Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Glia. 2019 Aug 16;68(3):472–494. doi: 10.1002/glia.23699

Roles of glial ion transporters in brain diseases

Shanshan Song 1,2, Lanxin Luo 1,2,3,4, Baoshan Sun 4,5, Dandan Sun 1,2,6
PMCID: PMC6957693  NIHMSID: NIHMS1064821  PMID: 31418931

Abstract

Glial ion transporters are important in regulation of ionic homeostasis, cell volume, and cellular signal transduction under physiological conditions of the central nervous system (CNS). In response to acute or chronic brain injuries, these ion transporters can be activated and differentially regulate glial functions, which has subsequent impact on brain injury or tissue repair and functional recovery. In this review, we summarized the current knowledge about major glial ion transporters, including Na+/H+ exchangers (NHE), Na+/Ca2+ exchangers (NCX), Na+–K+–Cl cotransporters (NKCC), and Na+–HCO3 cotransporters (NBC). In acute neurological diseases, such as ischemic stroke and traumatic brain injury (TBI), these ion transporters are rapidly activated and play significant roles in regulation of the intra- and extracellular pH, Na+, K+, and Ca2+ homeostasis, synaptic plasticity, and myelin formation. However, overstimulation of these ion transporters can contribute to glial apoptosis, demyelination, inflammation, and excitotoxicity. In chronic brain diseases, such as glioma, Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS), glial ion transporters are involved in the glioma Warburg effect, glial activation, neuroinflammation, and neuronal damages. These findings suggest that glial ion transporters are involved in tissue structural and functional restoration, or brain injury and neurological disease development and progression. A better understanding of these ion transporters in acute and chronic neurological diseases will provide insights for their potential as therapeutic targets.

Keywords: brain diseases, Na+/Ca2+ exchanger, Na+/H+ exchanger, Na+–HCO3 cotransporter, Na+–K+–Cl cotransporter

1. |. GLIAL ION TRANSPORT IN ACUTE NEUROLOGICAL DISEASES

1.1 |. Glial cells and glial ion transporters

Increasing evidence suggests that glial cells, including astrocytes, microglia, and oligodendrocytes, are not only neuron supporters, but also master regulators that control various aspects of brain functions (Zuchero & Barres, 2015). Astrocytes regulate normal brain functions through blood flow and vasculature support (Mishra, Pal, Gupta, & Carmel, 2017), blood-brain barrier (BBB) formation and function (Erdo, Denes, & de Lange, 2017), neurotrophic factor secretion (Jha et al., 2018), and formation of the tripartite synapses and neural circuit remodeling (Chai et al., 2017; Kim, Nabekura, & Koizumi, 2017). Microglia are the resident phagocytes in the central nervous system (CNS) and function as the first line of defense against injury (Ma, Wang, Wang, & Yang, 2017). Under physiological conditions, microglial cells extend their branching processes in a ramified morphology and constantly scan their surrounding environment for surveillance (Wolf, Boddeke, & Kettenmann, 2017). The microglial processes can also make direct contact with neuronal synapses (Wake, Moorhouse, Jinno, Kohsaka, & Nabekura, 2009) and plays an important role in synapse elimination and pruning, synapse formation, and neuronal network synchronization in both developing and adult brains (Akiyoshi et al., 2018; Miyamoto et al., 2016; Reshef et al., 2017). Oligodendrocyte precursor cells (OPCs) migrate within the brain parenchyma and differentiate into mature oligodendrocytes, which can extend their cell membrane protrusions to form myelinating sheaths around the neuronal axons for fast saltatory nerve conduction between nodes of Ranvier (Domingues et al., 2018). Myelination by oligodendrocytes is especially enriched in the white matter, and is regulated by neuronal activity (Gibson et al., 2014). Maintenance of oligodendrocytes and myelination homeostasis is vital for normal neurological functions, such as cognitive, perceptual, and sensorimotor functions (Wang, Liu, et al., 2016). Collectively, glial cells are required for physiological brain functions by providing structural and material support, as well as interacting with axons and synapses for signal transduction and neuronal network regulation.

Increasingly accumulated research shows that several major glial ion transporters, including Na+/H+ exchangers (NHE), Na+/Ca2+ exchangers (NCX), Na+–K+–Cl cotransporters (NKCC), and Na+–HCO3 cotransporters (NBC), play important roles in regulation of glial functions. In acute neurological diseases, such as ischemic stroke and traumatic brain injury (TBI), these ion transporters are rapidly activated to regulate the intra- and extracellular pH, Na+, K+, and Ca2+ homeostasis, synaptic plasticity, and myelin formation. However, pathological overstimulation of these ion transporters can contribute to glial apoptosis, demyelination, inflammation, and excitotoxicity (Figure 1). In this section, we will summarize the roles of these glial ion transporters in the functional changes of astrocytes, microglia, and oligodendrocytes. These findings reveal the potential of targeting these ion transporter proteins for therapeutic treatment for acute neurological diseases.

FIGURE 1.

FIGURE 1

Roles of glial ion transporters in ischemic brain damage. Ischemic stroke results in an immediate marked reductions of intracellular pH (pHi) as a consequence of oxygen depletion, which necessitates a switch from aerobic metabolism to anaerobic glycolysis, leading to generation of intracellular lactic acid as well as protons. Several ion transporters/exchangers in astrocytes, microglia, and/or oligodendrocytes are activated to prevent the pHi dysregulation. In astrocytes, Na+–HCO3 cotransporter (NBC) transports HCO3 to alkalinize pHi. Stimulation of Na+/H+ exchanger isoform-1 (NHE1) extrudes H+ in exchange of Na+ influx, which leads to astrocytic swelling and disruption of astrocytic end-feet function at the blood-brain barrier (BBB), and contributes to inflammation by secretion of various cytokines. The increased Na+ overload triggers the reversal operation of Na+/Ca2+ exchanger (NCXrev) and causes intracellular overload of Ca2+, and reduces glutamate uptake via the Na+-dependent excitatory amino acid transporter (EAAT). Astrocytic Na+–K+–Cl cotransporter (NKCC1) activation also contributes to astrocyte swelling and cascade events as described above. In microglia, NHE1 maintains the optimal pHi for NADPH oxidase (NOX2) activation, together with NCXrev activation, promoting the release of reactive oxygen species (ROS) and contributes to oxidative injury. In oligodendrocytes, NKCC1 is activated via the upstream WNK-SPAK/OSR1 pathway, and triggers NCXrev function and enhances excitotoxicity and apoptosis. Taken together, NHE1, NCX, NKCC1, and NBC play important roles in regulation of astrocytic, microglial, and oligodendrocytic functions in response to ischemic stroke

1.1.1 |. Ion transporters in reactive astrocytes after ischemic stroke

Ischemic stroke results from occlusions in major cerebral arteries, most commonly the middle cerebral artery, and leads to irreversible neuronal as well as glial cell death in the ischemic core due to lack of of glucose and oxygen (Puig, Brenna, & Magnus, 2018). Astrocytes, microglia, and oligodendrocytes in the brain tissue surrounding the ischemic core (the penumbra) alter their functions in response to the ischemic insult for restricting injury and restoring brain functions or accelerating cell death and ischemic infarct (Liu & Chopp, 2016; Ma et al., 2017; Shindo et al., 2016).

Within minutes after ischemic stroke, astrocytes in the penumbra are rapidly activated by cytokines produced by injured neurons in the ischemic lesion (Liu & Chopp, 2016; Pekny et al., 2016). Within a few days after the ischemic insult, these reactive astrocytes exhibit stimulated proliferation and elongated processes to form a glial scar, a physical wall surrounding the ischemic lesion, which functions in isolating the ischemic core and preventing the diffusion of detrimental factors into the nonlesioned remote area (Liu & Chopp, 2016; Pekny et al., 2016). The reactive astrocytes can be beneficial by secreting restorative growth factors such as brain derived neurotrophic factor (BDNF; Miyamoto et al., 2015), transforming growth factor (TGF)-β (Cekanaviciute et al., 2014), and ciliary neurotrophic factor (CNTF) for poststroke tissue repair (Kang, Keasey, Cai, & Hagg, 2012). However, the astrocytic glial scar can also secrete chondroitin sulfate proteoglycans (CSPG) that inhibits axonal regeneration and hinders the functional recovery during the late phase after ischemic stroke (Liu & Chopp, 2016). A transcriptome analysis of the reactive astrocytes isolated from the ischemic brains showed that the signal transducer and activator of transcription-3 (STAT3) pathway was activated and contributed to the increased secretion of detrimental molecules, such as galectin-3, lipocalin-2, and osteopontin (Rakers et al., 2019). Selective deletion of the Stat3 gene in astrocytes reduced ischemic infarction and improved motor functions in a mouse model of ischemic stroke (transient middle cerebral artery occlusion, tMCAO; Rakers et al., 2019).

These studies illustrate that reactive astrocytes have both detrimental and restorative functions, and transformation of astrocytes toward the protective phenotype is important for the resolution of ischemic brain injury. Multiple ion transporters have been reported to mediate the switch between beneficial or detrimental phenotypes of astrocytes.

Astrocytic NHE1 in pHi, homeostasis, BBB breakdown, ER stress, and inflammation

Among nine members of the NHE family (encoded by SLC9A1-SLC9A9 genes), NHE1 protein is the most abundantly expressed isoform in the CNS (Orlowski & Grinstein, 2004). NHE1 is ubiquitously expressed in all cell types in the brain and regulates pHi and cell volume homeostasis by mediating H+ extrusion in exchange of Na+ influx at a 1:1 ratio (Shi, Kim, Caldwell, & Sun, 2013).

The steady-state pHi in mouse cortical astrocytes are reported at 7.0–7.2 in CO2/HCO3 buffered saline when both NBC and NHE1 are functioning (Chesler, 2005; Theparambil & Deitmer, 2015). In the absence of HCO3, NHE1 functions as the major regulator for astrocytic pHi and maintains their resting pHi at about 6.86 (Kintner et al., 2004). Knockout of Nhe1 gene acidified the astrocytic pHi to 6.53 (Kintner et al., 2004). Under hypoxic conditions, in part mediated by the activation of extracellular signal regulated kinase (ERK) signaling pathway (Kintner, Look, Shull, & Sun, 2005), NHE1 in astrocytes was rapidly activated for extrusion of the excess cytosolic H+ and pHi recovery, which was abolished by either genetic knockout or pharmacological inhibition of NHE1 in astrocytes (Kintner et al., 2004; Wang, Li, et al., 2016).

On the other hand, NHE1-mediated Na+ influx led to astrocytic swelling and was associated with reduced astrocyte viability (Wang, Li, et al., 2016). Both genetic knockout and pharmacological inhibition of astrocytic NHE1 significantly reduced Na+i overload (Kintner et al., 2004), decreased astrocytic aquaporin (AQP)-4 expression, and increased resistance of astrocytes to swelling in response to oxygen glucose deprivation (OGD; Kintner et al., 2004; Rutkowsky, Wallace, Wise, & O’Donnell, 2011). Selective deletion of Nhe1 gene in astrocytes also reduced brain edema, along with alleviated BBB breakdown, swelling of endothelial cells, and structural interruption of tight junctions, altogether leading to an increased regional cerebral blood flow in mice after tMCAO (Begum et al., 2018). These astrocytic Nhe1-null mice also exhibited reduced astrogliosis and infarct volume, as well as improved neurological functions after tMCAO (Begum et al., 2018). In addition, the elevated Na+i led to activation of the reverse mode of the NCX (NCXrev) in astrocytes, which subsequently caused the overload of Ca2+ stores in the endoplasmic reticulum (ER) and the mitochondria, contributing to ER stress, mitochondrial dysfunction, and astrocytic apoptosis after OGD (Bondarenko, Svichar, & Chesler, 2005; Cengiz et al., 2014; Kintner et al., 2005). The NHE1-mediated Na+i increase also triggered reversal of the Na+-dependent excitatory amino acid transporter (EAAT), which mediated glutamate release from astrocytes, leading to excitotoxicity after OGD (Cengiz et al., 2014). Moreover, astrocytic NHE1 is involved in proinflammatory cytokine release after ischemia, such as interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and matrix metallopeptidase (MMP)-9 (Begum et al., 2018; Cengiz et al., 2014). Pharmacological inhibition or genetic knockout of astrocytic NHE1 protein significantly reduced these cytokine levels after ischemia (Begum et al., 2018; Cengiz et al., 2014).

Therefore, upon ischemic stroke, the overstimulated astrocytic NHE1 activity plays an important role in maintaining the astrocytic pHi homeostasis, but also mediates astrocytic end-feet swelling, BBB breakdown, and inflammation. Inhibition of astrocytic NHE1 activity reduces brain edema and inflammation for ischemic brain tissue repair.

Astrocytic NCX in synaptic plasticity, excitotoxicity, ER stress, and astrocyte apoptosis

NCX mediates three Na+ ion influx and one Ca2+ extrusion across the cell membrane under physiological conditions, whose stoichiometric and electrogenic characteristics regulate Na+- and Ca2+-dependent signaling, as well as membrane potentials in the brain (Khananshvili, 2013). Depending on the intracellular and extracellular Na+ and Ca2+ levels, NCX can operate either in forward mode, extruding Ca2+ in exchange of Na+ influx, or in reverse mode, coupling Na+ efflux to the Ca2+ influx (Khananshvili, 2013).

Astrocytes express all three isoforms of NCX (NCX1-3; Parpura, Sekler, & Fern, 2016; Rose & Verkhratsky, 2016), with NCX1 transcript being the most predominant (Sakaue et al., 2000). Glutamatergic neuronal activity caused drastic elevations of Na+i through the astrocytic NCX in neocortical but not hippocampal astrocytes (Ziemens, Oschmann, Gerkau, & Rose, 2019), which promoted astrocytic metabolism (Chatton, Magistretti, & Barros, 2016), and drove the Ca2+-mediated signaling (Ziemens et al., 2019). Astrocytic NCX in reverse mode (NCXrev) is the major contributor for maintaining Na+ and Ca2+ homeostasis in astrocytes both at resting states and with injury stimulus (Reyes, Verkhratsky, & Parpura, 2012). Specific blockade of NCXrev activity with KB-R7943 showed a significant decrease in the astrocytic cytosol Ca2+ concentration, while blockade of NCX with benzamil did not show such effect (Reyes et al., 2012).

Astrocytic NCX activation could play multiple roles in healthy and ischemic brains. On one hand, the astrocytic NCX is located on the plasma membrane directly opposing the ER (Blaustein, Juhaszova, Golovina, Church, & Stanley, 2002), indicating that NCX-mediated Ca2+ flux closely relates to ER functions in astrocytes. In fact, in cultured astrocytes, NCXrev activity can be induced by increased local Na+i concentration, either mediated by Na+ pump inhibition or elevated NKCC1 activity, and led to cytosolic Ca2+i elevation and subsequent augmentation of the adjacent ER Ca2+ stores, which contributed to ER stress in ischemic conditions (Blaustein et al., 2002; Golovina, Song, James, Lingrel, & Blaustein, 2003; Kintner et al., 2007; Lenart, Kintner, Shull, & Sun, 2004). On the other hand, the NCXrev-mediated Ca2+ influx can also be stored at the proximal processes of astrocytes called “branchlets” (Semyanov, 2019), where the release of the Ca2+ store is triggered by synaptic activity near the peri-synaptic distal end processes of astrocytes (“leaflets”) through inositol-3-phosphate (IP3)-dependent mechanisms, and in turn spreads the Ca2+-mediated events and regulated neuronal network and synaptic plasticity (Minelli et al., 2007; Semyanov, 2019). In addition, the mitochondrial subtype of NCX in astrocytes, primarily the Na+, Li+/Ca2+ exchanger (NCLX), which exchanges Na+ or Li+ for Ca2+ (Sharma, Roy, Sekler, & O’Halloran, 2017), can mediate Ca2+ release in post-tetanic potentiation, a form of short-term synaptic plasticity (Castaldo et al., 2009; Gobbi et al., 2007). We speculate that changes of NCX activity in different brain regions/astrocytic cellular locations could be involved in learning, memory, and information processing functions in the brain. These regulations are also Na+-dependent, and thus are possible to occur in the ischemic brains with prolonged Na+i elevation (Brazhe, Verisokin, Verveyko, & Postnov, 2018; Gerkau, Rakers, Durry, Petzold, & Rose, 2018; Verkhratsky, Trebak, Perocchi, Khananshvili, & Sekler, 2018).

The involvement of astrocytic NCX in synaptic transmission and plasticity is also through glutamate release/uptake from the astrocytes, as blockade of NCXrev by KB-R7943 in cultured astrocytes significantly reduced the glutamate release induced by mild polarization (Reyes et al., 2012). The NCX-mediated Ca2+ mobilization (increase) led to the internalization of the glutamate transporter GLT-1 in astrocytes, which reduced glutamate reuptake upon ischemic stroke injury (Ibanez, Bartolome-Martin, Piniella, Gimenez, & Zafra, 2019).

In addition, the NCXrev-mediated Ca2+ influx is responsible for the nitric oxide (NO)-induced cytotoxicity in cultured astrocytes via the ERK, c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) pathways (Kitao et al., 2010; Takuma, Ago, & Matsuda, 2013). Pharmacological inhibition of NCXrev attenuated the Ca2+-induced production of reactive oxygen species (ROS), DNA ladder formation, and nuclear condensation, increasing cell viability and reducing apoptosis in the cultured astrocytes (Matsuda et al., 2001). NCX activity contributed to ischemic brain damage, as pharmacological inhibition of NCX significantly reduced infarct volume and neuronal damage in tMCAO models (Matsuda et al., 2001; Shenoda, 2015).

Taken together, considering the multiple functions of astrocytic NCX in regulation of synapse and intracellular Ca2+ signaling, additional studies are needed to investigate their impacts on brain functions under both healthy and pathological conditions.

Astrocytic NKCC1 in cell volume regulation, excitotoxicity, and ER stress

Na+–K+–2Cl cotransporters (NKCCs) belong to the SLC12 superfamily of cation-chloride cotransporters (CCCs) and exist in two isoforms, NKCC1 and NKCC2 (Ben-Ari, 2017). NKCC1 is the predominant isoform in the brain, while NKCC2 is mostly expressed in the thick ascending limb of the loop of Henle in the kidney (Zeuthen & MacAulay, 2012). NKCC1 couples Na+, K+, and Cl import at a 1:1:2 ratio (Ben-Ari, 2017), and regulates these ion concentrations along with cell volume homeostasis by cotransporting 600 molecules of water influx per turnover (Zeuthen & MacAulay, 2012). NKCC1 counteracts the K+–Cl cotransporter (KCC), which is another member of the CCCs that mediates K+ and Cl export along with water efflux (Zhang et al., 2018). Upon ischemic stroke, both NKCC1 and KCCs are phosphorylated via the upstream with-no-lysine (K) kinase (WNK)-Ste20/SPS1-related proline-alanine-rich protein kinase (SPAK)/oxidative stress responsive kinase (OSR1) signaling pathway, leading to NKCC1 activation and KCC inhibition (Baudel, Poole, & Darlison, 2017; Zhang et al., 2018). The WNK-calcium binding protein (Cab39; Bhuiyan et al., 2017) or the MAPK (p38, ERK, JNK) pathways (Yu et al., 2018) have also been reported to be associated with NKCC1 activation after ischemia. The human subacute ischemic stroke brain tissues display increased expression of NKCC1 protein (Bhuiyan et al., 2017). NKCC1 protein activation contributed to ischemic brain damages, because genetic deletion of NKCC1 or its upstream regulator WNK3 exhibited reduced infarction, edema, and white matter damage in mouse tMCAO models (Begum et al., 2015; Chen, Luo, Kintner, Shull, & Sun, 2005). Pharmacological inhibition of NKCC1 with its potent inhibitor bumetanide or a novel inhibitor STS66 also reduced ischemic infarction and white matter disruption, and improved neurological functions in rodent models of ischemic stroke (Huang et al., 2019; Xu et al., 2017; Yu et al., 2018), especially comorbid with hypertension (Bhuiyan et al., 2017).

