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 31 毫秒
1.
Summary The voltage- and time-dependent K+ current,I K + out , elicited by depolarization of corn protoplasts, was inhibited by the addition of calcium channel antagonists (nitrendipine, nifedipine, verapamil, methoxyverapamil, bepridil, but not La3+) to the extracellular medium. These results suggested that the influx of external Ca2+ was necessary for K+ current activation. The IC50, concentration of inhibitor that caused 50% reduction of the current, for nitrendipine was 1 m at a test potential of +60 mV following a 20-min incubation period.In order to test whether intracellular Ca2+ actuated the K+ current, we altered either the Ca2+ buffering capacity or the free Ca2+ concentration of the intracellular medium (pipette filling solution). By these means,I K + out could be varied over a 10-fold range. Increasing the free Ca2+ concentration from 40 to 400nm also shifted the activation of the K+ current toward more negative potentials. Maintaining cytoplasmic Ca2+ at 500nm with 40nm EGTA resulted in a more rapid activation of the K+ current. Thus the normal rate of activation of this current may reflect changes in cytoplasmic Ca2+ on depolarization. Increasing intracellular Ca2+ to 500nm or 1 m also led to inactivation of the K+ current within a few minutes. It is concluded thatI K + out is regulated by cytosolic Ca2+, which is in turn controlled by Ca2+ influx through dihydropyridine-, and phenylalkylamine-sensitive channels.  相似文献   

2.
Outward and inward currents, mainly carried by K+, were detected in protoplasts of pollen grains (PG) and pollen tubes (PT) of Lilium longiflorum Thunb. by using the whole-cell configuration of the patch-clamp technique. The outward K+ current (IK+ out) was similar in both protoplast types, while the inward K+ current (IK+ in) was higher in pollen tube protoplasts. In PT but not in PG protoplasts, inward K+ currents were already detectable at negative membrane voltages usually monitored in lily pollen. IK+ in consisted of a slow and a fast current component, as revealed by fitting a sum of two exponential functions to the time-dependent current. The contribution of the fast component to the total inward current was higher in PT than in PG protoplasts, which was even more evident at acidic pH of the external medium. Therefore, based on the measured characteristics, the IK+ in of PT protoplasts may contribute to the endogenous K+ currents surrounding a growing pollen tube. Abbreviations: BS, bath solution; BTP, bis-Tris-propane; MES, 2-N-morpholinoethane sulfonic acid; Vact, activation voltage; VM, membrane voltage; Erev, reversal potential; IK+ in, inward K+ current; IK+ out, outward K+ current; PG, pollen grain; PT, pollen tube; PM, pipette medium  相似文献   

3.
During resorption of mineralized tissues, osteoclasts are exposed to marked changes in the concentration of extracellular Ca2+ and H+. We examined the effects of these cations on two types of K+ currents previously described in these cells. Whole-cell patch clamp recordings of membrane currents were made from osteoclasts freshly isolated from neonatal rats. In control saline (1 mm Ca2+, pH 7.4), the voltage-gated, outwardly rectifying K+ current activates at approximately 45 mV and the conductance is half-maximally activated at –29 mV (V 0.5). Increasing [Ca2+]out rapidly and reversibly shifted the current-voltage (I–V) relation to more positive potentials. Current at –29 mV decreased to 28 and 9% of control current at 5 and 10 mm [Ca2+]out, respectively. This effect of elevating [Ca2+]out was due to a positive shift of the K+ channel voltage activation range. Zn2+ or Ni2+ (5 to 500 m) also shifted the I–V relation to more positive potentials and had additional effects consistent with blockade of the K+ channel. Based on the extent to which these divalent cations affected the voltage activation range of the outwardly rectifying K+ current, the potency sequence was Zn2+ > Ni2+ > Ca2+. Lowering or raising extracellular pH also caused shifts of the voltage activation range to more positive or negative potentials, respectively. In contrast to their effects on the outwardly rectifying K+ current, changes in the concentration of extracellular H+ or Ca2+ did not shift the voltage activation range of the inwardly rectifying K+ current. These findings are consistent with Ca2+ and other cations affecting voltage-dependent gating of the osteoclast outwardly rectifying K+ channel through changes in surface charge.This work was supported by The Arthritis Society and the Medical Research Council of Canada. S.M.S. is supported by a Scientist Award and S.J.D. by a Development Grant from the Medical Research Council.  相似文献   

