556–562. doi: 10.1126/science.298.5593.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Finley SK, Kritzer MF. Immunoreactivity for intracellu-lar androgen receptors in identified subpopulations of neurons, as-trocytes and oligodendrocytes in primate prefrontal cortex. J Neu-robiol. 1999;40:446–457. [PubMed] [Google Scholar]
- 63.Follesa P, Biggio F, Caria S, Gorini G, Bigio G. Modulation of GABA-A receptor gene expression by allopreg-nanolone and ethanol. Eur J Pharmacol. 2004;500:413–425. doi: 10.1016/j.ejphar.2004.07.041. [DOI] [PubMed] [Google Scholar]
- 64.Fonnum F. Determination of transmitter amino acid turnover. In: Boulton AA, Baker GB, Wood JD, editors. Neuromethods. New Jersey: Humana Press; 1985. pp. 201–235. (Series I: Neurochemistry. 3. Amino acids). [Google Scholar]
- 65.Fraser DD, Duffy S, Angelides KJ, Perez-Velazquez JL, Kettenmann H, MacVicar BA. GABA-A benzodiazepine receptors in acutely isolated hippocampal astrocytes. J Neurosci. 1995;15:270–2732. doi: 10.1523/JNEUROSCI.15-04-02720.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fritschy JM, Mohler H. GABAA-receptor heterogeneity in the adult rat brain: differential regional and cellular distribution of seven major subunits. J Comp Neurol. 1995;359:154–194. doi: 10.1002/cne.903590111. [DOI] [PubMed] [Google Scholar]
- 67.Frye CA, Duncan JE, Basham M, Erkine MS. Behavioral effects of 3alpha-androstanediol. II: hypothalamic and preoptic area actions via a GABAergic mechanism. Behav Brain Res. 1996b;79:119–130. doi: 10.1016/0166-4328(96)00005-8. [DOI] [PubMed] [Google Scholar]
- 68.Frye CA, Van Keuren KR, Erkine MS. Behavioral effects of 3alpha-androstanediol. I: modulation of sexual receptivity and promotion of GABA-stimulated chloride flux. Behav Brain Res. 1996;79:109–118. doi: 10.1016/0166-4328(96)00004-6. [DOI] [PubMed] [Google Scholar]
- 69.Gago N, Akwa Y, Sananes N, Guennoun R, Baulieu EE, El Etr M, Schumacher M. Progesterone and the oligoden-droglial lineage: stage-dependent biosynthesis and metabolism. Glia. 2001;36:295–308. doi: 10.1002/glia.1117. [DOI] [PubMed] [Google Scholar]
- 70.Gago N, Avellana-Adalid V, Baron-Evercooren A, Schu-macher M. Control of cell survival and proliferation of postnatal PSA-NCAM progenitors. Mol Cell Neurosci. 2003;22:162–178. doi: 10.1016/s1044-7431(02)00030-1. [DOI] [PubMed] [Google Scholar]
- 71.Gago N, El-Etr M, Sananès N, Cadepond F, Samule D, Av-ellana-Adalid V, Baron-Van Evercooren A, Schumacher M. 3α,5α-Tetrahydroprogesterone (allopregnanolone) and γ-aminobutyric acid: autocrine/paracrine interactions in the control of neonatal PSA-NCAM+ progenitor proliferation. J Neurosci Res. 2004;78:770–783. doi: 10.1002/jnr.20348. [DOI] [PubMed] [Google Scholar]
- 72.Gallagher JP, Higashi H, Nishi S. Characterization and ionic basis of GABA-induced depolarization recorded in vitro from cat primary afferent neurons. J Physiol (Lond.) 1978;275:263–282. doi: 10.1113/jphysiol.1978.sp012189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gallo V, Suergiu R, Levi G. Kainic acid stimulates GABA release from a subpopulation of cerebellar astrocytes. Eur J Pharmacol. 1986;132:319–322. doi: 10.1016/0014-2999(86)90624-2. [DOI] [PubMed] [Google Scholar]
- 74.Gandolfo P, Patte C, Thoumas JL, Leprince J, Vaudry H, Tonon MC. The endozepine ODN stimulates [3H]thymidine incorporation in cultured rat astrocytes. Neuropharmacology. 1999;38:725–732. doi: 10.1016/s0028-3908(98)00231-7. [DOI] [PubMed] [Google Scholar]
- 75.Garcia Estrada J, Del Rio JA, Luquin S, Soriano E, Garcia-Segura LM. Gonadal hormones down-regulate reactive gliosis and astrocyte proliferation after a penetrating brain injury. Brain Res. 1993;628:271–278. doi: 10.1016/0006-8993(93)90964-o. [DOI] [PubMed] [Google Scholar]
- 76.Garcia Segura LM, Chowen JA, Duenas M, Torres-Aleman I, Naftolin F. Gonadal steroids as promoters of neuro-glial plasticity. Psychoneuroendocrinology. 1994;19:445–453. doi: 10.1016/0306-4530(94)90031-0. [DOI] [PubMed] [Google Scholar]
- 77.Garcia Segura LM, Luquin S, Parducz A, Naftolin F. Gonadal hormone regulation of glial fibrillary acidic protein im-munoreactivity and glial ultrastructure in the rat neuroendocrine hypothalamus. Glia. 1994;10:59–69. doi: 10.1002/glia.440100108. [DOI] [PubMed] [Google Scholar]
- 78.Ghoumari AM, Ibanez C, El-Etr M, Leclerc P, Eychenne P, O’Malley BW, Baulieu EE, Schumacher M. Progester-one and its metabolite increase myelin basic protein expression in organotypic slice cultures of rat cerebellum. J Neurochem. 2003;86:848–859. doi: 10.1046/j.1471-4159.2003.01881.x. [DOI] [PubMed] [Google Scholar]
- 79.Gibson KM, Donarum E, Stephn D, Gupta M, Senephansiri H, Larkin K, Snead OC, Switzer R, Murphy E, Jakobs C. Hypomyelination in murine succinate semialdehyde dehydro-genase (SSADH) deficiency. Soc For Neurosci. Annu. Meeting. 2005 Abs.132.11. [Google Scholar]
- 80.Gilbert P, Kettenmann H, Schachner M. Gamma-aminobutyric acid directly depolarizes cultured oligodendrocytes. J Neurosci. 1984;4:561–569. doi: 10.1523/JNEUROSCI.04-02-00561.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Goldman S. Glia as neural progenitor cells. Trends Neuro-sci. 2003;26:590–596. doi: 10.1016/j.tins.2003.09.011. [DOI] [PubMed] [Google Scholar]
- 82.Guarneri P, Guarneri R, Cascio C, Piccoli F, Papadopoulos V. γ-Aminobutyric acid type A/benzodiazepine receptors regulate rat retina neurosteroidogenesis. Brain Res. 1995;683:65–72. doi: 10.1016/0006-8993(95)00343-o. [DOI] [PubMed] [Google Scholar]
- 83.Guennoun R, Schumacher M, Robert F, Delespierre B, Goue-zou M, Eychenne B, Akwa Y, Robel P, Baulieu EE. Neurosteroids: expression of functional 3β-hydroxysteroid dehy-drogenase by rat sensory neurons and Schwann cells. J Neurosci. 1997;9:2236–2247. doi: 10.1111/j.1460-9568.1997.tb01642.x. [DOI] [PubMed] [Google Scholar]
- 84.Hanani M. Satellite glial cells in sensory ganglia: from form to function. Brain Res Rev. 2005;48:457–476. doi: 10.1016/j.brainresrev.2004.09.001. [DOI] [PubMed] [Google Scholar]
- 85.Hansson E, Ronnback L. Glial neuronal signaling in the central nervous system. FASEB J. 2003;17:341–348. doi: 10.1096/fj.02-0429rev. [DOI] [PubMed] [Google Scholar]