Astrocytic expression of NKCC1 was detected in cortex, corpus callosum, hippocampus, cerebellum, and optic nerves in rats (MacVicar, Feighan, Brown, & Ransom, 2002; Yan, Dempsey, & Sun, 2001). NKCC1 in astrocytes maintains Na+ homeostasis along with Na+/K+-ATPase and NBC (Rose & Karus, 2013; Rose & Verkhratsky, 2016). Astrocytic NKCC1 activity is the main cell volume regulatory mechanism to revert the osmotic stress-mediated astrocytic shrinkage during regulatory volume increase (RVI; Deng et al., 2016; Zhang et al., 2018). During ischemia, the astrocytic NKCC1 activation is dependent on the hypertonicity induced by Na+/K+-ATPase activity (Hertz et al., 2013; Hertz & Chen, 2016) via the β1-adrenergic (Hertz, Song, Xu, Peng, & Gibbs, 2015; Song et al., 2014; Song, Xu, Hertz, & Peng, 2015) or ERK1/2 MAPK signaling pathways (Cai et al., 2011; Yuen et al., 2014). Overstimulation of astrocytic NKCC1 activity contributed to brain edema formation under hypotonic osmotic stress or after ischemic stroke (Deng et al., 2016; Zhang et al., 2018). Both pharmacological inhibition and genetic deficiency of NKCC1 reduced astrocytic swelling, cerebral edema, and BBB breakdown during the reperfusion phase after ischemic stroke (Chen & Sun, 2005; Hertz et al., 2014; Kahle et al., 2009). Moreover, NKCC1-mediated elevation in Na+i can subsequently trigger NCXrev activity in astrocytes, which led to ER Ca2+ overload and mitochondrial dysfunction, contributing to the ischemic brain damage (Kintner et al., 2007; Lenart et al., 2004). Pharmacological inhibition or genetic knockout of NKCC1 abolished the increase in Na+i (Chen et al., 2005; Lenart et al., 2004), as well as the Ca2+i accumulation in astrocytes (Kintner et al., 2007) in OGD models.

The role of astrocytic NKCC1 activity in K+ homeostasis remains incompletely defined. First, it has been reported that knockout of NKCC1 in astrocytes completely abolished the K+ influx under hyperosmotic solution in response to high extracellular K+ stress (Chen & Sun, 2005; Su, Kintner, Flagella, Shull, & Sun, 2002; Su, Kintner, & Sun, 2002). The K+ uptake via NKCC1 requires glycogenolysis (Hertz et al., 2013; Xu et al., 2013). The astrocytic glycogenolysis mediates glutamate synthesis from astrocytes (Hertz et al., 2013), while the astrocytic NKCC1 is an important contributor for glutamate release, as genetic knockout of NKCC1 in astrocytes reduced 30% of glutamate release under hyperosmotic solution with high extracellular K+ (Chen & Sun, 2005; Su, Kintner, Flagella, et al., 2002; Su, Kintner, & Sun, 2002), collectively contributing to learning and memory functions (Hertz et al., 2013; Hertz & Chen, 2016; Xu et al., 2013). The astrocytic NKCC1-mediated glutamate release activated non-NMDA glutamate receptors on oligodendrocytes in the white matter, which resulted in oligodendrocyte death in a rat optic nerve model (Wilke, Thomas, Allcock, & Fern, 2004). On the other hand, however, recent studies showed that inhibition of NKCC1 failed to affect the rate of K+ removal from the extracellular space in rat hippocampal slices under normoxic conditions (Larsen et al., 2014). Inhibition of either Na+/K+-ATPase or Kir4.1 reduced the clearance of K+o transients in the hippocampal slices in response to increased extracellular K+, indicating that Na+/K+-ATPase and Kir4.1, but not NKCC1, played vital roles in extracellular K+ clearance (Larsen et al., 2014). The contribution of each astrocytic K+ regulatory mechanisms (NKCC1, Na+/K+-ATPase, and Kir4.1) in brain extracellular K+ clearance under ischemic pathological conditions warrants further study.

In addition to Na+, K+, and glutamate, NKCC1 also maintains Cl homeostasis, as the Cl influx was completely abolished in the NKCC1-null astrocytes (Chen & Sun, 2005; Su, Kintner, Flagella, et al., 2002; Su, Kintner, & Sun, 2002). The activation of glutamate N-methyl-d-aspartic acid (NMDA) receptors is dependent on the NKCC1-mediated Cl accumulation (Beck, Lenart, Kintner, & Sun, 2003). γ-aminobutyric acid (GABA) exerts its action via synaptic and extra-synaptic GABAA receptors, mediating phasic and tonic inhibition, respectively (Cellot & Cherubini, 2013). The Cl influx regulated by NKCC1 suppressed the inhibitory GABAergic responses and resulted in neuron depolarization, which mediated postsynaptic firing and contributed to excitotoxicity following ischemic stroke (Baudel et al., 2017; Ben-Ari, 2017; Schulte, Wierenga, & Bruining, 2018). Inhibition of NKCC1 using bumetanide promoted the inhibitory GABAergic transmission and eliminated seizures in a genetically induced mouse astrogliosis model (Robel et al., 2015). The astrocytic regulation of NKCC1 on GABAergic synaptic transmission was mediated by phospho-activation of the WNK-SPAK/OSR1 signaling pathways (Wilson & Mongin, 2019; Zhang et al., 2018).

In summary, astrocytic NKCC1 activity is the main cell volume regulatory mechanism mediated by conducting Na+, K+, and Cl influx. Overstimulation of NKCC1 activity affects astrocyte metabolism and GABAergic transmission regulation, which present NKCC1 as a therapeutic target for ischemic stroke, as well as epilepsy.

Astrocytic NBC in pHi, homeostasis, cell volume regulation, membrane potential, and energy metabolism

NBC mediates two or three HCO3 cotransport per Na+, thus eletrogenic, and maintains the astrocytic membrane potential at −70 to −80 mV (Rose & Verkhratsky, 2016). Among the three isoforms of NBC (NBC1-3), NBC1 is the most widely expressed in astrocytes throughout the CNS, including cerebral cortex, hippocampus, and cerebellum (Annunziato, Boscia, & Pignataro, 2013; Schmitt et al., 2000), and can operate in either inward or outward modes for regulations of Na+ signaling, pHi homeostasis, as well as glial membrane potential in the CNS (Boscia et al., 2016; Rose & Verkhratsky, 2016). Rose and Verkhratsky (2016) reported that the inward mode of NBC activation mediated influx of Na+ into astrocytes and contributed to setting the baseline [Na+]i under physiological conditions (Figure 1).

In addition, because NBC is electrogenic, it is activated by astrocytic membrane depolarization induced by neuronal activity and the subsequent K+ influx, which resulted in intracellular accumulation of K+, Na+, and HCO3, creating an osmotic gradient which drives water influx, the neuronal activity-dependent astrocytic swelling, and extracellular space shrinkage without affecting the K+ transients (Florence, Baillie, & Mulligan, 2012; Larsen & MacAulay, 2017; Ostby et al., 2009). The depolarization-induced alkalization of astrocytes also resulted in a stimulation of glycolysis through NBC activation, as the glycolytic response to increased K+ was absent when NBC was blocked by pharmacological inhibition or genetic knockout of NBC (Ruminot et al., 2011). NBC activation stimulated soluble adenylyl cyclase (sAC) and intracellular cyclic adenosine monophosphate (cAMP) levels, which enhanced glycolysis and the release of lactate into the extracellular space (Choi et al., 2012), whereas lactate can be transported through the monocarboxylate transporters and in turn stimulate NBC activity (Becker, Broer, & Deitmer, 2004; Choi et al., 2012), forming a positive feedback loop for metabolic communication between astrocytes and neurons (Becker et al., 2004; Choi et al., 2012). The NBC-mediated alkalization in astrocytes was also highly correlated with astrocytic membrane potential changes during seizure-like events in hippocampal astrocytes, confirming the role of the electrogenic NBC in tissue excitability through regulation of astrocytic membrane potential (Raimondo et al., 2016). This view has been further supported by a finding showing that elevated NBC immunoreactivity was detected in gerbil hippocampus on seizure onset, but not in the seizure-resistant gerbil hippocampus (Kang et al., 2002).

Oppositely, hyperpolarization induced by prompt reduction of the extracellular K+ concentration concurrently led to intracellular acidosis, which was dependent on extracellular Ca2+, as removal of Ca2+o led to an instant pHi recovery (increase) in mouse neural stem cell-derived astrocytes (Nordstrom, Andersson, & Akerman, 2019). This pHi recovery was mediated by the inward mode of NBC activation, since pharmacological inhibition of NBC prevented such pHi increase in the hyperpolarized astrocytes upon Ca2+o removal (Nordstrom et al., 2019). In addition, with the presence of high extracellular K+, the NBC-mediated pHi increase can override the Ca2+-mediated acidosis even in prehyperpolarized and acidified astrocytes (Nordstrom et al., 2019), further indicating the importance of NBC in astrocytic pHi regulation. The NBC-mediated regulation on pHi is essential for astrocytic development (Giffard et al., 2000), and has been confirmed in NBC knockout astrocytes (Theparambil, Ruminot, Schneider, Shull, & Deitmer, 2014). Upon ischemic stroke, NBC protein expression was upregulated in the ischemic penumbra in a rat pMCAO model (Jung, Choi, & Kwon, 2007), and in astrocytes in the CA1 region, but not in CA3 region of the hippocampus (Sohn et al., 2011), which played a vital role in astrocytic survival by buffering the ischemic stroke-induced intracellular acidosis (Yao et al., 2016). Silencing NBC expression rendered astrocytes more vulnerable to acidic injury (Giffard et al., 2000), and pharmacological inhibition of NBC by a generic NBC blocker S0859 exacerbated the ischemia-induced cell death in human stem cell-derived astrocytes (Yao et al., 2016).

In conclusion, NBC is important for maintaining pHi homeostasis, cell volume, membrane potential, and energy metabolism in astrocytes. While the upregulated NBC activity plays a positive role in astrocytic survival by alleviating the intracellular acidosis after ischemic stroke, increased NBC activity also exerts an effect on membrane potential, contributing to tissue excitability and seizure-like events.

1.1.2 |. Ion transporters in activated microglia after ischemic stroke

Upon brain injury, microglia are rapidly activated, migrate toward the lesion site and trigger proinflammatory responses by secreting cytokines, such as IL-1β, IL-6, and TNF-α (Bar & Barak, 2019; Liu et al., 2010). Alternatively, activated microglia transform their activity and contribute to tissue repair and remodeling by secretion of anti-inflammatory cytokines and growth factors, as well as phagocytic activity (Bar & Barak, 2019; Parkhurst et al., 2013; Song et al., 2018). Increased phagocytic activity of microglia is essential for clearance of tissue debris after brain injury, favoring oligodendrogenesis and remyelination (Bar & Barak, 2019; Olah et al., 2012; Rinaldi et al., 2016). Microglia also recognize and eliminate injured synapses in a phagocytosis-dependent manner, which subsequently promote synaptic remodeling and plasticity in ischemic brains (Bar & Barak, 2019; Wake et al., 2009). Increasing evidence suggests that microglial ion transporters play important roles in regulation of microglial function after ischemic stroke, which is further discussed in the following sections.

Microglial NHE1 in pHi, homeostasis and proinflammatory microglial activation

NHE1 maintains the basal pHi of microglia at about 7.19 in resting conditions, and pharmacological inhibition of NHE1 with its potent inhibitor HOE642 acidified the basal pHi to 6.82 (Liu et al., 2010). In response to lipopolysaccharides (LPS) or hypoxia stimulation, NHE1 protein is activated for excess H+ extrusion and maintains an alkaline optimal pHi at 7.2 during the microglial respiratory burst and superoxide production by NADPH oxidase (NOX; Lam et al., 2013;Liu et al., 2010 ; Shi, Kim, et al., 2013). This pHi homeostasis is important for microglial chemotaxis and migration (Shi et al., 2013). Pharmacological inhibition of NHE1 during this process failed to buffer the pHi changes, and led to sustained intracellular acidification and cessation of the NOX activity, and abolished microglial proinflammatory polarization (Liu et al., 2010; Shi, Chanana, Watters, Ferrazzano, & Sun, 2011). Selective deletion of the Nhe1 gene in microglia also prevented microglial proinflammatory activation as well as their proliferation in the ischemic brains (Song et al., 2018).

On the other hand, it is reported that increased expression of Na+ channels such as the voltage-gated Na+ channel (Nav; Pappalardo, Black, & Waxman, 2016) and the acid-sensing ion channels (ASICs; Yu et al., 2015), as well as the Na+ transporters/exchangers such as NHE1 (Liu et al., 2010) collectively contributed to the increased Na+ loading in microglia under pathological ischemic or inflammatory conditions. Activation of NHE1 caused cell swelling through increased Na+ influx, while inhibition of NHE1 caused microglial shrinkage (Nishioka et al., 2016). Importantly, increased Na+ loading activates NCXrev (Liu et al., 2010; Shi, Yuan, et al., 2013) and led to a two-fold increase of the Ca2+ level in microglial cells (Liu et al., 2010), which could subsequently trigger the Ca2+-dependent signaling pathways, such as the Ca2+-calcium/calmodulin-dependent protein kinase (CaMKII)/transforming growth factor β-activated kinase (TAK1)-nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) signaling, and contributed to increased inflammation (Huang et al., 2015). Pharmacological inhibition or genetic knockout of NHE1 in microglia reduced proinflammatory cytokines secretion, such as IL-1β, IL-6, and TNF-α (Liu et al., 2010; Shi et al., 2011; Song et al., 2018), as well as reduced CD86 protein expression (Song et al., 2018). Selective deletion of Nhe1 in microglia switched them to anti-inflammatory phenotypes with increased TGF-β and IL-10 secretion, along with elevated Ym1 protein expression after ischemic stroke (Song et al., 2018). Recent findings also revealed that NHE1 activation suppressed microglial phagocytic activity and contributed to sustained white matter demyelination and neurological dysfunction, possibly through reduced myelin debris clearance after ischemic stroke (Song et al., 2018).

Taken together, these findings collectively suggest that NHE1 plays an important role in maintaining the microglial pHi homeostasis, and is required for proinflammatory microglial activation and oxidative injury after ischemic stroke. Blocking NHE1 activity promotes transformation of activated microglia to restorative phenotypes for ischemic brain tissue repair.

Microglial NCX in proinflammatory microglial activation and microglial apoptosis

Microglia express all three isoforms of NCX, while NCX1 is the most abundant isoform (Newell, Stanley, & Schlichter, 2007; Parpura et al., 2016). NCX1 plays a major role in regulation of Ca2+ in microglia, as silencing NCX1 with small interfering RNA (siRNA) completely abolished the Ca2+ increase in cultured microglia after OGD (Boscia et al., 2009, 2013). NCX1rev activation can be induced by inflammatory factors such as interferon (IFN)-γ in cultured microglia, which was mediated by upregulation of protein kinase C (PKC), tyrosine kinase, and MAPK/ERK (Nagano, Kawasaki, Baba, Takemura, & Matsuda, 2004). The NCX1rev-mediated Ca2+ influx is important for microglial activation, migration, and phagocytosis after LPS stimulation in early postnatal mice (Sunkaria, Bhardwaj, Halder, Yadav, & Sandhir, 2016). However, the NCX1-dependent microglial migration is restricted to the bradykinin-induced chemotaxis, but not ATP-induced migration, as proven in the NCX1 knockout mouse model (Ifuku et al., 2007; Noda, Ifuku, Mori, & Verkhratsky, 2013). NCX plays a critical role in microglial phagocytic activity through the Ca2+-dependent activation of the purinergic receptors (P2Y2, P2Y6, and P2Y12) during postnatal development (Sunkaria et al., 2016), therefore might also be important in microglial phagocytosis of tissue debris after ischemic stroke. Indeed, upon ischemic stroke, NCX1rev activity was significantly enhanced in the round phagocytic microglia in the infarct core at 3–7 days after pMCAO, which was accompanied by downregulation of NCX2 and NCX3 protein expressions (Boscia et al., 2009, 2013). In addition, the microglia-mediated respiratory burst was suppressed by pharmacological inhibition of NCX1rev activity (Newell et al., 2007). This is at least partially through Na+-dependent mechanism and the activation of NHE1, since the reduced Ca2+ level with NCX inhibitor was similar to that with the NHE1 inhibitor in a microglial OGD model (Liu et al., 2010; Shi, Kim, et al., 2013). NCX can also couple with NHE1 activation in inflammatory responses, and contributes to the production of inducible nitric oxide synthase (iNOS) through the NHE1-NCX-CaMKN/TAK1-NFκB signaling in cultured microglia (Huang et al., 2015). Moreover, the NCX-mediated Ca2+ elevation concurrently led to ER stress and apoptosis in cultured microglia induced by NO, while pharmacological inhibition of NCX protected microglia from these events (Matsuda, Nagano, Takemura, & Baba, 2006; Nagano et al., 2005; Takuma et al., 2013). Thus, NCX is important for mediating Ca2+ signaling in microglia, which contributes to microglial proinflammatory activation and microglial apoptosis during ischemic stroke. Inhibition of NCX activity suppresses the microglia-mediated respiratory burst for tissue repair after ischemic stroke.

1.1.3 |. Ion transporters in oligodendrocytes after ischemic stroke

Due to limited collateral blood flow in the white matter, oligodendrocytes are the most vulnerable type of glial cells for ischemic damage (Wang, Liu, et al., 2016). Oligodendrocyte cell death can result from excessive release of glutamate (Doyle et al., 2018), increased oxidative stress (Takase et al., 2018), and elevated neuroinflammation (Choi et al., 2016), altogether leading to sustained loss of myelin sheath and prolonged neurological function deficits after ischemic stroke. Brain injury triggers immediate proliferation and migration of the OPCs, which can differentiate into the mature myelin-secreting oligodendrocytes (Hughes, Kang, Fukaya, & Bergles, 2013). However, concurrent activation of several signaling pathways (such as Nogo receptor-1) in OPCs prevents their maturation (Sozmen et al., 2016). Therefore, a better understanding of the regulatory mechanisms for white matter repair after ischemic stroke is important.

NHE1 and NBC have been reported to regulate pHi in cultured oligodendrocytes under homeostatic conditions (Ro & Carson, 2004). The NHE1-mediated H+ extrusion is the predominant alkalinizing mechanism in the perikaryon and proximal processes of oligodendrocytes, while the HCO3 efflux mediated by the outward mode of NBC activity functions as the predominant acidifying mechanism in the distal dendrites of oligodendrocytes (Ro & Carson, 2004). However, no studies on oligodendrocytic NHE1 or NBC under ischemic stroke conditions were reported. Oligodendrocytic NCX and NKCC1 have been shown to play important roles in oligodendrocyte damages and genesis after ischemic stroke.

NCX in oligodendrocytic survival, maturation, myelin formation

All three isoforms of NCX are expressed in oligodendrocytes (Parpura et al., 2016; Quednau, Nicoll, & Philipson, 1997), and can mediate myelin synthesis by modulating Ca2+ signaling in OPCs (Friess et al., 2016). Among the three isoforms, NCX1 and NCX3, but not NCX2, are involved in the OPC differentiation into mature oligodendrocytes during development, with NCX1 downregulated and NCX3 upregulated during this process (Boscia et al., 2012, 2013).

