Articles

  • KSBNS 2024

Article

Review Article

Exp Neurobiol 2016; 25(5): 222-232

Published online October 31, 2016

https://doi.org/10.5607/en.2016.25.5.222

© The Korean Society for Brain and Neural Sciences

Two-pore Domain Potassium Channels in Astrocytes

Kanghyun Ryoo and Jae-Yong Park*

School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea

Correspondence to: *To whom correspondence should be addressed.
TEL: 82-2-3290-5637, FAX: 82-2-921-7207
e-mail: jaeyong68@korea.ac.kr

Received: July 29, 2016; Revised: September 1, 2016; Accepted: September 15, 2016

Two-pore domain potassium (K2P) channels have a distinct structure and channel properties, and are involved in a background K+ current. The 15 members of the K2P channels are identified and classified into six subfamilies on the basis of their sequence similarities. The activity of the channels is dynamically regulated by various physical, chemical, and biological effectors. The channels are expressed in a wide variety of tissues in mammals in an isoform specific manner, and play various roles in many physiological and pathophysiological conditions. To function as channels, the K2P channels form dimers, and some isoforms form heterodimers that provide diversity in channel properties. In the brain, TWIK1, TREK1, TREK2, TRAAK, TASK1, and TASK3 are predominantly expressed in various regions, including the cerebral cortex, dentate gyrus, CA1-CA3, and granular layer of the cerebellum. TWIK1, TREK1, and TASK1 are highly expressed in astrocytes, where they play specific cellular roles. Astrocytes keep leak K+ conductance, called the passive conductance, which mainly involves TWIK1-TREK1 heterodimeric channel. TWIK1 and TREK1 also mediate glutamate release from astrocytes in an exocytosis-independent manner. The expression of TREK1 and TREK2 in astrocytes increases under ischemic conditions, that enhance neuroprotection from ischemia. Accumulated evidence has indicated that astrocytes, together with neurons, are involved in brain function, with the K2P channels playing critical role in these astrocytes.

Keywords: K2P channel, astrocyte, passive conductance, glutamate release

Potassium (K+) channels are membrane proteins that specifically transport K+ across the membrane and play important roles in cell volume regulation, hormone secretion, heart-beat, synaptic transmission and muscle contraction. The K+ channels are a superfamily of diverse members involved in K+ currents in various types of tissues. On the basis of structure, the K+ channels are categorized into voltage-gated K+ channel (Kv), inward rectifying K+ channel (Kir) and two-pore domain K+ channel (K2P) families [1,2,3].

The K2P channels were cloned by data mining from a genetic database, with 15 members currently identified. They induce leak background currents in heterologous systems as well as in endogenous tissues [4,5,6,7,8,9,10,11,12,13,14,15,16,17]. Loss-of-function studies using knockout animals have reported that the channels are expressed in various tissues and are involved in urine production, anesthesia, pain perception, depression, and neuroprotection [18]. Most studies on the K2P channels performed in nervous system have focused on neurons in the central and peripheral nervous systems, which have been thoroughly discussed in excellent reviews [19,20,21,22]. Although astrocytes are the most dominant types of cells in the brain and some K2P channels are highly expressed in these types of cells, their role in astrocytes have not been well studied. Accumulated evidence has recently suggested that astrocytic K2P channels play critical roles in brain function. The purpose of this review is therefore to provide an overview of the function of astrocytic K2P channels, and to discuss the direction of further studies of these channels in astrocytes.

Background K+ currents have been known since the early period of electrophysiology, but it took more than 50 years to identify the molecular components. Tandem of pore domains in a weak inward rectifying K+ channel 1 (TWIK1) was identified as an unconventional K+ channel, which showed very different electrophysiological properties from classical K+ channels such as Kv and Kir channels. This channel uniquely contains two pore (P) domains, whereas the Kv and Kir channels contain only one P domain [23]. Since TWIK1 was cloned, 14 additional K2P channels, sharing a highly conserved molecular architecture, have been identified in mammals. Based on the sequence similarity and functional characteristics, the 15 K2P channels are categorized into six subfamilies, including TWIK, TREK (TWIK-related K+ channel), TASK (TWIK-related acid-sensitive K+ channel), TALK (TWIK-related alkaline pH-activated K+ channel), THIK (tandem pore domain halothane-inhibited K+ channel) and TRESK (TWIK-related spinal cord K+ channel) families [19,24,25].

The K2P channels are not activated by a voltage change that induces pore openings of Kv channels, but mediate an instantaneous current at a wide range of membrane potentials [7,8,23,26]. They show a current-voltage (I-V) relationship similar to the one following the Goldman-Hodgkin-Katz (GHK) equation. They are also time-independent and do not show inactivation kinetics in their current, suggesting that the K2P channels have a distinct mechanism of pore gating [7]. However, the K2P channels have more or less tendency of rectifying currents. For example, TREK1 and TREK2 carry outward rectifying currents and TWIK1 and TASK3 carry slightly inward currents in heterologous systems that are induced by a specific range of membrane potentials, extracellular K+ concentrations, or divalent cations, such as magnesium (Mg2+) [5,8,10,13,15,23]. In addition, a recent study suggested that K2P channels have a non-canonical voltage sensing mechanism to gate their pores and show voltagedependent manners, although this possible process needs to be confirmed in physiological conditions [27]. Each K2P channel does not completely follow the electrophysiological properties of an ideal background K+ channel. However, the 15 isoforms have different voltage dependencies at different ranges of conditions and different expression patterns. It has been suggested that these characteristics enable the K2P channels to function as background channels in excitable and non-excitable cells.

Although the members of the K2P family have relatively low sequence homology, they share a very similar molecular architecture. They have 4 transmembrane (TM) domains, 2 P domains, a long extracellular loop between TM1 and P1, and the N- and the C-termini that are oriented into the intracellular space. The K2P channels form dimers to make functional K+ pore, which is composed of pseudo-tetrameric pore units and selectivity filter sequences in each P domain to provide the specificity for K+. The 4TM/2P structure of the K2P channel is well conserved from Caenorhabditis elegans and Drosophila melanogaster to mammals [28,29,30]. In the dimeric structure of TWIK1, the two extracellular loops in each subunit are located close to each other, and two cysteine residues in the extracellular loops form a disulfide bond [12]. In addition, TREK2 and TRAAK have cysteine residues in their external loops, which are involved in intersubunit disulfide bridges in a similar manner to TWIK1 [31,32,33]. In general, K2P channels form homodimers of two of the same isoforms, and the characteristics of the channels are determined by the single isoforms. Recently some K2P isoforms were discovered that form heterodimers between two different isoforms, and the heterodimers showed different channel properties from those of the homodimeric channels of each single isoform [34,35,36,37,38,39,40,41]. The heterodimerization of the channels therefore provides diversity in channel properties.