4.
Summary We examined the variability of occurrence and frequency of voltage-dependent whole-cell currents in human peripheral blood monocyte-derived macrophages (HMDM) maintained in culture for up to three weeks. An increase in cell capacitance from an average value of 9 pF on the day of isolation to 117 pF at 14 days accompanied growth and differentiation in culture. The average resting potential was approximately –34 mV for cells beyond two days in culture. Cells exhibited a voltage-and time-dependent outward current upon membrane depolarization above approximately –30 mV, which appeared to be composed of a number of separate currents with variable expression from donor to donor. Three of these currents are carried by K+. The frequency of each outward current type was calculated for 974 cells obtained from 36 donors. The HMDMs in these studies exhibited two 4-aminopyridine (4-AP) sensitive, time-dependent outward currents (I A andI B ) that could be differentiated on the basis of the presence or absence of steady-state inactivation in the physiological potential range, time course of inactivation during maintained depolarization, as well as threshold of activation. The 4-AP-insensitive outward current activated at approximately 10 mV. One component of the 4-AP insensitive-outward current (I C ) could be blocked by external TEA and by the exchange of internal Cs+ or Na+ for K+. The probability of observingI B andI C appeared to be donor dependent. Following total replacement of internal K+ with Cs+, two additional currents could be identified (i) a delayed component of outward current (I D ) remained which could be blocked by low concentrations of external Zn2+ (4 m) and was insensitive to anion replacement in the external solution and (ii) a Cl current with a reversal potential which shifted in the presence of external anion replacement and which was irreversibly inhibited by the stilbene SITS. The activation of a prominent time-independent inward currents was often observed with increasing hyperpolarization. This inward current was blocked by external Ba2+ and corresponded to the inwardly rectifying K+ current. Neither inward nor outward current expression appeared dependent on whether cells were differentiated in adherent or suspension culture nor was there demonstrable differential current expression observed upon transition from suspension to adherent form.  相似文献   

5.
The influence of cytosolic pH (pHi) in controlling K+-channel activity and its interaction with cytosolic-free Ca2+ concentration ([Ca2+]i) was examined in stomatal guard cells ofVicia faba L. Intact guard cells were impaled with multibarrelled microelectrodes and K+-channel currents were recorded under voltage clamp while pHi or [Ca2+]i was monitored concurrently by fluorescence ratio photometry using the fluorescent dyes 2,7-bis (2-carboxyethyl)-5(6)-carboxyfluorescein (BCECF) and Fura-2. In 10 mM external K+ concentration, current through inward-rectifying K+ channels (IK,in) was evoked on stepping the membrane from a holding potential of –100 mV to voltages from –120 to –250 mV. Challenge with 0.3-30 mM Na+-butyrate and Na+-acetate outside imposed acid loads, lowering pHi from a mean resting value of 7.64 ± 0.03 (n = 25) to values from 7.5 to 6.7. The effect on pHi was independent of the weak acid used, and indicated a H+-buffering capacity which rose from 90 mM H+/pH unit near 7.5 to 160 mM H+/pH unit near pHi 7.0. With acid-going pHi, (IK,in) was promoted in scalar fashion, the current increasing in magnitude with the acid load, but without significant effect on the current relaxation kinetics at voltages negative of –150 mV or the voltage-dependence for channel gating. Washout of the weak acid was followed by transient rise in pHi lasting 3–5 min and was accompanied by a reduction in (IK,in) before recovery of the initial resting pHi and current amplitude. The pHi-sensitivity of the current was consistent with a single, titratable site for H+ binding with a pKa near 6.3. Acid pHi loads also affected current through the outward-rectifying K+ channels (IK,out) in a manner antiparallel to (IK,in) The effect on IK, out was also scalar, but showed an apparent pKa of 7.4 and was best accommodated by a cooperative binding of two H+. Parallel measurements showed that Na+-butyrate loads were generally without significant effect on [Ca2+]i, except when pHi was reduced to 7.0 and below. Extreme acid loads evoked reversible increases in [Ca2+]i in roughly half the cells measured, although the effect was generally delayed with respect to the time course of pHi changes and K+-channel responses. The action on [Ca2+]i coincided with a greater variability in (IK,in) stimulation evident at pHi values around 7.0 and below, and with negative displacements in the voltage-dependence of (IK,in) gating. These results distinguish the actions of pHi and [Ca2+]i in modulating (IK,in) they delimit the effect of pHi to changes in current amplitude without influence on the voltage-dependence of channel gating; and they support a role for pHi as a second messenger capable of acting in parallel with, but independent of [Ca2+]i in controlling the K+ channels.Abbreviations BCECF 2,7-bis (2-carboxyethyl)-5(6)-carboxy fluorescein - [Ca2+]i cytosolic free Ca2+ concentration - gK ensemble (steady-state) K+-channel conductance - IK,out, IK,in outward-, inward-rectifying K+ channel (current) - IN current-voltage (relation) - Mes 2-(N-morpholinolethanesulfonic acid - pHi cytosolic pH - V membrane potential  相似文献   