- 86.Happola O, Paivarinta H, Soinila S, Wu JY, Panula P. Localization of L-glutamate decarboxylase and GABA transa-minase immunoreactivity in the sympathetic ganglia of the rat. Neuroscience. 1987;21:271–281. doi: 10.1016/0306-4522(87)90338-1. [DOI] [PubMed] [Google Scholar]
- 87.Harrison NL, Majewska MD, Harrington JW, Barker JL. Structure-activity relationships for steroid interaction with the gamma-aminobutyric acid-A receptor complex. J Pharmacol Exp Ther. 1987;241:346–353. [PubMed] [Google Scholar]
- 88.Harrison NL, Simmonds MA. Modulation of the GABA receptor complex by a steroid anaesthetic. Brain Res. 1984;323:287–292. doi: 10.1016/0006-8993(84)90299-3. [DOI] [PubMed] [Google Scholar]
- 89.Hayasaki H, Sohma Y, Kanbara K, Maemura K, Kubota T, Watanabe M. A local GABAergic system within rat trigeminal ganglion cells. Eur J Neurosci. 2006;23:745–757. doi: 10.1111/j.1460-9568.2006.04602.x. [DOI] [PubMed] [Google Scholar]
- 90.Haydar TF, Wang F, Schwartz ML, Rakic P. Differential modulation of proliferation in the neocortical ventricular and subventricular zones. J Neurosci. 2000;20:5764–5774. doi: 10.1523/JNEUROSCI.20-15-05764.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Haydon PG. Glia: listening and talking to the synapse. Nat Rev Neurosci. 2001;2:185–193. doi: 10.1038/35058528. [DOI] [PubMed] [Google Scholar]
- 92.Herbison AE, Pape JR. New evidence for estrogen receptors in gonadotropin-releasing hormone neurons. Front Neu-roendocrinol. 2001;22:292–308. doi: 10.1006/frne.2001.0219. [DOI] [PubMed] [Google Scholar]
- 93.Hertz L. Functional interactions between neurons and as-trocytes. I. Turnover and metabolism of putative aminoacid transmitters. Prog Neurobiol. 1979;13:277–323. doi: 10.1016/0301-0082(79)90018-2. [DOI] [PubMed] [Google Scholar]
- 94.Hoesli E, Hoesli L. Evidence for GABA-B receptors on cultured astrocytes of rat CNS: autoradiographic binding studies. Exp Brain Res. 1990;80:621–625. doi: 10.1007/BF00228002. [DOI] [PubMed] [Google Scholar]
- 95.Hosli E, Otten U, Hosli L. Expression of GABA(A) receptors by reactive astrocytes in explant and primary cultures of rat CNS. Int J Dev Neurosci. 1997;15:949–960. doi: 10.1016/s0736-5748(97)00041-5. [DOI] [PubMed] [Google Scholar]
- 96.Hosli L, Andrés PF, Hosli E. Neuron-glia interaction: indirect effect of GABA on cultured glial cells. Exp Brain Res. 1978;33:425–434. doi: 10.1007/BF00235564. [DOI] [PubMed] [Google Scholar]
- 97.Hosli L, Andrés PF, Hosli E. Effects of 4-aminopyridine and tetraethylammonium on the depolarization by GABA of cultured satellite glial cells. Neurosci Lett. 1979;11:193–196. doi: 10.1016/0304-3940(79)90126-5. [DOI] [PubMed] [Google Scholar]
- 98.Hosli L, Hosli E, Landolt H, Zehntner C. Efflux of potassium from neurons excited by glutamate and aspartate causes depolarization of cultured glial cells. Neurosci Lett. 1981;21:83–86. doi: 10.1016/0304-3940(81)90062-8. [DOI] [PubMed] [Google Scholar]
- 99.Hrabovszky E, Steinhauser A, Barabas K, Shughrue PJ, Petersen SL, Merchenthaler I, Liposits Z. Estrogen receptor-beta immunoreactivity in luteinizing hormone-releasing hormone neurons of the rat brain. Endocrinology. 2001;142:3261–3264. doi: 10.1210/endo.142.7.8176. [DOI] [PubMed] [Google Scholar]
- 100.Innocenti B, Parpura V, Haydon PG. Imaging extracel-lular waves of glutamate during calcium signaling in cultured as-trocytes. J Neurosci. 2000;20:1800–1808. doi: 10.1523/JNEUROSCI.20-05-01800.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Inoue M, Akaike N. Blockade of γ-aminobutyric acid-gated chloride current in frog sensory neurons by picrotoxin. Neu-rosci Res. 1988;5:380–394. doi: 10.1016/0168-0102(88)90024-7. [DOI] [PubMed] [Google Scholar]
- 102.Isomoto S, Kaibara M, Sakurai-Yamashita Y, Nagayama Y, Uezono Y, Yano K, Taniyama K. Cloning and tissue distribution of novel splice variants of the rat GABAB receptor. Biochem Biophys Res Commun. 1998;253:10–15. doi: 10.1006/bbrc.1998.9706. [DOI] [PubMed] [Google Scholar]
- 103.Israel JM, Schipke CG, Ohlemeyer C, Theodosis DT, Kettenmann H. GABA-A receptor-expressing astrocytes in the supraoptic nucleus lack glutamate uptake and receptor currents. Glia. 2003;44:102–110. doi: 10.1002/glia.10272. [DOI] [PubMed] [Google Scholar]
- 104.Jackson GL, Kuehl D. Gamma-aminobutyric acid (GABA) regulation of GnRH secretion in sheep. Reproduction. 2002;59:15–24. [PubMed] [Google Scholar]
- 105.Jahromi BS, Robitaille R, Charlton MP. Transmitter release increases intracellular calcium in perisynaptic Schwann cells in situ. Neuron. 1992;8:1069–1077. doi: 10.1016/0896-6273(92)90128-z. [DOI] [PubMed] [Google Scholar]
- 106.Jeftinija SD, Jeftinija KV, Stefanovic G, Liu F. Neu-roligand-evoked calcium-dependent release of excitatory amino acids from cultured astrocytes. J Neurochem. 1996;66:676–684. doi: 10.1046/j.1471-4159.1996.66020676.x. [DOI] [PubMed] [Google Scholar]
- 107.Jessen KR. GABAergic neurons in the myenteric plexus. In: Erdo SL, editor. GABA Outside the CNS. New-York: Springer-Verlag; 1992. pp. 19–27. [Google Scholar]
- 108.Jessen KR, Mirsky R, Dennison ME, Burnstock G. GABA may be a neurotransmitter in the vertebrate peripheral nervous system. Nature. 1979;281:71–74. doi: 10.1038/281071a0. [DOI] [PubMed] [Google Scholar]
- 109.Jones KA, Borowsky B, Tamm JA, Craig DA, Durkin MM, Dai M, Yao W-J, Johnson M, Gunwaldsen C, Huang L-Y, Tang C, Shen Q, Salon J-A, Morse K, Laz T, Smith KE, Nagarathnam D, Noble SA, Branchek TA, Gerald C. GABAB receptors function as a heteromeric assembly of the subunits GABABR1 and GABABR2. Nature. 1998;396:674–679. doi: 10.1038/25348. [DOI] [PubMed] [Google Scholar]
- 110.Jordan CL, Price RH Jr, Handa RJ. Androgen receptor messenger RNA and protein in adult rat sciatic nerve: implications for site of androgen action. J Neurosci Res. 2002;69:509–518. doi: 10.1002/jnr.10324. [DOI] [PubMed] [Google Scholar]
- 111.Jow F, Chiu D, Lim HK, Novak T, Lin S. Production of GABA by cultured hippocampal glial cells. Neurochem Int. 2004;45:273–283. doi: 10.1016/j.neuint.2003.11.021. [DOI] [PubMed] [Google Scholar]