The activity of K2P channels are regulated by various modulators, such as physical parameters, chemical signaling and biological signaling. TREK1, TREK2, and TRAAK are mechanosensitive channels, whose activities are enhanced if negative pressure is applied through a patch pipette during an electrophysiological recording [13,42,43]. TASK1 and TASK3 are inhibited by extracellular acidification but are insensitive to intracellular pH changes [7,10]. TREK1 and TRAAK are inhibited by extracellular acidification, while internal acidification activates TREK1 and TREK2 [44,45,46]. At alkaline pH, TALK1 and TALK2 are inhibited and TRAAK is activated by internal alkalization [47,48]. TREK1 activity is enhanced by heat between 22~42℃, which is a gradual and reversible process [49]. TREK2 and TRAAK are also controlled by temperature changes [50].

The K2P channels are also differentially modulated by lipid molecules. TREK1, TREK2, and TRAAK are activated by arachidonic acid (AA) and other polyunsaturated fatty acids, such as docosahexaenoic acid and linoleic acid [5,13,26,43,51]. TREK1 and TRAAK are also activated by lysophospholipids, including lysophosphatidylcholine and lysophosphatitylinositol [51,52]. In contrast, TRESK is inhibited by AA and docosahexaenoic acid [17]. The K+ currents mediated by TREK1 and TREK2 are dramatically increased by volatile anesthetics, including chloroform, halothane, isoflurane, and diethyl ether but TRAAK is insensitive to these anesthetics [13,53]. TASK1 activity is enhanced by halothane and isoflurane but is insensitive or decreased by chloroform and diethyl ether, respectively [53,54].

The K2P channels are regulated by various types of G-protein coupled receptors (GPCRs). TREK1 activity is inhibited by the activation of Gαq-coupled receptors, such as the thyrotropin-releasing hormone receptor 1 (TRHR1) and the Orexin receptor (Orx1R), but is enhanced by the Gαi-coupled receptors, including the metabotropic glutamate receptor 4 (mGluR4). A series of studies using pharmacological inhibitors and the mutants of TREK1 reported that the phosphorylation status of two serine residues in the C-terminal domain (S300 and S333) is critical in TREK1 regulation by GPCRs, and is dependent on protein kinase C (PKC) and protein kinase A (PKA) [55,56,57]. Recently, TREK1-mediated fast glutamate release from astrocytes has been reported to be triggered by TREK1 interaction with Gβγ subunits [38,58]. The TREK2 is also inhibited by the activation of Gαs- and Gαq-coupled receptors, such as 5HT4sR, mGluR1, and the M3 muscarinic receptor [13,59]. The Gαi-coupled receptors, such as the mGluR2, α2A adrenergic receptor and the GABAB receptor increase the current mediated by the TREK2 channel [13,60,61]. TASK1 and TASK3 are inhibited by activation of Gαq-coupled receptors, such as group I mGluRs, TRHR1, M3 muscarinic receptor, and angiotensin II (AT1a) receptors [37,62,63,64].

The binding partners of the K2P channels have been identified using yeast two-hybrid screening and immunoprecipitation combined with mass spectrophotometry. β-COP, 14-3-3β, and p11 are known to interact with TASK1 and regulate the trafficking of the channel. The β-COP, a subunit Coat Protein Complex I (COPI) keeps TASK1 in the endoplasmic reticulum (ER) by interaction with both the N- and C-termini of the channel, but the 14-3-3β induces forward trafficking of the channel to the cytoplasmic membrane [65,66]. The p11, a subunit of annexin II, which plays an important role in aldosterone secretion, allows TASK1 to release from ER retention by direct interaction with the channel [67]. A-kinase-anchoring protein 150 (AKAP150) and Mtap2, a microtubule-associated protein, were identified as the interacting proteins that bind to the C-terminal domain of TREK1 and TREK2. The AKAP150 increases the channel currents mediated by TREK1 and TREK2 and modulates the sensitivity of the channels to physical parameters and signaling mediated by Gαs-/Gαq-coupled receptors. The Mtap2 enhances localizations to the cytoplasmic membrane and the current densities of TREK1 and TREK2 [68,69]. The β-COP was identified as the interacting protein of TREK1, and increases both the surface expression and the current by binding to the N-terminal domain of the channel [70]. Intracellular Ca2+-induced activation of the TRESK channel is regulated by interaction with calcineurin and 14-3-3η [71,72].

The K2P channels have been reported to be expressed in many organs including the heart, kidney, lung, liver, pancreas, placenta, testis, spinal cord, and brain [5,6,7,8,13,23,26,73]. In the brain, TWIK1, TREK1, TREK2, TRAAK, TASK1, and TASK3 are preferentially expressed in specific regions in an isoform-dependent manner [22]. In situ hybridization showed that TWIK1 was expressed in the cerebral cortex, the dentate gyrus of the hippocampus, and the granular layer of the cerebellum [74,75]. TREK1 was detected in almost all areas of the rat brain including the cerebral cortex, basal ganglia, hippocampus, hypothalamus, mesencephalon and rhombencephalon [76]. TREK2 expression is high in the granule cell layer of the cerebellum [5,75]. TASK1 also shows a heterogeneous distribution in the brain, and its mRNA levels are high in the cerebral cortex, CA1-CA3, dentate gyrus, paraventricular thalamic nuclei, substantia nigra, and cerebellum [7,63,75]. In nervous systems, the channels are expressed in many kinds of neurons, including projection neurons, interneurons, motor neurons, and sensory neurons [75,76,77,78].

When compared with the number of neurons in the brain, the glia comprise a much larger percentage of the total number of cells in the brain, and astrocytes are one of the dominant cell types of glia [79,80]. Recently, many studies have reported that K2P channels are also expressed in various types of astrocytes. TASK1 was detected in the astrocytes of the rat hippocampus by immunohistochemistry [81,82]. A transcriptome analysis of astrocytes purified from mouse forebrains and a single-cell RT-PCR study with hippocampal astrocytes suggested that TWIK1 and TREK1 were expressed in astrocytes [83,84].

Passive conductance

The electrophysiological properties of astrocytes change during the maturation processes. Mature astrocytes typically show a very negative membrane potential (VM) and a linear I-V relationship. These features are thought to enable astrocytes homeostatic functions in the brain, such as extracellular K+ buffering and the uptake of released neurotransmitters from presynaptic neurons [85,86,87,88]. This linear I-V relationship, called passive conductance, results from leak K+ membrane conductance and is not affected by time and voltage. The molecular identity of the passive conductance is not associated with Kv channels, because is very similar to the linear I-V relationship described by the GHK equation [88,89,90,91].