6.
We have previously demonstrated the expression of BK channels in human glioma cells. There was a curious feature to the whole-cell currents of glioma cells seen during whole-cell patch-clamp: large, outward current transients accompanied repolarization of the cell membrane following an activating voltage step. This transient current, I transient, activated and inactivated rapidly (1 ms). The I-V relationship of I transient had features that were inconsistent with simple ionic current through open ion channels: (i) I transient amplitude peaked with a –80 mV voltage change and was invariant over a 200 mV range, and (ii) I transient remained large and outward at –140 mV. We provide evidence for a direct relationship of I transient to glioma BK currents. They had an identical time course of activation, identical pharmacology, identical voltage-dependence, and small, random variations in the amplitude of the steady-state BK current and I transient seen over time were often perfectly in phase. Substituting intracellular K+ with Cs+, Li+, or Na + ions reversibly reduced I transient and BK currents. I transient was not observed in recordings of other BK currents (hbr5 expressed in HEK cells and BK currents in rat neurons), suggesting I transient is unique to BK currents in human glioma cells. We conclude that I transient is generated by a mechanism related to the deactivation, and level of prior activation, of glioma BK channels. To account for these findings we propose that K+ ions are trapped within glioma BK channels during deactivation and are forced to exit to the extracellular side in a manner independent of membrane potential.  相似文献   

7.
Membrane potential and ionic currents were studied in cultured rabbit retinal pigment epithelial (RPE) cells using whole-cell patch clamp and perforated-patch recording techniques. RPE cells exhibited both outward and inward voltage-dependent currents and had a mean membrane capacitance of 26±12 pF (sd, n=92). The resting membrane potential averaged ?31±15 mV (n=37), but it was as high as ?60 mV in some cells. When K+ was the principal cation in the recording electrode, depolarization-activated outward currents were apparent in 91% of cells studied. Tail current analysis revealed that the outward currents were primarily K+ selective. The most frequently observed outward K+ current was a voltage- and time-dependent outward current (I K) which resembled the delayed rectifier K+ current described in other cells. I K was blocked by tetraethylammonium ions (TEA) and barium (Ba2+) and reduced by 4-aminopyridine (4-AP). In a few cells (3–4%), depolarization to ?50 mV or more negative potentials evoked an outwardly rectifying K+ current (I Kt) which showed more rapid inactivation at depolarized potentials. Inwardly rectifying K+ current (I KI) was also present in 41% of cells. I KI was blocked by extracellular Ba2+ or Cs+ and exhibited time-dependent decay, due to Na+ blockade, at negative potentials. We conclude that cultured rabbit RPE cells exhibit at least three voltage-dependent K+ currents. The K+ conductances reported here may provide conductive pathways important in maintaining ion and fluid homeostasis in the subretinal space.  相似文献   