- 112.Jung-Testas I, Renoir M, Bugnard H, Greene GL, Baulieu EE. Demonstration of steroid hormone receptors and steroid action in primary cultures of rat glial cells. J Steroid Biochem Mol Biol. 1992;41:815–821. doi: 10.1016/0960-0760(92)90394-x. [DOI] [PubMed] [Google Scholar]
- 113.Jung-Testas I, Schumacher M, Robel P, Baulieu EE. Actions of steroid hormones and growth factors on glial cells of the central and peripheral nervous system. J Steroid Biochem Mol Biol. 1994;48:145–154. doi: 10.1016/0960-0760(94)90261-5. [DOI] [PubMed] [Google Scholar]
- 114.Jung-Testas I, Schumacher M, Robel P, Baulieu EE. Demonstration of progesterone receptors in rat Schwann cells. J Steroid Biochem Mol Biol. 1996;58:77–82. doi: 10.1016/0960-0760(96)00009-x. [DOI] [PubMed] [Google Scholar]
- 115.Kafitz KW, Herth G, Bartsch U, Guttinger HR, Schachner M. Application of testosterone accelerates oligodendrocyte maturation in brain of zebra finches. Neuroreport. 1992;3:315–318. doi: 10.1097/00001756-199204000-00006. [DOI] [PubMed] [Google Scholar]
- 116.Kang J, Jiang L, Goldman SA, Nedergaard M. Astro-cyte mediated potentiation of inhibitory synaptic transmission. Nat Neurosci. 1998;1:683–692. doi: 10.1038/3684. [DOI] [PubMed] [Google Scholar]
- 117.Karhula T, Happola O, Joh T, Wu JY. Localization of L-glutamate decarboxylase immunoreactivity in the major pelvic ganglion and in the coeliac-superior mesenteric ganglion complex of the rat. Histochemistry. 1988;90:255–60. doi: 10.1007/BF00495968. [DOI] [PubMed] [Google Scholar]
- 118.Kaupmann K, Huggel K, Heid J, Flor PJ, Bischoff S, Mickel SJ, McMaster G, Angs C, Bittiger H, Froestl W, Bettler B. Expression cloning of GABAB receptors uncovers similarity to metabotropic glutamate receptors. Nature. 1997;386:239–246. doi: 10.1038/386239a0. [DOI] [PubMed] [Google Scholar]
- 119.Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B. GABAB-receptor subtypes assemble into functional heteromeric complexes. Nature. 1998;396:683–687. doi: 10.1038/25360. [DOI] [PubMed] [Google Scholar]
- 120.Keen KL, Burich AJ, Mitsushima D, Kasuya E, Terasawa E. Effects of pulsatile infusion of the GABA-A receptor blocker bicuculline on the onset of puberty in female rhesus monkeys. Endocrinology. 1999;140:5257–5266. doi: 10.1210/endo.140.11.7139. [DOI] [PubMed] [Google Scholar]
- 121.Keller AF, Breton JD, Schlichter R, Poisbeau P. Production of 5alpha-reduced neurosteroids is developmentally regulated and shapes GABA(A) miniature IPSCs in lamina II of the spinal cord. J Neurosci. 2004;24:907–915. doi: 10.1523/JNEUROSCI.4642-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kettenmann H, Gilbert P, Schnachner M. Depolarization of cultured oligodendrocytes by glutamate and GABA. Neurosci Lett. 1984;47:271–276. doi: 10.1016/0304-3940(84)90525-1. [DOI] [PubMed] [Google Scholar]
- 123.Kirchhoff F, Kettenmann H. GABA triggers [Ca2+]i increase in murine precursor cells of the oligodendrocyte lineage. Eur J Neurosci. 1992;4:1049–1058. doi: 10.1111/j.1460-9568.1992.tb00131.x. [DOI] [PubMed] [Google Scholar]
- 124.Koenig HL, Schumacher M, Ferzaz B, Do-Thi AN, Res-souches A, Guennoun R, Jung-Testas I, Robel P, Akwa Y, Baulieu EE. Progesterone synthesis and myelin formation by Schwann cells. Science. 1995;268:1500–1503. doi: 10.1126/science.7770777. [DOI] [PubMed] [Google Scholar]
- 125.Krnjevic K, Schwartz S. Some properties of unresponsive cells in the cerebral cortex. Exp Brain Res. 1967;3:306–319. doi: 10.1007/BF00237557. [DOI] [PubMed] [Google Scholar]
- 126.Kuhn SA, van Landeghem FK, Zacharias R, Farber K, Rap-pert A, Pavlovic S, Hoffmann A, Nolte C, Kettenmann H. Microglia express GABA(B) receptors to modulate inter-leukin release. Mol Cell Neurosci. 2004;25:312–322. doi: 10.1016/j.mcn.2003.10.023. [DOI] [PubMed] [Google Scholar]
- 127.Kulik A, Haentzsch A, Luckermann M, Reichelt W, Ballanyi K. Neuron-glia signaling via α1 adrenoceptor-mediated Ca2+ release in Bergmann glial cells in situ. J Neurosci. 1999;19:8401–8408. doi: 10.1523/JNEUROSCI.19-19-08401.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kuner R, Kohr G, Grunewald S, Eisenhardt G, Bach A, Kornau H-C. Role of heteromer formation in GABAB receptor function. Science. 1999;283:74–77. doi: 10.1126/science.283.5398.74. [DOI] [PubMed] [Google Scholar]
- 129.Kunkel DD, Hendrickson AE, Wu J-Y, Schwartzkroin PA. Glutamic acid decarboxylase (GAD) immunocytochemistry of developing rabbit hippocampus. J Neurosci. 1986;6:541–552. doi: 10.1523/JNEUROSCI.06-02-00541.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Lambert JJ, Belelli D, Harney SC, Peters JA, Frenguelli BG. Modulation of native and recombinant GABA(A) receptors by endogenous and synthetic neuroactive steroids. Brain Res Rev. 2001;37:68–80. doi: 10.1016/s0165-0173(01)00124-2. [DOI] [PubMed] [Google Scholar]
- 131.Lambert JJ, Belelli D, Peden E, Vardy H, Peters JA. Neurosteroid modulation of GABA-A receptors. Prog Neurobiol. 2003;71:67–80. doi: 10.1016/j.pneurobio.2003.09.001. [DOI] [PubMed] [Google Scholar]
- 132.Langub MC, Watson Jr R.E. Estrogen receptor-immunoreactive glia, endothelia, and ependyma in guinea pig pre-optic area and median eminence: electron microscopy. Endocrinol-ogy. 1992;130:364–372. doi: 10.1210/endo.130.1.1727710. [DOI] [PubMed] [Google Scholar]
- 133.Laping NJ, Teter B, Nichols NR, Rozovsky I, Finch CE. Glial fibrillary acidic protein: regulation by hormones, cy-tokines and growth factors. Brain Pathol. 1994;1:259–275. doi: 10.1111/j.1750-3639.1994.tb00841.x. [DOI] [PubMed] [Google Scholar]
- 134.Largo C, Cuevas P, Somjen GG, Martin del Rio R, Herreras O. The effect of depressing glial function in rat brain in situ on ion homeostasis, synaptic transmission, and neuron survival. J Neurosci. 1996;16:1219–1229. doi: 10.1523/JNEUROSCI.16-03-01219.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Levine JM, Card JP. Light and electron microscopic localization of a cell surface antigen (NG2) in the rat cerebellum: association with smooth protoplasmic astrocytes. J Neurosci. 1987;7:2711–2720. doi: 10.1523/JNEUROSCI.07-09-02711.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Levinson SW, Goldman JE. Both oligodendrocytes and astrocytes develop from progenitors in the subventricular zone of the postnatal rat forebrain. Neuron. 1993;10:201–212. doi: 10.1016/0896-6273(93)90311-e. [DOI] [PubMed] [Google Scholar]