Recent studies have suggested that TWIK1 and TREK1 predominantly mediate the astrocytic passive conductance in the hippocampus [38,92]. The passive conductance is significantly reduced by quinine that inhibits both TWIK1 and TREK1 channels, while not being affected by Ba2+, a blocker of classical K+ channels [92]. The gene silencing of one or both of TWIK1 and TREK1 by knockdown using shRNA markedly reduced the passive conductance [38]. In addition, TWIK1 and TREK1 form a heterodimeric channel involving an intermolecular disulfide bridge between two cysteine residues in each extracellular loop. The two K2P channels mutually regulate the surface expression of each other by heterodimerization in hippocampal astrocytes [32,33,38]. The inhibition of channel activity by inhibitors or gene silencing reduces most but not all of the passive conductance. TWIK1 and TREK1 are major isoforms of the K2P channels in hippocampal astrocytes and are responsible for at least 80 % of the entire passive conductance (Fig. 1, blue box) [38,92]. The expression profiles of the K2P channels are dependent on the region of the brain, so the passive conductance may be controlled by different K2P channels in different regions, suggesting that other K+ channels expressed in astrocytes, such as TREK2 or pHsensitive K2P channels such as TASKs may partially contribute to the passive conductance [84,93,94].

Glutamate release

In the brain, glutamate, one of the most important neurotransmitters, is released from excitatory presynaptic neurons and mediates synaptic transmission among neurons. Glutamate is also released from astrocytes and functions as a gliotransmitter, that modulates synaptic transmission and finely tunes synaptic plasticity [95]. There is controversy concerning the release mechanism of glutamate from astrocytes, while the release mechanism from neurons is well understood. The controversy regarding glutamate release from astrocytes involves whether it is mediated by vesicular exocytosis or by channels or transporters [96,97].

The observation that small vesicular organelles containing vesicular glutamate transporters were found in astrocytes and astrocyte-dependent synaptic potentiation was prevented by introduction of the active light-chain of tetanus neurotoxin (TeNT) into astrocytes suggested that astrocytic glutamate release is mediated by vesicular exocytosis [98,99]. However, these studies did not directly show that the glutamate release from astrocytes by astrocytic stimulation. In addition, another study reported that ATP- or hypotonicity-induced astrocytic glutamate release was not affected by TeNT [100]. Studies using pharmacological inhibitors reported that some anionic channels were involved in glutamate release from astrocytes without exocytotic machinery. Anionic channel blockers, such as 4,4'-diisothiocyanostilbene-2,2'-disulphonic acid, 5-nitro-2-(3-phenylpropylamino)-benzoic acid, flufenamic acid, and gossypol drastically inhibited astrocytic glutamate release [100,101].

A recent study using the sniffer-patch technique, a highly sensitive detection system for glutamate release from astrocytes revealed that TREK1 and Best1, an anion channel, mediated fast and slow glutamate release, respectively [58]. These two types of astrocytic glutamate release were independent of the exocytotic machinery, and the fast release mediated by TREK1 did not require an increase in intracellular Ca2+. Furthermore, to facilitate glutamate permeability, TREK1 needed to form a heterodimer with TWIK1, and gene silencing of only one of the two K2P isoforms inhibited the glutamate release from astrocytes [38]. Because the astrocytic glutamate release is triggered by the activation of GPCRs, such as PAR1 and CB1, the synaptic transmission can be fine-tuned by input from various synaptic environments (Fig. 1, orange box) [38,58].

The protection from ischemia

In the brain, ischemia elicits hypoxia and hypoglycemia that induce neuronal excitotoxicity due to glutamate and cause can severe brain damage resulting in disability and death [102]. Astrocytes play pivotal roles in the protection of neurons from toxicity induced by stress, while also supporting neuronal survival and general physiological functions. Astrocytes retain much hyperpolarized membrane potential and leak K+ conductance in the resting condition, which provides a major driving force for glutamate uptake and K+ spatial buffering [103,104]. Many studies have suggested that K2P channels contribute to the ability of astrocytes to protect neurons against ischemia.

TREK1-/- mice were more vulnerable to global ischemia following transient bilateral occlusion of common carotid arteries. Furthermore, TREK1+/+ mice showed an improved survival against ischemia after injection of linoleic acid or lysophosphatidylcholine, that are activators of TREK1, but the survival rate of TREK1-/- mice was not affected [105]. TREK1 expression was drastically and gradually increased after cerebral ischemia induced by middle cerebral artery occlusion in a time dependent manner in hippocampal CA1 and cerebral cortex [106,107]. The proliferation of cultured cortical astrocytes was increased by exposure to hypoxic condition, but was suppressed by treatment with quinine [107]. The clearance ability of extracellular glutamate by astrocytes was inhibited by blocking of TREK1 with quinine or bupivacaine under hypoxic conditions [108]. In addition, linoleic acid reduced apoptotic cell death, aquaporin 4 expression, and microglial activation induced by cerebral ischemia [106]. TREK2 expression were also increased by hypoxic exposure in cultured cortical astrocytes and hippocampal astrocytes in vivo, and the increase was mediated by de novo translation of TREK2 protein [109,110]. The increase of TREK channels by hypoxic conditions may therefore be a defense mechanism against glutamate toxicity induced by hypoxia.

The K2P channels are expressed in both of neurons and astrocytes. In general, excitable neurons and non-excitable astrocytes have different cellular contents and physiology, so it is expected that the function and regulation of K2P channels in astrocytes are different from those in neurons. However, most studies on K2P channels in the brain have been focused on neuron, and we do not well understand how the channels function in astrocytes. Our current knowledge provides some clues regarding future studies of K2P channels in astrocytes (Fig. 1, grey objects and dashed lines).

A series of knockout animals with deleted genes encoding the K2P channels were developed and used to investigate the loss-of-function of the channels. These studies made significant contributions to the current knowledge of K2P channels [18]. However, the whole body knockout animals did not simultaneously express the target channels in neurons and astrocytes, so it is not clear whether the effect of gene silencing originated from neurons or astrocytes. To definitively establish astrocyte-specific loss-of-function, the gene-silencing should be performed using astrocyte-specific promoters, such as glial fibrillary acidic protein (GFAP), glutamate-aspartate transporter (GLAST), or glutamate transporter-1 (GLT-1) promoters [111,112]. Conditional knockout mice where the K2P channel expression is removed only in astrocytes will provide a better system to assess the astrocyte-specific function of the channels.

The cellular localization and the activity of the K2P channels are regulated by various interacting proteins [65,66,67,68,69,70,71,72]. Although no interacting partner of the K2P channels in astrocytes has not been identified yet, there is a strong possibility that the channels can be regulated by interacting proteins in astrocytes. Importantly, some of the known proteins interacting with K2P channels in other types of cells are also expressed in astrocytes [113,114,115], and should be the objects of future studies. In addition, new interacting partners expressed in astrocytes should be identified to learn more about the regulatory mechanisms of the channels.

