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 15 毫秒
1.
Initial clearance of extracellular K+ ([K+]o) following neuronal excitation occurs by astrocytic uptake, because elevated [K+]o activates astrocytic but not neuronal Na+,K+-ATPases. Subsequently, astrocytic K+ is re-released via Kir4.1 channels after distribution in the astrocytic functional syncytium via gap junctions. The dispersal ensures widespread release, preventing renewed [K+]o increase and allowing neuronal Na+,K+-ATPase-mediated re-uptake. Na+,K+-ATPase operation creates extracellular hypertonicity and cell shrinkage which is reversed by the astrocytic cotransporter NKCC1. Inhibition of Kir channels by activation of specific PKC isotypes may decrease syncytial distribution and enable physiologically occurring [K+]o increases to open L-channels for Ca2+, activating [K+]o-stimulated gliotransmitter release and regulating gap junctions. Learning is impaired when [K+]o is decreased to levels mainly affecting astrocytic membrane potential or Na+,K+-ATPase or by abnormalities in its α2 subunit. It is enhanced by NKCC1-mediated ion and water uptake during the undershoot, reversing neuronal inactivity, but impaired in migraine with aura in which [K+]o is highly increased. Vasopressin augments NKCC1 effects and facilitates learning. Enhanced myelination, facilitated by astrocytic-oligodendrocytic gap junctions also promotes learning.  相似文献   

2.
Nanna MacAulay 《Glia》2020,68(11):2192-2211
Neuronal signaling in the central nervous system (CNS) associates with release of K+ into the extracellular space resulting in transient increases in [K+]o. This elevated K+ is swiftly removed, in part, via uptake by neighboring glia cells. This process occurs in parallel to the [K+]o elevation and glia cells thus act as K+ sinks during the neuronal activity, while releasing it at the termination of the pulse. The molecular transport mechanisms governing this glial K+ absorption remain a point of debate. Passive distribution of K+ via Kir4.1-mediated spatial buffering of K+ has become a favorite within the glial field, although evidence for a quantitatively significant contribution from this ion channel to K+ clearance from the extracellular space is sparse. The Na+/K+-ATPase, but not the Na+/K+/Cl cotransporter, NKCC1, shapes the activity-evoked K+ transient. The different isoform combinations of the Na+/K+-ATPase expressed in glia cells and neurons display different kinetic characteristics and are thereby distinctly geared toward their temporal and quantitative contribution to K+ clearance. The glia cell swelling occurring with the K+ transient was long assumed to be directly associated with K+ uptake and/or AQP4, although accumulating evidence suggests that they are not. Rather, activation of bicarbonate- and lactate transporters appear to lead to glial cell swelling via the activity-evoked alkaline transient, K+-mediated glial depolarization, and metabolic demand. This review covers evidence, or lack thereof, accumulated over the last half century on the molecular mechanisms supporting activity-evoked K+ and extracellular space dynamics.  相似文献   

3.
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   

4.
The cotransporter of Na+, K+, 2Cl, and water, NKKC1, is activated under two conditions in the brain, exposure to highly elevated extracellular K+ concentrations, causing astrocytic swelling, and regulatory volume increase in cells shrunk in response to exposure to hypertonic medium. NKCC1‐mediated transport occurs as secondary active transport driven by Na+/K+‐ATPase activity, which establishes a favorable ratio for NKCC1 operation between extracellular and intracellular products of the concentrations of Na+, K+, and Cl × Cl. In the adult brain, astrocytes are the main target for NKCC1 stimulation, and their Na+/K+‐ATPase activity is stimulated by elevated K+ or the β‐adrenergic agonist isoproterenol. Extracellular K+ concentration is normal during regulatory volume increase, so this study investigated whether the volume increase occurred faster in the presence of isoproterenol. Measurement of cell volume via live cell microscopic imaging fluorescence to record fluorescence intensity of calcein showed that this was the case at isoproterenol concentrations of ≥1 µM in well‐differentiated mouse astrocyte cultures incubated in isotonic medium with 100 mM sucrose added. This stimulation was abolished by the β1‐adrenergic antagonist betaxolol, but not by ICI118551, a β2‐adrenergic antagonist. A large part of the β1‐adrenergic signaling pathway in astrocytes is known. Inhibitors of this pathway as well as the glycogenolysis inhibitor 1,4‐dideoxy‐1,4‐imino‐D‐arabinitol hydrochloride and the NKCC1 inhibitors bumetanide and furosemide abolished stimulation by isoproterenol, and it was weakened by the Na+/K+‐ATPase inhibitor ouabain. These observations are of physiological relevance because extracellular hypertonicity occurs during intense neuronal activity. This might trigger a regulatory volume increase, associated with the post‐excitatory undershoot. © 2014 Wiley Periodicals, Inc.  相似文献   