- 137.Levison SW, Young GM, Goldman JE. Cycling cells in the adult rat neocortex preferentially generate oligodendroglia. J Neurosci Res. 1999;57:435–446. [PubMed] [Google Scholar]
- 138.Li R, Shen Y, Yang LB, Lue LF, Finch C, Rogers J. Estrogen enhances uptake of amyloid beta-protein by microglia derived from the human cortex. J Neurochem. 2000;75:1447–1454. doi: 10.1046/j.1471-4159.2000.0751447.x. [DOI] [PubMed] [Google Scholar]
- 139.Lin SC, Bergles DE. Physiological characteristic of NG2-expressing glial cells. J Neurocytol. 2002;31:537–549. doi: 10.1023/a:1025799816285. [DOI] [PubMed] [Google Scholar]
- 140.Lin SC, Bergles DE. Synaptic signalling between neurons and glia. Glia. 2004;47:290–298. doi: 10.1002/glia.20060. [DOI] [PubMed] [Google Scholar]
- 141.Lin SC, Bergles DE. Synaptic signaling between GABAergic interneurons and oligodendrocyte precursor cells in the hippocampus. Nat Neurosci. 2004;7:24–32. doi: 10.1038/nn1162. [DOI] [PubMed] [Google Scholar]
- 142.Liske S, Morris ME. Extrasynaptic effects of GABA (gamma-aminobutyric acid) agonists on myelinated axons of peripheral nerve. Can J Physiol Pharmacol. 1994;72:368–374. doi: 10.1139/y94-054. [DOI] [PubMed] [Google Scholar]
- 143.Liu QY, Schaffner AE, Chang YH, Maric D, Barker JL. Persistent activation of GABA(A) receptor/Cl- channels by astrocyte-derived GABA in cultured embryonic rat hippocampal neurons. J Neurophysiol. 2000;84:1392–1403. doi: 10.1152/jn.2000.84.3.1392. [DOI] [PubMed] [Google Scholar]
- 144.Liu X, Wang Q, Haydar TF, Bordey A. Nonsynaptic GABA signaling in postnatal subventricular zone controls proliferation of GFAP-expressing progenitors. Nat Neurosci. 2005;8:1179–1187. doi: 10.1038/nn1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.LoTurco JJ, Owens DF, Health MJS, Davies MBE, Krieg-stein AR. GABA and glutamate depolarise cortical progenitor cells and inhibit DNA synthesis. Neuron. 1995;15:1287–1298. doi: 10.1016/0896-6273(95)90008-x. [DOI] [PubMed] [Google Scholar]
- 146.Lujan R, Shigemoto R. Localization of metabotropic GABA receptor subunits GABA and GABA relative to synaptic sites in the rat developing cerebellum. Eur J Neurosci. 2006;23:1479–1490. doi: 10.1111/j.1460-9568.2006.04669.x. [DOI] [PubMed] [Google Scholar]
- 147.MacVicar BA, Tse FW, Crichton A, Kettenmann H. GABAactivated Cl channels in astrocytes of hippocampal slices. J Neurosci. 1989;9:3577–3583. doi: 10.1523/JNEUROSCI.09-10-03577.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Magnaghi V, Ballabio M, Cavarretta ITC, Froestl W, Lambert JJ, Zucchi I, Melcangi RC. GABA-B receptors in Schwann cells influence proliferation and myelin protein expression. Eur J Neurosci. 2004;19:2641–2649. doi: 10.1111/j.0953-816X.2004.03368.x. [DOI] [PubMed] [Google Scholar]
- 149.Magnaghi V, Ballabio M, Consoli A, Lambert JJ, Roglio I, Melcangi RC. GABA-receptor mediated effects in the peripheral nervous system: a cross-interaction with neuroactive steroids. J Mol Neurosci. 2006;28:89–102. doi: 10.1385/jmn:28:1:89. [DOI] [PubMed] [Google Scholar]
- 150.Magnaghi V, Ballabio M, Gonzalez LC, Leonelli E, Motta M, Melcangi RC. The synthesis of glycoprotein Po and peripheral myelin protein 22 in sciatic nerve of male rats is modulated by testosterone metabolites. Mol Brain Res. 2000;126:67–73. doi: 10.1016/j.molbrainres.2004.03.009. [DOI] [PubMed] [Google Scholar]
- 151.Magnaghi V, Cavarretta I, Galbiati M, Martini L, Melcangi RC. Neuroactive steroids and peripheral myelin proteins. Brain Res Rev. 2001;37:360–371. doi: 10.1016/s0165-0173(01)00140-0. [DOI] [PubMed] [Google Scholar]
- 152.Magnaghi V, Cavarretta I, Zucchi I, Susani L, Rupprecht R, Hermann B, Martini L, Melcangi RC. Po gene expression is modulated by androgens in the sciatic nerve of adult male rats. Mol Brain Res. 1999;70:36–44. doi: 10.1016/s0169-328x(99)00124-2. [DOI] [PubMed] [Google Scholar]
- 153.Maguire JL, Stell BM, Rafizadeh M, Mody I. Ovarian cycle-linked changes in GABA(A) receptors mediating tonic inhibition alter seizure susceptibility and anxiety. Nat Neurosci. 2005;8:797–804. doi: 10.1038/nn1469. [DOI] [PubMed] [Google Scholar]
- 154.Majewska MD, Harrison NL, Schwartz RD, Barker JL, Paul SM. Steroid hormone metabolites are barbiturate-like modulators of the GABA receptor. Science. 1986;232:1004–1007. doi: 10.1126/science.2422758. [DOI] [PubMed] [Google Scholar]
- 155.Mantelas A, Stamatakis A, Kazanis I, Philippidis H, Stylianopoulou F. Control of neuronal nitric oxide synthase and brain-derived neurotrophic factor levels by GABA-A receptors in the developing rat cortex. Dev Brain Res. 2003;145:185–195. doi: 10.1016/j.devbrainres.2003.08.001. [DOI] [PubMed] [Google Scholar]
- 156.Margeta-Mitrovic M, Mitrovic I, Riley RC, Jan LY, Bas-baum AI. Immunohistochemical localization of GABAB receptors in the rat central nervous system. J Comp Neurol. 1999;405:299–321. doi: 10.1002/(sici)1096-9861(19990315)405:3<299::aid-cne2>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- 157.Marshall FH, Jones KA, Kaupmann K, Bettler B. GABAB receptors - the first 7TM heterodimers. Trends Pharmacol Sci. 1999;20:396–399. doi: 10.1016/s0165-6147(99)01383-8. [DOI] [PubMed] [Google Scholar]
- 158.Masson J, Sagné C, Hamon M, El Mestikawy S. Neu-rotransmitter transporters in the central nervous system. Pharma-col Rev. 1999;51:439–464. [PubMed] [Google Scholar]
- 159.Matsutami S, Yamamoto N. GABAergic neuron-to-astrocyte signalling regulates dendritic branching in cocolture. J Neurobiol. 1998;37:251–264. doi: 10.1002/(sici)1097-4695(19981105)37:2<251::aid-neu5>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 160.McCarthy KD, De Vellis J. Preparation of separate as-troglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol. 1980;85:890–902. doi: 10.1083/jcb.85.3.890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.McCarthy MM, Amateau SK, Mong JA. Steroid modulation of astrocytes in the neonatal brain: implications for adult reproductive function. Biol Reprod. 2002;67:691–698. doi: 10.1095/biolreprod.102.003251. [DOI] [PubMed] [Google Scholar]