8.
Proton (H+) conductive pathways are suggested to play roles in the regulation of intracellular pH. We characterized temperature-sensitive whole cell currents in mouse bone marrow–derived mast cells (BMMC), immature proliferating mast cells generated by in vitro culture. Heating from 24 to 36°C reversibly and repeatedly activated a voltage-dependent outward conductance with Q10 of 9.9 ± 3.1 (mean ± SD) (n = 6). Either a decrease in intracellular pH or an increase in extracellular pH enhanced the amplitude and shifted the activation voltage to more negative potentials. With acidic intracellular solutions (pH 5.5), the outward current was detected in some cells at 24°C and Q10 was 6.0 ± 2.6 (n = 9). The reversal potential was unaffected by changes in concentrations of major ionic constituents (K+, Cl, and Na+), but depended on the pH gradient, suggesting that H+ (equivalents) is a major ion species carrying the current. The H+ current was featured by slow activation kinetics upon membrane depolarization, and the activation time course was accelerated by increases in depolarization, elevating temperature and extracellular alkalization. The current was recorded even when ATP was removed from the intracellular solution, but the mean amplitude was smaller than that in the presence of ATP. The H+ current was reversibly inhibited by Zn2+ but not by bafilomycin A1, an inhibitor for a vacuolar type H+-ATPase. Macroscopic measurements of pH using a fluorescent dye (BCECF) revealed that a rapid recovery of intracellular pH from acid-load was attenuated by lowering temperature, addition of Zn2+, and depletion of extracellular K+, but not by bafilomycin A1. These results suggest that the H+ conductive pathway contributes to intracellular pH homeostasis of BMMC and that the high activation energy may be involved in enhancement of the H+ conductance.  相似文献   

9.
The activation by abscisic acid (ABA) of current through outward-rectifying K+ channels and its dependence on cytoplasmic pH (pHi) was examined in stomatal guard cells of Vicia faba L. Intact guard cells were impaled with multibarrelled and H+-selective microelectrodes to record membrane potentials and pHi during exposures to ABA and the weak acid butyrate. Potassium channel currents were monitored under voltage clamp and, in some experiments, guard cells were loaded with pH buffers by iontophoresis to suppress changes in pHi. Following impalements, stable pHi values ranged between 7.53 and 7.81 (7.67±0.04, n = 17). On adding 20 M ABA, pHi rose over periods of 5–8 min to values 0.27±0.03 pH units above the pHi before ABA addition, and declined slowly thereafter. Concurrent voltage-clamp measurements showed a parallel rise in the outward-rectifying K+ channel current (IK, out) and, once evoked, both pHi and IK, out responses were unaffected by ABA washout. Acid loads, imposed with external butyrate, abolished the ABA-evoked rise in IK, out. Butyrate concentrations of 10 and 30 mM (pH0 6.1) caused pHi to fall to values near 7.0 and below, both before and after adding ABA, consistent with a cytoplasmic buffer capacity of 128±12 mM per pH unit (n = 10) near neutrality. Butyrate washout was characterised by an appreciable alkaline overshoot in pHi and concomitant swell in the steady-state conductance of IK, out. The rise in pHi and iK, out in ABA were also virtually eliminated when guard cells were first loaded with pH buffers to raise the cytoplasmic buffer capacity four- to sixfold; however, buffer loading was without appreciable effect on the ABA-evoked inactivation of a second, inward-rectifying class of K+ channels (IK, in). The pHi dependence of IK, out was consistent with a cooperative binding of at least 2H+ (apparent pKa = 8.3) to achieve a voltage-independent block of the channel. These results establish a causal link previously implicated between cytoplasmic alkalinisation and the activation of IK, out in ABA and, thus, affirm a role for H+ in signalling and transport control in plants distinct from its function as a substrate in H+-coupled transport. Additional evidence implicates a coordinate control of IK, in by cytoplasmic-free [Ca2+] and pHi.Abbreviations ABA abscisic acid - [Ca2+]i cytoplasmic free [Ca2+]i - EK K+ equilibrium potential - IK, out, IK, in outward-, inward-rectifying K+ channel (current) - I-V current-voltage (relation) - Mes 2-(N-morpholino)ethanesulfonic acid - pHi cytoplasmic pH - Tes 2-{[2-hydroxy-1,1-bis(hydroxymethyl)ethyl]-amino}ethanesulfonic acid - Vm membrane potential We are grateful to G. Thiel (Pflanzenphysiologisches Institut, Universität Göttingen, Germany) for helpful discussions. This work was possible with equipment grants-in-aid from the Gatsby Charitable Foundation, the Royal Society and the University of London Central Research Fund. F.A. holds a Sainsbury Studentship.  相似文献   