Oligodendrocytic NCX3 activation is beneficial and promotes myelination. In response to OGD, only NCX3 mRNA level in cultured rat OPCs was significantly decreased, but not NCX1 or NCX2 (Cai, Ma, Tian, Li, & Li, 2018). Pharmacological inhibition of NCX3 led to elevated lactate dehydrogenase (LDH) release in these OPCs, which correlated with increased OPC apoptosis, mitochondrial damage, and the activation of ERK1/2 and poly [ADP-ribose] polymerase (PARP1) pathways (Cai et al., 2018). Knockdown of NCX3 using siRNA in OPC cultures inhibited the expression of myelin proteins 2′,3′-cyclic-nucleotide 3′-phosphodiesterase (CNPase) and myelin basic protein (MBP), while overexpression of NCX3 promoted these myelin protein expressions (Boscia et al., 2012, 2013). Genetic knockout of NCX3 increased NG2+ OPC counts in the spinal cord in mice, but concurrently exhibited reduced size of the spinal cord as well as hypomyelination with decreased MBP and axonal neurofilament (NF200) protein expression (Boscia et al., 2012, 2013). These findings indicate that loss of NCX3 protein arrests OPC differentiation and myelination, but not OPC proliferation. The NCX3-mediated modulation on myelin synthesis is through Ca2+ signaling, and thus can be induced by increased Na+i level which subsequently triggers the NCXrev activity, such as through the inhibition of the α2 isoform of Na+–K+–ATPase (Hammann, Bassetti, White, Luhmann, & Kirischuk, 2018). Increased resting Ca2+i level stimulated MBP synthesis in cultured OPCs, while blockade of NCXrev reduced resting Ca2+i level and led to less MBP synthesis in acute slices of the corpus callosum from mice at P20-30 but not P10 (Friess et al., 2016). In addition, a recent study also demonstrated that NCX3 played an important role in the glutamate-mediated remyelination in a cuprizone-induced demyelination model in mice (de Rosa et al., 2019). This could relate to the study that optic nerve myelin responded to axonal action potentials with a rise in Ca2+ levels, which was mediated by glutamatergic NMDA receptors, and played a role in modulating oligodendrocyte myelination in an activity-dependent manner (Micu et al., 2016).

On the contrary, unlike NCX3, increased NCX1 activity in axons exerted detrimental effects on axonal injury and myelination through increased Ca2+i overload and elevated Na+ influx, as ~73% of the injured axons expressing β-amyloid precursor protein (β-APP) co-expressed NCX1 and the voltage-gated Na+ channel Nav1.6, while only ~4% coexpression was found in the β-APP negative axons in the spinal cord from a demyelination model of EAE in mice (Craner, Hains, Lo, Black, & Waxman, 2004). Moreover, oligodendrocytic NCX activity is also induced by NKCC1-mediated Na+ influx, and the NCX-mediated Ca2+i increase is involved in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-induced excitotoxicity for mitochondrial damage and oligodendrocyte cell death in cultured oligodendrocytes (Chen et al., 2007). Whether the NCX1 isoform plays a role in this damaging process remains unknown.

Therefore, different isoforms of NCX play extinct roles in regulation of white matter injury and repair, with NCX3 promotes oligodendrocytic survival, maturation, and myelin formation, while NCX1 induces demyelination and excitotoxicity. Inhibition of NCX1 and enhancement of NCX3 may be beneficial for ischemic white matter repair.

NKCC1 in oligodendrocytic survival, proliferation, maturation, and myelin formation

NKCC1 is present in oligodendrocytes (Chen & Sun, 2005), and regulates the formation of OPCs at the early stage of neurogenesis at E14.5, as genetic ablation of NKCC1 displayed a significantly reduced number of Olig2+ oligodendrocytes at this embryonic stage (Magalhaes & Rivera, 2016). The postnatal expression of oligodendrocytic NKCC1 started to increase from P3-7 (Jantzie et al., 2015; Wang, Yan, Kintner, Lytle, & Sun, 2003), and this was in parallel with the increase of MBP expression from P6 to P21 (Wang et al., 2003), indicating its role in myelination during development. Selective deletion of Nkcc1 gene from the PDGFRα+ OPCs showed increased proliferation of NG2+Ki67+ OPC counts, but suppressed differentiation of OPCs with reduced CC1+ mature oligodendrocyte counts and decreased MBP expression in the cerebellum white matter of neonatal mice (Zonouzi et al., 2015). These effects resembled the results from administration of GABAA receptor antagonists in neonatal wild-type mice (Zonouzi et al., 2015), suggesting that the NKCC1-mediated regulation on neonatal OPC maturation and white matter myelination could relate to the activation of GABAA receptors. Indeed, NKCC1 activity was stimulated by the Cl efflux and cell shrinkage mediated by the activation of GABAA receptors in oligodendrocytes (Wang et al., 2003). The upregulated NKCC1 activity in turn maintained a high Cli to allow for the GABAergic trophic effects, which played an important role in oligodendrocytic survival following the withdrawal of growth factors (Wang et al., 2003). Inhibition of NKCC1 with bumetanide completely abolished the GABAA receptor-mediated trophic effects and resulted in higher oligodendrocyte mortality (Wang et al., 2003).

However, activation of NKCC1 concurrently mediates Na+ influx, which triggers Ca2+ overload through NCXrev and non-NMDA type glutamate receptors in oligodendrocytes, eliciting compromised mitochondrial functions, enhanced glutamergic excitotoxicity induced by AMPA, and increased oligodendrocyte death (Chen et al., 2007; Wilke et al., 2004). During ischemia, elevated oligodendrocytic NKCC1 activity increased the vulnerability to ischemic white matter injury (Jantzie et al., 2015). Blockade of NKCC1 activity using bumetanide reverted the ischemic stroke-induced MBP loss and white matter damage in neonatal rats (Jantzie et al., 2015). NKCC1 inhibition also promoted OPC proliferation through increased DNA synthesis and cell cycle progression, with elevated expressions of cyclin D1, CDK4, and cyclin E in an OGD model (Fu et al., 2015). Similarly, bumetanide induced OPC proliferation in a bilateral common carotid artery stenosis model in adult mice, along with improved white matter integrity and working memory functions (Yu et al., 2018). The detrimental role of NKCC1 on oligodendrocytes and white matter damages after ischemic stroke was through the upstream regulation of WNK3-SPAK/OSR1 signaling pathway, since genetic knockout of WNK3 decreased the phospho-activation and overall protein expression of NKCC1, along with reduced axonal demyelination and improved neurological functions after ischemic stroke (Begum et al., 2015). Genetic knockout of WNK3 or knockdown of SPAK/OSR1 using siRNA also illustrated an increased tolerance of oligodendrocytes after OGD (Begum et al., 2015). However, regarding relationship between WNK-SAPK/OSR1-NKCC1 complex and ERK signaling pathway, inconsistent results have been reported: bumetanide reduced p-ERK and elevated p-p38 MAPK expressions in the OPCs in response to OGD (Fu et al., 2015), but elevated p-ERK and decreased p-JNK and p-p38 MAPK signaling were detected in the poststroke corpus callosum after bumetanide treatment (Yu et al., 2018).

Hence, NKCC1 is essential for oligodendrocytic survival, maturation, and myelin formation during development. However, upon ischemic stroke, the overstimulated NKCC1 plays harmful roles in stimulating excitotoxicity, which increases oligodendrocyte apoptosis, as well as suppressing OPC proliferation and causing white matter demyelination, which contributes to cognitive dysfunctions. The regulative mechanisms of the WNK3-SPAK/OSR1-NKCC1 signaling pathway in oligodendrogenesis during development and postinjury remyelination are not well defined and require more study. This will shed light on their potentials as new therapeutic targets.

1.2 |. Glial ion transporters in TBI

TBI directly causes primary injury in neurons, and differential polarization of astrocytes (Burda, Bernstein, & Sofroniew, 2016), microglia (Jassam, Izzy, Whalen, McGavern, & El Khoury, 2017), and oligodendrocyte (Takase et al., 2018) can either sustain the damage through a cascade of events, causing secondary injury, or accelerate tissue repair, leading to lesion resolving. Increasing evidence suggest that glial ion transporters play important roles in the regulation of these glial functions for tissue damage or repair after TBI.

1.2.1 |. Ion transporters in reactive astrocytes for astrogliosis, brain edema, astrocyte apoptosis, and synaptic transmission after TBI

Astrocytes are pivotal responders to TBI and can form reactive astrogliosis in areas of injured, but surviving and functioning neural tissue (Burda et al., 2016). Similar to that in the ischemic stroke, the reactive astrocytes can form an astroglial scar that surrounds and restricts the spread of inflammatory response from the lesion core to the remote nonlesioned area (Burda et al., 2016). The secretion of restorative factors from the astroglial scar, such as BDNF, TGF-β, and CNTF, are important for synaptogenesis and axonal outgrowth after TBI or spinal cord injury (SCI; Bei et al., 2016; Chen et al., 2017; Diniz, Matias, Siqueira, Stipursky, & Gomes, 2018), while its concurrent secretion of CSPG is detrimental for neurite outgrowth in a mouse TBI model (Galindo et al., 2018).

Astrocytic NCXrev activity is involved in astrocyte migration and proliferation, which contributes to astrogliosis in a mechanical injury model using cultured primary astrocytes (Pappalardo, Samad, Black, & Waxman, 2014). This is dependent on the increased Na+i mediated by Nav1.5, as knockdown of Nav1.5 mRNA attenuated the NCX-mediated Ca2+ rise as well as astrogliosis (Pappalardo et al., 2014). However, brain astrocytes exhibit a significant regional heterogeneity that hippocampal astrocytes are different from neocortical astrocytes in multiple ways (Ziemens et al., 2019). In response to brain injury, hippocampal astrocytes expressed reduced NCX mRNA and protein expression, which was opposite to that in the neocortical astrocytes (Hwang et al., 2006; Matsuda & Baba, 1998; Zhao, Gorin, Berman, & Lyeth, 2008).

Astrocytic NKCC1 activity contributes to brain edema following TBI. In an in vitro fluid percussion (FPI) injury model, the cultured astrocytes showed extensive swelling with an upregulation of AQP4 expression in the plasma membrane, which was dependent on the astrocytic NKCC1 activation (Rao, Reddy, Curtis, & Norenberg, 2011) via the MAPKs and oxidative/nitrosative stress pathways (Jayakumar et al., 2011). The astrocytic swelling was also triggered by glutamate and proinflammatory cytokines, such as TNF-α and IL-1β, released from microglia after TBI (Jayakumar et al., 2018). Pharmacological inhibition of NKCC1 with bumetanide or silencing NKCC1 with siRNA significantly reduced the astrocytic swelling (Jayakumar et al., 2011). In addition, elevated expression of NKCC1 was detected along with increased astrocyte apoptosis in a stretch injury model using cultured astrocytes, with increased Bax level and cleaved caspase-3 activity, enhanced proinflammatory astrocytic responses of elevated IL-1β, IL-6, and TNF-α, and the upregulation of the NF-κB pathway (Zhang, Cui, Cui, Wang, & Zhong, 2017). Inhibition of NF-κB significantly reduced the astrocytic NKCC1 activation, while inhibition of NKCC1 using bumetanide exhibited significantly decreased cleaved caspase-3 activity (Zhang et al., 2017). On the other hand, astrocytic NKCC1 can be beneficial and plays an important role in K+ uptake after TBI, which reverses the neuronal inactivity, enhances synaptic transmission and neuronal excitability, and facilitates learning and memory functions (Hertz & Chen, 2016; Sajja, Hlavac, & VandeVord, 2016).

1.2.2 |. Ion transporters in activated microglia for microglial migration, inflammation, and hypersensitivity after TBI

Microglia are rapidly activated and migrate toward the lesion site in response to CNS injury, such as TBI (Bar & Barak, 2019). Microglial migration can be induced by bradykinin, mainly through the B1 bradykinin receptor, as B1 receptor antagonist markedly inhibited the microglial motility in cultured microglia, and genetic knockout of B1 receptor suppressed microglial chemotaxis in response to bradykinin (Ifuku et al., 2007). This bradykinin-induced microglial migration is dependent on Ca2+ influx via the NCXrev activity, since both pharmacological blockade and genetic knockout of NCX abolished the bradykinin-induced microglial migration (Ifuku et al., 2007). NCX also plays a role in microglial migration and phagocytosis induced by the active form of thyroid hormone T3 (3, 3′, 5-triiodothyronine) in a stab wound model in mice (Mori et al., 2015). However, the subsequently activated channels are different between these NCX-mediated mechanisms, as the bradykinin-triggered NCX activation resulted in the intermediate-conductance (IK) type of Ca2+-dependent K+ channel activation, while the T3-induced NCX activation led to the apaminsensitive small-conductance (SK) type of activation in microglia (Ifuku et al., 2007; Mori et al., 2015). The chemoattracted microglia can exert inflammatory responses in the lesion site, where overexpression of NKCC1 and downregulation of KCCs are needed, for microglial secretions of cytotoxic cytokines and neurotrophic factors, such as TNF-α and BDNF, as well as MMPs and ROS, altogether contributing to hypersensitivity and neuropathic pain in rodent SCI or peripheral nerve injury models (Lopez-Alvarez, Modol, Navarro, & Cobianchi, 2015; Schomberg, Ahmed, Miranpuri, Olson, & Resnick, 2012). In addition, NKCC1 also plays a role in increasing the seizure susceptibility induced by TBI using the controlled cortical impact (CCI) procedures in awake un-anesthetized mice (Wang et al., 2017). Microglial secretion of TGF-β can upregulate NKCC1 expressions in the cortex and hippocampus, and leads to increased latency and duration of seizure after TBI, but it remains unclear about the specific role of microglial NKCC1 during this process (Wang et al., 2017). Recent findings also suggest that microglia-derived but not astrocyte-derived cytokines (such as TNF-α and IL-1β) upregulated NKCC1 activity in cultured neurons and promoted neuronal swelling in a severe FPI model of TBI, while no swelling in microglia themselves was observed (Jayakumar et al., 2018). Exposure of microglia to the conditioned media from traumatized neurons or astrocytes also had no effects on the microglial cell volume (Jayakumar et al., 2018).

In conclusion, a few recent studies reveal the roles of NCX and NKCC1 in astrogliosis, astrocyte apoptosis, brain edema formation, synaptic transmission, as well as microglial migration, inflammation, and hypersensitivity after TBI. However, extensive studies on glial ion transporters in the pathogenesis and progression of TBI are lacking. Further investigations are needed for a better understanding of the roles of these ion transporters in reactive astrocytes, microglia-mediated inflammation and phagocytosis for tissue repair, and oligodendrocyte-mediated remyelination after TBI.

2 |. GLIAL ION TRANSPORT IN CHRONIC NEUROLOGICAL DISEASES

2.1 |. Glial ion transporters in glioma tumors

Glioma is the malignant tumor that starts in the glial cells of the brain or the spine. Based on the specific cell types, glioma includes malignancy of astrocytes (astrocytoma and glioblastoma), oligodendrocytes (oligodendroglioma), and ependymal cells (ependymoma; Ahmed, Gull, Khuroo, Aqil, & Sultana, 2017). The World Health Organization (WHO) classifies glioma into four grades (I, II, III, and IV). Grade I and II are low-grade gliomas (LGGs), whereas Grade III and IV are high-grade gliomas (HGGs; Claus et al., 2015; Delgado-Lopez, Corrales-Garcia, Martino, Lastra-Aras, & Duenas-Polo, 2017). Standard of treatment in both LGGs and HGGs includes maximal safe surgical resection followed by radiation and chemotherapy (Oberheim Bush & Chang, 2016; Weller, 2011). Patients with LGGs have an indolent course with longer-term survival (~7 years) in comparison with HGGs. Moreover, the majority of LGGs patients (over 80%) present seizures, and 35% continue to have spontaneous seizure recurrence, which is termed as tumor-associated epilepsy (Campbell et al., 2015; Schiff, 2017). Thus, LGGs are considered malignant because of their invasive growth, resistance to therapy, and high risk of transforming into HGGs (Kohanbash et al., 2017).

Glioblastoma (GBM) belongs to Grade IV and is characterized as the most aggressive, primary malignant tumor (Louis et al., 2016). Under the standard care, GBM patients have a mean 5-year survival rate of less than 10% (Hide, Shibahara, & Kumabe, 2019). The difficulty in effectively treating GBM patients is largely due to the heterogeneous nature of these tumors and to the complex network of interactions with the tumor microenvironment (TME; Oliveira et al., 2017). The GBM microenvironment consists of a complex network of a variety of glioma-associated stromal cell types within the brain, including microglia, macrophages, astrocytes, and oligodendrocytes (Oliveira et al., 2017; van der Vos et al., 2016). Presence of tumor-associated stromal cells in the microenvironment of glioma promotes tumor progression, driven by paracrine signals and secreting trophic factors (Oliveira et al., 2017), and they consequently increase glioma resistance to current therapies (Brandao, Simon, Critchley, & Giamas, 2018; O’Brien, Howarth, & Sibson, 2013). Among various cells of the TME, glial cells, including astrocytes and microglia/macrophages (TAMs), are the most common cellular entities that interact with GBM (Matias et al., 2018). The TAM population is usually the dominant glioma-infiltrating immune cells, occupied around 30% of glioma mass (Miller et al., 2014). Astrocytes, which comprise 40–50% of the cells in TME, become reactive, resulting in a layer of reactive astrocytes surrounding the tumor (Guan, Hasan, Maniar, Jia, & Sun, 2018; Placone, Quinones-Hinojosa, & Searson, 2016). Therefore, better understanding of the TME is important for developing effective therapies for GBM patients.

2.1.1 |. Ion transporters in glioma proliferation and migration

New studies show that ion transporter proteins play a role in glioma pathophysiology. They are involved in cell proliferation, migration and invasion (Guan et al., 2018). Recent bioinformatics analysis of the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) datasets revealed that SLC9A1 mRNA expression (encoding for NHE1 protein) is significantly higher in gliomas and is associated with glioma malignancy. Worsened survival probabilities were also correlated with the elevated SLC9A1 mRNA levels in glioma (Guan et al., 2018). The underlying mechanisms of elevated NHE1 protein in glioma progression may include promoting angiogenesis, and extracellular matrix remodeling (Guan, Luo, Begum, et al., 2018). NHE1 protein in gliomas maintains alkaline pHi of 7.3–7.5 by extruding H+ in exchange of Na+ influx (Boedtkjer, Bunch, & Pedersen, 2012), which is a driving force for glycolytic metabolism (Stock & Pedersen, 2017; Figure 2). Cancer cells rely on oxidative glycolysis by increasing glucose uptake and lactate production even in the presence of oxygen and fully functioning mitochondria, known as Warburg effect (Guan, Hasan, Begum, et al., 2018). Upregulation of NHE1 protein expression in glioma also plays a vital role in glioma migration, survival and their resistance to TMZ-mediated apoptosis via activation of ERK signaling pathways (Cong et al., 2014; Figure 2). Moreover, ezrin-radixinmoesin (ERM) proteins play an important role in glioma migration and invasion and interact with NHE1 protein (Figure 2). NHE1 is partially colocalized with ezrin and increased colocalization of NHE1 and ezrin was detected in glioma cells which was abolished in cells treated with NHE1 inhibitor HOE642 (Cong et al., 2014). Furthermore, direct interactions between NHE1 and ERM can recruit phosphoinositide 3-kinase and Akt to regulate cell survival (Cong et al., 2014). In addition, glioma cells migrate through narrow and tortuous extracellular spaces posing significant demands on cell volume changes, which can also be mediated by Cl channels and transporters (Ernest & Sontheimer, 2007; Habela, Ernest, Swindall, & Sontheimer, 2009). The Cl channels, including ClC-2, ClC-3, ClC-5, ClC-6, and ClC-7, carry Cl influx or efflux along with obligated water across the cell membrane and are involved in glioma cell volume dynamics and glioma cell migration (Ernest, Weaver, Van Duyn, & Sontheimer, 2005). Among these Cl channels and transporters, NKCC1 protein was also detected in human gliomas and correlated with the tumor malignancy (Schiapparelli et al., 2017) and plays a fundamental role in regulation of intracellular volume by transport of Na+, K+, and Cl ions across the plasma membrane into glioma cells. Expression of NKCC1 protein localized to the leading edge of glioma cells which contributes to tumor invasion through interacting with ERM cytoskeleton proteins (Lin, Liu, Ling, & Xu, 2016; Zhu et al., 2014; Figure 2). NKCC1 protein also promotes epithelial mesenchymal transformation-like process via facilitating the binding of Rac1 and RhoA to GTP during glioma cell invasion (Ma et al., 2019). Ilkhanizadeh et al. (2018) recently found that anti-secretory factor (AF) inhibited cell volume regulation of GBM cells, an effect that was phenocopied in vitro by the NKCC1 inhibitor bumetanide (BMT) through reducing interstitial fluid pressure (IFP) and increasing drug uptake (Ilkhanizadeh et al., 2018). In short, these new findings indicate that ion transporters play an important role in tumorigenesis and emerge as new therapeutic targets for glioma.