- 162.McQueen JK. Glial cells and neuroendocrine function. J Endocrinol. 1994;143:411–415. doi: 10.1677/joe.0.1430411. [DOI] [PubMed] [Google Scholar]
- 163.McQueen JK, Wright AK, Arbuthnott GW, Fink G. Glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes are increased in the hypothalamus of androgen-insensitive testicular feminized (Tfm) mice. Neurosci Lett. 1990;118:77–81. doi: 10.1016/0304-3940(90)90252-5. [DOI] [PubMed] [Google Scholar]
- 164.Melcangi RC, Azcoitia I, Ballabio M, Cavarretta I, Gonzalez LC, Leonelli E, Magnaghi V, Veiga S, Garcia-Segura LM. Neuroactive steroids influence peripheral myelination: a promising opportunity for preventing or treating age-dependent disfunctions of peripheral nerves. Prog Neurobiol. 2003;71:57–66. doi: 10.1016/j.pneurobio.2003.09.003. [DOI] [PubMed] [Google Scholar]
- 165.Melcangi RC, Cavarretta I, Ballabio M, Leonelli E, Sche-none A, Azcoitia I, Garcia-Segura LM, Magnaghi V. Peripheral nerves: a target for the action of neuroactive steroids. Brain Res Rev. 2005;48:328–338. doi: 10.1016/j.brainresrev.2004.12.021. [DOI] [PubMed] [Google Scholar]
- 166.Melcangi RC, Celotti F, Castano P, Martini L. Is the 5α-reductase-3α-hydroxysteroid dehydrogenase complex associated with the myelin in the peripheral nervous system of young and old male rats? Endocr Reg. 1992;26:119–125. [PubMed] [Google Scholar]
- 167.Melcangi RC, Magnaghi V, Cavarretta I, Martini L, Piva F. Age induced decrease of glycoprotein Po and myelin basic protein gene expression in the rat sciatic nerve. Repair by steroid derivatives. Neuroscience. 1998;85:569–578. doi: 10.1016/s0306-4522(97)00628-3. [DOI] [PubMed] [Google Scholar]
- 168.Melcangi RC, Magnaghi V, Cavarretta I, Riva MA, Martini L. Corticosteroid effects on gene expression of myelin basic protein in oligodendrocytes and of glial fibrillary acidic protein in type 1 astrocytes. J Neuroendocrinol. 1997;9:729–733. doi: 10.1046/j.1365-2826.1997.00621.x. [DOI] [PubMed] [Google Scholar]
- 169.Melcangi RC, Magnaghi V, Cavarretta I, Zucchi I, Bovolin P, D’Urso D, Martini L. Progesterone derivatives are able to influence peripheral myelin protein 22 and Po gene expression: possible mechanisms of action. J Neurosci Res. 1999;56:349–357. doi: 10.1002/(SICI)1097-4547(19990515)56:4<349::AID-JNR3>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
- 170.Melcangi RC, Magnaghi V, Galbiati M, Ghelarducci B, Sebastiani L, Martini L. The action of steroid hormones on peripheral myelin proteins: A possible new tool for the rebuilding of myelin? J Neurocytol. 2000;29:327–339. doi: 10.1023/a:1007105121765. [DOI] [PubMed] [Google Scholar]
- 171.Melcangi RC, Magnaghi V, Martini L. Steroid metabolism and effects in central and peripheral glial cells. J Neurobiol. 1999;40:471–483. [PubMed] [Google Scholar]
- 172.Melcangi RC, Magnaghi V, Martini L. Aging in peripheral nerves: regulation of myelin protein genes by steroid hormones. Prog Neurobiol. 2000;60:291–308. doi: 10.1016/s0301-0082(99)00028-3. [DOI] [PubMed] [Google Scholar]
- 173.Melcangi RC, Riva MA, Fumagalli F, Magnaghi V, Racagni G, Martini L. Effect of progesterone, testosterone and their 5α-reduced metabolites on GFAP gene expression in type 1 astro-cytes. Brain Res. 1996;711:10–15. doi: 10.1016/0006-8993(95)01302-4. [DOI] [PubMed] [Google Scholar]
- 174.Mennerick S, Zorumski CF. Glial contribution to excitatory neurotransmission in cultured hippocampal cells. Nature. 1994;368:59–62. doi: 10.1038/368059a0. [DOI] [PubMed] [Google Scholar]
- 175.Mensah-Nyagan AG, Do-Rego JL, Beaujean D, Luu-The V, Pelletier G, Vaudry H. Regulation of neurosteroi biosyn-theis in the frog diencephalons by GABA and endozepines. Horm Behav. 2001;40:218–225. doi: 10.1006/hbeh.2001.1689. [DOI] [PubMed] [Google Scholar]
- 176.Mihalek RM, Banerjee PK, Korpi ER, Quinlan JJ, Fire-stone LL, Mi ZP, Lagenaur C, Tretter V, Sieghart W, Anagnostaras SG, Sage J R, Fanselow MS, Guidotti A, Spigelman I, Li Z, DeLorey T M, Olsen R W, Homanics GE. Attenuated sensitivity to neuroactive steroids in gamma-aminobutyrate type A receptor delta subunit knockout mice. Proc Natl Acad Sci USA. 1999;96:12905–12910. doi: 10.1073/pnas.96.22.12905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Minchen MC, Iverson LL. Release of ‘H-GABA from glial cells in rat dorsal root ganglia. J Neurochem. 1974;23:533–540. doi: 10.1111/j.1471-4159.1974.tb06056.x. [DOI] [PubMed] [Google Scholar]
- 178.Mor G, Nilsen J, Horvath T, Bechmann I, Brown S, Garcia-Segura LM, Naftolin F. Estrogen and microglia: A regulatory system that affects the brain. J Neurobiol. 1999;40:484–496. doi: 10.1002/(sici)1097-4695(19990915)40:4<484::aid-neu6>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 179.Morris ME, Di Costanzo GA, Fox S, Werman R. Depolarizing action of GABA (γ-aminobutyric acid) on myelinated fibers of peripheral nerves. Brain Res. 1983;278:117–126. doi: 10.1016/0006-8993(83)90230-5. [DOI] [PubMed] [Google Scholar]
- 180.Mudrick-Donnon LA, Williams PJ, Pittman QJ, MacVicar BA. Postsynaptyc potentials mediated by GABA and do-pamine evoked in stellate glial cells of the pituitary pars intermedia. J Neurosci. 1993;13:4660–4668. doi: 10.1523/JNEUROSCI.13-11-04660.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Muller T, Fritschy JM, Grosche J, Pratt GD, Mohler H, Kettenmann H. Developmental regulation of voltage-gated K+ channel and GABAA receptor expression in Bergmann glial cells. J Neurosci. 1994;14:2503–2514. doi: 10.1523/JNEUROSCI.14-05-02503.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Nagai T, Delay RJ, Welton J, Roper SD. Uptake and release of neurotransmitter candidates, [3H]serotonin, [3H]glutamate, and [3H]gamma-aminobutyric acid, in taste buds of the mudpuppy, Necturus maculosus. J Comp Neurol. 1998;392:199–208. [PubMed] [Google Scholar]
- 183.Nedergaard M, Ransom B, Goldman SA. New roles for astrocytes: redefining the functional architecture of the brain. Trends Neurosci. 2003;26:523–530. doi: 10.1016/j.tins.2003.08.008. [DOI] [PubMed] [Google Scholar]
- 184.Neuberger TJ, Kalimi O, Regelson W, Kalimi M, De Vreis GH. Glucocorticoids enhance the potency of Schwann cell mitogens. J Neurosci Res. 1994;38:300–313. doi: 10.1002/jnr.490380308. [DOI] [PubMed] [Google Scholar]