10.
Simultaneous net uptake of Na+ and net extrusion of H+, both inhibited by amiloride, could be stimulated in red blood cells of the frog, Rana temporaria, either by intracellular acidification or cellular shrinkage. Net transports of Na+ and H+ were transient, dying out after 10–20 min (20°C) when stimulated by intracellular acidification but developing more slowly and proceeding for more than 60 min (20°C) when stimulated by cellular shrinkage. Evidence is presented suggesting a coupling between the transports of Na+ and H+ with an exchange ratio of 1:1 Na+/H+ exchange, stimulated by intracellular acidification, was able to readjust intracellular pH also when operating in parallel to a fully working anion exchanger in CO2/HCO 3 - -buffered media. Inhibition of anion exchange resulted in reduced cellular net uptake of Na+.Abbreviations DIDS 4,4-diisothiocyanatostilbene-2,2-disulphonate - DMSO dimethylsulphoxide - IU international unit - pH e extracellular pH - pH i intracellular pH - RBC red blood cell  相似文献   

11.
Extracellular pH was measured with a microelectrode positioned over the lower surface of singleLemna gibba plants. Upon addition of glucose, a transient extracellular alkalinization occurred. Saturated extracellular pH changes were observed with 5 mM glucose. Simultaneously, the membrane potential difference of –250 mV in the dark measured with intracellular glass micropipettes, trnasiently decreased by 105 mV. Uptake of [14C]glucose and extracellular alkalinization was enhanced by light whereas glucose-induced membrane-potential changes were reduced in the light and became even smaller with increasing the preillumination time. Glucose uptake was optimal at pH 6. The results are taken as further evidence in favor of H+-glucose cotransport inLemna.Dedicated to Professor W. Simonis on the occasion of his 70th birthdayUniversity of Missouri Agricultural Experiment Station Journal Series, paper No. 8266  相似文献   

12.
Summary Using intracellular microelectrode technique, we investigated the changes in membrane voltage (V) of cultured bovine pigmented ciliary epithelial cells induced by different extracellular solutions. (1)V in 213 cells under steady-state conditions averaged –46.1±0.6 mV (sem). (2) Increasing extracellular K+ concentration ([K+] o ) depolarizedV. Addition of Ba2+ could diminish this response. (3) Depolarization on doubling [K+] o was increased at higher [K+] o (or low voltage). (4) Removing extracellular Ca2+ decreasedV and reduced theV amplitude on increasing [K+] o . (5)V was pH sensitive. Extra-and intracellular acidification depolarizedV; alkalinization induced a hyperpolarization.V responses to high [K+] o were reduced at acidic extracellular pH. (6) Removing K o + depolarized, K o + readdition after K+ depletion transiently hyperpolarizedV. These responses were insensitive to Ba2+ but were abolished in the presence of ouabain or in Na+-free medium. (7) Na+ readdition after Na+ depletion transiently hyperpolarizedV. This reaction was markedly reduced in the presence of ouabain or in K+-free solution but unchanged by Ba2+. It is concluded that in cultured bovine pigmented ciliary epithelial cells K+ conductance depends on Ca2+, pH and [K+] o (or voltage). An electrogenic Na+/K+-transport is present, which is stimulated during recovery from K+ or Na+ depletion. This transport is inhibited by ouabain and in K+-or Na+-free medium.  相似文献   