FIGURE 2.

FIGURE 2

Schematic illustration of ion transporters in glioma-TAM interaction. NHE1 is expressed by both glioma cells and tumor-associated microglia/macrophages (TAMs). NHE1 functions to maintain an alkaline pHi and drives the Warburg effect for glycolysis and glioma proliferation. Moreover, NHE1 and NKCC1 localize to the leading edge of glioma cells and promote glioma migration by interacting with ezrin-radixin-moesin (ERM) cytoskeleton proteins, together with the Cl channels (ClCs), K+ channels (e.g., Kv channels), and water channels (e.g., AQP4) in conducting K+ and Cl along with obligated water efflux across the cell membrane. Soluble factors secreted by glioma cells can stimulate microglial NHE1 activation and induce release of various cytokines (TGF-β, IL-10, and Arg-1). NHE1 activation also leads to upregulation of ERK pathways, which triggers pro-survival signal transduction in glioma, and creates an acidic extracellular environment, which enhances activation of membrane Type 1 metalloprotease (MT1-MMP) and MMP9 for matrix degradation and tumor invasion. Taken together, ion transporters have multiple functions in promoting glioma progression

2.1.2 |. Ion transporters in TAMs for glioma proliferation and invasion

Microglia are considered to share macrophages characteristic of acquiring different phenotypes, like M1/M2 polarization (Matias et al., 2018). TAMs can display polarized M1-like phenotypes that contribute to both innate and adaptive anti-tumor immunity by presenting antigen to adaptive immune cells, producing proinflammatory cytokines and phagocytosing tumor cells (Wei, Gabrusiewicz, & Heimberger, 2013). Whereas in M2 state, TAMs become tumor supportive and secrete chemokines and immunosuppressive cytokines, such as IL-10, TGF-β, and arginase-1 (Arg-1). Tumor-supportive TAMs play a prominent role in angiogenesis, activating matrix-metalloproteases (MMPs) and remodeling extracellular matrix, therefore, promote tumor growth (Coniglio & Segall, 2013; Li & Graeber, 2012).

Increased mRNA expression of SLC9A1 gene encoding NHE1 protein is related to TAM accumulation (Guan, Luo, Begum, et al., 2018). TAMs exhibited increased NHE1 expression and positively correlated with Iba1 level in glioma xenografts and human GBM tissue microarray (Zhu et al., 2016). NHE1 activity is involved in upregulation of iNOS, Arg-1, and microglial cytokine IL-6, IL-10, and TGF-β expression, indicating an important role of NHE1 in glioma-mediated microglia activation (Zhu et al., 2016). Interestingly, increased transcriptional expression and release of TGF-β1 by glioma-stimulated microglia can be abolished when NHE1 activity was attenuated either by inhibition with its inhibitor HOE642 or by siRNA knockdown, indicating that microglial NHE1 protein may promote glioma cell proliferation through the production and release of TGF-β (Zhu et al., 2016). In addition, MMPs are involved in the expansion of malignant gliomas by facilitating their penetration of anatomical barriers and migration within the neuropil (Valastyan & Weinberg, 2011). Zhu et al. (2016) observed that GC22 glioma xenograft tissues exhibited abundant expression of membrane Type 1 metalloprotease (MT1-MMP), MMP2, and MMP9, whereas MT1-MMP appears more abundant in Iba1+ cells (Zhu et al., 2016). When applied glioma-conditioned media to microglia resulted in increased expression of MT1-MMP and MMP9 proteins, but not MMP2. Interestingly, this increased MT1-MMP and MMP9 expression were abolished after exposing microglia to NHE1 inhibitor HOE642, indicating that NHE1 activity is required for these changes (Zhu et al., 2016; Figure 2).

2.1.3 |. Ion transporters of TAAs in tumor-associated epilepsy and peritumoral edema formation

The roles of TAMs on brain tumor progression have been actively studied, while astrocytes, which comprise nearly half of the total healthy brain cells (Brandao et al., 2018), have received less attention (Placone et al., 2016). As glioma development involves healthy tissue invasion and destruction, astrocytes undergo a number of molecular, cellular, and functional changes to form tumor-associated reactive astrocytes (TAAs; Brandao et al., 2018). These reactive astrocytes initially aim to fight the progression of the tumor and repair the brain tissue. Some signaling mechanisms, such as NF-κB signaling, may then stimulate astrocytes into TAAs that subsequently support tumor growth (Brandao et al., 2018). TAAs have been shown to be involved in tumor cell proliferation (Bonavia et al., 2003), invasion (Rath, Fair, Jamal, Camphausen, & Tofilon, 2013), apoptosis evasion (Oliveira et al., 2017), immunoprotection (Brandao et al., 2018) and chemoprotection (Chen et al., 2015) by releasing different cytokines. For instance, TAAs-released IL-6 appears to be at the crossroads of multiple TAAs’ effects on GBM development, including promoting proliferation, invasion, and also modulating immune response to tumor progression (Brandao et al., 2018).

The interactions between glioma cells and TAAs in TME are facilitated via ion channels and ion transporters (Guan, Hasan, Maniar, et al., 2018). Among the ions, K+ homeostasis in astrocytes and/or neurons plays an important role in tumor-associated epilepsy. In normal conditions, astrocytes show a very negative resting membrane potential (between −80 and −90 mV) and prefer K+ uptake, caused by inwardly rectifying currents (Guan, Hasan, Maniar, et al., 2018), and that astrocytes are essential in maintaining the cerebral ionic homeostasis by clearance of excess K+ from the extracellular space (Schousboe, Sarup, Bak, Waagepetersen, & Larsson, 2004). The initial clearance of extracellular K+ following neuronal excitation and increase in the [K+]o is mediated by astrocytic uptake, because elevated [K+]o activates astrocytic but not neuronal Na+, K+-ATPases (Hertz & Chen, 2016). Subsequently, astrocytic K+ is re-released via Kir4.1 channel after distribution in the astrocytic functional syncytium via gap junctions. This K+ uptake by astrocytes serves a crucial purpose to prevent sustained rise in [K+]o in neural tissues, which would cause neurons to remain depolarized and unable to fire action potentials for signaling (Guan, Hasan, Maniar, et al., 2018). However, glioma cells have a more positive resting membrane potential than astrocytes, between −20 and −40 mV (Guan, Hasan, Maniar, et al., 2018; Molenaar, 2011), which leads to accumulation of K+o and may explain the prevalence of epileptic seizures seen in glioma patients (Guan, Hasan, Maniar, et al., 2018). Moreover, the less negative resting potential in glioma cells impaired the ability of Na+ gradient across the plasma membrane. Therefore, decreased Na+-dependent glutamate transporters causing a higher glutamate presence extracellularly lead to epilepsy in glioma patients (Campbell et al., 2015; Molenaar, 2011). Furthermore, the increased [K+]o leading to abnormal K+ uptake through Na–K–ATPases or stimulated Na–K–Cl cotransporters produces cell swelling and depolarization of astrocytes (Coulter & Steinhauser, 2015). The dysfunction of astrocytes in epilepsy revealed alterations in expression, subcellular localization, and function of astrocytic K+ channels and gap junctions, resulting in impaired K+ buffering and disturbance of delivery of energetic substrates to neurons (Steinhauser, Seifert, & Bedner, 2012).

In addition, high [K+]o appears to play an important role in tumor cell volume and could be involved in peritumoral edema. Peritumoral brain edema is defined as an increase in brain water content around the tumor (Blystad et al., 2017). Peritumoral brain edema is a leading cause of morbidity and mortality in patients with brain tumors. There are two types of brain edema: breakdown of the structural integrity of the BBB and the subsequent increase in interstitial fluid (vasogenic brain edema); and intracellular ionic dysregulation-mediated cell swelling (cytotoxic edema; Murayi & Chittiboina, 2016). The cytotoxic brain edema was believed to be mainly caused by swelling of astrocytes, but also includes neuronal swell (Zhang et al., 2019). High [K+]o induces swelling in astrocytes, leading to cytotoxic edema and cell death in the brain, such as after stroke and TBI (Murakami & Kurachi, 2016). The underlying mechanism involves in enhanced activity of Na, K-ATPases, and NKCC1 and results in the absorption of excess extracellular K+ and water into astrocytes, whereas, the efflux of K+ through K–Cl cotransporters (KCCs) and Kir channels was suppressed following the elevation of [K+]o (Murakami & Kurachi, 2016). Astrocytic swelling has been studied in nontumor brain edema, however, its contribution to peri-tumoral brain edema and the underlying mechanisms remain unknown, which should be further investigated.

2.2 |. Glial ion transporters in neurodegenerative diseases

The degeneration of the CNS is characterized by chronic, progressive loss of the structure and functions of neurons, leading to neurological function deficits (Chen, Zhang, & Huang, 2016). Various neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS), involve lesions in different brain regions and display different etiology. Cumulative data suggest common cellular and molecular mechanisms (Volkman & Offen, 2017). A growing number of evidences suggest that glial cells play an important role in neural survival, nerve repair and regeneration, synaptic transmission and immune inflammation, and associated with progression of neurodegenerative diseases (Xu, Wang, Song, Jiang, & Xie, 2016). Neuroinflammatory processes, mainly mediated by reactive astrocytes and microglia, are observed in almost all neurodegenerative diseases (Ettle, Schlachetzki, & Winkler, 2016). In addition, the involvement of astrocytes in the neuropathology of these diseases is likely a consequence of both the loss of normal homeostatic functions and gain of toxic functions (Phatnani & Maniatis, 2015). Furthermore, oligodendrocytes and their widely distributed progenitor cells profoundly influence and control processes known to be frequently dysregulated in neurodegenerative diseases, including ionic homeostasis and nerve impulse conduction (Ettle et al., 2016). Studies of glial ion transporters relating to these neurodegenerative diseases will be discussed in the following sections.

2.2.1 |. Astrocytic ion transporters are involved in AD pathology

AD, the most prevalent cause of dementia, is pathologically characterized by extracellular amyloid β (Aβ) plaques and intraneuronal deposits of neurofibrillary tangles (NFTs; Wang & Colonna, 2019). However, several investigations suggest that inflammation may be the key neuropathological event leading to neurodegeneration in AD (Fakhoury, 2018). Astrocytes in AD become reactive as a result of deposition of Aβ, which has detrimental consequences, including decreased glutamate uptake due to decreased expression of glutamate transporters, altered energy metabolism, altered ion homeostasis (K+ and Ca2+), increased tonic GABA-mediated inhibition, and increased release of cytokines and inflammatory mediators (Assefa, Gebre, & Altaye, 2018). It was recently shown that exposing cultured astrocytes to Aβ25-35 causes a significant increase in intracellular Na+ levels, which is associated with reduced Na+/K+-ATPase protein containing the α2 subunit expression and activity (Boscia et al., 2016). Moreover, elevated NHE1 activity was detected in cultured astrocytes in response to HIV-linked gp-120 and cytokines, likely resulting in increased pHi and enhanced release of glutamate via reversal of Na+-dependent glutamate transporters in the development of AIDS-mediated dementia (Boscia et al., 2016). In addition, it has been hypothesized that disturbance in Ca2+ homeostatic cascades may progressively affect cellular homeostasis, synaptic transmission, and ultimately cell survival (Verkhratsky, Rodriguez-Arellano, Parpura, & Zorec, 2017). Generation of astrocytic Ca2+ signals is mainly due to Ca2+ release from the ER, which is mediated by a plethora of ionotropic glutamatergic, purinergic and cholinergic receptors, transient receptor potential (TRP) channels, and NCXrev (Verkhratsky et al., 2017). Alberdi et al. (2013) found that exposure Aβ (Aβ 1–42) oligomers to cultured astrocytes led to a cytoplasmic Ca2+ increase and triggered increased expression of GFAP, as well as oxidative and ER stress, as indicated by eIF2α phosphorylation and overexpression of chaperone GRP78 (Alberdi et al., 2013). These effects were decreased by inhibitors of phospholipase C and inhibitors of ryanodine receptors and InsP3 receptors, respectively (Alberdi et al., 2013; Lim et al., 2013). These results provide evidence that Aβ disrupts ER Ca2+ homeostasis and induces ER stress that leads to astrogliosis, which may be relevant to AD pathophysiology (Alberdi et al., 2013). Taken together, astrocytic ion homeostasis is important in AD progression and future study is needed to further examine roles of ion transporters in reactive astrocytes in AD brains.

2.2.2 |. Astrocytic ion transporters are involved in dopaminergic neuron degeneration in PD

PD, the common progressive neurodegenerative movement disorder, is pathologically characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNc; Booth, Hirst, & Wade-Martins, 2017) and intraneuronal aggregates of α-synuclein in neuronal cell bodies (Lewy body) and axons (Lewy neurites; Lees, Hardy, & Revesz, 2009). Although the cellular mechanisms of dopaminergic loss in PD is not completely understood, several abnormalities including inflammation, mitochondrial dysfunction, iron accumulation, and oxidative stress are likely involved in the cascade of events (Aquilano, Baldelli, Rotilio, & Ciriolo, 2008; Lee, Kim, & Lee, 2017). It has been shown that astrocytes accumulate nonfibrillized α-synuclein in their cytoplasm during the initiation and progression of PD (Halliday & Stevens, 2011). Accumulated α-synuclein in astrocytes have been shown to produce proinflammatory cytokines and chemokines which lead to microglia activation and the dysregulation of other astrocyte functions, such as glutamate uptake and BBB integrity (Booth et al., 2017), and further cause loss of dopaminergic and motor neurons (Gu et al., 2010; Halliday & Stevens, 2011; Phatnani & Maniatis, 2015).

Oxidative stress is increasingly recognized as a central event contributing to the degeneration of dopaminergic neurons in the pathogenesis of PD (Guo, Zhao, Li, Li, & Liu, 2018). Moreover, the role of NO in the pathogenesis and progression of neurodegenerative illnesses such as PD and AD has become prominent over the years (Hannibal, 2016). Much of the oxidative damage of proteins due to the overproduction of NO by nitric oxide synthases (NOS) and its subsequent reactivity with ROS(Hannibal, 2016). High level of NOS was observed in the nigrostriatal region and basal ganglia in postmortem brains with PD (Aquilano et al., 2008). Astrocytic NCX plays an important role in the regulation of intracellular Ca2+ concentration and can be stimulated by NO in astrocytes (Ago et al., 2011). SEA0400, the specific NCX inhibitor, protects against MPTP-induced neurotoxicity by inhibiting NCX-mediated Ca2+ influx and blocking ERK phosphorylation and lipid peroxidation (Ago et al., 2011). Additionally, NCX1 mRNA is detected in the rat midbrain, where dopaminergic cell bodies are localized (Canitano et al., 2002), and in the rat striatum, where the terminal projections of dopaminergic nigrostriatal neurons are found (Papa et al., 2003). Moreover, NCX in mitochondria also plays a role in PD. PTEN-induced kinase 1 (PINK1) is a mitochondrial kinase that regulates Ca2+ efflux from the mitochondria via the mitochondrial NCX (Gandhi et al., 2009). The cause of mitochondrial Ca2+ accumulation in PINK1 deficiency is a direct impairment of Ca2+ efflux from the mitochondria secondary to dysfunction of the NCX (Gandhi et al., 2009). Ca2+ overload stimulates ROS production via NADPH oxidase, which inhibits the glucose transporter, resulting in reduced substrate delivery and impaired respiration (Gandhi et al., 2009). Taken together, NCX is an important protein in PD but the underlying mechanism of astrocytic NCX in reactive astrocyte dysfunction and its contribution to PD pathology need to be further studied.

2.2.3 |. Microglial ion transporters in inflammation and neurodegeneration in PD

In PD, neuroinflammation and microglia activation are considered neuropathological hallmarks (Joers, Tansey, Mulas, & Carta, 2017). Activated microglia were reported to cluster around dead or dystrophic, pigmented dopamine neurons in the PD substantia nigra and elevated cytokines and other inflammatory mediators were subsequently found both in the nigra and striatum of PD patient autopsy (Rogers, Mastroeni, Leonard, Joyce, & Grover, 2007). Microglia is recognized as a double-edged sword, depending on the activation state of microglia and showing both neurotoxic and neuroprotective effects (Okolie et al., 2016). In the early stage of the PD, microglia can be activated directly or indirectly by a range of misfolded proteins or pathogens, such as α-synuclein, and produce a variety of proinflammatory mediators such as cytokines, chemokines, which contribute to engulf infectious organisms or invading pathogens, and clearing toxic proteins and cell debris from the injury site by phagocytosis (Okolie et al., 2016). However, prolonged or excessive activation of microglia may result in pathological forms of inflammation that contribute to the progression of neurodegeneration, causing the loss of dopaminergic neurons (Cao, Standaert, & Harms, 2012; Ramirez et al., 2017). Among the three different NCX isoforms (NCX1, NCX2, and NCX3), NCX1 is the most highly expressed isoform in microglia (Boscia et al., 2009). Upregulation of microglial NCX1 plays a crucial role in regulation of microglia activation (Boscia et al., 2009). Sirabella et al. (2018) found that high levels of NCX1 protein expression are detected in IBA-1-positive microglial cells in the striatum of A53T mice (an animal model mimicking a familial form of PD). Whereas decreased NCX3 protein was observed in tyrosine hydroxylase (TH)-positive neurons in SNc of A53T mice, a phenomenon that was accompanied by an increase in mitochondrial Ca2+ concentrations and neuronal death (Sirabella et al., 2018). These new findings demonstrated that changes in NCXs expression and activity in the midbrain and striatum of A53T mice contribute to the loss of dopaminergic neurons and also suggested that an imbalance of the mechanisms involved in the regulation of cytosolic and mitochondrial Ca2+ homeostasis might be associated with the degeneration of dopaminergic neurons in the presence of a mutated form of α-synuclein A53T, thus revealing new potential players in the pathophysiology of PD (Sirabella et al., 2018). Limited information is available on the underlying mechanism of NCXs in astrocytes and microglia involved in PD pathology by regulating Ca2+ homeostasis. Future studies are needed to investigate glial ion transporters in specific processes of the pathological development of PD.