- 185.Newman EA. New roles for astrocytes: regulation of synaptic transmission. Trends Neurosci. 2003;26:452–536. doi: 10.1016/S0166-2236(03)00237-6. [DOI] [PubMed] [Google Scholar]
- 186.Ng GYK, Clark J, Coulombe N, Ethier N, Hebert TE, Sullivan R, Kargman S, Chateauneuf A, Tsukamoto N, McDonald T, Whiting P, Mezey E, Johnson MP, Liu Q, Ko-lakowski JrLF, Evans JF, Bonner TI, O’Neill GP. Identification of a GABAB receptor subunit, gb2, required for functional GABAB receptor activity. J Biol Chem. 1999;274:7607–7610. doi: 10.1074/jbc.274.12.7607. [DOI] [PubMed] [Google Scholar]
- 187.Nguyen L, Malgrange B, Breuskin I, Bettendorff L, Moonen G, Belachew S, Rigo JM. Autocrine/paracrine activation of the GABA(A) receptor inhibits the proliferation of neurogenic polysialylated neural cell adhesion molecule-positive (PSA-NCAM) precursor cells from postnatal striatum. J Neurosci. 2003;23:3278–3294. doi: 10.1523/JNEUROSCI.23-08-03278.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Nilsson M, Eriksson PS, Ronnback L, Hansson E. GABA induces Ca2+ transients in astrocytes. Neuroscience. 1993;54:605–614. doi: 10.1016/0306-4522(93)90232-5. [DOI] [PubMed] [Google Scholar]
- 189.Nishiyama A, Chang A, Trapp BD. NG2-glial cells: a novel glial cell population in the adult brain. J Neuropathol Exp Neurol. 1999;58:1113–1124. doi: 10.1097/00005072-199911000-00001. [DOI] [PubMed] [Google Scholar]
- 190.Nishiyama A, Lin XH, Giese N, Heldin CH, Stallcup WB. Co-localization of NG2 proteoglycan and PDGF alpha-receptor on O2A progenitor cells in the developing rat brain. J Neurosci Res. 1996;43:299–314. doi: 10.1002/(SICI)1097-4547(19960201)43:3<299::AID-JNR5>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- 191.Nusser Z, Sieghart W, Somogyi P. Segregation of differing GABA-A receptors to synaptic and extrasynaptic membranes of cerebellar granule cells. J Neurosci. 1998;18:1693–1703. doi: 10.1523/JNEUROSCI.18-05-01693.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Obrietan K, Gao XB, Van Den Pol AN. Excitatory actions of GABA increase BDNF expression via a MAPK-CREB-dependent mechanism; a positive feedback circuit in developing neurons. J Neurophysiol. 2002;88:1005–1015. doi: 10.1152/jn.2002.88.2.1005. [DOI] [PubMed] [Google Scholar]
- 193.Olsen RW, Snowhill EW, Wamsley JK. Autoradiographic localization of low affinity GABA receptors with [3H]bicuculline methochloride. Eur J Pharmacol. 1984;99:247–248. doi: 10.1016/0014-2999(84)90249-8. [DOI] [PubMed] [Google Scholar]
- 194.Owens DF, Kriegstein AR. Is there more to GABA than synaptic inhibition? Nat Rev Neurosci. 2002;3:715–727. doi: 10.1038/nrn919. [DOI] [PubMed] [Google Scholar]
- 195.Pannese E. The satellite cells of the sensory ganglia. Adv Anat Embryol Cell Biol. 1981;65:1–1111. doi: 10.1007/978-3-642-67750-2. [DOI] [PubMed] [Google Scholar]
- 196.Park-Chung M, Malayev A, Purdy RH, Gibbs TT, Farb DH. Sulfated and unsulfated steroids modulate gamma-aminobutyric acidA receptor function through distinct sites. Brain Res. 1999;830:72–87. doi: 10.1016/s0006-8993(99)01381-5. [DOI] [PubMed] [Google Scholar]
- 197.Park-Chung M, Wu FS, Farb DH. 3alpha-hydroxy-5beta-pregnan-20-one sulfate: a negative modulator of the NMDA-induced current in cultured neurons. Mol Pharmacol. 1994;46:146–150. [PubMed] [Google Scholar]
- 198.Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG. Glutamate-mediated astrocyte-neuron signalling. Nature. 1994;369:744–747. doi: 10.1038/369744a0. [DOI] [PubMed] [Google Scholar]
- 199.Pastor A, Chvatal A, Sykova E, Kettenmann H. Glycine- and GABA-activated currents in identified glial cells of the developing rat spinal cord slice. Eur J Nurosci. 1995;7:1188–1198. doi: 10.1111/j.1460-9568.1995.tb01109.x. [DOI] [PubMed] [Google Scholar]
- 200.Pellerin L, Magistretti P. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci USA. 1994;91:10625–10629. doi: 10.1073/pnas.91.22.10625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Peters A, Palay SL, deF Webster H. The fine structure of the nervous system: Neurons and their supporting cells. Third edition. New York: Oxford Univ. Press; 1991. [Google Scholar]
- 202.Pfaff T, Malitschek B, Kaupmann K, Prézeau L, Pin J-P, Bettler B, Karschin A. Alternative splicing generates a novel isoform of the rat metabotropic GABABR1 receptor. Eur J Neurosci. 1999;11:2874–2882. doi: 10.1046/j.1460-9568.1999.00704.x. [DOI] [PubMed] [Google Scholar]
- 203.Poisbeau P, Feltz P, Schlichter R. Modulation of GABA-A receptor-mediated IPSCs by neuroactive steroids in a rat hypo-thalamo-hypophyseal coculture model. J Physiol. 1997;500:475–485. doi: 10.1113/jphysiol.1997.sp022034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Porter JT, McCarthy KD. Hippocampal astrocytes in situ respond to glutamate released from synaptic terminals. J Neuro-sci. 1996;16:5073–5081. doi: 10.1523/JNEUROSCI.16-16-05073.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Porter JT, McCarthy KD. Astrocytic neurotransmitter receptors in situ and in vivo. Prog Neurobiol. 1997;51:439–455. doi: 10.1016/s0301-0082(96)00068-8. [DOI] [PubMed] [Google Scholar]
- 206.Pow DV, Robinson SR. Glutamate in some retinal neurons is derived solely from glia. Neuroscience. 1994;60:355–366. doi: 10.1016/0306-4522(94)90249-6. [DOI] [PubMed] [Google Scholar]
- 207.Puia G, Santi MR, Vicini S, Pritchett DB, Purdy RH, Paul SM, Seeburg PH, Costa E. Neurosteroids act on recombinant human GABAA receptors. Neuron. 1990;4:759–765. doi: 10.1016/0896-6273(90)90202-q. [DOI] [PubMed] [Google Scholar]
- 208.Raff MC, Abney ER, Cohen J, Lindsay R, Noble M. Two types of astrocytes in cultures of developing rat white matter: differences in morphology, surface gangliosides, and growth characteristics. J Neurosci. 1983;3:1289–1300. doi: 10.1523/JNEUROSCI.03-06-01289.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Raff MC, Miller RH, Noble M. A glial progenitor cell that develops in vitro into an astrocyte or an oligodendrocyte depending on culture medium. Nature. 1983;303:390–396. doi: 10.1038/303390a0. [DOI] [PubMed] [Google Scholar]