The K2P channel activity is dynamically and diversely regulated by signals via GPCRs [55,56,57,59,60,61,62,63,64]. In particular, PAR1 and CB1 are already known to induce glutamate release from astrocytes by interaction with TREK1 [38,58]. In astrocytes, various kinds of GPCRs are expressed and contribute to cell-tocell communication and functional regulation of the cells [116,117]. These results suggest that there may be many regulatory mechanisms of K2P channels involving GPCRs, which can provide new therapeutic targets for the treatment of neurodegenerative and psychiatric diseases.

The K2P channels have distinct characteristics as K+ channels that differ from Kv or Kir channels. The K2P channels mediate leak K+ conductance independently of time or voltage and are not characterized by inactivation kinetics. The K2P channels therefore function as background K+ channels that are evolutionarily conserved from C. elegance to mammals. The 15 members of the mammalian K2P family are specifically expressed in a wide variety of tissues in an isoform-dependent manner, and are involved in various physiological and pathophysiological phenomena. In astrocytes, TWIK1, TREK1, TREK2, TASK1, and TASK3 are the dominantly expressed isoforms that contribute to the functions of astrocytes. Furthermore, K2P channels play critical roles in the induction of passive conductance, in glutamate release, and in neuroprotection from hypoxia. Recently, the K2P channels have been reported to be involved in hippocampal epilepsy and cellular volume regulation [118,119,120,121,122]. These findings suggest that the K2P channels can be new therapeutic targets for treatment of neurodegenerative and psychiatric diseases in astrocytes.

Because astrocytes are physically and functionally associated with neurons, and the K2P channels are expressed in both types of cells, it is difficult to study the K2P channels in astrocytes using conventional strategies. To investigate the function of the K2P channels in astrocytes, it is therefore necessary to use astrocyte-specific conditional knockout animals, high-throughput screening to find the interaction proteins with the channels in astrocytes, and small molecular modulators. The new approaches should provide novel strategies to better understand the K2P channels in astrocytes.

