首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
The release of glutamine from astrocytes adjacent to synapses in the central nervous system is thought to play a vital role in the mechanism of glutamate recycling and is therefore important for maintaining excitatory neurotransmission. Here we investigate the nature of astrocytic membrane transport of glutamine in rat brainstem slices, using electrophysiological recording and fluorescent imaging of pHi and . Glutamine application to perisynaptic astrocytes induced a membrane current, caused by activation of system A (SA) family transporters. A significant electroneutral component was also observed, which was mediated by the system N (SN) family transporters. This response was stimulated by glutamine (KM of 1.57 mM), histidine, and asparagine, but not by leucine or serine, indicating activation of the SNAT3 isoform of SN. We hypothesized that increasing the [Na+]i would alter the SNAT3 transporter equilibrium, thereby stimulating glutamine release. In support of this hypothesis, we show that SNAT3 transport can be driven by changing cation concentration and that manipulations to raise [Na+]i (activation of excitatory amino acid transporters (EAATs), SA transporters or AMPA receptors) all directly influence SNAT3 transport rate. A kinetic model of glutamine fluxes is presented, which shows that EAAT activation causes the release of glutamine, driven mainly by the increased [Na+]i. These data demonstrate that SNAT3 is functionally active in perisynaptic astrocytes in situ. As a result, astrocytic signaling, as would be stimulated by neighboring synaptic activity, has the capacity to stimulate astrocytic glutamine release to support glutamate recycling.  相似文献   

2.
The clearance of extracellular glutamate is mainly mediated by pH‐ and sodium‐dependent transport into astrocytes. During hepatic encephalopathy (HE), however, elevated extracellular glutamate concentrations are observed. The primary candidate responsible for the toxic effects observed during HE is ammonium (NH4+/NH3). Here, we examined the effects of NH4+/NH3 on steady‐state intracellular pH (pHi) and sodium concentration ([Na+]i) in cultured astrocytes in two different age groups. Moreover, we assessed the influence of NH4+/NH3 on glutamate transporter activity by measuring D ‐aspartate‐induced pHi and [Na+]i transients. In 20–34 days in vitro (DIV) astrocytes, NH4+/NH3 decreased steady‐state pHi by 0.19 pH units and increased [Na+]i by 21 mM. D ‐Aspartate‐induced pHi and [Na+]i transients were reduced by 80–90% in the presence of NH4+/NH3, indicating a dramatic reduction of glutamate uptake activity. In 9–16 DIV astrocytes, in contrast, pHi and [Na+]i were minimally affected by NH4+/NH3, and D ‐aspartate‐induced pHi and [Na+]i transients were reduced by only 30–40%. Next we determined the contribution of Na+, K+, Cl?‐cotransport (NKCC). Immunocytochemical stainings indicated an increased expression of NKCC1 in 20–34 DIV astrocytes. Moreover, inhibition of NKCC with bumetanide prevented NH4+/NH3‐evoked changes in steady‐state pHi and [Na+]i and attenuated the reduction of D ‐aspartate‐induced pHi and [Na+]i transients by NH4+/NH3 to 30% in 20–34 DIV astrocytes. Our results suggest that NH4+/NH3 decreases steady‐state pHi and increases steady‐state [Na+]i in astrocytes by an age‐dependent activation of NKCC. These NH4+/NH3‐evoked changes in the transmembrane pH and sodium gradients directly reduce glutamate transport activity, and may, thus, contribute to elevated extracellular glutamate levels observed during HE. © 2008 Wiley‐Liss, Inc.  相似文献   