- 210.Reichenbach A. Glial K+ permeability and CNS K+ clearance by diffusion and spatial buffering. Ann NY Acad Sci. 1991;633:272–286. doi: 10.1111/j.1749-6632.1991.tb15620.x. [DOI] [PubMed] [Google Scholar]
- 211.Represa A, Ben-Ari Y. Trophic actions of GABA on neuronal development. Trends Neurosci. 2005;28:278–283. doi: 10.1016/j.tins.2005.03.010. [DOI] [PubMed] [Google Scholar]
- 212.Roberts E, Kuriyama K. Biochemical-physiological correlations in studies of the γ-aminobutyric acid system. Brain Res. 1968;8:1–35. doi: 10.1016/0006-8993(68)90170-4. [DOI] [PubMed] [Google Scholar]
- 213.Robitaille R. Modulation of synaptic efficacy and synaptic depression by glial cells at the frog neuromuscular junction. Neuron. 1998;21:847–855. doi: 10.1016/s0896-6273(00)80600-5. [DOI] [PubMed] [Google Scholar]
- 214.Rochon D, Rousse I, Robitaille R. Synapse-glia interactions at the mammalian neuromuscular junction. J Neurosci. 2001;21:3819–3829. doi: 10.1523/JNEUROSCI.21-11-03819.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Roy G, Philippe E, Gaulin F, Guay G. Peripheral projections of the chick primary sensory neurons expressing gamma-aminobutyric acid immunoreactivity. Neuroscience. 1991;45:177–183. doi: 10.1016/0306-4522(91)90113-3. [DOI] [PubMed] [Google Scholar]
- 216.Rozovsky I, Laping NJ, Krohn K, Teter B, O’Callaghan JP, Finch CE. Transcriptional regulation of glial fibrillary acidic protein by corticosterone in rat astrocytes in vitro is influenced by the duration of time in culture and by astrocyte-neuron interactions. Endocrinology. 1995;136:2066–2073. doi: 10.1210/endo.136.5.7720656. [DOI] [PubMed] [Google Scholar]
- 217.Runquist M, Alonso G. Gabaergic signaling mediates the morphological organization of astrocytes in the adult rat forebrain. Glia. 2003;41:137–151. doi: 10.1002/glia.10166. [DOI] [PubMed] [Google Scholar]
- 218.Rupprecht R, Holsboer F. Neuroactive steroids: mechanisms of action and neuropsychopharmacological perspectives. Trends Neurosci. 1999;22:410–416. doi: 10.1016/s0166-2236(99)01399-5. [DOI] [PubMed] [Google Scholar]
- 219.Sanna E, Talani G, Busonero F, Pisu MG, Purdy RH, Serra M, Biggio G. Brain steroidogenesis mediates ethanol modulation of GABA-A receptor activity in rat hippocampus. J Neurosci. 2004;24:6521–6530. doi: 10.1523/JNEUROSCI.0075-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Santagati S, Melcangi RC, Celotti F, Martini L, Maggi A. Estrogen receptor is expressed in different types of glial cells in culture. J Neurochem. 1994;63:2058–2064. doi: 10.1046/j.1471-4159.1994.63062058.x. [DOI] [PubMed] [Google Scholar]
- 221.Schlichter R, Keller AF, De Roo M, Breton JD, Inquimbert P, Poisbeau P. Fast non-genomic effects of steroids on synaptic transmission and role of endogenous neurosteroids in spinal pain pathways. J Mol Neurosci. 2005;28:33–52. doi: 10.1385/jmn:28:1:33. [DOI] [PubMed] [Google Scholar]
- 222.Schon F, Kelly JS. Autoradigraphic localisation of [3H]GABA and [3H]glutamate over satellite glial cells. Brain Res. 1974;66:275–288. [Google Scholar]
- 223.Schousboe A, Larsson OM, Wood JD, Krogsgaard-Larsen P. Transport and metabolism of gamma-aminobutyric acid in neurons and glia: implications for epilepsy. Epilepsia. 1983;24:531–538. doi: 10.1111/j.1528-1157.1983.tb03417.x. [DOI] [PubMed] [Google Scholar]
- 224.Schumacher M, Guennoun R, Mercier G, Désarnaud F, Lacor P, Bénavides J, Ferzaz B, Robert F, Baulieu EE. Pro-gesterone synthesis and myelin formation in peripheral nerves. Brain Res Rev. 2001;37:343–359. doi: 10.1016/s0165-0173(01)00139-4. [DOI] [PubMed] [Google Scholar]
- 225.Seifert G, Steinhausen C. Ionotropic glutamate receptors in astrocytes. Prog Brain Res. 2001;132:287–299. doi: 10.1016/S0079-6123(01)32083-6. [DOI] [PubMed] [Google Scholar]
- 226.Semyanov A, Walker MC, Kullmann DM. GABA uptake regulates cortical excitability via cell type-specific tonic inhibition. Nat Neurosci. 2003;6:484–490. doi: 10.1038/nn1043. [DOI] [PubMed] [Google Scholar]
- 227.Shao Y, McCarthy KD. Plasticity of astrocytes. Glia. 1994;11:147–155. doi: 10.1002/glia.440110209. [DOI] [PubMed] [Google Scholar]
- 228.Shelton MK, McCarthy KD. Hippocampal astrocytes exhibit Ca2+-elevating muscarinic cholinergic and histaminergic receptors in situ. J Neurochem. 2000;74:555–563. doi: 10.1046/j.1471-4159.2000.740555.x. [DOI] [PubMed] [Google Scholar]
- 229.Shen H, Gong QH, Yuan M, Smith SS. Short-term steroid treatment increases δ GABA-A receptor subunit expression in rat CA1 hippocampus: Pharmacological and behavioral effects. Neuropharmacology. 2005;49:573–586. doi: 10.1016/j.neuropharm.2005.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Shu H-J, Eisenman LN, Jinadasa D, Covey DF, Zorumski CF, Mennerick S. Slow actions of neuroactive steroids at GABA-A receptors. J Neurosci. 2004;24:6667–6675. doi: 10.1523/JNEUROSCI.1399-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Sieghart W, Sperk G. Subunit composition, distribution and function of GABA(A) receptor subtypes. Curr Top Med Chem. 2002;2:795–816. doi: 10.2174/1568026023393507. [DOI] [PubMed] [Google Scholar]
- 232.Slezak M, Pfrieger FW. New roles for astrocytes: regulation of CNS synaptogenesis. Trends Neurosci. 2003;26:531–535. doi: 10.1016/j.tins.2003.08.005. [DOI] [PubMed] [Google Scholar]
- 233.Somjen GG. Electrophysiology of neuroglia. Annu Rev Physiol. 1975;37:163–191. doi: 10.1146/annurev.ph.37.030175.001115. [DOI] [PubMed] [Google Scholar]
- 234.Somjen GG. Nervenkitt: notes on the history of the concept of neuroglia. Glia. 1988;1:2–9. doi: 10.1002/glia.440010103. [DOI] [PubMed] [Google Scholar]
- 235.Spivak CE. Desensitization and noncompetitive blockade of GABA-A receptors in ventral midbrain neurons by a neuroster-oid dehydroepiandrosterone sulfate. Synapse. 1994;16:113–122. doi: 10.1002/syn.890160205. [DOI] [PubMed] [Google Scholar]
- 236.Stevens B, Fields RD. Response of Schwann cells to action potentials in development. Science. 2000;287:2267–2271. doi: 10.1126/science.287.5461.2267. [DOI] [PubMed] [Google Scholar]