5.
Hypotonicity following water intoxication and/or salt loss leads to mainly astrocytic brain swelling. Astrocytic swelling also occurs following brain trauma or ischemia, together with an increase in extracellular K+ ([K+]o), stimulating a bumetanide/furosemide/ethacrynic acid‐inhibitable cotransporter, NKCC1, that accumulates Na+ and K+ together with 2 Cl and osmotically obliged water. Either type of swelling may become fatal and is associated with phosphorylation of extracellular regulated kinases 1 and 2 (ERK1/2). Only the swelling associated with elevated [K+]o, leads to an increase in astrocytic proliferation and in expression of the astrocytic marker, glial fibrillary acidic protein. These differences prompted us to investigate key aspects of the molecular pathways between hypotonicity‐induced and high‐K+‐mediated swelling in primary cultures of mouse astrocytes. In the latter Ca2+‐mediated, AG1478‐inhibitable transactivation of the epidermal growth factor (EGF) receptor leads, via bumetanide‐inhibitable activation of the mitogen activated protein (MAP) kinase pathway to ERK phosphorylation and to NKCC1‐mediated swelling. In the former, inhibition of the MAP kinase pathway, but not of EGF receptor activation, abolishes ERK phosphorylation, but has no effect on swelling, indicating that activation of ERK is a result, not a cause, of the swelling.  相似文献   

6.
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   

7.
Changes in extracellular potassium ([K+]e) modulate neuronal networks via changes in membrane potential, voltage-gated channel activity, and alteration to transmission at the synapse. Given the limited extracellular space in the central nervous system, potassium clearance is crucial. As activity-induced potassium transients are rapidly managed by astrocytic Kir4.1 and astrocyte-specific Na+/K+-ATPase, any neurotransmitter/neuromodulator that can regulate their function may have indirect influence on network activity. Neuromodulators differentially affect cortical/thalamic networks to align sensory processing with differing behavioral states. Given serotonin (5HT), norepinephrine (NE), and acetylcholine (ACh) differentially affect spike frequency adaptation and signal fidelity (“signal-to-noise”) in somatosensory cortex, we hypothesize that [K+]e may be differentially regulated by the different neuromodulators to exert their individual effects on network function. This study aimed to compare effects of individually applied 5HT, NE, and ACh on regulating [K+]e in connection to effects on cortical-evoked response amplitude and adaptation in male mice. Using extracellular field and K+ ion-selective recordings of somatosensory stimulation, we found that differential effects of 5HT, NE, and ACh on [K+]e regulation mirrored differential effects on amplitude and adaptation. 5HT effects on transient K+ recovery, adaptation, and field post-synaptic potential amplitude were disrupted by barium (200 µM), whereas NE and ACh effects were disrupted by ouabain (1 µM) or iodoacetate (100 µM). Considering the impact [K+]e can have on many network functions; it seems highly efficient that neuromodulators regulate [K+]e to exert their many effects. This study provides functional significance for astrocyte-mediated buffering of [K+]e in neuromodulator-mediated shaping of cortical network activity.  相似文献   