3.
The transmembrane Na+ concentration gradient is an important source of energy required not only to enable the generation of action potentials in excitable cells, but also for various transmembrane transporters both in excitable and non‐excitable cells, like astrocytes. One of the vital functions of astrocytes in the central nervous system (CNS) is to regulate neurotransmitter concentrations in the extracellular space. Most neurotransmitters in the CNS are removed from the extracellular space by Na+‐dependent neurotransmitter transporters (NeuTs) expressed both in neurons and astrocytes. Neuronal NeuTs control mainly phasic synaptic transmission, i.e., synaptically induced transient postsynaptic potentials, while astrocytic NeuTs contribute to the termination of phasic neurotransmission and modulate the tonic tone, i.e., the long‐lasting activation of extrasynaptic receptors by neurotransmitter that has diffused out of the synaptic cleft. Consequently, local intracellular Na+ ([Na+]i) transients occurring in astrocytes, for example via the activation of ionotropic neurotransmitter receptors, can affect the driving force for neurotransmitter uptake, in turn modulating the spatio‐temporal profiles of neurotransmitter levels in the extracellular space. As some NeuTs are close to thermodynamic equilibrium under resting conditions, an increase in astrocytic [Na+]i can stimulate the direct release of neurotransmitter via NeuT reversal. In this review we discuss the role of astrocytic [Na+]i changes in the regulation of uptake/release of neurotransmitters. It is emphasized that an activation of one neurotransmitter system, including either its ionotropic receptor or Na+‐coupled co‐transporter, can strongly influence, or even reverse, other Na+‐dependent NeuTs, with potentially significant consequences for neuronal communication. GLIA 2016;64:1655–1666  相似文献   

4.
During neuronal activity in the mammalian brain, the K+ released into the synaptic space is initially buffered by the astrocytic compartment. In parallel, the extracellular space (ECS) shrinks, presumably due to astrocytic cell swelling. With the Na+/K+/2Cl? cotransporter and the Kir4.1/AQP4 complex not required for the astrocytic cell swelling in the hippocampus, the molecular mechanisms underlying the activity‐dependent ECS shrinkage have remained unresolved. To identify these molecular mechanisms, we employed ion‐sensitive microelectrodes to measure changes in ECS, [K+]o and [H+]o/pHo during electrical stimulation of rat hippocampal slices. Transporters and receptors responding directly to the K+ and glutamate released into the extracellular space (the K+/Cl? cotransporter, KCC, glutamate transporters and G protein‐coupled receptors) did not modulate the extracellular space dynamics. The ‐transporting mechanism, which in astrocytes mainly constitutes the electrogenic Na+/ cotransporter 1 (NBCe1), is activated by the K+‐mediated depolarization of the astrocytic membrane. Inhibition of this transporter reduced the ECS shrinkage by ~25% without affecting the K+ transients, pointing to NBCe1 as a key contributor to the stimulus‐induced astrocytic cell swelling. Inhibition of the monocarboxylate cotransporters (MCT), like‐wise, reduced the ECS shrinkage by ~25% without compromising the K+ transients. Isosmotic reduction of extracellular Cl? revealed a requirement for this ion in parts of the ECS shrinkage. Taken together, the stimulus‐evoked astrocytic cell swelling does not appear to occur as a direct effect of the K+ clearance, as earlier proposed, but partly via the pH‐regulating transport mechanisms activated by the K+‐induced astrocytic depolarization and the activity‐dependent metabolism.  相似文献   

5.
Joel A. Black  Stephen G. Waxman 《Glia》2014,62(7):1162-1175
Astrogliosis is a prominent feature of many, if not all, pathologies of the brain and spinal cord, yet a detailed understanding of the underlying molecular pathways involved in the transformation from quiescent to reactive astrocyte remains elusive. We investigated the contribution of voltage‐gated sodium channels to astrogliosis in an in vitro model of mechanical injury to astrocytes. Previous studies have shown that a scratch injury to astrocytes invokes dual mechanisms of migration and proliferation in these cells. Our results demonstrate that wound closure after mechanical injury, involving both migration and proliferation, is attenuated by pharmacological treatment with tetrodotoxin (TTX) and KB‐R7943, at a dose that blocks reverse mode of the Na+/Ca2+ exchanger (NCX), and by knockdown of Nav1.5 mRNA. We also show that astrocytes display a robust [Ca2+]i transient after mechanical injury and demonstrate that this [Ca2+]i response is also attenuated by TTX, KB‐R7943, and Nav1.5 mRNA knockdown. Our results suggest that Nav1.5 and NCX are potential targets for modulation of astrogliosis after injury via their effect on [Ca2+]i. GLIA 2014;62:1162–1175  相似文献   