13.
Voltage-gated whole-cell currents were recorded from cultured microglial cells which had been developed in the presence of the macrophage/microglial growth factor granulocyte/macrophage colony-stimulating factor. Outward K+ currents (I K) were most prominent in these cells. I Kcould be activated at potentials more positive than –40 mV. Half-maximal activation of I Kwas achieved at –13.8 mV and half-maximal inactivation of I Kwas determined at –33.8 mV. The recovery of I Kfrom inactivation was described by a time constant of 7.9 sec. For a tenfold change in extracellular K+ concentration the reversal potential of I Kshifted by 54 mV.Extracellularly applied 10 mm tetraethylammonium chloride reduced I K by about 50%, while 5 mm 4-aminopyridine almost completely abolished I K. Several divalent cations (Ba2+, Cd2+, Co2+, Zn2+) reduced current amplitudes and shifted the activation curve of I Kto more positive values. Charybdotoxin (IC50 = 1.14 nm) and noxiustoxin (IC50=0.89 nm) blocked I Kin a concentration-dependent manner, whereas dendrotoxin and mast cell degranulating peptide had no effect on the current amplitudes.  相似文献   

14.
Summary Patch-clamp studies of whole-cell ionic currents were carried out in parietal cells obtained by collagenase digestion of the gastric fundus of the guinea pig stomach. Applications of positive command pulses induced outward currents. The conductance became progressively augmented with increasing command voltages, exhibiting an outwardly rectifying current-voltage relation. The current displayed a slow time course for activation. In contrast, inward currents were activated upon hyperpolarizing voltage applications at more negative potentials than the equilibrium potential to K+ (E K). The inward currents showed time-dependent inactivation and an inwardly rectifying current-voltage relation. Tail currents elicited by voltage steps which had activated either outward or inward currents reversed at nearE K, indicating that both time-dependent and voltagegated currents were due to K+ conductances. Both outward and inward K+ currents were suppressed by extracellular application of Ba2+, but little affected by quinine. Tetraethylammonium inhibited the outward current without impairing the inward current, whereas Cs+ blocked the inward current but not the outward current. The conductance of inward K+ currents, but not outward K+ currents, became larger with increasing extracellular K+ concentration. A Ca2+-mobilizing acid secretagogue, carbachol, and a Ca2+ ionophore, ionomycin, brought about activation of another type of outward K+ currents and voltage-independent cation currents. Both currents were abolished by cytosolic Ca2+ chelation. Quinine preferentially inhibited this K+ current. It is concluded that resting parietal cells of the guinea pig have two distinct types of voltage-dependent K+ channels, inward rectifier and outward rectifier, and that the cells have Ca2+-activated K+ channels which might be involved in acid secretion under stimulation by Ca2+-mobilizing secretagogues.  相似文献   

15.
The ionic fluxes associated with the ATP-dependent acidification of endocytic vesicles were studied in a preparation isolated from rabbit reticulocytes enriched for transferrin-transferrin receptor complexes. No vesicle acidification was observed in the absence of intra- and extravesicular ions (sucrosein/sucroseout), while maximal acidification was observed with NaClin/KClout·K in + was a poor substitute for Na in + , and Cl out could be replaced by other anions with the following efficacy of acidification: Cl>Br>I>PO 4 3– >gluconate>SO 4 2– . Flux studies using36Cl and22Na+ showed that the vesicles had a permeability for Cl and Na+, and that ATP-dependent H+ pumping was accompanied by a net influx of Cl and a net efflux of Na+ provided that there was a Na+ concentration gradient. After 3 mins, the time necessary to maximal acidification, the electrical charge generated by the entrance of H+ was countered to about 45% by the Cl influx and to about 42% by the Na+ efflux. These studies demonstrated that both Cl and Na+ fluxes are necessary for optimal endocytic vesicle acidification.  相似文献   