8.
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   

9.
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   

10.
Ion-sensitive microelectrodes were used to study the behavior of extracellular ions in rat sciatic nerve during and following activity. Nerve stimulation produced increases in [K+]o that were dependent upon the frequency and duration of stimulation; no change in extracellular pH occurred with stimulation. Increases in [K+]o dependent on axonal discharge since they were blocked by inhibiting sodium channels with tetrodotoxin. At 22°C, stimulation could induce increases in [K+]o of several mM; at 36°C, stimulation rarely produced increases in [K+]o greater than 1mM. Stimulated increases in [K+]o dissipated very slowly (i.e. t1/2 = 50–100s) and the rate of dissipation was not significantly affected by anoxia, changes in temperature, changes in extracellular pH, or the application of a blocker of Na+, K+-ATPase (ouabain) or a K+ channel blocker (Ba2+). In comparison to the central nervous system, neural activity in rat sciatic nerve produced smaller increases in [K+]o and these increases dissipated much more slowly. The primary mechanism of K+ dissipation appeared to be diffusion, probably facilitated by the larger extracellular space in peripheral nerve compared to the central nervous system, but impeded by diffusion barriers imposed by the blood-nerve barrier.  相似文献   

11.
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   

12.
We examined the hypotheses that aging alters the capacity of brain tissue to regulate extracellular K+activity (Ko+), and that age-related decreases in glucose metabolism may underlie these alterations. Hippocampal slices from young adult (6–9 months old), middle-aged (16–19 months old), and aged (26–29 months old) Fischer 344 rats were exposed to physiological solutions containing 5–20 mM glucose maintained at 36–37°C. Schaffer collaterals in each slice were stimulated at 40 Hz for 2 s, and the resulting changes in Ko+ were recorded with Ko+-sensitive microelectrodes placed in stratum pyramidale of hippocampal subfield CA1. We found that K+ clearance from the extracellular space of hippocampal slices was significantly slowed in the middle-aged group compared with the young adult group in physiological solutions containing 5 and 10 mM glucose. Age-related differences in Ko+ clearance disappeared in 20 mM glucose. Also, the rate of Ko+ clearance was modified by glucose concentration. These results suggest that K+ transport rates are modified by age, and that age-related alterations in glucose metabolism may be involved.  相似文献   

13.
Sodium homeostasis is at the center stage of astrocyte (and brain) physiology because the large inwardly directed Na+ gradient provides the energy for transport of ions, neurotransmitters, amino acids and many other molecules across the plasmalemma and endomembranes. Cell imaging with commercially available chemical indicators allows analysis of dynamic changes in intracellular Na+ concentration (Na+]i), albeit further technological developments, such as genetically‐controlled or membrane targeted indicators or dyes usable for advanced microscopy (such as fluorescence‐lifetime imaging microscopy) are urgently needed. Thus, important questions related to the existence of Na+ gradients between different cellular compartments or occurrence of localised Na+ micro/nanodomains at the plasma membrane remain debatable. Extrusion of Na+ (and hence Na+ homeostasis) in astrocytes is mediated by the ubiquitously expressed Na+/K+‐ATPase (NKA), the major energy consumer of the brain. The activity of the NKA is counteracted by constant constitutive influx of Na+ through transporters such as the NKCC1 (Na+‐K+?2Cl‐co‐transport) or the NBC (Na+?2 ‐co‐transport). In addition, Na+‐permeable ion channels at the plasma membrane as well as Na+‐dependent solute carrier transporters provide for Na+ influx into astrocytes. Activation of these pathways in response to neuronal activity results in an increase of [Na+]i in astrocytes and there is manifold evidence for diverse signalling functions of these [Na+]i transients. Thus, in addition to its established homeostatic functions, activity dependent fluctuations of astrocyte [Na+]i regulate signalling cascades by feeding back on Na+‐dependent transporters. The Na+ signalling system may be ideally placed for fast coordinating signalling between neuronal activity and glial “homeostatic” Na+‐dependent transporters. GLIA 2016;64:1611–1627  相似文献   