6.
Sodium dynamics are essential for regulating functional processes in glial cells. Indeed, glial Na+ signaling influences and regulates important glial activities, and plays a role in neuron‐glia interaction under physiological conditions or in response to injury of the central nervous system (CNS). Emerging studies indicate that Na+ pumps and Na+‐dependent ion transporters in astrocytes, microglia, and oligodendrocytes regulate Na+ homeostasis and play a fundamental role in modulating glial activities in neurological diseases. In this review, we first briefly introduced the emerging roles of each glial cell type in the pathophysiology of cerebral ischemia, Alzheimer's disease, epilepsy, Parkinson's disease, Amyotrophic Lateral Sclerosis, and myelin diseases. Then, we discussed the current knowledge on the main roles played by the different glial Na+‐dependent ion transporters, including Na+/K+ ATPase, Na+/Ca2+ exchangers, Na+/H+ exchangers, Na+‐K+‐Cl? cotransporters, and Na+‐ cotransporter in the pathophysiology of the diverse CNS diseases. We highlighted their contributions in cell survival, synaptic pathology, gliotransmission, pH homeostasis, and their role in glial activation, migration, gliosis, inflammation, and tissue repair processes. Therefore, this review summarizes the foundation work for targeting Na+‐dependent ion transporters in glia as a novel strategy to control important glial activities associated with Na+ dynamics in different neurological disorders. GLIA 2016;64:1677–1697  相似文献   

7.
Influx of sodium ions into active neurons is a highly energy‐expensive process which must be strictly limited. Astrocytes could play an important role herein because they take up glutamate and potassium from the extracellular space, thereby dampening neuronal excitation. Here, we performed sodium imaging in mouse hippocampal slices combined with field potential and whole‐cell patch‐clamp recordings and measurement of extracellular potassium ([K+]o). Network activity was induced by Mg2+‐free, bicuculline‐containing saline, during which neurons showed recurring epileptiform bursting, accompanied by transient increases in [K+]o and astrocyte depolarizations. During bursts, neurons displayed sodium increases by up to 22 mM. Astrocyte sodium concentration increased by up to 8.5 mM, which could be followed by an undershoot below baseline. Network sodium oscillations were dependent on action potentials and activation of ionotropic glutamate receptors. Inhibition of glutamate uptake caused acceleration, followed by cessation of electrical activity, irreversible sodium increases, and swelling of neurons. The gliotoxin NaFAc (sodium‐fluoroacetate) resulted in elevation of astrocyte sodium concentration and reduced glial uptake of glutamate and potassium uptake through Na+/K+‐ATPase. Moreover, NaFAc extended epileptiform bursts, caused elevation of neuronal sodium, and dramatically prolonged accompanying sodium signals, most likely because of the decreased clearance of glutamate and potassium by astrocytes. Our experiments establish that recurrent neuronal bursting evokes sodium transients in neurons and astrocytes and confirm the essential role of glutamate transporters for network activity. They suggest that astrocytes restrict discharge duration and show that an intact astrocyte metabolism is critical for the neurons' capacity to recover from sodium loads during synchronized activity. GLIA 2015;63:936–957  相似文献   