16.
The initial response of coleoptile cells to growth hormones and light is a rapid change in plasma-membrane polarization. We have isolated protoplasts from the cortex of maize (Zea mays L.) coleoptiles to study the electrical properties of their plasma membrane by the patch-clamp techniqueUsing the whole-cell configuration and cell-free membrane patches we could identify an H+-ATPase, hyperpolarizing the membrane potential often more negative than -150 mV, and a voltage-dependent, inward-rectifying K+ channel (unit conductance 5–7 pS) as the major membrane conductan-ces Potassium currents through this channel named CKC1in (for Coleoptile K + Channel inward rectifier) were elicited upon voltage steps negative to -80 mV, characterized by a half-activation potential of -112 mV. The kinetics of activation, well described by a double-exponential process, were strongly dependent on the degree of hyperpolarization and the cytoplasmic Ca2+ level. Whereas at nanomolar Ca2+ concentrations K+ currents increased with a t1/2=16 ms (at -180 mV), higher calcium levels slowed the activation process about fourto fivefoldUpon changes in the extracellular K+ concentration the reversal potential of the K+ channel followed the Nernst potential for potassium with a 56-mV shift for a tenfold increaseThe absence of a measurable conductance for Na+, Rb+, Cs+ and a permeability ratio PNH 4 + /PK+ around 0.25 underlines the high selectivity of CKC1in for K+In contrast to Cs+, which at submillimolar concentration blocks the channel in a voltage-dependent manner, Rb+, often used as a tracer for K+, does not permeate this type of K+ channelThe lack of Rb+ permeability is unique with respect to other K+ transporters. Therefore, future molecular analysis of CKC1in, considered as a unique variation of plant inward rectifiers, might help to understand the permeation properties of K+ channels in general.Abbreviations CKC1in Coleoptile K + Channel inward rectifier - U membrane voltage - Iss steady-state currents - Itail tail currents Experiments were conducted in the laboratory of F.G. during the stay of RHas a guest professor sponsored by Special Project RAISA, subproject N2.1, paper N2155.  相似文献   

17.
Previous studies in chick embryo cardiac myocytes have shown that the inhibition of Na+/K+-ATPase with ouabain induces cell shrinkage in an isosmotic environment (290 mOsm). The same inhibition produces an enhanced RVD (regulatory volume decrease) in hyposmotic conditions (100 mOsm). It is also known that submitting chick embryo cardiomyocytes to a hyperosmotic solution induces shrinkage and a concurrent intracellular alkalization. The objective of this study was to evaluate the involvement of intracellular pH (pHi), intracellular Ca2+ ([Ca2+]i) and Na+/K+-ATPase inhibition during hyposmotic swelling. Changes in intracellular pH and Ca2+ were monitored using BCECF and fura-2, respectively. The addition of ouabain (100 M) under both isosmotic and hyposmotic stimuli resulted in a large increase in [Ca2+]i (200%). A decrease in pHi (from 7.3 ± 0.09 to 6.4 ± 0.08, n = 6; p < 0.05) was only observed when ouabain was applied during hyposmotic swelling. This acidification was prevented by the removal of extracellular Ca2+. Inhibition of Na+/H2+ exchange with amiloride (1 mM) had no effect on the ouabain-induced acidification. Preventing the mitochondrial accumulation of Ca2+ using CCCP (10 M) resulted in a blockade of the progressive acidification normally induced by ouabain. The inhibition of mitochondrial membrane K+/H+ exchange with DCCD (1 mM) also completely prevented the acidification. Our results suggest that intracellular acidification upon cell swelling is mediated by an initial Ca2+ influx via Na+/Ca2+ exchange, which under hyposmotic conditions activates the K+ and Ca2+ mitochondrial exchange systems (K+/H+ and Ca2+/H+).Deceased  相似文献   