2.2.4 |. Glial ion transporters in axonal damage in MS

MS is a chronic demyelinating disease of the CNS, characterized by immune-mediated inflammatory and degenerative changes (Lemus, Warrington, & Rodriguez, 2018). This axonal demyelination and neurodegeneration play an important role in accumulating nonremitting neurologic deficits in chronic lesions, showing a variety of clinical presentations and unclear pathogenesis (Lemus et al., 2018; Ludwin, Rao, Moore, & Antel, 2017; Plemel, Liu, & Yong, 2017). Although the mechanisms involved in MS are incompletely understood, myelin membranes and oligodendrocytes are destroyed through chronic inflammation in the CNS (Dulamea, 2017). In general, a multifactorial interplay of genetic and environmental factors leads to a chronic activation of the immune cells and cerebral tissue injury in MS (Dulamea, 2017). NCX3, identified as a myelin membrane component, regulates [Na+]i and [Ca2+]i in oligodendrocytes during physiological and demyelinating conditions (Boscia et al., 2016). Ca2+ signaling mediated by the NCX3 (forward mode) is involved in oligodendrocyte maturation and myelin formation. Knockdown of NCX3 isoform in OPCs prevented expressions of two major myelinating oligodendrocyte markers 2′,3′-cyclic nucleotide-3′-phosphodiesterase (CNPase) and myelin basic protein (MBP), while overexpression of NCX3 isoform triggered an upregulation of CNPase and MBP expression (Boscia et al., 2012). Furthermore, NCX3 knockout mice showed a reduced size of spinal cord with marked hypo-myelination, a reduced diameter of axons and a dramatic decrease in OPC and premyelinating cells (Boscia et al., 2012; Casamassa et al., 2016). Moreover, during the chronic phase of an EAE model, elevated NCX3 protein expression was detected and coexpressed with the marker NG2 and CNPase while knockout of NCX3 accelerated the early onset of symptoms (Casamassa et al., 2016). In addition, astrocytes function as CNS immune cells and release inflammatory molecules in the pathogenesis of MS (Guerrero-Garcia, 2017). Astrocytes exert active, dual, and paradoxical roles during disease development in MS (Williams, Piaton, & Lubetzki, 2007). They could contribute to MS through the following mechanisms: (a) as part of the innate immune system, (b) as a source of cytotoxic factors, (c) inhibiting remyelination and axonal regeneration by forming a glial scar, and (d) contributing to axonal mitochondrial dysfunction (Correale & Farez, 2015). Chronic MS plaques contain demyelinated axons that are often surrounded by a dense meshwork of astrocytic processes (Holley, Gveric, Newcombe, Cuzner, & Gutowski, 2003). These astrocytic processes exhibited immunofluorescence staining signals of Nav1.2 and NCX (no specific isomer is determined). Astrocytic NCX can contribute to the regulation of Ca2+i levels via forward Ca2+ extrusion or reverse Ca2+ influx operation. Black, Newcombe, Trapp, and Waxman (2007) examined the expression of Nav1.2 and NCX in chronic MS plaques within the spinal cord. They found Nav1.2 was not detected along demyelinated axons in chronic lesions but was expressed by scar and reactive astrocytes within the plaque (Black et al., 2007). Moreover, NCX protein was clearly present within the scar astrocytes surrounding the demyelinated axons (Black et al., 2007). Although the functional significance of NCX and Nav1.2 in these astrocytes is not clear at this time, the presence of NCX in these cells implies that these molecules may be involved in Ca2+ flux into or out of glial cells within chronic lesions. Future studies are needed to understand function of astrocytic NCX and other ion transporters in peri-plaque scars and demyelination in MS.

3. |. CONCLUSION

The significance of glial cells (astrocytes, microglia, and oligodendrocytes) in neurological diseases has now been widely recognized. Several major ion transporters (NCX, NHE1, and NKCC1, and so on) are well documented to play critical and distinct roles in regulation of glial functions under physiological and pathophysiological conditions. During acute brain injuries (such as ischemic stroke and TBI), these ion transporters are rapidly activated in glial cells and affect excitotoxicity, ion homeostasis, inflammation, and myelination in experimental models. However, in comparison to ischemic stroke research, studies of these glial ion transporters in post-TBI brains and their impact on tissue repair and chronic functional deficits are lacking. Some newly emerged studies suggest that ion transporters (NHE1 and NKCC1) play important roles in activation and transformation of TAMs and TAAs in developing protumoral microenvironment of glioma. Glial NCX (in astrocytes and microglia) is shown to regulate Ca2+ signaling and glutamate uptake in experimental AD, MS, and PD models. Considering the significant roles of glial cells in both acute and chronic neurological diseases, our knowledge of glial ion transporters in transformation of glial functions and in disease pathogenesis is limited, which warrants further exploration, especially for chronic neurodegenerative diseases.