Fig. 1. The K2P channels in astrocytes. The scheme summarizes the known functions and regulatory mechanisms of K2P channels in astrocytes, and suggests the strategies for future studies. In astrocytes, TWIK1 (green) and TREK1 (blue) are highly expressed and form heterodimeric channels. The TWIK1-TREK1 channel mainly contributes to passive conductance that is a leak K+ background current in astrocytes (blue box). The heterodimeric channel also mediates glutamate release from astrocytes that does not require exocytotic machinery proteins, such as SNARE components. The release process is triggered by a direct interaction between a TREK1 subunit and the Gβγ subunits after the activation of Gαi-protein coupled receptors (orange box). βCOP, PKA, and PKC are already known to control TREK1 by protein-protein interaction or by phosphorylation after GPCR activation in non-astrocytic cells. However, the effector proteins are also expressed in astrocytes, so they may regulate the TWIK1-TREK1 channel by similar mechanisms (gray objects and dashed lines). In addition, other novel interacting partners of the K2P channels that can be novel regulators are expected to be identified in astrocytes in future studies. As suggested by some studies, there is the possibility that other K2P channels, such as TASK1 may be expressed and could contribute to the function of astrocytes (K2P channel in orange).
  1. Choe S. Potassium channel structures. Nat Rev Neurosci 2002;3:115-121.
    Pubmed
  2. Coetzee WA, Amarillo Y, Chiu J, Chow A, Lau D, McCormack T, Moreno H, Nadal MS, Ozaita A, Pountney D, Saganich M, Vega-Saenz de Miera E, Rudy B. Molecular diversity of K+ channels. Ann N Y Acad Sci 1999;868:233-285.
    Pubmed
  3. Maljevic S, Lerche H. Potassium channels: a review of broadening therapeutic possibilities for neurological diseases. J Neurol 2013;260:2201-2211.
    Pubmed
  4. Ashmole I, Goodwin PA, Stanfield PR. TASK-5, a novel member of the tandem pore K+ channel family. Pflugers Arch 2001;442:828-833.
    Pubmed
  5. Bang H, Kim Y, Kim D. TREK-2, a new member of the mechanosensitive tandem-pore K+ channel family. J Biol Chem 2000;275:17412-17419.
    Pubmed
  6. Chavez RA, Gray AT, Zhao BB, Kindler CH, Mazurek MJ, Mehta Y, Forsayeth JR, Yost CS. TWIK-2, a new weak inward rectifying member of the tandem pore domain potassium channel family. J Biol Chem 1999;274:7887-7892.
    Pubmed
  7. Duprat F, Lesage F, Fink M, Reyes R, Heurteaux C, Lazdunski M. TASK, a human background K+ channel to sense external pH variations near physiological pH. EMBO J 1997;16:5464-5471.
    Pubmed
  8. Fink M, Duprat F, Lesage F, Reyes R, Romey G, Heurteaux C, Lazdunski M. Cloning, functional expression and brain localization of a novel unconventional outward rectifier K+ channel. EMBO J 1996;15:6854-6862.
    Pubmed
  9. Girard C, Duprat F, Terrenoire C, Tinel N, Fosset M, Romey G, Lazdunski M, Lesage F. Genomic and functional characteristics of novel human pancreatic 2P domain K(+) channels. Biochem Biophys Res Commun 2001;282:249-256.
    Pubmed
  10. Kim Y, Bang H, Kim D. TASK-3, a new member of the tandem pore K(+) channel family. J Biol Chem 2000;275:9340-9347.
    Pubmed
  11. Lesage F, Maingret F, Lazdunski M. Cloning and expression of human TRAAK, a polyunsaturated fatty acids-activated and mechano-sensitive K(+) channel. FEBS Lett 2000;471:137-140.
    Pubmed
  12. Lesage F, Reyes R, Fink M, Duprat F, Guillemare E, Lazdunski M. Dimerization of TWIK-1 K+ channel subunits via a disulfide bridge. EMBO J 1996;15:6400-6407.
    Pubmed
  13. Lesage F, Terrenoire C, Romey G, Lazdunski M. Human TREK2, a 2P domain mechano-sensitive K+ channel with multiple regulations by polyunsaturated fatty acids, lysophospholipids, and Gs, Gi, and Gq protein-coupled receptors. J Biol Chem 2000;275:28398-28405.
    Pubmed
  14. Rajan S, Wischmeyer E, Karschin C, Preisig-Müller R, Grzeschik KH, Daut J, Karschin A, Derst C. THIK-1 and THIK-2, a novel subfamily of tandem pore domain K+ channels. J Biol Chem 2001;276:7302-7311.
    Pubmed
  15. Rajan S, Wischmeyer E, Xin Liu G, Preisig-Müller R, Daut J, Karschin A, Derst C. TASK-3, a novel tandem pore domain acid-sensitive K+ channel. An extracellular histiding as pH sensor. J Biol Chem 2000;275:16650-16657.
    Pubmed
  16. Salinas M, Reyes R, Lesage F, Fosset M, Heurteaux C, Romey G, Lazdunski M. Cloning of a new mouse two-P domain channel subunit and a human homologue with a unique pore structure. J Biol Chem 1999;274:11751-11760.
    Pubmed
  17. Sano Y, Inamura K, Miyake A, Mochizuki S, Kitada C, Yokoi H, Nozawa K, Okada H, Matsushime H, Furuichi K. A novel two-pore domain K+ channel, TRESK, is localized in the spinal cord. J Biol Chem 2003;278:27406-27412.
    Pubmed
  18. Sabbadini M, Yost CS. Molecular biology of background K channels: insights from K(2P) knockout mice. J Mol Biol 2009;385:1331-1344.
    Pubmed
  19. Enyedi P, Czirják G. Molecular background of leak K+ currents: two-pore domain potassium channels. Physiol Rev 2010;90:559-605.
    Pubmed
  20. Franks NP, Honoré E. The TREK K2P channels and their role in general anaesthesia and neuroprotection. Trends Pharmacol Sci 2004;25:601-608.
    Pubmed
  21. Honoré E. The neuronal background K2P channels: focus on TREK1. Nat Rev Neurosci 2007;8:251-261.
    Pubmed
  22. Talley EM, Sirois JE, Lei Q, Bayliss DA. Two-pore-Domain (KCNK) potassium channels: dynamic roles in neuronal function. Neuroscientist 2003;9:46-56.
    Pubmed
  23. Lesage F, Guillemare E, Fink M, Duprat F, Lazdunski M, Romey G, Barhanin J. TWIK-1, a ubiquitous human weakly inward rectifying K+ channel with a novel structure. EMBO J 1996;15:1004-1011.
    Pubmed
  24. Feliciangeli S, Chatelain FC, Bichet D, Lesage F. The family of K2P channels: salient structural and functional properties. J Physiol 2015;593:2587-2603.
    Pubmed
  25. Lesage F, Lazdunski M. Molecular and functional properties of two-pore-domain potassium channels. Am J Physiol Renal Physiol 2000;279:F793-F801.
    Pubmed
  26. Fink M, Lesage F, Duprat F, Heurteaux C, Reyes R, Fosset M, Lazdunski M. A neuronal two P domain K+ channel stimulated by arachidonic acid and polyunsaturated fatty acids. EMBO J 1998;17:3297-3308.
    Pubmed
  27. Schewe M, Nematian-Ardestani E, Sun H, Musinszki M, Cordeiro S, Bucci G, de Groot BL, Tucker SJ, Rapedius M, Baukrowitz T. A non-canonical voltage-sensing mechanism controls gating in k2p k(+) channels. Cell 2016;164:937-949.
    Pubmed