14.
Seigneur J  Timofeev I 《Epilepsia》2011,52(1):132-139
Purpose: Seizures are associated with a reduction in extracellular Ca2+ concentration ([Ca2+]o) and an increase in extracellular K+ concentration ([K+]o). The long‐range synchrony observed between distant electrodes during seizures is weak. We hypothesized that changes in extracellular ionic conditions during seizures are sufficient to alter synaptic neuronal responses and synchrony in the neocortex. Methods: We obtained in vivo and in vitro electrophysiologic recordings combined with microstimulation from cat/rat neocortical neurons during seizures and seizure‐like ionic conditions. In vitro the [K+]o was 2.8, 6.25, 8.0, and 12 mm and the [Ca2+]o was 1.2 and 0.6 mm . Key Findings: During seizures recorded in vivo, we observed abolition of evoked synaptic responses. In vitro, the membrane potential of both regular‐spiking and fast‐spiking neurons was depolarized in high [K+]o conditions and hyperpolarized in high [Ca2+]o conditions. During high [K+]o conditions, changes in [Ca2+]o did not affect membrane potential. The synaptic responsiveness of both regular‐spiking and fast‐spiking neurons was reduced during seizure‐like ionic conditions. A reduction in [Ca2+]o to 0.6 mm increased failure rates but did not abolish responses. However, an increase in [K+]o to 12 mm abolished postsynaptic responses, which depended on a blockade in axonal spike propagation. Significance: We conclude that concomitant changes in [K+]o and [Ca2+]o observed during seizures contribute largely to the alterations of synaptic neuronal responses and to the decrease in long‐range synchrony during neocortical seizures.  相似文献   

15.
16.
Purpose: KCNJ10 encodes subunits of inward rectifying potassium (Kir) channel Kir4.1 found predominantly in glial cells within the brain. Genetic inactivation of these channels in glia impairs extracellular K+ and glutamate clearance and produces a seizure phenotype. In both mice and humans, polymorphisms and mutations in the KCNJ10 gene have been associated with seizure susceptibility. The purpose of the present study was to determine whether there are differences in Kir channel activity and potassium‐ and glutamate‐buffering capabilities between astrocytes from seizure resistant C57BL/6 (B6) and seizure susceptible DBA/2 (D2) mice that are consistent with an altered K+ channel activity as a result of genetic polymorphism of KCNJ10. Methods: Using cultured astrocytes and hippocampal brain slices together with whole‐cell patch‐clamp, we determined the electrophysiologic properties, particularly K+ conductances, of B6 and D2 mouse astrocytes. Using a colorimetric assay, we determined glutamate clearance capacity by B6 and D2 astrocytes. Results: Barium‐sensitive Kir currents elicited from B6 astrocytes are substantially larger than those elicited from D2 astrocytes. In addition, potassium and glutamate buffering by D2 cortical astrocytes is impaired, relative to buffering by B6 astrocytes. Discussion: In summary, the activity of Kir4.1 channels differs between seizure‐susceptible D2 and seizure‐resistant B6 mice. Reduced activity of Kir4.1 channels in astrocytes of D2 mice is associated with deficits in potassium and glutamate buffering. These deficits may, in part, explain the relatively low seizure threshold of D2 mice.  相似文献   

17.
Cultured astrocytes and cell lines derived therefrom maintain a high energy level ([ATP]/[ADP]) through operation of oxidative phosphorylation and glycolysis. The contribution from the latter to total ATP production is 25–32%. A powerful Na+/K+ pump maintains potassium, sodium, and calcium gradients out of equilibrium. [Na+]i is about 20 mM, [K+]i is 130 mM and [Ca2+]i is less than 100 nM. Under non-stimulated conditions, the Na+/K+ ATPase consumes 20% of astrocytic ATP production. Inhibition of the pump by ouabain decreases energy expenditure, raises [creatine phosphate]/[creatine], and leads to a leakage of sodium, potassium, and calcium ions. Decrease in the pump function via a fall in [ATP] also collapses ion gradients; the rate and extent of the fall correlates positively with cellular energy state. Under “normal” conditions (i.e., when ATP production pathways are not inhibited), there appears to be no preferential utilization of energy produced by either glycolysis or oxidative phosphorylation for the support of pump function. GLIA 21:35–45, 1997. © 1997 Wiley-Liss, Inc.  相似文献   