8.
Gap junctions between glial cells allow intercellular exchange of ions and small molecules. We have investigated the influence of gap junction coupling on regulation of intracellular Na+ concentration ([Na+]i) in cultured rat hippocampal astrocytes, using fluorescence ratio imaging with the Na+ indicator dye SBFI (sodium-binding benzofuran isophthalate). The [Na+]i in neighboring astrocytes was very similar (12.0 ± 3.3 mM) and did not fluctuate under resting conditions. During uncoupling of gap junctions with octanol (0.5 mM), baseline [Na+]i was unaltered in 24%, increased in 54%, and decreased in 22% of cells. Qualitatively similar results were obtained with two other uncoupling agents, heptanol and α-glycyrrhetinic acid (AGA). Octanol did not alter the recovery from intracellular Na+ load induced by removal of extracellular K+, indicating that octanol's effects on baseline [Na+]i were not due to inhibition of Na+, K+-ATPase activity. Under control conditions, increasing [K+]o from 3 to 8 mM caused similar decreases in [Na+]i in groups of astrocytes, presumably by stimulating Na+, K+-ATPase. During octanol application, [K+]o-induced [Na+]i decreases were amplified in cells with increased baseline [Na+]i, and reduced in cells with decreased baseline [Na+]i. This suggests that baseline [Na+]i in astrocytes “sets” the responsiveness of Na+, K+-ATPase to increases in [K+]o. Our results indicate that individual hippocampal astrocytes in culture rapidly develop different levels of baseline [Na+]i when they are isolated from one another by uncoupling agents. In astrocytes, therefore, an apparent function of coupling is the intercellular exchange of Na+ ions to equalize baseline [Na+]i, which serves to coordinate physiological responses that depend on the intracellular concentration of this ion. GLIA 20:299–307, 1997. © 1997 Wiley-Liss, Inc.  相似文献   

9.
D‐serine is an endogenous coagonist of N‐methyl‐D‐aspartate (NMDA) receptors that plays an important role in synaptic function, neuronal development, and excitotoxicity. Mechanisms of D‐serine transport are important in regulation of extracellular D‐serine concentration and therefore of these critical processes. D‐serine can be transported with low affinity through the Na+‐dependent amino acid transporter termed ASCT2, whereas high‐affinity D‐serine uptake has been reported through the Na+‐independent transporter termed asc‐1. We investigated immunoreactivity for ASCT2 and asc‐1 and D‐serine transport kinetics in cultured cortical neurons and astrocytes to gain insight into how D‐serine transporters regulate CNS D‐serine levels. Both neurons and astrocytes exhibited low‐affinity Na+‐dependent D‐serine uptake (KT > 1 mM) with broad substrate selectivity that was consistent with uptake through ASCT2. Both neurons and astrocytes also stained positively for ASCT2 in immunocytochemistry studies. Neurons but not astrocytes stained positively for the high‐affinity D‐serine transporter asc‐1, but no evidence of functional asc‐1 could be detected in neurons with conditions that produced such activity in cortical synaptosomes. These data support ASCT2 function in both neuron and astrocyte cultures and identify a discrepancy between observed asc‐1 immunoreactivity and lack of functional asc‐1 activity in neuron cultures. Together these findings further our knowledge of the processes that govern D‐serine regulation. © 2009 Wiley‐Liss, Inc.  相似文献   

10.
Jun Li  Sien-Yao Chow 《Epilepsia》1994,35(1):20-26
Summary: Effects of chronic treatment of dibutyryl cyclic AMP (db-cyclic AMP) on Na+, K+-ATPase activity in cell homogenates and intracellular N a f and K+ contents [(Na+)i and (K+)i] were studied in primary cultures of astrocytes derived from cerebral cortex of neonatal audiogenic seizure-susceptible DBA and audiogenic seizure-resistant C57 mice. Na+, K+-ATPase activity in cell homogenates was greater and (Na+)i was less in DBA astrocytes than in C57 astrocytes. There was no difference in (K+)i between astrocytes from DBA and C57 mice. Addition of db-cyclic AMP to the medium from day 14 to day 21 in culture (final concentration 0.25 mM) increased Na+, K+-ATPase activity in cell homogenates and decreased (Na+)i, but had no significant effect on (K+)i in astrocytes from either DBA or C57 mice. Chronic treatment with db-cyclic AMP altered cell growth. Protein and DNA content of cultured astrocytes from both DBA and C57 mice was decreased. DNA was more affected than protein. Modifying K+ and Na+ concentration in medium altered Na+, K+-ATPase activity in cell homogenates as well as (Na+)i and (K+)i in cultured astrocytes of both DBA and C57 mice. Changes in (Na+)i and (K+)i at different K+ concentrations in medium paralleled those in Na+, K+-ATPase activity in cell homogenates. Results indicate that the ability to transport Na+ across the cell membrane and the response of Na+, K+-ATPase to db-cyclic AMP and to the changes in K + in medium of cultured astrocytes from audiogenic seizure-susceptible DBA mice are sufficient.  相似文献   