Acknowledgments

Funding information

NIH, Grant/Award Numbers: NSR0148216, R0138118

REFERENCES

  1. Ago Y, Kawasaki T, Nashida T, Ota Y, Cong Y, Kitamoto M, … Matsuda T (2011). SEA0400, a specific Na+/Ca2+ exchange inhibitor, prevents dopaminergic neurotoxicity in an MPTP mouse model of Parkinson’s disease. Neuropharmacology, 61(8), 1441–1451. 10.1016/j.neuropharm.2011.08.041 [DOI] [PubMed] [Google Scholar]
  2. Ahmed S, Gull A, Khuroo T, Aqil M, & Sultana Y (2017). Glial cell: A potential target for cellular and drug based therapy in various CNS diseases. Current Pharmaceutical Design, 23(16), 2389–2399. 10.2174/1381612823666170316124500 [DOI] [PubMed] [Google Scholar]
  3. Akiyoshi R, Wake H, Kato D, Horiuchi H, Ono R, Ikegami A, … Nabekura J. (2018). Microglia enhance synapse activity to promote local network synchronization. eNeuro, 5(5), e0088–18. 10.1523/ENEURO.0088-18.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alberdi E, Wyssenbach A, Alberdi M, Sanchez-Gomez MV, Cavaliere F, Rodriguez JJ, … Matute C (2013). Ca(2+)-dependent endoplasmic reticulum stress correlates with astrogliosis in oligomeric amyloid beta-treated astrocytes and in a model of Alzheimer’s disease. Aging Cell, 12(2), 292–302. 10.1111/acel.12054 [DOI] [PubMed] [Google Scholar]
  5. Annunziato L, Boscia F, & Pignataro G (2013). Ionic transporter activity in astrocytes, microglia, and oligodendrocytes during brain ischemia. Journal of Cerebral Blood Flow and Metabolism, 33(7), 969–982. 10.1038/jcbfm.2013.44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Aquilano K, Baldelli S, Rotilio G, & Ciriolo MR (2008). Role of nitric oxide synthases in Parkinson’s disease: A review on the antioxidant and anti-inflammatory activity of polyphenols. Neurochemical Research, 33(12), 2416–2426. 10.1007/s11064-008-9697-6 [DOI] [PubMed] [Google Scholar]
  7. Assefa B, Gebre A, & Altaye B (2018). Reactive astrocytes as drug target in Alzheimer’s disease. BioMed Research International, 2018, 4160210–4160247. 10.1155/2018/4160247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bar E, & Barak B (2019). Microglia roles in synaptic plasticity and myelination in homeostatic conditions and neurodevelopmental disorders. Glia. 10.1002/glia.23637 [DOI] [PubMed] [Google Scholar]
  9. Baudel MASMA, Poole AV, & Darlison MG (2017). Chloride cotransporters as possible therapeutic targets for stroke. Journal of Neurochemistry, 140(2), 195–209. 10.1111/jnc.13901 [DOI] [PubMed] [Google Scholar]
  10. Beck J, Lenart B, Kintner DB, & Sun D (2003). Na-K-Cl cotransporter contributes to glutamate-mediated excitotoxicity. The Journal of Neuroscience, 23(12), 5061–5068 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12832529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Becker HM, Broer S, & Deitmer JW (2004). Facilitated lactate transport by MCT1 when coexpressed with the sodium bicarbonate cotransporter (NBC) in Xenopus oocytes. Biophysical Journal, 86(1), 235–247. 10.1016/S0006-3495(04)74099-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Begum G, Song SS, Wang SX, Zhao HS, Bhuiyan MIH, Li E, … Sun DD (2018). Selective knockout of astrocytic Na+/H+ exchanger isoform 1 reduces astrogliosis, BBB damage, infarction, and improves neurological function after ischemic stroke. Glia, 66(1), 126–144. 10.1002/glia.23232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Begum G, Yuan H, Kahle KT, Li L, Wang S, Shi Y, … Sun D (2015).Inhibition of WNK3 kinase signaling reduces brain damage and accelerates neurological recovery after stroke. Stroke, 46(7), 1956–1965. 10.1161/STROKEAHA.115.008939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bei F, Lee HHC, Liu X, Gunner G, Jin H, Ma L, … He Z (2016). Restoration of visual function by enhancing conduction in regenerated axons. Cell, 164(1–2), 219–232. 10.1016/j.cell.2015.11.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Ben-Ari Y (2017). NKCC1 chloride importer antagonists attenuate many neurological and psychiatric disorders. Trends in Neurosciences, 40(9), 536–554. 10.1016/j.tins.2017.07.001 [DOI] [PubMed] [Google Scholar]
  16. Bhuiyan MIH, Song SS, Yuan H, Begum G, Kofler J, Kahle KT, … Sun D (2017). WNK-Cab39-NKCC1 signaling increases the susceptibility to ischemic brain damage in hypertensive rats. Journal of Cerebral Blood Flow and Metabolism, 37(8), 2780–2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Black JA, Newcombe J, Trapp BD, & Waxman SG (2007). Sodium channel expression within chronic multiple sclerosis plaques. Journal of Neuropathology and Experimental Neurology, 66(9), 828–837. 10.1097/nen.0b013e3181462841 [DOI] [PubMed] [Google Scholar]
  18. Blaustein MP, Juhaszova M, Golovina VA, Church PJ, & Stanley EF (2002). Na/Ca exchanger and PMCA localization in neurons and astrocytes: Functional implications. Annals of the New York Academy of Sciences, 976, 356–366 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12502582. [DOI] [PubMed] [Google Scholar]
  19. Blystad I, Warntjes JBM, Smedby O, Lundberg P, Larsson EM, & Tisell A (2017). Quantitative MRI for analysis of peritumoral edema in malignant gliomas. PLoS One, 12(5), e0177135 10.1371/journal.pone.0177135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Boedtkjer E, Bunch L, & Pedersen SF (2012). Physiology, pharmacology and pathophysiology of the pH regulatory transport proteins NHE1 and NBCn1: Similarities, differences, and implications for cancer therapy. Current Pharmaceutical Design, 18(10), 1345–1371 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/22360557. [DOI] [PubMed] [Google Scholar]
  21. Bonavia R, Bajetto A, Barbero S, Pirani P, Florio T, & Schettini G (2003). Chemokines and their receptors in the CNS: Expression of CXCL12/SDF-1 and CXCR4 and their role in astrocyte proliferation. Toxicology Letters, 139(2–3), 181–189. 10.1016/S0378-4274(02)00432-0 [DOI] [PubMed] [Google Scholar]
  22. Bondarenko A, Svichar N, & Chesler M (2005). Role of Na+-H+ and Na+-Ca2+ exchange in hypoxia-related acute astrocyte death. Glia, 49 (1), 143–152. 10.1002/glia.20107 [DOI] [PubMed] [Google Scholar]
  23. Booth HDE, Hirst WD, & Wade-Martins R (2017). The role of astrocyte dysfunction in Parkinson’s disease pathogenesis. Trends in Neurosciences, 40(6), 358–370. 10.1016/j.tins.2017.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Boscia F, Begum G, Pignataro G, Sirabella R, Cuomo O, Casamassa A, … Annunziato L (2016). Glial Na(+)-dependent ion transporters in pathophysiological conditions. Glia, 64(10), 1677–1697. 10.1002/glia.23030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Boscia F, D’Avanzo C, Pannaccione A, Secondo A, Casamassa A, Formisano L, … Annunziato L (2012). Silencing or knocking out the Na+/Ca2+ exchanger-3 (NCX3) impairs oligodendrocyte differentiation. Cell Death and Differentiation, 19(4), 562–572. 10.1038/cdd.2011.125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Boscia F, D’Avanzo C, Pannaccione A, Secondo A, Casamassa A, Formisano L, … Annunziato L (2013). New roles of NCX in glial cells: Activation of microglia in ischemia and differentiation of oligodendrocytes. Advances in Experimental Medicine and Biology, 961, 307–316. 10.1007/978-1-4614-4756-6_26 [DOI] [PubMed] [Google Scholar]
  27. Boscia F, Gala R, Pannaccione A, Secondo A, Scorziello A, Di Renzo G, & Annunziato L (2009). NCX1 expression and functional activity increase in microglia invading the infarct core. Stroke, 40(11), 3608–3617. 10.1161/STROKEAHA.109.557439 [DOI] [PubMed] [Google Scholar]
  28. Brandao M, Simon T, Critchley G, & Giamas G (2018). Astrocytes, the rising stars of the glioblastoma microenvironment. Glia, 67, 779–790. 10.1002/glia.23520 [DOI] [PubMed] [Google Scholar]
  29. Brazhe AR, Verisokin AY, Verveyko DV, & Postnov DE (2018). Sodium-calcium exchanger can account for regenerative Ca2+ entry in thin astrocyte processes. Frontiers in Cellular Neuroscience, 12, 250 10.3389/fncel.2018.00250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Burda JE, Bernstein AM, & Sofroniew MV (2016). Astrocyte roles in traumatic brain injury. Experimental Neurology, 275(Pt 3), 305–315. 10.1016/j.expneurol.2015.03.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Cai L, Du T, Song D, Li B, Hertz L, & Peng L (2011). Astrocyte ERK phosphorylation precedes K(+)-induced swelling but follows hypotonicity-induced swelling. Neuropathology, 31(3), 250–264. 10.1111/j.1440-1789.2010.01172.x [DOI] [PubMed] [Google Scholar]
  32. Cai Q, Ma T, Tian Y, Li C, & Li H (2018). Catalpol inhibits ischemia-induced premyelinating oligodendrocyte damage through regulation of intercellular calcium homeostasis via Na+/Ca2+ exchanger 3. International Journal of Molecular Sciences, 19(7), 1925 10.3390/ijms19071925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Campbell SL, Robel S, Cuddapah VA, Robert S, Buckingham SC, Kahle KT, & Sontheimer H (2015). GABAergic Disinhibition and impaired KCC2 Cotransporter activity underlie tumor-associated epilepsy. Glia, 63(1), 23–36. 10.1002/glia.22730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Canitano A, Papa M, Boscia F, Castaldo P, Sellitti S, Taglialatela M, & Annunziato L (2002). Brain distribution of the Na+/Ca2+ exchangerencoding genes NCX1, NCX2, and NCX3 and their related proteins in the central nervous system. Cellular and Molecular Physiology of Sodium-Calcium Exchange, 976, 394 Retrieved from <Go to ISI>://WOS:000180979900058. [DOI] [PubMed] [Google Scholar]
  35. Cao S, Standaert D, & Harms A (2012). The gamma chain subunit of fc receptors is required for alpha-synuclein-induced pro-inflammatory signaling in microglia. Journal of Neuroinflammation, 9, 259 10.1186/1742-2094-9-259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Casamassa A, La Rocca C, Sokolow S, Herchuelz A, Matarese G, Annunziato L, & Boscia F (2016). Ncx3 gene ablation impairs oligodendrocyte precursor response and increases susceptibility to experimental autoimmune encephalomyelitis. Glia, 64(7), 1124–1137. 10.1002/glia.22985 [DOI] [PubMed] [Google Scholar]
  37. Castaldo P, Cataldi M, Magi S, Lariccia V, Arcangeli S, & Amoroso S (2009). Role of the mitochondrial sodium/calcium exchanger in neuronal physiology and in the pathogenesis of neurological diseases. Progress in Neurobiology, 87(1), 58–79. 10.1016/j.pneurobio.2008.09.017 [DOI] [PubMed] [Google Scholar]
  38. Cekanaviciute E, Fathali N, Doyle KP, Williams AM, Han J, & Buckwalter MS (2014). Astrocytic transforming growth factor-beta signaling reduces subacute neuroinflammation after stroke in mice. Glia, 62(8), 1227–1240. 10.1002/glia.22675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cellot G, & Cherubini E (2013). Functional role of ambient GABA in refining neuronal circuits early in postnatal development. Frontiers in Neural Circuits, 7, 136 10.3389/fncir.2013.00136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cengiz P, Kintner DB, Chanana V, Yuan H, Akture E, Kendigelen P, … Sun D (2014). Sustained Na+/H+ exchanger activation promotes gliotransmitter release from reactive hippocampal astrocytes following oxygen-glucose deprivation. PLoS One, 9(1), e84294 10.1371/journal.pone.0084294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Chai H, Diaz-Castro B, Shigetomi E, Monte E, Octeau JC, Yu X, … Khakh BS (2017). Neural circuit-specialized astrocytes: Trans-criptomic, proteomic, morphological, and functional evidence. Neuron, 95(3), 531–549 e539 10.1016/j.neuron.2017.06.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Chatton JY, Magistretti PJ, & Barros LF (2016). Sodium signaling and astrocyte energy metabolism. Glia, 64(10), 1667–1676. 10.1002/glia.22971 [DOI] [PubMed] [Google Scholar]
  43. Chen H, Kintner DB, Jones M, Matsuda T, Baba A, Kiedrowski L, & Sun D (2007). AMPA-mediated excitotoxicity in oligodendrocytes: Role for Na(+)-K(+)-cl(−) co-transport and reversal of Na(+)/Ca(2+) exchanger. Journal of Neurochemistry, 102(6), 1783–1795. 10.1111/j.1471-4159.2007.04638.x [DOI] [PubMed] [Google Scholar]
  44. Chen H, Luo J, Kintner DB, Shull GE, & Sun D (2005). Na(+)-dependent chloride transporter (NKCC1)-null mice exhibit less gray and white matter damage after focal cerebral ischemia. Journal of Cerebral Blood Flow and Metabolism, 25(1), 54–66. 10.1038/sj.jcbfm.9600006 [DOI] [PubMed] [Google Scholar]
  45. Chen H, & Sun D (2005). The role of Na-K-Cl co-transporter in cerebral ischemia. Neurological Research, 27(3), 280–286. 10.1179/016164105X25243 [DOI] [PubMed] [Google Scholar]
  46. Chen T, Wu Y, Wang Y, Zhu J, Chu H, Kong L, … Ma H (2017). Brain-derived Neurotrophic factor increases synaptic protein levels via the MAPK/Erk signaling pathway and Nrf2/Trx Axis following the transplantation of neural stem cells in a rat model of traumatic brain injury. Neurochemical Research, 42(11), 3073–3083. 10.1007/s11064-017-2340-7 [DOI] [PubMed] [Google Scholar]
  47. Chen W, Wang D, Du X, He Y, Chen SY, Shao QQ, … Li XG (2015). Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Medical Oncology, 32(3), 43 10.1007/s12032-015-0487-0 [DOI] [PubMed] [Google Scholar]
  48. Chen W, Zhang X, & Huang W (2016). Role of neuroinflammation in neurodegenerative diseases (review). Molecular Medicine Reports, 13 (4), 3391–3396. 10.3892/mmr.2016.4948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Chesler M (2005). Failure and function of intracellular pH regulation in acute hypoxic-ischemic injury of astrocytes. Glia, 50(4), 398–406. 10.1002/glia.20141 [DOI] [PubMed] [Google Scholar]
  50. Choi B, Kim D, Back D, Kang C, Moon W, Han J, … Kim H (2016).Characterization of white matter injury in a rat model of chronic cerebral hypoperfusion. Stroke, 47(2), 542–547. 10.1161/STROKEAHA.115.011679 [DOI] [PubMed] [Google Scholar]
  51. Choi H, Gordon GRJ, Zhou N, Tai C, Rungta RL, Martinez J, … MacVicar BA (2012). Metabolic communication between astrocytes and neurons via bicarbonate-responsive soluble adenylyl cyclase. Neuron, 75(6), 1094–1104. 10.1016/j.neuron.2012.08.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Claus EB, Walsh KM, Wiencke JK, Molinaro AM, Wiemels JL, Schildkraut JM, … Wrensch M (2015). Survival and low-grade glioma: The emergence of genetic information. Neurosurgical Focus, 38 (1), E6 10.3171/2014.10.FOCUS12367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Cong D, Zhu W, Shi Y, Pointer KB, Clark PA, Shen H, … Sun D(2014). Upregulation of NHE1 protein expression enables glioblastoma cells to escape TMZ-mediated toxicity via increased H(+) extrusion, cell migration and survival. Carcinogenesis, 35(9), 2014–2024. 10.1093/carcin/bgu089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Coniglio SJ, & Segall JE (2013). Review: Molecular mechanism of microglia stimulated glioblastoma invasion. Matrix Biology, 32(7–8), 372–380. 10.1016/j.matbio.2013.07.008 [DOI] [PubMed] [Google Scholar]
  55. Correale J, & Farez MF (2015). The role of astrocytes in multiple sclerosis progression. Frontiers in Neurology, 6, 180 10.3389/fneur.2015.00180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Coulter DA, & Steinhauser C (2015). Role of astrocytes in epilepsy. Cold Spring Harbor Perspectives in Medicine, 5(3), a022434 10.1101/cshperspect.a022434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Craner MJ, Hains BC, Lo AC, Black JA, & Waxman SG (2004). Co-localization of sodium channel Na(v)1.6 and the sodium-calcium exchanger at sites of axonal injury in the spinal cord in EAE. Brain, 127, 294–303. 10.1093/brain/awh032 [DOI] [PubMed] [Google Scholar]
  58. de Rosa V, Secondo A, Pannaccione A, Ciccone R, Formisano L, Guida N, … Boscia F (2019). D-aspartate treatment attenuates myelin damage and stimulates myelin repair. EMBO Molecular Medicine, 11 (1), e9278 10.15252/emmm.201809278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Delgado-Lopez PD, Corrales-Garcia EM, Martino J, Lastra-Aras E, & Duenas-Polo MT (2017). Diffuse low-grade glioma: A review on the new molecular classification, natural history and current management strategies. Clinical & Translational Oncology, 19(8), 931–944. 10.1007/s12094-017-1631-4 [DOI] [PubMed] [Google Scholar]
  60. Deng YY, Shen FC, Xie D, Han QP, Fang M, Chen CB, & Zeng HK (2016). Progress in drug treatment of cerebral edema. Mini Reviews in Medicinal Chemistry, 16(11), 917–925 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/26948324. [DOI] [PubMed] [Google Scholar]
  61. Diniz LP, Matias I, Siqueira M, Stipursky J, & Gomes FCA (2018). Astrocytes and the TGF-beta1 pathway in the healthy and diseased brain: A double-edged sword. Molecular Neurobiology, 56, 4653–4679. 10.1007/s12035-018-1396-y [DOI] [PubMed] [Google Scholar]
  62. Domingues HS, Cruz A, Chan JR, Relvas JB, Rubinstein B, & Pinto IM (2018). Mechanical plasticity during oligodendrocyte differentiation and myelination. Glia, 66(1), 5–14. 10.1002/glia.23206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Doyle S, Hansen DB, Vella J, Bond P, Harper G, Zammit C, … Fern R (2018). Vesicular glutamate release from central axons contributes to myelin damage. Nature Communications, 9(1), 1032 10.1038/s41467-018-03427-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Dulamea AO (2017). Role of oligodendrocyte dysfunction in demyelination, remyelination and neurodegeneration in multiple sclerosis. Advances in Experimental Medicine and Biology, 958, 91–127. 10.1007/978-3-319-47861-6_7 [DOI] [PubMed] [Google Scholar]
  65. Erdo F, Denes L, & de Lange E (2017). Age-associated physiological and pathological changes at the blood-brain barrier: A review. Journal of Cerebral Blood Flow and Metabolism, 37(1), 4–24. 10.1177/0271678x16679420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Ernest NJ, & Sontheimer H (2007). Extracellular glutamine is a critical modulator for regulatory volume increase in human glioma cells. Brain Research, 1144, 231–238. 10.1016/j.brainres.2007.01.085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Ernest NJ, Weaver AK, Van Duyn LB, & Sontheimer HW (2005). Relative contribution of chloride channels and transporters to regulatory volume decrease in human glioma cells. American Journal of Physiology-Cell Physiology, 288(6), C1451–C1460. 10.1152/ajpcell.00503.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Ettle B, Schlachetzki JCM, & Winkler J (2016). Oligodendroglia and myelin in neurodegenerative diseases: More than just bystanders? Molecular Neurobiology, 53(5), 3046–3062. 10.1007/s12035-015-9205-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Fakhoury M (2018). Microglia and astrocytes in Alzheimer’s disease: Implications for therapy. Current Neuropharmacology, 16(5), 508–518. 10.2174/1570159X15666170720095240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Florence CM, Baillie LD, & Mulligan SJ (2012). Dynamic volume changes in astrocytes are an intrinsic phenomenon mediated by bicarbonate ion flux. PLoS One, 7(11), e51124 10.1371/journal.pone.0051124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Friess M, Hammann J, Unichenko P, Luhmann HJ, White R, & Kirischuk S (2016). Intracellular ion signaling influences myelin basic protein synthesis in oligodendrocyte precursor cells. Cell Calcium, 60 (5), 322–330. 10.1016/j.ceca.2016.06.009 [DOI] [PubMed] [Google Scholar]
  72. Fu PC, Tang RH, Yu ZY, Huang SS, Xie MJ, Luo X, & Wang W (2015). Bumetanide-induced NKCC1 inhibition attenuates oxygen-glucose deprivation-induced decrease in proliferative activity and cell cycle progression arrest in cultured OPCs via p-38 MAPKs. Brain Research, 1613, 110–119. 10.1016/j.brainres.2015.04.006 [DOI] [PubMed] [Google Scholar]
  73. Galindo LT, Mundim MTVV, Pinto AS, Chiarantin GMD, Almeida MES, Lamers ML, … Porcionatto M (2018). Chondroitin sulfate impairs neural stem cell migration through ROCK activation. Molecular Neurobiology, 55(4), 3185–3195. 10.1007/s12035-017-0565-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Gandhi S, Wood-Kaczmar A, Yao Z, Plun-Favreau H, Deas E, Klupsch K, … Abramov AY (2009). PINK1-associated Parkinson’s disease is caused by neuronal vulnerability to calcium-induced cell death. Molecular Cell, 33(5), 627–638. 10.1016/j.molcel.2009.02.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Gerkau NJ, Rakers C, Durry S, Petzold GC, & Rose CR (2018). Reverse NCX attenuates cellular sodium loading in metabolically compromised cortex. Cerebral Cortex, 28(12), 4264–4280. 10.1093/cercor/bhx280 [DOI] [PubMed] [Google Scholar]
  76. Gibson EM, Purger D, Mount CW, Goldstein AK, Lin GL, Wood LS, … Monje M (2014). Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science, 344(6183), 1252304 10.1126/science.1252304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Giffard RG, Papadopoulos MC, van Hooft JA, Xu LJ, Giuffrida R, & Monyer H (2000). The electrogenic sodium bicarbonate cotransporter: Developmental expression in rat brain and possible role in acid vulnerability. Journal of Neuroscience, 20(3), 1001–1008. 10.1523/Jneurosci.20-03-01001.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Gobbi P, Castaldo P, Minelli A, Salucci S, Magi S, Corcione E, & Amoroso S (2007). Mitochondrial localization of Na+/Ca2+ exchangers NCX1–3 in neurons and astrocytes of adult rat brain in situ. Pharmacological Research, 56(6), 556–565. 10.1016/j.phrs.2007.10.005 [DOI] [PubMed] [Google Scholar]
  79. Golovina V, Song H, James P, Lingrel J, & Blaustein M (2003). Regulation of Ca2+ signaling by Na+ pump alpha-2 subunit expression. Annals of the New York Academy of Sciences, 986, 509–513 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12763872. [DOI] [PubMed] [Google Scholar]
  80. Gu XL, Long CX, Sun L, Xie C, Lin X, & Cai H (2010). Astrocytic expression of Parkinson’s disease-related A53T alpha-synuclein causes neurodegeneration in mice. Molecular Brain, 3, 12 10.1186/1756-6606-3-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Guan X, Hasan MN, Begum G, Kohanbash G, Carney KE, Pigott VM, … Sun D (2018). Blockade of Na/H exchanger stimulates glioma tumor immunogenicity and enhances combinatorial TMZ and anti-PD-1 therapy. Cell Death & Disease, 9(10), 1010 10.1038/s41419-018-1062-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Guan X, Hasan MN, Maniar S, Jia W, & Sun D (2018). Reactive astrocytes in glioblastoma multiforme. Molecular Neurobiology, 55(8), 6927–6938. 10.1007/s12035-018-0880-8 [DOI] [PubMed] [Google Scholar]
  83. Guan X, Luo L, Begum G, Kohanbash G, Song QK, Rao A, … Jia W(2018). Elevated Na/H exchanger 1 (SLC9A1) emerges as a marker for tumorigenesis and prognosis in gliomas. Journal of Experimental & Clinical Cancer Research, 37, 255 10.1186/s13046-018-0923-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Guerrero-Garcia JJ (2017). The role of astrocytes in multiple sclerosis pathogenesis. Neurologia. 10.1016/j.nrl.2017.07.021 [DOI] [PubMed] [Google Scholar]
  85. Guo JD, Zhao X, Li Y, Li GR, & Liu XL (2018). Damage to dopaminergic neurons by oxidative stress in Parkinson’s disease (review). International Journal of Molecular Medicine, 41(4), 1817–1825. 10.3892/ijmm.2018.3406 [DOI] [PubMed] [Google Scholar]