18.
Patch-clamp recordings were made on rat hippocampal neurons maintained in culture. In cell-attached and excised inside-out and outside-out patches a large single-channel current was observed. This channel had a conductance of 220 and 100 pS in 140 mM [K+]i/140 mM [K+]o and 140 mM [K+]i/3 mM [K+]o respectively. From the reversal potential the channel was highly selective for K+, the PK+/Pna+ ratio being 50/1. Channel activity was voltage-dependent, the open probability at 100 mM [Ca2+]i increasing by e-fold for a 22 mV depolarization. It was also dependent on [Ca2+]i at both resting and depolarized membrane potentials. Channel open states were best described by the sum of two exponentials with time constants that increased as the membrane potential became more positive. Channel activity was sensitive to both external (500 μM) and internal (5 mM) tetraethylammonium chloride. These data are consistent with the properties of maxi-K+ channels described in other preparations, and further suggest a role for maxi-channel activity in regulating neuronal excitability at the resting membrane potential. Channel activity was not altered by 8-chlorophenyl thio cAMP, concanavalin A, pH reduction or neuraminidase. In two of five patches lemakalim (BRL 38227) increased channel activity. Internal ruthenium red (10 μM) blocked the channel by shortening the duration of both open states. This change in channel gating was distinct from the ‘mode switching’ seen in two patches, where a channel switched spontaneously from normal activity typified by two open states to a mode where only short openings were represented.  相似文献   

19.
We have recently demonstrated that endothelin (ET) is functionally coupled to Nax, a Na+ concentration‐sensitive Na+ channel for lactate release via ET receptor type B (ETBR) and is involved in peripheral nerve regeneration in a sciatic nerve transection–regeneration mouse model. Nax is known to interact directly with Na+/K+‐ATPase, leading to lactate production in the brain. To investigate the role of Na+/K+‐ATPase in peripheral nerve regeneration, in this study, we applied ouabain, a Na+/K+‐ATPase inhibitor, to the cut site for 4 weeks with an osmotic pump. While functional recovery and nerve reinnervation to the toe started at 5 weeks after axotomy and were completed by 7 weeks, ouabain delayed them by 2 weeks. The delay by ouabain was improved by lactate, and its effect was blocked by α‐cyano‐4‐hydroxy‐cinnamic acid (CIN), a broad monocarboxylate transporter (MCT) inhibitor. In primary cultures of dorsal root ganglia, neurite outgrowth of neurons and lactate release into the culture medium was inhibited by ouabain. Conversely, lactate enhanced the neurite outgrowth, which was blocked by CIN, but not by AR‐C155858, a MCT1/2‐selective inhibitor. ET‐1 and ET‐3 increased neurite outgrowth of neurons, which was attenuated by an ETBR antagonist, ouabain and 2 protein kinase C inhibitors. Taken together with the finding that ETBR was expressed in Schwann cells, these results demonstrate that ET enhanced neurite outgrowth of neurons mediated by Na+/K+‐ATPase via ETBR in Schwann cells. This study suggests that Na+/K+‐ATPase coupled to the ET‐ETBR system plays a critical role in peripheral nerve regeneration via lactate signalling.  相似文献   

20.
High-frequency fatigue (HFF), the decline of force during continuous tetanic stimulation (lasting 4–40 s), was studied in isolated bundles of rat skeletal muscle fibers. HFF was slower in slow-twitch soleus fibers than in fast-twitch red or white sternomastoid fibers; denervation accelerated fatigue in soleus. Maximal 200-mmol/L potassium contractures of normal amplitude were induced in fatigued fibers, suggesting that crossbridge cycling and the voltage activation of excitation–contraction coupling could still occur maximally, but that activation by action potentials was impaired. An increase in [Na+]o slowed HFF, while a small increase in [K+]o or reduction in [Cl?]o accelerated HFF. Increasing the tetanic stimulation frequency exacerbated fatigue. Recovery from HFF proceeded rapidly since force increased markedly within a few seconds when stimulation ceased. These results support the hypothesis that a redistribution of Na+, K+, and Cl? across the transverse tubular membranes during repeated action potential activity induces fatigue by reducing the amplitude and conduction of action potentials. © 1995 John Wiley & Sons, Inc.  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