  28. de la Cruz IP, Levin JZ, Cummins C, Anderson P, Horvitz HR. sup-9, sup-10, and unc-93 may encode components of a two-pore K+ channel that coordinates muscle contraction in Caenorhabditis elegans. J Neurosci 2003;23:9133-9145.
    Pubmed
  29. Goldstein SA, Price LA, Rosenthal DN, Pausch MH. ORK1, a potassium-selective leak channel with two pore domains cloned from Drosophila melanogaster by expression in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 1996;93:13256-13261.
    Pubmed
  30. Kunkel MT, Johnstone DB, Thomas JH, Salkoff L. Mutants of a temperature-sensitive two-P domain potassium channel. J Neurosci 2000;20:7517-7524.
    Pubmed
  31. Brohawn SG, del Mármol J, MacKinnon R. Crystal structure of the human K2P TRAAK, a lipid- and mechano-sensitive K+ ion channel. Science 2012;335:436-441.
    Pubmed
  32. Dong YY, Pike AC, Mackenzie A, McClenaghan C, Aryal P, Dong L, Quigley A, Grieben M, Goubin S, Mukhopadhyay S, Ruda GF, Clausen MV, Cao L, Brennan PE, Burgess-Brown NA, Sansom MS, Tucker SJ, Carpenter EP. K2P channel gating mechanisms revealed by structures of TREK-2 and a complex with Prozac. Science 2015;347:1256-1259.
    Pubmed
  33. Miller AN, Long SB. Crystal structure of the human two-pore domain potassium channel K2P1. Science 2012;335:432-436.
    Pubmed
  34. Berg AP, Talley EM, Manger JP, Bayliss DA. Motoneurons express heteromeric TWIK-related acid-sensitive K+ (TASK) channels containing TASK-1 (KCNK3) and TASK-3 (KCNK9) subunits. J Neurosci 2004;24:6693-6702.
    Pubmed
  35. Bittner S, Ruck T, Fernández-Orth J, Meuth SG. TREK-king the blood-brain-barrier. J Neuroimmune Pharmacol 2014;9:293-301.
    Pubmed
  36. Blin S, Chatelain FC, Feliciangeli S, Kang D, Lesage F, Bichet D. Tandem pore domain halothane-inhibited K+ channel subunits THIK1 and THIK2 assemble and form active channels. J Biol Chem 2014;289:28202-28212.
    Pubmed
  37. Czirják G, Enyedi P. Formation of functional heterodimers between the TASK-1 and TASK-3 two-pore domain potassium channel subunits. J Biol Chem 2002;277:5426-5432.
    Pubmed
  38. Hwang EM, Kim E, Yarishkin O, Woo DH, Han KS, Park N, Bae Y, Woo J, Kim D, Park M, Lee CJ, Park JY. A disulphide-linked heterodimer of TWIK-1 and TREK-1 mediates passive conductance in astrocytes. Nat Commun 2014;5:3227.
    Pubmed
  39. Levitz J, Royal P, Comoglio Y, Wdziekonski B, Schaub S, Clemens DM, Isacoff EY, Sandoz G. Heterodimerization within the TREK channel subfamily produces a diverse family of highly regulated potassium channels. Proc Natl Acad Sci U S A 2016;113:4194-4199.
    Pubmed
  40. Plant LD, Zuniga L, Araki D, Marks JD, Goldstein SA. SUMOylation silences heterodimeric TASK potassium channels containing K2P1 subunits in cerebellar granule neurons. Sci Signal 2012;5:ra84.
    Pubmed
  41. Lengyel M, Czirják G, Enyedi P. Formation of functional heterodimers by trek-1 and trek-2 two-pore domain potassium channel subunits. J Biol Chem 2016;291:13649-13661.
    Pubmed
  42. Maingret F, Fosset M, Lesage F, Lazdunski M, Honoré E. TRAAK is a mammalian neuronal mechano-gated K+ channel. J Biol Chem 1999;274:1381-1387.
    Pubmed
  43. Patel AJ, Honoré E, Maingret F, Lesage F, Fink M, Duprat F, Lazdunski M. A mammalian two pore domain mechano-gated S-like K+ channel. EMBO J 1998;17:4283-4290.
    Pubmed
  44. Kim Y, Gnatenco C, Bang H, Kim D. Localization of TREK-2 K+ channel domains that regulate channel kinetics and sensitivity to pressure, fatty acids and pHi. Pflugers Arch 2001;442:952-960.
    Pubmed
  45. Maingret F, Patel AJ, Lesage F, Lazdunski M, Honoré E. Mechano- or acid stimulation, two interactive modes of activation of the TREK-1 potassium channel. J Biol Chem 1999;274:26691-26696.
    Pubmed
  46. Sandoz G, Douguet D, Chatelain F, Lazdunski M, Lesage F. Extracellular acidification exerts opposite actions on TREK1 and TREK2 potassium channels via a single conserved histidine residue. Proc Natl Acad Sci U S A 2009;106:14628-14633.
    Pubmed
  47. Kang D, Kim D. Single-channel properties and pH sensitivity of two-pore domain K+ channels of the TALK family. Biochem Biophys Res Commun 2004;315:836-844.
    Pubmed
  48. Kim Y, Bang H, Gnatenco C, Kim D. Synergistic interaction and the role of C-terminus in the activation of TRAAK K+ channels by pressure, free fatty acids and alkali. Pflugers Arch 2001;442:64-72.
    Pubmed
  49. Maingret F, Lauritzen I, Patel AJ, Heurteaux C, Reyes R, Lesage F, Lazdunski M, Honoré E. TREK-1 is a heat-activated background K(+) channel. EMBO J 2000;19:2483-2491.
    Pubmed
  50. Kang D, Choe C, Kim D. Thermosensitivity of the two-pore domain K+ channels TREK-2 and TRAAK. J Physiol 2005;564:103-116.
    Pubmed
  51. Danthi S, Enyeart JA, Enyeart JJ. Modulation of native TREK-1 and Kv1.4 K+ channels by polyunsaturated fatty acids and lysophospholipids. J Membr Biol 2003;195:147-164.
    Pubmed
  52. Maingret F, Patel AJ, Lesage F, Lazdunski M, Honoré E. Lysophospholipids open the two-pore domain mechano-gated K(+) channels TREK-1 and TRAAK. J Biol Chem 2000;275:10128-10133.
    Pubmed
  53. Patel AJ, Honoré E, Lesage F, Fink M, Romey G, Lazdunski M. Inhalational anesthetics activate two-pore-domain background K+ channels. Nat Neurosci 1999;2:422-426.
    Pubmed
  54. Sirois JE, Lei Q, Talley EM, Lynch C, Bayliss DA. The TASK-1 two-pore domain K+ channel is a molecular substrate for neuronal effects of inhalation anesthetics. J Neurosci 2000;20:6347-6354.
    Pubmed
  55. Cain SM, Meadows HJ, Dunlop J, Bushell TJ. mGlu4 potentiation of K(2P)2.1 is dependant on C-terminal dephosphorylation. Mol Cell Neurosci 2008;37:32-39.
    Pubmed
  56. Chemin J, Girard C, Duprat F, Lesage F, Romey G, Lazdunski M. Mechanisms underlying excitatory effects of group I metabotropic glutamate receptors via inhibition of 2P domain K+ channels. EMBO J 2003;22:5403-5411.
    Pubmed
  57. Murbartián J, Lei Q, Sando JJ, Bayliss DA. Sequential phosphorylation mediates receptor- and kinase-induced inhibition of TREK-1 background potassium channels. J Biol Chem 2005;280:30175-30184.
    Pubmed
  58. Woo DH, Han KS, Shim JW, Yoon BE, Kim E, Bae JY, Oh SJ, Hwang EM, Marmorstein AD, Bae YC, Park JY, Lee CJ. TREK-1 and Best1 channels mediate fast and slow glutamate release in astrocytes upon GPCR activation. Cell 2012;151:25-40.
    Pubmed
  59. Kang D, Han J, Kim D. Mechanism of inhibition of TREK-2 (K2P10.1) by the Gq-coupled M3 muscarinic receptor. Am J Physiol Cell Physiol 2006;291:C649-C656.
    Pubmed