11.
Summary. Glutamate uptake into astrocytes and the resulting increase in intracellular Na+ (Na+i) have been identified as a key signal coupling excitatory neuronal activity to increased glucose utilization. Arguments based mostly on mathematical modeling led to the conclusion that physiological concentrations of glutamate more than double astrocytic Na+/K+-ATPase activity, which should proportionally increase its ATP hydrolysis rate. This hypothesis was tested in the present study by fluorescence monitoring of free Mg2+ (Mg2+i), a parameter that inversely correlates with ATP levels. Glutamate application measurably increased Mg2+i (i.e. decreased ATP), which was reversible after glutamate washout. Na+i and ATP changes were then directly compared by simultaneous Na+i and Mg2+ imaging. Glutamate increased both parameters with different rates and blocking the Na+/K+-ATPase during the glutamate-evoked Na+i response, resulted in a drop of Mg2+i levels (i.e. increased ATP). Taken together, this study demonstrates the tight correlation between glutamate transport, Na+ homeostasis and ATP levels in astrocytes.  相似文献   

12.
Network activity in the brain is associated with a transient increase in extracellular K+ concentration. The excess K+ is removed from the extracellular space by mechanisms proposed to involve Kir4.1‐mediated spatial buffering, the Na+/K+/2Cl? cotransporter 1 (NKCC1), and/or Na+/K+‐ATPase activity. Their individual contribution to [K+]o management has been of extended controversy. This study aimed, by several complementary approaches, to delineate the transport characteristics of Kir4.1, NKCC1, and Na+/K+‐ATPase and to resolve their involvement in clearance of extracellular K+ transients. Primary cultures of rat astrocytes displayed robust NKCC1 activity with [K+]o increases above basal levels. Increased [K+]o produced NKCC1‐mediated swelling of cultured astrocytes and NKCC1 could thereby potentially act as a mechanism of K+ clearance while concomitantly mediate the associated shrinkage of the extracellular space. In rat hippocampal slices, inhibition of NKCC1 failed to affect the rate of K+ removal from the extracellular space while Kir4.1 enacted its spatial buffering only during a local [K+]o increase. In contrast, inhibition of the different isoforms of Na+/K+‐ATPase reduced post‐stimulus clearance of K+ transients. The astrocyte‐characteristic α2β2 subunit composition of Na+/K+‐ATPase, when expressed in Xenopus oocytes, displayed a K+ affinity and voltage‐sensitivity that would render this subunit composition specifically geared for controlling [K+]o during neuronal activity. In rat hippocampal slices, simultaneous measurements of the extracellular space volume revealed that neither Kir4.1, NKCC1, nor Na+/K+‐ATPase accounted for the stimulus‐induced shrinkage of the extracellular space. Thus, NKCC1 plays no role in activity‐induced extracellular K+ recovery in native hippocampal tissue while Kir4.1 and Na+/K+‐ATPase serve temporally distinct roles. GLIA 2014;62:608–622  相似文献   