- 237.Stevens B, Porta S, Haak LL, Gallo V, Fields RD. Adenosine: a neuron-glial transmitter promoting myelination in the CNS in response to action potentials. Neuron. 2002;36:855–868. doi: 10.1016/s0896-6273(02)01067-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Stone DJ, Song Y, Anderson CP, Krohn KK, Finch CE, Rozovsky I. Bidirectional transcription regulation of glial fibrillary acidic protein by estradiol in vivo and in vitro. Endocri-nology. 1998;139:3202–3209. doi: 10.1210/endo.139.7.6084. [DOI] [PubMed] [Google Scholar]
- 239.Sun BB, Chiu SY. N-type calcium channels and their regulation by GABA-B receptors in axons of neonatal rat optic nerve. J Neurosci. 1999;19:5185–5194. doi: 10.1523/JNEUROSCI.19-13-05185.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Swope SL, Moss SJ, Blackstone CD, Huganir RL. Phosphorylation of ligand-gated ion channels: a possible mode of synaptic plasticity. FASEB J. 1992;6:2514–23. [PubMed] [Google Scholar]
- 241.Thi AD, Jung-Testas I, Baulieu EE. Neuronal signals are required for estrogen-mediated induction of progesterone receptor in cultured rat Schwann cells. J Steroid Biochem Mol Biol. 1998;67:201–211. doi: 10.1016/s0960-0760(98)00116-2. [DOI] [PubMed] [Google Scholar]
- 242.Towers S, Princivalle A, Billinton A, Edmunds M, Bettler B, Urban L, Castro-Lopes J, Bowery NG. GABAB receptor protein and mRNA distribution in rat spinal cord and dorsal root ganglia. Eur J Neurosci. 2000;12:3201–3210. doi: 10.1046/j.1460-9568.2000.00237.x. [DOI] [PubMed] [Google Scholar]
- 243.Tsuneishi S, Takada S, Motoike T, Ohashi T, Sano K, Na-kamura H. Effects of dexamethasone on the expression of myelin basic protein, proteolipid protein, and glial fibrillary acidic protein genes in developing rat brain. Dev Brain Res. 1991;61:117–123. doi: 10.1016/0165-3806(91)90121-x. [DOI] [PubMed] [Google Scholar]
- 244.Tunnicliff G. 4-aminobutyrate transaminase. In: Boulton AA, Baker GB, Yu PH, editors. Neuromethods. Clifton, N.J.: Humana Press; 1986. pp. 389–419. (Series I: Neuro-chemistry. 5. Neurotransmitter Enzymes). [Google Scholar]
- 245.Valeyev AY, Hackman JC, Holohean AM, Wood PM, Katz JL, Davidoff RA. GABA-induced Cl- current in cultured embryonic human dorsal root ganglion neurons. J Neuro-physiol. 1999;82:1–9. doi: 10.1152/jn.1999.82.1.1. [DOI] [PubMed] [Google Scholar]
- 246.Vegeto E, Bonincontro C, Pollio G, Sala A, Viappiani S, Nardi F, Brusadelli A, Viviani B, Ciana P, Maggi A. Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. J Neurosci. 2001;21:1809–1818. doi: 10.1523/JNEUROSCI.21-06-01809.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Verkhratsky A, Orkand RK, Kettenmann H. Glial calcium: homeostasis and signaling function. Physiol Rev. 1998;78:99–141. doi: 10.1152/physrev.1998.78.1.99. [DOI] [PubMed] [Google Scholar]
- 248.Vielkind U, Walencewicz A, Levine JM, Bohn MC. Type II glucocorticoid receptors are expressed in oligodendrocytes and astrocytes. J Neurosci Res. 1990;27:360–373. doi: 10.1002/jnr.490270315. [DOI] [PubMed] [Google Scholar]
- 249.Villegas SN, Poletta FA, Carri NG. Glia: a reassessment based on novel data on the developing and mature central nervous system. Cell Biol Int. 2003;27:599–609. doi: 10.1016/s1065-6995(03)00119-7. [DOI] [PubMed] [Google Scholar]
- 250.Vitry S, Lachapelle F, Avellana-Adalid V, Baron-Van Everco-oren A. Migration and multipotentiality of PSA-NCAM neural precursors transplanted in the developing brain. Mol Cell Neurosci. 2001;17:983–1000. doi: 10.1006/mcne.2001.0987. [DOI] [PubMed] [Google Scholar]
- 251.Von Blankenfeld G, Trotter J, Kettenmann H. Expression and developmental regulation of a GABAA receptor in cultured murine cells of the oligodendrocyte lineage. Eur J Neuro-sci. 1991;3:310–316. doi: 10.1111/j.1460-9568.1991.tb00817.x. [DOI] [PubMed] [Google Scholar]
- 252.Wang Z, Haydon PG, Yeung ES. Direct observation of calcium-independent intercellular ATP signalling in astrocytes. Anal Chem. 2000;72:2001–2007. doi: 10.1021/ac9912146. [DOI] [PubMed] [Google Scholar]
- 253.Watanabe M, Maemura K, Kanbara K, Tamayama T, Hayasaki H. GABA and GABA receptors in the central nervous system and other organs. Int Rev Cytol. 2002;213:1–47. doi: 10.1016/s0074-7696(02)13011-7. [DOI] [PubMed] [Google Scholar]
- 254.Watkins LR, Maier SF. Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiol Rev. 2002;82:981–1011. doi: 10.1152/physrev.00011.2002. [DOI] [PubMed] [Google Scholar]
- 255.White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, Barnes AA, Emson P, Foord SM, Marshall FH. Heterodimerization is required for the formation of a functional GABAB receptor. Nature. 1998;396:679–682. doi: 10.1038/25354. [DOI] [PubMed] [Google Scholar]
- 256.Whiting PJ, McAllister G, Vasilatis D, Bonnert TP, Heavens RP, Smith DW, Hewson L, O’Donnell R, Rigby MR, Sirinathainghji DJS, Marshall G, Thompson SA, Wafford KA. Neuronally restricted splicing regulates the expression of a novel GABAA receptor subunit conferring atypical functional properties. J Neurosci. 1997;17:5027–5037. doi: 10.1523/JNEUROSCI.17-13-05027.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Whiting PJ, McKernan RM, Wafford KA. Structure and pharmacology of vertebrate GABAA receptor subtypes. Int Rev Neurobiol. 1995;38:95–138. doi: 10.1016/s0074-7742(08)60525-5. [DOI] [PubMed] [Google Scholar]
- 258.Wilkin GP, Marriott DR, Cholewinski AJ. Astrocyte heterogeneity. Trends Neurosci. 1990;13:43–46. doi: 10.1016/0166-2236(90)90065-i. [DOI] [PubMed] [Google Scholar]
- 259.Williamson AV, Mellor JR, Grant AL, Randall AD. Properties of GABA-A receptors in cultured rat oligodendrocyte progenitor cells. Neuropharmacology. 1998;37:859–873. doi: 10.1016/s0028-3908(98)00016-1. [DOI] [PubMed] [Google Scholar]
- 260.Wu FS, Gibbs TT, Farb DH. Pregnenolone sulfate: a positive allosteric modulator at the N-methyl-D-aspartate receptor. Mol Pharmacol. 1991;40:333–336. [PubMed] [Google Scholar]
- 261.Xilouri M, Papazafiri P. Anti-apoptotic effects of allo-pregnanolone on P19 neurons. Eur J Neurosci. 2006;23:43–54. doi: 10.1111/j.1460-9568.2005.04548.x. [DOI] [PubMed] [Google Scholar]
- 262.Zagorodnyuk VP, D’Antona G, Brookes SJ, Costa M. Functional GABA-B receptors are present in guinea pig nodose ganglion cell bodies but not in peripheral mechanosensitive endings. Auton Neurosci. 2002;29:20–29. doi: 10.1016/s1566-0702(02)00183-2. [DOI] [PubMed] [Google Scholar]