  60. Deng PY, Xiao Z, Yang C, Rojanathammanee L, Grisanti L, Watt J, Geiger JD, Liu R, Porter JE, Lei S. GABA(B) receptor activation inhibits neuronal excitability and spatial learning in the entorhinal cortex by activating TREK-2 K+ channels. Neuron 2009;63:230-243.
    Pubmed
  61. Xiao Z, Deng PY, Rojanathammanee L, Yang C, Grisanti L, Permpoonputtana K, Weinshenker D, Doze VA, Porter JE, Lei S. Noradrenergic depression of neuronal excitability in the entorhinal cortex via activation of TREK-2 K+ channels. J Biol Chem 2009;284:10980-10991.
    Pubmed
  62. Chen X, Talley EM, Patel N, Gomis A, McIntire WE, Dong B, Viana F, Garrison JC, Bayliss DA. Inhibition of a background potassium channel by Gq protein alpha-subunits. Proc Natl Acad Sci U S A 2006;103:3422-3427.
    Pubmed
  63. Millar JA, Barratt L, Southan AP, Page KM, Fyffe RE, Robertson B, Mathie A. A functional role for the two-pore domain potassium channel TASK-1 in cerebellar granule neurons. Proc Natl Acad Sci U S A 2000;97:3614-3618.
    Pubmed
  64. Talley EM, Lei Q, Sirois JE, Bayliss DA. TASK-1, a two-pore domain K+ channel, is modulated by multiple neurotransmitters in motoneurons. Neuron 2000;25:399-410.
    Pubmed
  65. O'Kelly I, Butler MH, Zilberberg N, Goldstein SA. Forward transport. 14-3-3 binding overcomes retention in endoplasmic reticulum by dibasic signals. Cell 2002;111:577-588.
    Pubmed
  66. Rajan S, Preisig-Müller R, Wischmeyer E, Nehring R, Hanley PJ, Renigunta V, Musset B, Schlichthörl G, Derst C, Karschin A, Daut J. Interaction with 14-3-3 proteins promotes functional expression of the potassium channels TASK-1 and TASK-3. J Physiol 2002;545:13-26.
    Pubmed
  67. Girard C, Tinel N, Terrenoire C, Romey G, Lazdunski M, Borsotto M. p11, an annexin II subunit, an auxiliary protein associated with the background K+ channel, TASK-1. EMBO J 2002;21:4439-4448.
    Pubmed
  68. Sandoz G, Tardy MP, Thümmler S, Feliciangeli S, Lazdunski M, Lesage F. Mtap2 is a constituent of the protein network that regulates twik-related K+ channel expression and trafficking. J Neurosci 2008;28:8545-8552.
    Pubmed
  69. Sandoz G, Thümmler S, Duprat F, Feliciangeli S, Vinh J, Escoubas P, Guy N, Lazdunski M, Lesage F. AKAP150, a switch to convert mechano-, pH- and arachidonic acid-sensitive TREK K(+) channels into open leak channels. EMBO J 2006;25:5864-5872.
    Pubmed
  70. Kim E, Hwang EM, Yarishkin O, Yoo JC, Kim D, Park N, Cho M, Lee YS, Sun CH, Yi GS, Yoo J, Kang D, Han J, Hong SG, Park JY. Enhancement of TREK1 channel surface expression by protein-protein interaction with beta-COP. Biochem Biophys Res Commun 2010;395:244-250.
    Pubmed
  71. Czirják G, Tóth ZE, Enyedi P. The two-pore domain K+ channel, TRESK, is activated by the cytoplasmic calcium signal through calcineurin. J Biol Chem 2004;279:18550-18558.
    Pubmed
  72. Czirják G, Vuity D, Enyedi P. Phosphorylation-dependent binding of 14-3-3 proteins controls TRESK regulation. J Biol Chem 2008;283:15672-15680.
    Pubmed
  73. Kim D, Gnatenco C. TASK-5, a new member of the tandem-pore K(+) channel family. Biochem Biophys Res Commun 2001;284:923-930.
    Pubmed
  74. Lesage F, Lauritzen I, Duprat F, Reyes R, Fink M, Heurteaux C, Lazdunski M. The structure, function and distribution of the mouse TWIK-1 K+ channel. FEBS Lett 1997;402:28-32.
    Pubmed
  75. Talley EM, Solorzano G, Lei Q, Kim D, Bayliss DA. Cns distribution of members of the two-pore-domain (KCNK) potassium channel family. J Neurosci 2001;21:7491-7505.
    Pubmed
  76. Hervieu GJ, Cluderay JE, Gray CW, Green PJ, Ranson JL, Randall AD, Meadows HJ. Distribution and expression of TREK-1, a two-pore-domain potassium channel, in the adult rat CNS. Neuroscience 2001;103:899-919.
    Pubmed
  77. Cadaveira-Mosquera A, Pérez M, Reboreda A, Rivas-Ramírez P, Fernández-Fernández D, Lamas JA. Expression of K2P channels in sensory and motor neurons of the autonomic nervous system. J Mol Neurosci 2012;48:86-96.
    Pubmed
  78. Medhurst AD, Rennie G, Chapman CG, Meadows H, Duckworth MD, Kelsell RE, Gloger II, Pangalos MN. Distribution analysis of human two pore domain potassium channels in tissues of the central nervous system and periphery. Brain Res Mol Brain Res 2001;86:101-114.
    Pubmed
  79. Kimelberg HK, Nedergaard M. Functions of astrocytes and their potential as therapeutic targets. Neurotherapeutics 2010;7:338-353.
    Pubmed
  80. Oberheim NA, Wang X, Goldman S, Nedergaard M. Astrocytic complexity distinguishes the human brain. Trends Neurosci 2006;29:547-553.
    Pubmed
  81. Kindler CH, Pietruck C, Yost CS, Sampson ER, Gray AT. Localization of the tandem pore domain K+ channel TASK-1 in the rat central nervous system. Brain Res Mol Brain Res 2000;80:99-108.
    Pubmed
  82. Kim JE, Kwak SE, Choi SY, Kang TC. Region-specific alterations in astroglial TWIK-related acid-sensitive K+-1 channel immunoreactivity in the rat hippocampal complex following pilocarpine-induced status epilepticus. J Comp Neurol 2008;510:463-474.
    Pubmed
  83. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson KS, Xing Y, Lubischer JL, Krieg PA, Krupenko SA, Thompson WJ, Barres BA. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 2008;28:264-278.
    Pubmed
  84. Seifert G, Hüttmann K, Binder DK, Hartmann C, Wyczynski A, Neusch C, Steinhäuser C. Analysis of astroglial K+ channel expression in the developing hippocampus reveals a predominant role of the Kir4.1 subunit. J Neurosci 2009;29:7474-7488.
    Pubmed
  85. Kafitz KW, Meier SD, Stephan J, Rose CR. Developmental profile and properties of sulforhodamine 101--labeled glial cells in acute brain slices of rat hippocampus. J Neurosci Methods 2008;169:84-92.
    Pubmed
  86. Kimelberg HK. Functions of mature mammalian astrocytes: a current view. Neuroscientist 2010;16:79-106.
    Pubmed
  87. Wang DD, Bordey A. The astrocyte odyssey. Prog Neurobiol 2008;86:342-367.
    Pubmed
  88. Zhou M, Schools GP, Kimelberg HK. Development of GLAST(+) astrocytes and NG2(+) glia in rat hippocampus CA1: mature astrocytes are electrophysiologically passive. J Neurophysiol 2006;95:134-143.
    Pubmed
  89. D'Ambrosio R, Wenzel J, Schwartzkroin PA, McKhann GM, Janigro D. Functional specialization and topographic segregation of hippocampal astrocytes. J Neurosci 1998;18:4425-4438.
    Pubmed
  90. Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir4.1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation. J Neurosci 2007;27:11354-11365.
    Pubmed
  91. Kang J, Jiang L, Goldman SA, Nedergaard M. Astrocyte-mediated potentiation of inhibitory synaptic transmission. Nat Neurosci 1998;1:683-692.
    Pubmed
  92. Zhou M, Xu G, Xie M, Zhang X, Schools GP, Ma L, Kimelberg HK, Chen H. TWIK-1 and TREK-1 are potassium channels contributing significantly to astrocyte passive conductance in rat hippocampal slices. J Neurosci 2009;29:8551-8564.
    Pubmed
  93. Gnatenco C, Han J, Snyder AK, Kim D. Functional expression of TREK-2 K+ channel in cultured rat brain astrocytes. Brain Res 2002;931:56-67.
    Pubmed
  94. Chu KC, Chiu CD, Hsu TT, Hsieh YM, Huang YY, Lien CC. Functional identification of an outwardly rectifying pH- and anesthetic-sensitive leak K(+) conductance in hippocampal astrocytes. Eur J Neurosci 2010;32:725-735.
    Pubmed
  95. Haydon PG, Carmignoto G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiol Rev 2006;86:1009-1031.
    Pubmed
  96. Hamilton NB, Attwell D. Do astrocytes really exocytose neurotransmitters?. Nat Rev Neurosci 2010;11:227-238.
    Pubmed
  97. Nedergaard M, Takano T, Hansen AJ. Beyond the role of glutamate as a neurotransmitter. Nat Rev Neurosci 2002;3:748-755.
    Pubmed
  98. Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhäuser C, Pilati E, Volterra A. Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate. Nat Neurosci 2004;7:613-620.
    Pubmed
  99. Jourdain P, Bergersen LH, Bhaukaurally K, Bezzi P, Santello M, Domercq M, Matute C, Tonello F, Gundersen V, Volterra A. Glutamate exocytosis from astrocytes controls synaptic strength. Nat Neurosci 2007;10:331-339.
    Pubmed
  100. Takano T, Kang J, Jaiswal JK, Simon SM, Lin JH, Yu Y, Li Y, Yang J, Dienel G, Zielke HR, Nedergaard M. Receptor-mediated glutamate release from volume sensitive channels in astrocytes. Proc Natl Acad Sci U S A 2005;102:16466-16471.
    Pubmed
  101. Cavelier P, Attwell D. Tonic release of glutamate by a DIDS-sensitive mechanism in rat hippocampal slices. J Physiol 2005;564:397-410.
    Pubmed
  102. Lipton P. Ischemic cell death in brain neurons. Physiol Rev 1999;79:1431-1568.
    Pubmed
  103. Anderson CM, Swanson RA. Astrocyte glutamate transport: review of properties, regulation, and physiological functions. Glia 2000;32:1-14.
    Pubmed
  104. Kofuji P, Newman EA. Potassium buffering in the central nervous system. Neuroscience 2004;129:1045-1056.
    Pubmed
  105. Heurteaux C, Guy N, Laigle C, Blondeau N, Duprat F, Mazzuca M, Lang-Lazdunski L, Widmann C, Zanzouri M, Romey G, Lazdunski M. TREK-1, a K+ channel involved in neuroprotection and general anesthesia. EMBO J 2004;23:2684-2695.
    Pubmed
  106. Liu Y, Sun Q, Chen X, Jing L, Wang W, Yu Z, Zhang G, Xie M. Linolenic acid provides multi-cellular protective effects after photothrombotic cerebral ischemia in rats. Neurochem Res 2014;39:1797-1808.
    Pubmed
  107. Wang M, Song J, Xiao W, Yang L, Yuan J, Wang W, Yu Z, Xie M. Changes in lipid-sensitive two-pore domain potassium channel TREK-1 expression and its involvement in astrogliosis following cerebral ischemia in rats. J Mol Neurosci 2012;46:384-392.
    Pubmed
  108. Wu X, Liu Y, Chen X, Sun Q, Tang R, Wang W, Yu Z, Xie M. Involvement of TREK-1 activity in astrocyte function and neuroprotection under simulated ischemia conditions. J Mol Neurosci 2013;49:499-506.
    Pubmed
  109. Kucheryavykh LY, Kucheryavykh YV, Inyushin M, Shuba YM, Sanabria P, Cubano LA, Skatchkov SN, Eaton MJ. Ischemia increases trek-2 channel expression in astrocytes: Relevance to glutamate clearance. Open Neurosci J 2009;3:40-47.
    Pubmed
  110. Rivera-Pagán AF, Rivera-Aponte DE, Melnik-Martínez KV, Zayas-Santiago A, Kucheryavykh LY, Martins AH, Cubano LA, Skatchkov SN, Eaton MJ. Up-regulation of TREK-2 potassium channels in cultured astrocytes requires de novo protein synthesis: relevance to localization of TREK-2 channels in astrocytes after transient cerebral ischemia. PLoS One 2015;10:e0125195.
    Pubmed
  111. Lee Y, Messing A, Su M, Brenner M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 2008;56:481-493.
    Pubmed
  112. Regan MR, Huang YH, Kim YS, Dykes-Hoberg MI, Jin L, Watkins AM, Bergles DE, Rothstein JD. Variations in promoter activity reveal a differential expression and physiology of glutamate transporters by glia in the developing and mature CNS. J Neurosci 2007;27:6607-6619.
    Pubmed
  113. Kawamoto Y, Akiguchi I, Tomimoto H, Shirakashi Y, Honjo Y, Budka H. Upregulated expression of 14-3-3 proteins in astrocytes from human cerebrovascular ischemic lesions. Stroke 2006;37:830-835.
    Pubmed
  114. Megias L, Guerri C, Fornas E, Azorin I, Bendala E, Sancho-Tello M, Durán JM, Tomás M, Gomez-Lechon MJ, Renau-Piqueras J. Endocytosis and transcytosis in growing astrocytes in primary culture. Possible implications in neural development. Int J Dev Biol 2000;44:209-221.
    Pubmed
  115. Satoh J, Yamamura T, Arima K. The 14-3-3 protein epsilon isoform expressed in reactive astrocytes in demyelinating lesions of multiple sclerosis binds to vimentin and glial fibrillary acidic protein in cultured human astrocytes. Am J Pathol 2004;165:577-592.
    Pubmed
  116. Bradley SJ, Challiss RA. G protein-coupled receptor signalling in astrocytes in health and disease: a focus on metabotropic glutamate receptors. Biochem Pharmacol 2012;84:249-259.
    Pubmed
  117. Porter JT, McCarthy KD. Astrocytic neurotransmitter receptors in situ and in vivo. Prog Neurobiol 1997;51:439-455.
    Pubmed
  118. Benesova J, Rusnakova V, Honsa P, Pivonkova H, Dzamba D, Kubista M, Anderova M. Distinct expression/function of potassium and chloride channels contributes to the diverse volume regulation in cortical astrocytes of GFAP/EGFP mice. PLoS One 2012;7:e29725.
    Pubmed
  119. Kim DS, Kim JE, Kwak SE, Choi HC, Song HK, Kimg YI, Choi SY, Kang TC. Up-regulated astroglial TWIK-related acid-sensitive K+ channel-1 (TASK-1) in the hippocampus of seizure-sensitive gerbils: a target of anti-epileptic drugs. Brain Res 2007;1185:346-358.
    Pubmed
  120. Kim JE, Kwak SE, Kang TC. Upregulated TWIK-related acid-sensitive K+ channel-2 in neurons and perivascular astrocytes in the hippocampus of experimental temporal lobe epilepsy. Epilepsia 2009;50:654-663.
    Pubmed
  121. Kim JE, Yeo SI, Ryu HJ, Chung CK, Kim MJ, Kang TC. Changes in TWIK-related acid sensitive K+-1 and -3 channel expressions from neurons to glia in the hippocampus of temporal lobe epilepsy patients and experimental animal model. Neurochem Res 2011;36:2155-2168.
    Pubmed
  122. Minieri L, Pivonkova H, Caprini M, Harantova L, Anderova M, Ferroni S. The inhibitor of volume-regulated anion channels DCPIB activates TREK potassium channels in cultured astrocytes. Br J Pharmacol 2013;168:1240-1254.
    Pubmed