13.
Astrocytic volume regulation and neurotransmitter uptake are critically dependent on the intracellular anion concentration, but little is known about the mechanisms controlling internal anion homeostasis in these cells. Here we used fluorescence lifetime imaging microscopy (FLIM) with the chloride‐sensitive dye MQAE to measure intracellular chloride concentrations in murine Bergmann glial cells in acute cerebellar slices. We found Bergmann glial [Cl?]int to be controlled by two opposing transport processes: chloride is actively accumulated by the Na+‐K+‐2Cl? cotransporter NKCC1, and chloride efflux through anion channels associated with excitatory amino acid transporters (EAATs) reduces [Cl?]int to values that vary upon changes in expression levels or activity of these channels. EAATs transiently form anion‐selective channels during glutamate transport, and thus represent a class of ligand‐gated anion channels. Age‐dependent upregulation of EAATs results in a developmental chloride switch from high internal chloride concentrations (51.6 ± 2.2 mM, mean ± 95% confidence interval) during early development to adult levels (35.3 ± 0.3 mM). Simultaneous blockade of EAAT1/GLAST and EAAT2/GLT‐1 increased [Cl?]int in adult glia to neonatal values. Moreover, EAAT activation by synaptic stimulations rapidly decreased [Cl?]int. Other tested chloride channels or chloride transporters do not contribute to [Cl?]int under our experimental conditions. Neither genetic removal of ClC‐2 nor pharmacological block of K+‐Cl? cotransporter change resting Bergmann glial [Cl?]int in acute cerebellar slices. We conclude that EAAT anion channels play an important and unexpected role in adjusting glial intracellular anion concentration during maturation and in response to cerebellar activity. GLIA 2017;65:388–400  相似文献   

14.
Christine R. Rose  Claudia Karus 《Glia》2013,61(8):1191-1205
The intracellular sodium concentration of astrocytes is classically viewed as being kept under tight homeostatic control and at a relatively stable level under physiological conditions. Indeed, the steep inwardly directed electrochemical gradient for sodium, generated by the Na+/K+‐ATPase, contributes to maintain the electrochemical gradient of K+ and the highly K+‐based negative membrane potential, and is a central element in energizing membrane transport. As such it is tightly coupled to the homeostasis of extra‐ and intracellular potassium, calcium or pH and to the reuptake of transmitters such as glutamate. Recent studies, however, have demonstrated that this picture is far too simplistic. It is now firmly established that transmitters, most notably glutamate, and excitatory neuronal activity evoke long‐lasting sodium transients in astrocytes, the properties of which are distinctly different from those of activity‐related glial calcium signals. From these studies, it emerges that sodium homeostasis and signaling are two sides of the same coin: sodium‐dependent transporters, primarily known for their role in ion regulation and homeostasis, also generate relevant ion signals during neuronal activity. The functional consequences of activity‐related sodium transients are manifold and are just coming into view, enabling surprising and important new insights into astrocyte function and neuron‐glia interaction in the brain. The present review will highlight current knowledge about the mechanisms that contribute to sodium homeostasis in astrocytes, present recent data on the spatial and temporal properties of activity‐related glial sodium signals and discuss their functional consequences with a special emphasis on pathophysiological conditions. GLIA 2013;61:1191–1205  相似文献   

15.
Cultured mouse cortical astrocytes of the stellate type were studied by using the patch-clamp technique in whole-cell configuration. The astrocytes express at least two types of outwardly rectifying K+ channels which mediate a transient and a sustained current. Activation of AMPA receptors by kainate leads to a substantial blockade of both types of K+ currents. The blockade is absent when Na+ is withdrawn from the external medium, suggesting that it is caused by constant Na+ influx through AMPA receptors. The presence of high Na+ solutions in the pipette induces a blockade of both K+ currents which is very similar to the blockade induced by kainate, supporting thus the view that the mechanism of the blockade of K+ channels by kainate involves Na+ increases in the submembrane area. The blockade occurs between 20 and 40 mM [Na+]i, which is within the physiological range of [Na+]i in astrocytes. The data may suggest that the blockade of K+ channels by high [Na+]i conditions could provide a mechanism to prevent K+ leakage from the astrocytes into the extracellular space during periods of intense neuronal activity. GLIA 20:38-50, 1997. © 1997 Wiley-Liss, Inc.  相似文献   

16.
Astrocytes express a variety of purinergic (P2) receptors, involved in astrocytic communication through fast increases in [Ca2+]i. Of these, the metabotropic ATP receptors (P2Y) regulate cytoplasmic Ca2+ levels through the PLC‐PKC pathway. GABA transporters are a substrate for a number of Ca2+‐related kinases, raising the possibility that calcium signalling in astrocytes impact the control of extracellular levels of the major inhibitory transmitter in the brain. To access this possibility we tested the influence of P2Y receptors upon GABA transport into astrocytes. Mature primary cortical astroglial‐enriched cultures expressed functional P2Y receptors, as evaluated through Ca2+ imaging, being P2Y1 the predominant P2Y receptor subtype. ATP (100 μM, for 1 min) caused an inhibition of GABA transport through either GAT‐1 or GAT‐3 transporters, decreasing the Vmax kinetic constant. ATP‐induced inhibition of GATs activity was still evident in the presence of adenosine deaminase, precluding an adenosine‐mediated effect. This, was mimicked by a specific agonist for the P2Y1,12,13 receptor (2‐MeSADP). The effect of 2‐MeSADP on GABA transport was blocked by the P2 (PPADS) and P2Y1 selective (MRS2179) receptor antagonists, as well as by the PLC inhibitor (U73122). 2‐MeSADP failed to inhibit GABA transport in astrocytes where intracellular calcium had been chelated (BAPTA‐AM) or where calcium stores were depleted (α‐cyclopiazonic acid, CPA). In conclusion, P2Y1 receptors in astrocytes inhibit GABA transport through a mechanism dependent of P2Y1‐mediated calcium signalling, suggesting that astrocytic calcium signalling, which occurs as a consequence of neuronal firing, may operate a negative feedback loop to enhance extracellular levels of GABA. GLIA 2014;62:1211–1226  相似文献   

17.
We investigated the effect of changes in membrane-voltage on intracellular sodium concentration ([Na+]i) of dopamine-sensitive neurons of the substantia nigra pars compacta in a slice preparation of rat mesencephalon. Whole-cell patch-clamp techniques were combined with microfluorometric measurements of [Na+]i using the Na+-sensitive probe, sodium-binding benzofuran isophthalate (SBFI). Hyperpolarization of spontaneously active dopamine neurons (recorded in current-clamp mode) caused the cessation of action potential firing accompanied by an elevation in [Na+]i. In dopamine neurons voltage-clamped at a holding potential of ?60 mV elevations of [Na+]i were induced by long-lasting (45–60 s) voltage jumps to more negative membrane potentials (–90 to ?120 mV) but not by corresponding voltage jumps to ?30 mV. These hyperpolarization-induced elevations of [Na+]i were depressed during inhibition of Ih, a hyperpolarization-activated inward current, by Cs+. Hyperpolarization-induced elevations in [Na+]i might occur also in other cell types which express a powerful Ih and might signal lack of postsynaptic activity.  相似文献   

18.
We used the pH-sensitive fluorescent dye BCECF to study intracellular pH (pHi) regulation in primary cultures of rat astrocytes and C6 glioma cells. Both cell types contain three pH-regulating transporters: (1) alkalinizing Na+/H+ exchange; (2) alkalinizing Na+ + HCO3 ?/Cl? exchange; and (3) acidifying Cl?/HCO3 ? exchange. Na+/H+ exchange was most evident in the absence of CO2; recovery from acidification was Na+ dependent and amiloride sensitive. Exposure to CO2 caused a cell alkalinization that was inhibited by DIDS, dependent on external Na+, and inhibited 75% in the absence of Cl? (thus mediated by Na+ + HCO3 ?/Cl? exchange). When pHi was increased above the normal steady-state pHi, a DIDS-inhibitable and Na+ -independent acidifying recovery was evident, indicating the presence of Cl? /HCO3 ? exchange. Astrocytes, but not C6 cells, contain a fourth pH-regulating transporter, Na+ ?HCO3 ? cotransport; in the presence of CO2, depolarization caused an alkalinization of 0.12 +? 0.01 (n = 8) and increased the rate of CO2-induced alkalinization from 0.23 ± 0.02 to 0.42 ± 0.03 pH unit/min. Since C6 cells lack the Na+ -HCO3 + cotransporter, they are an inferior model of pHi regulation in glia. Our results differ from previous observations in glia in that: (1) Na+ /H+ exchange was entirely inhibited by amiloride; (2) Na+ + HCO3 ?/Cl? exchange was present and largely responsible for CO2?induced alkalinization; (3) Cl? /HCO3 ? exchange was only active at pHi values above steady state; and (4) depolarization-induced alkalinization of astrocytes was seen only in the presence of CO2.  相似文献   

19.
Cytosolic H+ buffering plays a major role for shaping intracellular H+ shifts and hence for the availability of H+ for biochemical reactions and acid/base‐coupled transport processes. H+ buffering is one of the prime means to protect the cell from large acid/base shifts. We have used the H+ indicator dye BCECF and confocal microscopy to monitor the cytosolic H+ concentration, [H+]i, in cultured cortical astrocytes of wild‐type mice and of mice deficient in sodium/bicarbonate cotransporter NBCe1 (NBCe1‐KO) or in carbonic anhydrase isoform II (CAII‐KO). The steady‐state buffer strength was calculated from the amplitude of [H+]i transients as evoked by CO2/HCO3? and by butyric acid in the presence and absence of CO2/HCO3?. We tested the hypotheses if, in addition to instantaneous physicochemical H+ buffering, rapid acid/base transport across the cell membrane contributes to the total, “effective” cytosolic H+ buffering. In the presence of 5% CO2/26 mM HCO3?, H+ buffer strength in astrocytes was increased 4–6 fold, as compared with that in non‐bicarbonate, HEPES‐buffered solution, which was largely attributable to fast HCO3? transport into the cells via NBCe1, supported by CAII activity. Our results show that within the time frame of determining physiological H+ buffering in cells, fast transport and equilibration of CO2/H+/HCO3? can make a major contribution to the total “effective” H+ buffer strength. Thus, “effective” cellular H+ buffering is, to a large extent, attributable to membrane transport of base equivalents rather than a purely passive physicochemical process, and can be much larger than reported so far. Not only physicochemical H+ buffering, but also rapid import of HCO3? via the electrogenic sodium‐bicarbonate cotransporter NBCe1, supported by carbonic anhydrase II (CA II), was identified to enhance cytosolic H+ buffer strength substantially. GLIA 2015;63:1581–1594  相似文献   

20.
During early development, cortical neurons migrate from their places of origin to their final destinations where they differentiate and establish synaptic connections. During corticogenesis, radially migrating cells move from deeper zone to the marginal zone, but they do not invade the latter. This “stop” function of the marginal zone is mediated by a number of factors, including glutamate and γ‐aminobutyric acid (GABA), two main neurotransmitters in the central nervous system. In the marginal zone, GABA has been shown to be released via GABA transporters (GAT)‐2/3, whereas glutamate transporters (EAATs) operate in the uptake mode. In this study, GABAergic postsynaptic currents (GPSCs) were recorded from Cajal‐Retzius cells in the marginal zone of murine neonatal neocortex using a whole‐cell patch‐clamp technique. Minimal electrical stimulation was applied to elicit evoked GPSCs using a paired‐pulse protocol. EAAT blockade with dl ‐threo‐b‐benzyloxyaspartic acid (dl ‐TBOA), a specific non‐transportable EAAT antagonist, abolishes constitutive GAT‐2/3‐mediated GABA release. In contrast to dl ‐TBOA, d ‐aspartate, an EAAT substrate, fails to block GAT‐2/3‐mediated GABA release. SNAP‐5114, a specific GAT‐2/3 antagonist, induced an elevation of intracellular sodium concentration ([Na+]i) under resting conditions and in the presence of d ‐aspartate, indicating that GAT‐2/3 operates in reverse mode. In the presence of dl ‐TBOA, however, SNAP‐5114 elicited a [Na+]i decrease, demonstrating that GAT‐2/3 operates in uptake mode. We conclude that EAATs via intracellular Na+ signaling and/or cell depolarization can govern the strength/direction of GAT‐mediated GABA transport.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号