18.
Using the two-microelectrode voltage clamp technique in Xenopus laevis oocytes, we estimated Na+-K+-ATPase activity from the dihydroouabain-sensitive current (I DHO) in the presence of increasing concentrations of tetraethylammonium (TEA+; 0, 5, 10, 20, 40 mm), a well-known blocker of K+ channels. The effects of TEA+ on the total oocyte currents could be separated into two distinct parts: generation of a nonsaturating inward current increasing with negative membrane potentials (V M) and a saturable inhibitory component affecting an outward current easily detectable at positive V M. The nonsaturating component appears to be a barium-sensitive electrodiffusion of TEA+ which can be described by the Goldman-Hodgkin-Katz equation, while the saturating component is consistent with the expected blocking effect of TEA+ on K+ channels. Interestingly, this latter component disappears when the Na+-K+-ATPase is inhibited by 10 m DHO. Conversely, TEA+ inhibits a component of I DHO with a k d of 25±4 mm at +50 mV. As the TEA+-sensitive current present in I DHO reversed at –75 mV, we hypothesized that it could come from an inhibition of K+ channels whose activity varies in parallel with the Na+-K+-ATPase activity. Supporting this hypothesis, the inward portion of this TEA+-sensitive current can be completely abolished by the addition of 1 mm Ba2+ to the bath. This study suggests that, in X. laevis oocytes, a close link exists between the Na-K-ATPase activity and TEA+-sensitive K+ currents and indicates that, in the absence of effective K+ channel inhibitors, I DHO does not exclusively represent the Na+-K+-ATPase-generated current.  相似文献   

19.
Using the voltage-clamp technique, we investigated transmembrane ion currents in isolated smooth muscle cells of the guinea pigtaenia coli. In our study, we identified and studied a charibdotoxin-sensitive component of Ca2+-dependent K+ current carried through the channels of high conductance (in most publications called “big conductance,”I BK(Ca)). This component was completely blocked by 100 nM charibdotoxin and by tetraethylammonium in concentrations as low as 1 mM.I BK(Ca) demonstrated fast kinetics of inactivation, which nearly coincided with that of Ca2+ current. In addition to the dependence on Ca2+ concentration, this current also showed voltage-dependent properties: with a rise in the level of depolarization its amplitude increased. In many cells, depolarizing shifts in the membrane potential evoke spontaneous outward currents. Such currents probably represent the secondary effect of cyclic Ca2+ release from the caffeine-sensitive intracellular stores that result in short-term activation of charibdotoxin-sensitive Ca2+-dependent K+ channels.  相似文献   

20.
The efficacy and mechanism of -dendrotoxin (DTX) block of K+ channel currents in Vicia stomatal guard cells was examined. Currents carried by inward- and outward-rectifying K+ channels were determined under voltage clamp in intact guard cells, and block was characterized as a function of DTX and external K+ (K+) concentrations. Added to the bath, 0.1-30 nM DTX blocked the inward-rectifying K+ current (IK,in), but was ineffective in blocking current through the outward-rectifying K+ channels (IK,out) even at concentrations of 30 nM. DTX block was independent of clamp voltage and had no significant effect on the voltage-dependent kinetics for IK,in, neither altering its activation at voltages negative of –120 mV nor its deactivation at more positive voltages. No evidence was found for a use dependence to DTX action. Block of IK,in followed a simple titration function with an apparent K1/2 for block of 2.2 nM in 3 mm K o + . However, DTX block was dependent on the external K+ concentration. Raising K+ from 3 to 30 mm slowed block and resulted in a 60–70% reduction in its efficacy (apparent K i = 10 mm in 10 nm DTX). The effect of K+ in protecting I K,in was competitive with DTX and specific for permeant cations. A joint analysis of IK,in block with DTX and K+ concentration was consistent with a single class of binding sites with a K d for DTX of 240 pm. A K d of 410 m for extracellular K+ was also indicated. These results complement previous studies implicating a binding site requiring extracellular K+ (K1/2 1 mm) for IK,in activation; they parallel features of K+ channel block by DTX and related peptide toxins in many animal cells, demonstrating the sensitivity of plant plasma membrane K+ channels to nanomolar toxin concentrations under physiological conditions; the data also highlight one main difference: in the guard cells, DTX action appears specific to the K+ inward rectifier.We thank J.O. Dolly (Imperial, London) and S.M. Jarvis (University of Kent, Canterbury) for several helpful discussions. This work was supported by SERC grant GR/H07696 and was aided by equipment grants from the Gatsby Foundation, the Royal Society and the University of London Central Research Fund. G.O. was supported by an Ausbildungsstipendium (OB 85/1-1) from the Deutsche Forschungsgemeinschaft. F.A. holds a Sainsbury Studentship.  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