  86. Habela CW, Ernest NJ, Swindall AF, & Sontheimer H (2009). Chloride accumulation drives volume dynamics underlying cell proliferation and migration. Journal of Neurophysiology, 101(2), 750–757. 10.1152/jn.90840.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Halliday GM, & Stevens CH (2011). Glia: Initiators and progressors of pathology in Parkinson’s disease. Movement Disorders, 26(1), 6–17. 10.1002/mds.23455 [DOI] [PubMed] [Google Scholar]
  88. Hammann J, Bassetti D, White R, Luhmann HJ, & Kirischuk S (2018). alpha2 isoform of Na(+),K(+)-ATPase via Na(+),Ca(2+) exchanger modulates myelin basic protein synthesis in oligodendrocyte lineage cells in vitro. Cell Calcium, 73, 1–10. 10.1016/j.ceca.2018.03.003 [DOI] [PubMed] [Google Scholar]
  89. Hannibal L (2016). Nitric oxide homeostasis in neurodegenerative diseases. Current Alzheimer Research, 13(2), 135–149 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/26391043. [DOI] [PubMed] [Google Scholar]
  90. Hertz L, & Chen Y (2016). Importance of astrocytes for potassium ion (K (+)) homeostasis in brain and glial effects of K(+) and its transporters on learning. Neuroscience and Biobehavioral Reviews, 71, 484–505. 10.1016/j.neubiorev.2016.09.018 [DOI] [PubMed] [Google Scholar]
  91. Hertz L, Song D, Xu JN, Peng L, & Gibbs ME (2015). Role of the astrocytic Na+, K+-ATPase in K+ homeostasis in brain: K+ uptake, signaling pathways and substrate utilization. Neurochemical Research, 40 (12), 2505–2516. 10.1007/s11064-014-1505-x [DOI] [PubMed] [Google Scholar]
  92. Hertz L, Xu J, Chen Y, Gibbs ME, Du T, Hertz L, … Du T (2014).Antagonists of the vasopressin V1 receptor and of the beta(1)-adrenoceptor inhibit cytotoxic brain Edema in stroke by effects on astrocytes - but the mechanisms differ. Current Neuropharmacology, 12(4), 308–323. 10.2174/1570159X12666140828222723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Hertz L, Xu JN, Song D, Yan EZ, Gu L, & Peng L (2013). Astrocytic and neuronal accumulation of elevated extracellular K+ with a 2/3 K+/Na+ flux ratio-consequences for energy metabolism, osmolarity and higher brain function. Frontiers in Computational Neuroscience, 7, 114 10.3389/fncom.2013.00114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Hide T, Shibahara I, & Kumabe T (2019). Novel concept of the border niche: Glioblastoma cells use oligodendrocytes progenitor cells (GAOs) and microglia to acquire stem cell-like features. Brain Tumor Pathology, 36, 63–73. 10.1007/s10014-019-00341-2 [DOI] [PubMed] [Google Scholar]
  95. Holley JE, Gveric D, Newcombe J, Cuzner ML, & Gutowski NJ (2003). Astrocyte characterization in the multiple sclerosis glial scar. Neuropathology and Applied Neurobiology, 29(5), 434–444 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/14507335. [DOI] [PubMed] [Google Scholar]
  96. Huang C, Lu X, Wang J, Tong LJ, Jiang B, & Zhang W (2015). Inhibition of endogenous heat shock protein 70 attenuates inducible nitric oxide synthase induction via disruption of heat shock protein 70/Na+/H+ exchanger 1-Ca2+-calcium-calmodulin-dependent protein kinase II/transforming growth factor beta-activated kinase 1-nuclear factor-kappa B signals in BV-2 microglia. Journal of Neuroscience Research, 93(8), 1192–1202. 10.1002/jnr.23571 [DOI] [PubMed] [Google Scholar]
  97. Huang H, Bhuiyan MIH, Jiang T, Song S, Shankar S, Taheri T, … Sun D (2019). A novel Na(+)-K(+)-cl(−) cotransporter 1 inhibitor STS66* reduces brain damage in mice after ischemic stroke. Stroke, 50 (4), 1021–1025. 10.1161/STROKEAHA.118.024287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Hughes EG, Kang SH, Fukaya M, & Bergles DE (2013). Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nature Neuroscience, 16(6), 668–676. 10.1038/nn.3390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hwang IK, Yoo KY, Kim DW, Kang TC, Choi SY, Kwon YG, … Won MH (2006). Na+/Ca2+ exchanger 1 alters in pyramidal cells and expresses in astrocytes of the gerbil hippocampal CA1 region after ischemia. Brain Research, 1086(1), 181–190. 10.1016/j.brainres.2006.02.048 [DOI] [PubMed] [Google Scholar]
  100. Ibanez I, Bartolome-Martin D, Piniella D, Gimenez C, & Zafra F (2019). Activity dependent internalization of the glutamate transporter GLT-1 requires calcium entry through the NCX sodium/calcium exchanger. Neurochemistry International, 123, 125–132. 10.1016/j.neuint.2018.03.012 [DOI] [PubMed] [Google Scholar]
  101. Ifuku M, Farber K, Okuno Y, Yamakawa Y, Miyamoto T, Nolte C, … Noda M (2007). Bradykinin-induced microglial migration mediated by B1-bradykinin receptors depends on Ca2+ influx via reverse-mode activity of the Na+/Ca2+ exchanger. The Journal of Neuroscience, 27(48), 13065–13073. 10.1523/JNEUROSCI.3467-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Ilkhanizadeh S, Sabelstrom H, Miroshnikova YA, Frantz A, Zhu W, Idilli A, … Persson AI (2018). Antisecretory factor-mediated inhibition of cell volume dynamics produces antitumor activity in glioblastoma. Molecular Cancer Research, 16(5), 777–790. 10.1158/1541-7786.MCR-17-0413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Jantzie LL, Hu MY, Park HK, Jackson MC, Yu J, Maxwell JR, & Jensen FE (2015). Chloride cotransporter NKCC1 inhibitor bumetanide protects against white matter injury in a rodent model of periventricular leukomalacia. Pediatric Research, 77(4), 554–562. 10.1038/pr.2015.9 [DOI] [PubMed] [Google Scholar]
  104. Jassam YN, Izzy S, Whalen M, McGavern DB, & El Khoury J (2017). Neuroimmunology of traumatic brain injury: Time for a paradigm shift. Neuron, 95(6), 1246–1265. 10.1016/j.neuron.2017.07.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Jayakumar AR, Panickar KS, Curtis KM, Tong XY, Moriyama M, & Norenberg MD (2011). Na-K-Cl cotransporter-1 in the mechanism of cell swelling in cultured astrocytes after fluid percussion injury. Journal of Neurochemistry, 117(3), 437–448. 10.1111/j.1471-4159.2011.07211.x [DOI] [PubMed] [Google Scholar]
  106. Jayakumar AR, Taherian M, Panickar KS, Shamaladevi N, Rodriguez ME, Price BG, & Norenberg MD (2018). Differential response of neural cells to trauma-induced swelling in vitro. Neurochemical Research, 43(2), 397–406. 10.1007/s11064-017-2434-2 [DOI] [PubMed] [Google Scholar]
  107. Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, … Suk K (2018). Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Progress in Neurobiology, 162, 37–69. 10.1016/j.pneurobio.2017.12.003 [DOI] [PubMed] [Google Scholar]
  108. Joers V, Tansey MG, Mulas G, & Carta AR (2017). Microglial phenotypes in Parkinson’s disease and animal models of the disease. Progress in Neurobiology, 155, 57–75. 10.1016/j.pneurobio.2016.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Jung YW, Choi IJ, & Kwon TH (2007). Altered expression of sodium transporters in ischemic penumbra after focal cerebral ischemia in rats. Neuroscience Research, 59(2), 152–159. 10.1016/j.neures.2007.06.1470 [DOI] [PubMed] [Google Scholar]
  110. Kahle KT, Simard JM, Staley KJ, Nahed BV, Jones PS, & Sun D (2009). Molecular mechanisms of ischemic cerebral edema: Role of electroneutral ion transport. Physiology (Bethesda), 24, 257–265. 10.1152/physiol.00015.2009 [DOI] [PubMed] [Google Scholar]
  111. Kang S, Keasey M, Cai J, & Hagg T (2012). Loss of neuron-astroglial interaction rapidly induces protective CNTF expression after stroke in mice. The Journal of Neuroscience, 32(27), 9277–9287. 10.1523/JNEUROSCI.1746-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Kang T, An S, Park S, Hwang I, Suh J, Oh Y, … Won M (2002). Alterations in Na+/H+ exchanger and Na+/HCO3− cotransporter immunoreactivities within the gerbil hippocampus following seizure. Molecular Brain Research, 109(1–2), 226–232. 10.1016/S0169-328x(02)00559-4 [DOI] [PubMed] [Google Scholar]
  113. Khananshvili D (2013). The SLC8 gene family of sodium-calcium exchangers (NCX)--Structure, function, and regulation in health and disease. Molecular Aspects of Medicine, 34(2–3), 220–235. 10.1016/j.mam.2012.07.003 [DOI] [PubMed] [Google Scholar]
  114. Kim SK, Nabekura J, & Koizumi S (2017). Astrocyte-mediated synapse remodeling in the pathological brain. Glia, 65(11), 1719–1727. 10.1002/glia.23169 [DOI] [PubMed] [Google Scholar]
  115. Kintner DB, Look A, Shull GE, & Sun DD (2005). Stimulation of astrocyte Na+/H+ exchange activity in response to in vitro ischemia depends in part on activation of ERK1/2. American Journal of Physiology-Cell Physiology, 289(4), C934–C945. 10.1152/ajpcell.00092.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Kintner DB, Luo J, Gerdts J, Ballard AJ, Shull GE, & Sun D (2007). Role of Na+–K+–Cl cotransport and Na+/Ca2+ exchange in mitochondrial dysfunction in astrocytes following in vitro ischemia. American Journal of Physiology-Cell Physiology, 292(3), C1113–C1122. 10.1152/ajpcell.00412.2006 [DOI] [PubMed] [Google Scholar]
  117. Kintner DB, Su G, Lenart B, Ballard AJ, Meyer JW, Ng LL, … Sun DD (2004). Increased tolerance to oxygen and glucose deprivation in astrocytes from Na+/H+ exchanger isoform 1 null mice. American Journal of Physiology-Cell Physiology, 287(1), C12–C21. 10.1152/ajpcell.00560.2003 [DOI] [PubMed] [Google Scholar]
  118. Kitao T, Takuma K, Kawasaki T, Inoue Y, Ikehara A, Nashida T, … Matsuda T (2010). The Na+/Ca2+ exchanger-mediated Ca2+ influx triggers nitric oxide-induced cytotoxicity in cultured astrocytes. Neurochemistry International, 57(1), 58–66. 10.1016/j.neuint.2010.04.016 [DOI] [PubMed] [Google Scholar]
  119. Kohanbash G, Carrera DA, Shrivastav S, Ahn BJ, Jahan N, Mazor T, … Okada H (2017). Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. The Journal of Clinical Investigation, 127(4), 1425–1437. 10.1172/JCI90644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Lam TI, Brennan-Minnella AM, Won SJ, Shen YG, Hefner C, Shi YJ, … Swanson RA (2013). Intracellular pH reduction prevents excitotoxic and ischemic neuronal death by inhibiting NADPH oxidase. Proceedings of the National Academy of Sciences of the United States of America, 110(46), E4362–E4368. 10.1073/pnas.1313029110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Larsen BR, Assentoft M, Cotrina ML, Hua SZ, Nedergaard M, Kaila K, MacAulay N (2014). Contributions of the Na+/K+-ATPase, NKCC1, and Kir4.1 to hippocampal K+ clearance and volume responses. Glia, 62(4), 608–622. 10.1002/glia.22629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Larsen BR, & MacAulay N (2017). Activity-dependent astrocyte swelling is mediated by pH-regulating mechanisms. Glia, 65(10), 1668–1681. 10.1002/glia.23187 [DOI] [PubMed] [Google Scholar]
  123. Lee Y, Kim MS, & Lee J (2017). Neuroprotective strategies to prevent and treat Parkinson’s disease based on its pathophysiological mechanism. Archives of Pharmacal Research, 40(10), 1117–1128. 10.1007/s12272-017-0960-8 [DOI] [PubMed] [Google Scholar]
  124. Lees AJ, Hardy J, & Revesz T (2009). Parkinson’s disease. Lancet, 373 (9680), 2055–2066. 10.1016/S0140-6736(09)60492-X [DOI] [PubMed] [Google Scholar]
  125. Lemus HN, Warrington AE, & Rodriguez M (2018). Multiple sclerosis: mechanisms of disease and strategies for myelin and axonal repair. Neurologic Clinics, 36(1), 1–11. 10.1016/j.ncl.2017.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Lenart B, Kintner DB, Shull GE, & Sun D (2004). Na-K-Cl cotransporter-mediated intracellular Na+ accumulation affects Ca2+ signaling in astrocytes in an in vitro ischemic model. The Journal of Neuroscience, 24(43), 9585–9597. 10.1523/JNEUROSCI.2569-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Li W, & Graeber MB (2012). The molecular profile of microglia under the influence of glioma. Neuro-Oncology, 14(8), 958–978. 10.1093/neuonc/nos116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Lim D, Iyer A, Ronco V, Grolla AA, Canonico PL, Aronica E, & Genazzani AA (2013). Amyloid beta deregulates astroglial mGluR5-mediated calcium signaling via calcineurin and Nf-kB. Glia, 61 (7), 1134–1145. 10.1002/glia.22502 [DOI] [PubMed] [Google Scholar]
  129. Lin Q, Liu Z, Ling F, & Xu G (2016). Astrocytes protect glioma cells from chemotherapy and upregulate survival genes via gap junctional communication. Molecular Medicine Reports, 13(2), 1329–1335. 10.3892/mmr.2015.4680 [DOI] [PubMed] [Google Scholar]
  130. Liu Y, Kintner DB, Chanana V, Algharabli J, Chen X, Gao Y, … Sun D (2010). Activation of microglia depends on Na+/H+ exchange-mediated H+ homeostasis. The Journal of Neuroscience, 30(45), 15210–15220. 10.1523/JNEUROSCI.3950-10.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Liu Z, & Chopp M (2016). Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Progress in Neurobiology, 144, 103–120. 10.1016/j.pneurobio.2015.09.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Lopez-Alvarez VM, Modol L, Navarro X, & Cobianchi S (2015). Early increasing-intensity treadmill exercise reduces neuropathic pain by preventing nociceptor collateral sprouting and disruption of chloride cotransporters homeostasis after peripheral nerve injury. Pain, 156(9), 1812–1825. 10.1097/j.pain.0000000000000268 [DOI] [PubMed] [Google Scholar]
  133. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, … Ellison DW (2016). The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathologica, 131(6), 803–820. 10.1007/s00401-016-1545-1 [DOI] [PubMed] [Google Scholar]
  134. Ludwin SK, Rao VTS, Moore CS, & Antel JP (2017). Astrocytes in multiple sclerosis. Multiple Sclerosis Journal, 22(9), 1114–1124. [DOI] [PubMed] [Google Scholar]
  135. Ma H, Li T, Tao Z, Hai L, Tong L, Yi L, … Yang X (2019). NKCC1 promotes EMT-like process in GBM via RhoA and Rac1 signaling pathways. Journal of Cellular Physiology, 234(2), 1630–1642. 10.1002/jcp.27033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Ma Y, Wang J, Wang Y, & Yang GY (2017). The biphasic function of microglia in ischemic stroke. Progress in Neurobiology, 157, 247–272. 10.1016/j.pneurobio.2016.01.005 [DOI] [PubMed] [Google Scholar]
  137. MacVicar BA, Feighan D, Btrn A, & Ransom B (2002). Intrinsic optical signals in the rat optic nerve: Role for K+ uptake via NKCC1 and swelling of astrocytes. Glia, 37(2), 114–123. 10.1002/glia.10023 [DOI] [PubMed] [Google Scholar]
  138. Magalhaes AC, & Rivera C (2016). NKCC1-deficiency results in abnormal proliferation of neural progenitor cells of the lateral ganglionic eminence. Frontiers in Cellular Neuroscience, 10, 200 10.3389/fncel.2016.00200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Matias D, Balca-Silva J, da Graca GC, Wanjiru CM, Macharia LW, Nascimento CP, … Moura-Neto V (2018). Microglia/astrocytes-glioblastoma crosstalk: Crucial molecular mechanisms and microenvironmental factors. Frontiers in Cellular Neuroscience, 12, 235 10.3389/fncel.2018.00235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Matsuda T, Arakawa N, Takuma K, Kishida Y, Kawasaki Y, Sakaue M, … Baba A (2001). SEA0400, a novel and selective inhibitor of the Na+-Ca2+ exchanger, attenuates reperfusion injury in the in vitro and in vivo cerebral ischemic models. Journal of Pharmacology and Experimental Therapeutics, 298(1), 249–256. [PubMed] [Google Scholar]
  141. Matsuda T, & Baba A (1998). Response of Na+/Ca2+ antiporter to ischemia and glial/neuronal death. Nihon Yakurigaku Zasshi, 111(1), 13–19 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/9551468. [DOI] [PubMed] [Google Scholar]
  142. Matsuda T, Nagano T, Takemura M, & Baba A (2006). Topics on the Na+/Ca2+ exchanger: Responses of Na+/Ca2+ exchanger to interferon-gamma and nitric oxide in cultured microglia. Journal of Pharmacological Sciences, 102(1), 22–26. 10.1254/jphs.FMJ06002X4 [DOI] [PubMed] [Google Scholar]
  143. Micu I, Plemel JR, Lachance C, Proft J, Jansen AJ, Cummins K, … Stys PK (2016). The molecular physiology of the axo-myelinic synapse. Experimental Neurology, 276, 41–50. 10.1016/j.expneurol.2015.10.006 [DOI] [PubMed] [Google Scholar]
  144. Miller IS, Didier S, Murray DW, Turner TH, Issaivanan M, Ruggieri R, … Symons M (2014). Semapimod sensitizes glioblastoma tumors to ionizing radiation by targeting microglia. PLoS One, 9(5), e95885 10.1371/journal.pone.0095885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Minelli A, Castaldo P, Gobbi P, Salucci S, Magi S, & Amoroso S (2007). Cellular and subcellular localization of Na+-Ca2+ exchanger protein isoforms, NCX1, NCX2, and NCX3 in cerebral cortex and hippocampus of adult rat. Cell Calcium, 41(3), 221–234. 10.1016/j.ceca.2006.06.004 [DOI] [PubMed] [Google Scholar]
  146. Mishra AM, Pal A, Gupta D, & Carmel JB (2017). Paired motor cortex and cervical epidural electrical stimulation timed to converge in the spinal cord promotes lasting increases in motor responses. The Journal of Physiology, 595(22), 6953–6968. 10.1113/JP274663 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Miyamoto A, Wake H, Ishikawa AW, Eto K, Shibata K, Murakoshi H, … Nabekura J (2016). Microglia contact induces synapse formation in developing somatosensory cortex. Nature Communications, 7, 12540 10.1038/ncomms12540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Miyamoto N, Maki T, Shindo A, Liang AC, Maeda M, Egawa N, … Arai K (2015). Astrocytes promote oligodendrogenesis after white matter damage via brain-derived neurotrophic factor. The Journal of Neuroscience, 35(41), 14002–14008. 10.1523/JNEUROSCI.1592-15.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Molenaar RJ (2011). Ion channels in glioblastoma. ISRN Neurology, 2011, 590247–590249. 10.5402/2011/590249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Mori Y, Tomonaga D, Kalashnikova A, Furuya F, Akimoto N, Ifuku M, … Noda M (2015). Effects of 3,3’,5-triiodothyronine on microglial functions. Glia, 63(5), 906–920. 10.1002/glia.22792 [DOI] [PubMed] [Google Scholar]
  151. Murakami S, & Kurachi Y (2016). Mechanisms of astrocytic K(+) clearance and swelling under high extracellular K(+) concentrations. The Journal of Physiological Sciences, 66(2), 127–142. 10.1007/s12576-015-0404-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Murayi R, & Chittiboina P (2016). Glucocorticoids in the management of peritumoral brain edema: A review of molecular mechanisms. Child’s Nervous System, 32(12), 2293–2302. 10.1007/s00381-016-3240-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Nagano T, Kawasaki Y, Baba A, Takemura M, & Matsuda T (2004). Up-regulation of Na(+)-Ca2+ exchange activity by interferon-gamma in cultured rat microglia. Journal of Neurochemistry, 90(4), 784–791. 10.1111/j.1471-4159.2004.02511.x [DOI] [PubMed] [Google Scholar]
  154. Nagano T, Osakada M, Ago Y, Koyama Y, Baba A, Maeda S, … Matsuda T (2005). SEA0400, a specific inhibitor of the Na+-Ca2+ exchanger, attenuates sodium nitroprusside-induced apoptosis in cultured rat microglia. British Journal of Pharmacology, 144(5), 669–679. 10.1038/sj.bjp.0706104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Newell EW, Stanley EF, & Schlichter LC (2007). Reversed Na+/Ca2+ exchange contributes to Ca2+ influx and respiratory burst in microglia. Channels (Austin, Tex.), 1(5), 366–376. 10.4161/chan.5391 [DOI] [PubMed] [Google Scholar]
  156. Nishioka R, Sugimoto K, Aono H, Mise A, Choudhury ME, Miyanishi K, … Tanaka J (2016). Treadmill exercise ameliorates ischemia-induced brain edema while suppressing Na+/H+ exchanger 1 expression. Experimental Neurology, 277, 150–161. 10.1016/j.expneurol.2015.12.016 [DOI] [PubMed] [Google Scholar]
  157. Noda M, Ifuku M, Mori Y, & Verkhratsky A (2013). Calcium influx through reversed NCX controls migration of microglia. Advances in Experimental Medicine and Biology, 961, 289–294. 10.1007/978-1-4614-4756-6_24 [DOI] [PubMed] [Google Scholar]
  158. Nordstrom T, Andersson LC, & Akerman KEO (2019). Regulation of intracellular pH by electrogenic Na+/HCO3− co-transporters in embryonic neural stem cell-derived radial glia-like cells. Biochimica et Biophysica Acta - Biomembranes, 1861(6), 1037–1048. 10.1016/j.bbamem.2019.03.007 [DOI] [PubMed] [Google Scholar]
  159. O’Brien ER, Howarth C, & Sibson NR (2013). The role of astrocytes in CNS tumors: Pre-clinical models and novel imaging approaches. Frontiers in Cellular Neuroscience, 7, 40 10.3389/fncel.2013.00040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Oberheim Bush NA, & Chang S (2016). Treatment strategies for low-grade Glioma in adults. Journal of Oncology Practice/American Society of Clinical Oncology, 12(12), 1235–1241. 10.1200/JOP.2016.018622 [DOI] [PubMed] [Google Scholar]
  161. Okolie O, Bago JR, Schmid RS, Irvin DM, Bash RE, Miller CR, & Hingtgen SD (2016). Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model. Neuro-Oncology, 18(12), 1622–1633. 10.1093/neuonc/now117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Olah M, Amor S, Brouwer N, Vinet J, Eggen B, Biber K, & Boddeke HWGM (2012). Identification of a microglia phenotype supportive of remyelination. Glia, 60(2), 306–321. 10.1002/glia.21266 [DOI] [PubMed] [Google Scholar]
  163. Oliveira AI, Anjo SI, Vieira de Castro J, Serra SC, Salgado AJ, Manadas B, & Costa BM (2017). Crosstalk between glial and glioblastoma cells triggers the “go-or-gtr” phenotype of tumor cells. Cell Communication and Signaling, 15(1), 37 10.1186/s12964-017-0194-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Orlowski J, & Grinstein S (2004). Diversity of the mammalian sodium/proton exchanger SLC9 gene family. Pflugers Archiv, 447(5), 549–565. 10.1007/s00424-003-1110-3 [DOI] [PubMed] [Google Scholar]
  165. Ostby I, Oyehaug L, Einevoll GT, Nagelhus EA, Plahte E, Zeuthen T, … Omholt SW (2009). Astrocytic mechanisms explaining neural-activity-induced shrinkage of Extraneuronal space. PLoS Computational Biology, 5(1), e1000272 10.1371/journal.pcbi.1000272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Papa M, Canitano A, Boscia F, Castaldo P, Sellitti S, Porzig H, … Annunziato L (2003). Differential expression of the Na+-Ca2+ exchanger transcripts and proteins in rat brain regions. The Journal of Comparative Neurology, 461(1), 31–48. 10.1002/cne.10665 [DOI] [PubMed] [Google Scholar]
  167. Pappalardo LW, Black JA, & Waxman SG (2016). Sodium channels in Astroglia and microglia. Glia, 64(10), 1628–1645. 10.1002/glia.22967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Pappalardo LW, Samad OA, Black JA, & Waxman SG (2014). Voltage-gated sodium channel Na(v)1.5 contributes to astrogliosis in an in vitro model of glial injury via reverse Na+/Ca2+ exchange. Glia, 62(7), 1162–1175. 10.1002/glia.22671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR, Lafaille JJ, … Gan WB (2013). Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell, 155(7), 1596–1609. 10.1016/j.cell.2013.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Parpura V, Sekler I, & Fern R (2016). Plasmalemmal and mitochondrial Na(+)-Ca(2+) exchange in neuroglia. Glia, 64(10), 1646–1654. 10.1002/glia.22975 [DOI] [PubMed] [Google Scholar]
  171. Pekny M, Pekna M, Messing A, Steinhauser C, Lee JM, Parpura V, … Verkhratsky A (2016). Astrocytes: A central element in neurological diseases. Acta Neuropathologica, 131(3), 323–345. 10.1007/s00401-015-1513-1 [DOI] [PubMed] [Google Scholar]
  172. Phatnani H, & Maniatis T (2015). Astrocytes in neurodegenerative disease. Cold Spring Harbor Perspectives in Biology, 7(6), a020628 10.1101/cshperspect.a020628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Placone AL, Quinones-Hinojosa A, & Searson PC (2016). The role of astrocytes in the progression of brain cancer: Complicating the picture of the tumor microenvironment. Tumour Biology, 37(1), 61–69. 10.1007/s13277-015-4242-0 [DOI] [PubMed] [Google Scholar]
  174. Plemel JR, Liu WQ, & Yong VW (2017). Remyelination therapies: A new direction and challenge in multiple sclerosis. Nature Reviews. Drug Discovery, 16(9), 617–634. 10.1038/nrd.2017.115 [DOI] [PubMed] [Google Scholar]
  175. Puig B, Brenna S, & Magnus T (2018). Molecular communication of a dying neuron in stroke. International Journal of Molecular Sciences, 19 (9), 2834 10.3390/ijms19092834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Quednau BD, Nicoll DA, & Philipson KD (1997). Tissue specificity and alternative splicing of the Na+/Ca2+ exchanger isoforms NCX1, NCX2, and NCX3 in rat. The American Journal of Physiology, 272(4 Pt 1), C1250–C1261. 10.1152/ajpcell.1997.272.4.C1250 [DOI] [PubMed] [Google Scholar]
  177. Raimondo JV, Tomes H, Irkle A, Kay L, Kellaway L, Markram H, … Akerman CJ (2016). Tight coupling of astrocyte pH dynamics to Epileptiform activity revealed by genetically encoded pH sensors. Journal of Neuroscience, 36(26), 7002–7013. 10.1523/Jneurosci.0664-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Rakers C, Schleif M, Blank N, Matuskova H, Ulas T, Handler K, … Petzold GC (2019). Stroke target identification guided by astrocyte transcriptome analysis. Glia, 67(4), 619–633. 10.1002/glia.23544 [DOI] [PubMed] [Google Scholar]
  179. Ramirez AI, de Hoz R, Salobrar-Garcia E, Salazar JJ, Rojas B, Ajoy D, … Ramirez JM (2017). The role of microglia in retinal neurodegeneration: Alzheimer’s disease, Parkinson, and glaucoma. Frontiers in Aging Neuroscience, 9, 214 10.3389/fnagi.2017.00214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Rao KV, Reddy PV, Curtis KM, & Norenberg MD (2011). Aquaporin-4 expression in cultured astrocytes after fluid percussion injury. Journal of Neurotrauma, 28(3), 371–381. 10.1089/neu.2010.1705 [DOI] [PubMed] [Google Scholar]
  181. Rath BH, Fair JM, Jamal M, Camphausen K, & Tofilon PJ (2013). Astrocytes enhance the invasion potential of Glioblastoma stem-like cells. PLoS One, 8(1), e54752 10.1371/journal.pone.0054752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Reshef R, Kudryavitskaya E, Shani-Narkiss H, Isaacson B, Rimmerman N, Mizrahi A, & Yirmiya R (2017). The role of microglia and their CX3CR1 signaling in adult neurogenesis in the olfactory bulb. eLife, 6, e30809 10.7554/eLife.30809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Reyes RC, Verkhratsky A, & Parpura V (2012). Plasmalemmal Na+/Ca2+ exchanger modulates Ca2+-dependent exocytotic release of glutamate from rat cortical astrocytes. ASN Neuro, 4(1), AN20110059. 10.1042/AN20110059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Rinaldi M, Thomas L, Mathieu P, Carabias P, Troncoso MF, Pasquini JM, … Pasquini LA (2016). Galectin-1 circumvents lysolecithin-induced demyelination through the modulation of microglial polarization/phagocytosis and oligodendroglial differentiation. Neurobiology of Disease, 96, 127–143. 10.1016/j.nbd.2016.09.003 [DOI] [PubMed] [Google Scholar]
  185. Ro HA, & Carson JH (2004). pH microdomains in oligodendrocytes. Journal of Biological Chemistry, 279(35), 37115–37123. 10.1074/jbc.M403099200 [DOI] [PubMed] [Google Scholar]
  186. Robel S, Buckingham SC, Boni JL, Campbell SL, Danbolt NC, Riedemann T, … Sontheimer H (2015). Reactive astrogliosis causes the development of spontaneous seizures. The Journal of Neuroscience, 35 (8), 3330–3345. 10.1523/JNEUROSCI.1574-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Rogers J, Mastroeni D, Leonard B, Joyce J, & Grover A (2007). Neuroinflammation in Alzheimer’s disease and Parkinson’s disease: Are microglia pathogenic in either disorder? International Review of Neurobiology, 82, 235–246. [DOI] [PubMed] [Google Scholar]
  188. Rose CR, & Karus C (2013). Two sides of the same coin: Sodium homeostasis and signaling in astrocytes under physiological and pathophysiological conditions. Glia, 61(8), 1191–1205. 10.1002/glia.22492 [DOI] [PubMed] [Google Scholar]
  189. Rose CR, & Verkhratsky A (2016). Principles of sodium homeostasis and sodium signalling in Astroglia. Glia, 64(10), 1611–1627. 10.1002/glia.22964 [DOI] [PubMed] [Google Scholar]
  190. Ruminot I, Gutierrez R, Pena-Munzenmayer G, Anazco C, Sotelo-Hitschfeld T, Lerchundi R, … Barros LF (2011). NBCe1 mediates the acute stimulation of astrocytic glycolysis by extracellular K+. Journal of Neuroscience, 31(40), 14264–14271. 10.1523/Jneurosci.2310-11.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Rutkowsky JM, Wallace BK, Wise PM, & O’Donnell ME (2011). Effects of estradiol on ischemic factor-induced astrocyte swelling and AQP4 protein abundance. American Journal of Physiology. Cell Physiology, 301(1), C204–C212. 10.1152/ajpcell.00399.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Sajja VSSS, Hlavac N, & VandeVord PJ (2016). Role of glia in memory deficits following traumatic brain injury: Biomarkers of glia dysfunction. Frontiers in Integrative Neuroscience, 10, 7 10.3389/fnint.7016.00007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Sakaue M, Nakamura H, Kaneko I, Kawasaki Y, Arakawa N, Hashimoto H, … Matsuda T (2000). Na+-Ca2+ exchanger isoforms in rat neuronal preparations: Different changes in their expression during postnatal development. Brain Research, 881(2), 212–216. 10.1016/S0006-8993(00)02808-0 [DOI] [PubMed] [Google Scholar]
  194. Schiapparelli P, Guerrero-Cazares H, Magana-Maldonado R, Hamilla SM, Ganaha S, Goulin Lippi Fernandes E, … Quinones-Hinojosa A (2017). NKCC1 regulates migration ability of Glioblastoma cells by modulation of actin dynamics and interacting with Cofilin. eBioMedicine, 21, 94–103. 10.1016/j.ebiom.2017.06.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Schiff D (2017). Low-grade gliomas. Continuum (Minneap Minn), 23(6), 1564–1579. 10.1212/CON.0000000000000537 [DOI] [PubMed] [Google Scholar]
  196. Schmitt BM, Berger UV, Douglas RM, Bevensee MO, Hediger MA, Haddad GG, & Boron WF (2000). Na/HCO3 cotransporters in rat brain: Expression in glia, neurons, and choroid plexus. The Journal of Neuroscience, 20(18), 6839–6848 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/10995828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Schomberg D, Ahmed M, Miranpuri G, Olson J, & Resnick DK (2012). Neuropathic pain: Role of inflammation, immune response, and ion channel activity in central injury mechanisms. Annals of Neurosciences, 19(3), 125–132. 10.5214/ans.0972.7531.190309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Schousboe A, Sarup A, Bak LK, Waagepetersen HS, & Larsson OM (2004). Role of astrocytic transport processes in glutamatergic and GABAergic neurotransmission. Neurochemistry International, 45(4), 521–527. 10.1016/j.neuint.2003.11.001 [DOI] [PubMed] [Google Scholar]
  199. Schulte JT, Wierenga CJ, & Bruining H (2018). Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neuroscience and Biobehavioral Reviews, 90, 260–271. 10.1016/j.neubiorev.2018.05.001 [DOI] [PubMed] [Google Scholar]
  200. Semyanov A (2019). Spatiotemporal pattern of calcium activity in astrocytic network. Cell Calcium, 78, 15–25. 10.1016/j.ceca.2018.12.007 [DOI] [PubMed] [Google Scholar]
  201. Sharma V, Roy S, Sekler I, & O’Halloran DM (2017). The NCLX-type Na+/Ca2+ exchanger NCX-9 is required for patterning of neural circuits in Caenorhabditis elegans. Journal of Biological Chemistry, 292(13), 5364–5377. 10.1074/jbc.M116.758953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Shenoda B (2015). The role of Na+/Ca2+ exchanger subtypes in neuronal ischemic injury. Translational Stroke Research, 6(3), 181–190. 10.1007/s12975-015-0395-9 [DOI] [PubMed] [Google Scholar]
  203. Shi YJ, Chanana V, Watters JJ, Ferrazzano P, & Sun DD (2011). Role of sodium/hydrogen exchanger isoform 1 in microglial activation and proinflammatory responses in ischemic brains. Journal of Neurochemistry, 119(1), 124–135. 10.1111/jM471-4159.2011.07403.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Shi YJ, Kim D, Caldwell M, & Sun DD (2013). The role of Na+/H+ exchanger isoform 1 in inflammatory responses: Maintaining H+ homeostasis of immune cells. Advances in Experimental Medicine and Biology, 961, 411–418. 10.1007/978-1-4614-4756-6_35 [DOI] [PubMed] [Google Scholar]
  205. Shi YJ, Yuan H, Kim D, Chanana V, Baba A, Matsuda T, … Sun DD (2013). Stimulation of Na+/H+ exchanger isoform 1 promotes microglial migration. PLoS One, 8(8), e74201 10.1371/journal.pone.0074201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Shindo A, Liang AC, Maki T, Miyamoto N, Tomimoto H, Lo EH, & Arai K (2016). Subcortical ischemic vascular disease: Roles of oligodendrocyte function in experimental models of subcortical white-matter injury. Journal of Cerebral Blood Flow and Metabolism, 36(1), 187–198. 10.1038/jcbfm.2015.80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Sirabella R, Sisalli MJ, Costa G, Omura K, Ianniello G, Pinna A, … Scorziello A (2018). NCX1 and NCX3 as potential factors contributing to neurodegeneration and neuroinflammation in the A53T transgenic mouse model of Parkinson’s disease. Cell Death & Disease, 9(7), 725 10.1038/s41419-018-0775-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Sohn Y, Yoo KY, Park OK, Kwon SH, Lee CH, Choi JH, … Won MH (2011). Na+/HCO3-cotransporter immunoreactivity changes in neurons and expresses in astrocytes in the gerbil hippocampal CA1 region after ischemia/reperfusion. Neurochemical Research, 36 (12), 2459–2469. 10.1007/s11064-011-0572-5 [DOI] [PubMed] [Google Scholar]
  209. Song D, Xu J, Du T, Yan E, Hertz L, Walz W, & Peng L (2014). Inhibition of brain swelling after ischemia-reperfusion by beta-adrenergic antagonists: Correlation with increased K+ and decreased Ca2+ concentrations in extracellular fluid. BioMed Research International, 2014, 873510–873590. 10.1155/2014/873590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Song D, Xu J, Hertz L, & Peng L (2015). Regulatory volume increase in astrocytes exposed to hypertonic medium requires beta1 -adrenergic Na(+)/K(+)-ATPase stimulation and glycogenolysis. Journal of Neuroscience Research, 93(1), 130–139. 10.1002/jnr.23469 [DOI] [PubMed] [Google Scholar]
  211. Song S, Wang S, Pigott V, Jiang T, Foley LM, Mishra A, … Sun DD (2018). Selective role of Na+/H+ exchanger in Cx3cr1(+) microglial activation, white matter demyelination, and post-stroke function recovery. Glia, 66(11), 2279–2298. 10.1002/glia.23456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Sozmen EG, Rosenzweig S, Llorente IL, DiTullio DJ, Machnicki M, Vinters HV, … Carmichael ST (2016). Nogo receptor blockade overcomes remyelination failure after white matter stroke and stimulates functional recovery in aged mice. Proceedings of the National Academy of Sciences of the United States of America, 113(52), E8453–E8462. 10.1073/pnas.1615322113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Steinhauser C, Seifert G, & Bedner P (2012). Astrocyte dysfunction in temporal lobe epilepsy: K+ channels and gap junction coupling. Glia, 60 (8), 1192–1202. 10.1002/glia.22313 [DOI] [PubMed] [Google Scholar]
  214. Stock C, & Pedersen SF (2017). Roles of pH and the Na+/H+ exchanger NHE1 in cancer: From cell biology and animal models to an emerging translational perspective? Seminars in Cancer Biology, 43, 5–16. https://doi.org/10.10167i.semcancer.2016.12.001 [DOI] [PubMed] [Google Scholar]
  215. Su G, Kintner DB, Flagella M, Shull GE, & Sun D (2002). Astrocytes from Na(+)-K(+)-cl(−) cotransporter-null mice exhibit absence of swelling and decrease in EAA release. American Journal of Physiology. Cell Physiology, 282(5), C1147–C1160. 10.1152/ajpcell.00538.2001 [DOI] [PubMed] [Google Scholar]
  216. Su G, Kintner DB, & Sun D (2002). Contribution of Na(+)-K(+)-cl(−) cotransporter to high-[K(+)](o)-induced swelling and EAA release in astrocytes. American Journal of Physiology. Cell Physiology, 282(5), C1136–C1146. 10.1152/ajpcell.00478.2001 [DOI] [PubMed] [Google Scholar]
  217. Sunkaria A, Bhardwaj S, Halder A, Yadav A, & Sandhir R (2016). Migration and phagocytic ability of activated microglia during postnatal development is mediated by calcium-dependent Purinergic signalling. Molecular Neurobiology, 53(2), 944–954. 10.1007/s12035-014-9064-3 [DOI] [PubMed] [Google Scholar]
  218. Takase H, Liang AC, Miyamoto N, Hamanaka G, Ohtomo R, Maki T, … Arai K (2018). Protective effects of a radical scavenger edaravone on oligodendrocyte precursor cells against oxidative stress. Neuroscience Letters, 668, 120–125. 10.1016/j.neulet.2018.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Takase H, Washida K, Hayakawa K, Arai K, Wang X, Lo EH, & Lok J (2018). Oligodendrogenesis after traumatic brain injury. Behavioural Brain Research, 340, 205–211. 10.1016/j.bbr.2016.10.042 [DOI] [PubMed] [Google Scholar]
  220. Takuma K, Ago Y, & Matsuda T (2013). The glial sodium-calcium exchanger: A new target for nitric oxide-mediated cellular toxicity. Current Protein & Peptide Science, 14(1), 43–50 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/23441899. [DOI] [PubMed] [Google Scholar]
  221. Theparambil SM, & Deitmer JW (2015). High effective cytosolic H+ buffering in mouse cortical astrocytes attributable to fast bicarbonate transport. Glia, 63(9), 1581–1594. 10.1002/glia.22829 [DOI] [PubMed] [Google Scholar]
  222. Theparambil SM, Ruminot I, Schneider HP, Shull GE, & Deitmer JW (2014). The Electrogenic sodium bicarbonate Cotransporter NBCe1 is a high-affinity bicarbonate carrier in cortical astrocytes. Journal of Neuroscience, 34(4), 1148–1157. 10.1523/Jneurosci.2377-13.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Valastyan S, & Weinberg RA (2011). Tumor metastasis: Molecular insights and evolving paradigms. Cell, 147(2), 275–292. 10.1016/j.cell.2011.09.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, … Breakefield XO (2016). Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology, 18(1), 58–69. 10.1093/neuonc/nov244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Verkhratsky A, Rodriguez-Arellano JJ, Parpura V, & Zorec R (2017). Astroglial calcium signalling in Alzheimer’s disease. Biochemical and Biophysical Research Communications, 483(4), 1005–1012. 10.1016/j.bbrc.2016.08.088 [DOI] [PubMed] [Google Scholar]
  226. Verkhratsky A, Trebak M, Perocchi F, Khananshvili D, & Sekler I (2018). Crosslink between calcium and sodium signalling. Experimental Physiology, 103(2), 157–169. 10.1113/Ep086534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Volkman R, & Offen D (2017). Concise review: Mesenchymal stem cells in neurodegenerative diseases. Stem Cells, 35(8), 1867–1880. 10.1002/stem.2651 [DOI] [PubMed] [Google Scholar]
  228. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, & Nabekura J (2009). Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. Journal of Neuroscience, 29(13), 3974–3980. 10.1523/Jneurosci.4363-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Wang F, Wang X, Shapiro LA, Cotrina ML, Liu W, Wang EW, … Huang JH (2017). NKCC1 up-regulation contributes to early post-traumatic seizures and increased post-traumatic seizure susceptibility. Brain Structure & Function, 222(3), 1543–1556. 10.1007/s00429-016-1292-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Wang H, Yan Y, Kintner DB, Lytle C, & Sun D (2003). GABA-mediated trophic effect on oligodendrocytes requires Na-K-2Cl cotransport activity. Journal of Neurophysiology, 90(2), 1257–1265. 10.1152/jn.01174.2002 [DOI] [PubMed] [Google Scholar]
  231. Wang P, Li L, Zhang ZX, Kan QC, Gao F, & Chen SY (2016). Time-dependent activity of Na+/H+ exchanger isoform 1 and homeostasis of intracellular pH in astrocytes exposed to CoCl2 treatment. Molecular Medicine Reports, 13(5), 4443–4450. 10.3892/mmr.2016.5067 [DOI] [PubMed] [Google Scholar]
  232. Wang S, & Colonna M (2019). Microglia in Alzheimer’s disease: A target for immunotherapy. Journal of Leukocyte Biology, 106(1), 219–227. 10.1002/JLB.MR0818-319R [DOI] [PubMed] [Google Scholar]
  233. Wang Y, Liu G, Hong DD, Chen FH, Ji XM, & Cao GD (2016). White matter injury in ischemic stroke. Progress in Neurobiology, 141, 45–60. 10.1016/j.pneurobio.2016.04.00 [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Wei J, Gabrusiewicz K, & Heimberger A (2013). The controversial role of microglia in malignant gliomas. Clinical & Developmental Immunology, 2013, 285212–285246. 10.1155/2013/285246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Weller M (2011). Novel diagnostic and therapeutic approaches to malignant glioma. Swiss Medical Weekly, 141, w13210 10.4414/smw.2011.13210 [DOI] [PubMed] [Google Scholar]
  236. Wilke SR, Thomas R, Allcock N, & Fern R (2004). Mechanism of acute ischemic injury of oligodendroglia in early myelinating white matter: The importance of astrocyte injury and glutamate release. Journal of Neuropathology and Experimental Neurology, 63(8), 872–881. 10.1093/jnen/63.8.872 [DOI] [PubMed] [Google Scholar]
  237. Williams A, Piaton G, & Lubetzki C (2007). Astrocytes--Friends or foes in multiple sclerosis. Glia, 55(13), 1300–1312. 10.1002/glia.20546 [DOI] [PubMed] [Google Scholar]
  238. Wilson CS, & Mongin AA (2019). The signaling role for chloride in the bidirectional communication between neurons and astrocytes. Neuroscience Letters, 689, 33–44. 10.1016/j.neulet.2018.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Wolf SA, Boddeke HW, & Kettenmann H (2017). Microglia in physiology and disease. Annual Review of Physiology, 79, 619–643. 10.1146/annurev-physiol-022516-034406 [DOI] [PubMed] [Google Scholar]
  240. Xu H, Wang J, Song N, Jiang H, & Xie J (2016). Glial cells are involved in iron accumulation and degeneration of dopamine neurons in Parkinson’s disease. Sheng Li Xue Bao, 68(4), 455–463 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/27546505. [PubMed] [Google Scholar]
  241. Xu J, Song D, Xue Z, Gu L, Hertz L, & Peng L (2013). Requirement of glycogenolysis for uptake of increased extracellular K+ in astrocytes: Potential implications for K+ homeostasis and glycogen usage in brain. Neurochemical Research, 38(3), 472–485. 10.1007/s11064-012-0938-3 [DOI] [PubMed] [Google Scholar]
  242. Xu W, Mu X, Wang H, Song C, Ma W, Jolkkonen J, & Zhao C (2017). Chloride co-transporter NKCC1 inhibitor Bumetanide enhances neurogenesis and Behavioral recovery in rats after experimental stroke. Molecular Neurobiology, 54(4), 2406–2414. 10.1007/s12035-016-9819-0 [DOI] [PubMed] [Google Scholar]
  243. Yan Y, Dempsey RJ, & Sun D (2001). Expression of Na(+)-K(+)-cl(−) cotransporter in rat brain during development and its localization in mature astrocytes. Brain Research, 911(1), 43–55 Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/11489443. [DOI] [PubMed] [Google Scholar]
  244. Yao H, Azad P, Zhao HW, Wang J, Poulsen O, Freitas BC, … Haddad GG (2016). The Na(+)/Hco3-co-transporter is protective during ischemia in astrocytes. Neuroscience, 339, 329–337. 10.1016/j.neuroscience.2016.09.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Yu XW, Hu ZL, Ni M, Fang P, Zhang PW, Shu Q, … Wang F (2015). Acid-sensing ion channels promote the inflammation and migration of cultured rat microglia. Glia, 63(3), 483–496. 10.1002/glia.22766 [DOI] [PubMed] [Google Scholar]
  246. Yu Y, Fu P, Yu Z, Xie M, Wang W, & Luo X (2018). NKCC1 inhibition attenuates chronic cerebral hypoperfusion-induced white matter lesions by enhancing progenitor cells of Oligodendrocyte proliferation. Journal of Molecular Neuroscience, 64(3), 449–458. 10.1007/s12031-018-1043-0 [DOI] [PubMed] [Google Scholar]
  247. Yuen N, Lam TI, Wallace BK, Klug NR, Anderson SE, & O’Donnell ME (2014). Ischemic factor-induced increases in cerebral microvascular endothelial cell Na/H exchange activity and abundance: Evidence for involvement of ERK1/2 MAP kinase. American Journal of Physiology. Cell Physiology, 306(10), C931–C942. 10.1152/ajpcell.00021.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Zeuthen T, & MacAulay N (2012). Cotransport of water by Na+-K+−2Cl cotransporters expressed in Xenopus oocytes: NKCC1 versus NKCC2. Journal of Physiology-London, 590(5), 1139–1154. 10.1113/jphysiol.2011.226316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Zhang J, Wang Y, Zheng Z, Sun X, Chen T, Li C, … Guo J (2019). Intracellular ion and protein nanoparticle-induced osmotic pressure modify astrocyte swelling and brain edema in response to glutamate stimuli. Redox Biology, 21, 101112 10.1016/j.redox.2019.101112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Zhang M, Cui Z, Cui H, Wang Y, & Zhong C (2017). Astaxanthin protects astrocytes against trauma-induced apoptosis through inhibition of NKCC1 expression via the NF-kappa B signaling pathway. BMC Neuroscience, 18, 42 10.1186/s12868-017-0358-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Zhang Z, Sun D, Kahle KT, Zhang J, Jiang T, Banerjee S, … Huang H (2018). The WNK-SPAK/OSR1 kinases and the cation-chloride cotransporters as therapeutic targets for neurological diseases. Aging and Disease, 10, 626–636. 10.14336/ad.2018.0928 [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Zhao XR, Gorin FA, Berman RF, & Lyeth BG (2008). Differential hippocampal protection when blocking intracellular sodium and calcium entry during traumatic brain injury in rats. Journal of Neurotrauma, 25(10), 1195–1205. 10.1089/neu.2008.0635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Zhu W, Begum G, Pointer K, Clark PA, Yang SS, Lin SH, … Sun D (2014). WNK1-OSR1 kinase-mediated phospho-activation of Na+-K+−2Cl cotransporter facilitates glioma migration. Molecular Cancer, 13, 31 10.1186/1476-4598-13-31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Zhu W, Carney KE, Pigott VM, Falgoust LM, Clark PA, Kuo JS, & Sun D (2016). Glioma-mediated microglial activation promotes glioma proliferation and migration: Roles of Na+/H+ exchanger isoform 1. Carcinogenesis, 37(9), 839–851. 10.1093/carcin/bgw068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Ziemens D, Oschmann F, Gerkau NJ, & Rose CR (2019). Heterogeneity of activity-induced sodium transients between astrocytes of the mouse hippocampus and neocortex: Mechanisms and consequences. Journal of Neuroscience, 39(14), 2620–2634. 10.1523/Jneurosci.2029-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Zonouzi M, Scafidi J, Li PJ, McEllin B, Edwards J, Dupree JL, … Gallo V (2015). GABAergic regulation of cerebellar NG2 cell development is altered in perinatal white matter injury. Nature Neuroscience, 18(5), 674–682. 10.1038/nn.3990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Zuchero JB, & Barres BA (2015). Glia in mammalian development and disease. Development, 142(22), 3805–3809. 10.1242/dev.129304 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES