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.
Potassium channels are highly selective for K+ over the smaller Na+. Intriguingly, they are permeable to larger monovalent cations such as Rb+ and Cs+ but are specifically blocked by the similarly sized Ba2+. In this study, we used structural analysis to determine the binding profiles for these permeant and blocking ions in the selectivity filter of the potassium-selective NaK channel mutant NaK2K and also performed permeation experiments using single-channel recordings. Our data revealed that some ion binding properties of NaK2K are distinct from those of the canonical K+ channels KcsA and MthK. Rb+ bound at sites 1, 3, and 4 in NaK2K, as it does in KcsA. Cs+, however, bound predominantly at sites 1 and 3 in NaK2K, whereas it binds at sites 1, 3, and 4 in KcsA. Moreover, Ba2+ binding in NaK2K was distinct from that which has been observed in KcsA and MthK, even though all of these channels show similar Ba2+ block. In the presence of K+, Ba2+ bound to the NaK2K channel at site 3 in conjunction with a K+ at site 1; this led to a prolonged block of the channel (the external K+-dependent Ba2+ lock-in state). In the absence of K+, however, Ba2+ acts as a permeating blocker. We found that, under these conditions, Ba2+ bound at sites 1 or 0 as well as site 3, allowing it to enter the filter from the intracellular side and exit from the extracellular side. The difference in the Ba2+ binding profile in the presence and absence of K+ thus provides a structural explanation for the short and prolonged Ba2+ block observed in NaK2K.  相似文献   

2.
The movement and interaction of multiple ions passing through in single file underlie various fundamental K+ channel properties, from the effective conduction of K+ ions to channel blockade by Ba2+ ions. In this study, we used single-channel electrophysiology and x-ray crystallography to probe the interactions of Ba2+ with permeant ions within the ion conduction pathway of the MthK K+ channel. We found that, as typical of K+ channels, the MthK channel was blocked by Ba2+ at the internal side, and the Ba2+-blocking effect was enhanced by external K+. We also obtained crystal structures of the MthK K+ channel pore in both Ba2+–Na+ and Ba2+–K+ environments. In the Ba2+–Na+ environment, we found that a single Ba2+ ion remained bound in the selectivity filter, preferably at site 2, whereas in the Ba2+–K+ environment, Ba2+ ions were predominantly distributed between sites 3 and 4. These ionic configurations are remarkably consistent with the functional studies and identify a molecular basis for Ba2+ blockade of K+ channels.  相似文献   

3.
Ba2+ block of large conductance Ca2+-activated K+ channels was studied in patches of membrane excised from cultures of rat skeletal muscle using the patch clamp technique. Under conditions in which a blocking Ba2+ ion would dissociate to the external solution (150 mM N-methyl-d-glucamine+ o, 500 mM K+ i, 10 μM Ba2+ i, +30 mV, and 100 μM Ca2+ i to fully activate the channel), Ba2+ blocks with a mean duration of ∼2 s occurred, on average, once every ∼100 ms of channel open time. Of these Ba2+ blocks, 78% terminated with a single step in the current to the fully open level and 22% terminated with a transition to a subconductance level at ∼0.26 of the fully open level (preopening) before stepping to the fully open level. Only one apparent preclosing was observed in ∼10,000 Ba2+ blocks. Thus, the preopenings represent Ba2+-induced time-irreversible subconductance gating. The fraction of Ba2+ blocks terminating with a preopening and the duration of preopenings (exponentially distributed, mean = 0.75 ms) appeared independent of changes in [Ba2+]i or membrane potential. The fractional conductance of the preopenings increased from 0.24 at +10 mV to 0.39 at +90 mV. In contrast, the average subconductance level during normal gating in the absence of Ba2+ was independent of membrane potential, suggesting different mechanisms for preopenings and normal subconductance levels. Preopenings were also observed with 10 mM Ba2+ o and no added Ba2+ i. Adding K+, Rb+, or Na+ to the external solution decreased the fraction of Ba2+ blocks with preopenings, with K+ and Rb+ being more effective than Na+. These results are consistent with models in which the blocking Ba2+ ion either induces a preopening gate, and then dissociates to the external solution, or moves to a site located on the external side of the Ba2+ blocking site and acts directly as the preopening gate.  相似文献   

4.
Potassium channels selectively conduct K+ ions across cell membranes and have key roles in cell excitability. Their opening and closing can be spontaneous or controlled by membrane voltage or ligand binding. We used Ba2+ as a probe to determine the location of the ligand-sensitive gate in an inwardly rectifying K+ channel (Kir6.2). To a K+ channel, Ba2+ and K+ are of similar sizes, but Ba2+ blocks the pore by binding within the selectivity filter. We found that internal Ba2+ could still access its binding site when the channel was shut, which indicates that the ligand-sensitive gate lies above the Ba2+-block site, and thus within or above the selectivity filter. This is in marked contrast to the voltage-dependent gate of KV channels, which is located at the intracellular mouth of the pore.  相似文献   

5.
Summary Efflux of42K+ was measured in frog sartorius muscles equilibrated in depolarizing solutions with external K+ concentrations ([K+] o ) between 75 and 300mm and NaCl concentrations of 60, 120, or 240mm. For several combinations of KCl and NaCl, steady-state internal potentials (V i) were the same for different [K+] o . For the range ofV i examined, K+ efflux occurs principally through the K+ inward rectifier channels. When external K+ is removedV i remains constant for 2 to 3 hr because of the high membrane conductance to Cl, but K+ efflux drops by about one order of magnitude.External Ba2+ in the presence or absence of external K+ produces an inhibition of K+ efflux described by a relation of the formu=(u1/(1+C)[Ba2+] o ))+u 2, whereu is the uninhibited fraction of K+ efflux;u 1, u2 andC are constants; andu 1+u2=1.C depends both on [K+] o andV i. When [K+] o 75mm, increasing [K+] o at constantV i reduces Ba2+ sensitivity. For constantV i–30 mV, Ba2+ sensitivity is less when [K+] o =0 than when [K+] o 75mm. When [K+] o =0, Ba2+ sensitivity decreases asV i is made more positive. The dependence of the Ba2+ sensitivity onV i at constant [K+] o is greater when [K+] o =0 than when [K+] o 75mm.Both the activation of K+ efflux by external K+ and the Ba2+ inhibition of K+ efflux can be explained on the basis of two membrane control sites associated with each channel. When both sites are occupied by K+, the channels are in a high flux state. When one or both sites are empty, the channels are in a low, nonzero flux state. When Ba2+ occupies either site, K+ efflux is further reduced. The reduction of Ba2+-sensitivity by increasing [K+] o at high [K+] o is attributable to the displacement of Ba2+ from the control sites by K+. The increased Ba2+ sensitivity produced by going from [K+] o =0 to [K+] o >-75mm whenV i–30 mV is attributable to states in which Ba2+ occupies one site and K+ the other when [K+] o 0. The smallerV i dependence of the Ba2+ sensitivity when [K+] o 75mm compared to [K+] o =0 is attributable to the necessity that Ba2+ displace K+ at the control sites when [K+] o is high but not when [K+] o =0.  相似文献   

6.
The ability of biological ion channels to conduct selected ions across cell membranes is critical for the survival of both animal and bacterial cells. Numerous investigations of ion selectivity have been conducted over more than 50 years, yet the mechanisms whereby the channels select certain ions and reject others are not well understood. Here we report a new application of Jarzynski’s Equality to investigate the mechanism of ion selectivity using non-equilibrium molecular dynamics simulations of Na+ and K+ ions moving through the KcsA channel. The simulations show that the selectivity filter of KcsA adapts and responds to the presence of the ions with structural rearrangements that are different for Na+ and K+. These structural rearrangements facilitate entry of K+ ions into the selectivity filter and permeation through the channel, and rejection of Na+ ions. A mechanistic model of ion selectivity by this channel based on the results of the simulations relates the structural rearrangement of the selectivity filter to the differential dehydration of ions and multiple-ion occupancy and describes a mechanism to efficiently select and conduct K+. Estimates of the K+/Na+ selectivity ratio and steady state ion conductance for KcsA from the simulations are in good quantitative agreement with experimental measurements. This model also accurately describes experimental observations of channel block by cytoplasmic Na+ ions, the “punch through” relief of channel block by cytoplasmic positive voltages, and is consistent with the knock-on mechanism of ion permeation.  相似文献   

7.
Summary Efflux of42K+ was measured in frog sartorius muscles equilibrated in hyperosmotic depolarizing solutions. At the internal potentials obtained, K+ passes mainly through the inward rectifier potassium channels.Inhibition of K+ efflux by external Zn2+ (0.25 to 15mm) differs in three significant ways from inhibition by Ba2+. (1) The dose-response relation does not correspond to action at a single site. (2) The Zn2+-sensitivity of K+ efflux does not depend on [K+] o at constant internal potential. (3) Zn2+ inhibition is reduced by hydrogen ions, while Ba2+ inhibition is unaffected. Further, the Ba2+-sensitivity of K+ efflux is not altered by a half-inhibiting Zn2+ concentration, suggesting that the two ions do not interact at a common site.The histidine-modifying reagent diethylpyrocarbonate (DEPC) reduces Zn2+ inhibition. After DEPC treatment Zn2+ inhibition is further reduced by low pH. DEPC has little effect on Ba2+ inhibition. Zn2+ inhibition is not altered by treatment with the sulfhydryl reagents 5,5-dithio-bis(2-nitrobenzoic acid) or dithiothreitol.The results can be described by either of two models in which two sites can bind Zn2+ and one or both of the sites may also bind H+. When both sites bind Zn2+, K+ efflux is inhibited, and a third site may then bind H+. The effects of DEPC can be accounted for by a decrease in H+ affinity of the first two sites by a factor of 50, and a decrease in Zn2+ affinity of these sites and of the H+ affinity of the third site by about one order of magnitude.  相似文献   

8.
Potassium channels allow the selective flux of K+ excluding the smaller, and more abundant in the extracellular solution, Na+ ions. Here we show that Shab is a typical K+ channel that excludes Na+ under bi-ionic, Nao/Ki or Nao/Rbi, conditions. However, when internal K+ is replaced by Cs+ (Nao/Csi), stable inward Na+ and outward Cs+ currents are observed. These currents show that Shab selectivity is not accounted for by protein structural elements alone, as implicit in the snug-fit model of selectivity. Additionally, here we report the block of Shab channels by external Ca2+ ions, and compare the effect that internal K+ replacement exerts on both Ca2+ and TEA block. Our observations indicate that Ca2+ blocks the channels at a site located near the external TEA binding site, and that this pore region changes conformation under conditions that allow Na+ permeation. In contrast, the latter ion conditions do not significantly affect the binding of quinidine to the pore central cavity. Based on our observations and the structural information derived from the NaK bacterial channel, we hypothesize that Ca2+ is probably coordinated by main chain carbonyls of the pore´s first K+-binding site.  相似文献   

9.
Summary We studied the influence of mucosal Ba2+ ions on the recently described (Zeiske & Van Driessche, 1979a, J. Membrane Biol. 47:77) transepithelial, mucosa towards serosa directed K+ transport in the skin ofRana temporaria. The transport parametersG (conductance), PD (potential difference),I sc (short-circuit current, K+ current), as well as the noise ofI sc were recorded. Addition of millimolar concentrations of Ba2+ to the mucosal K+-containing solution resulted in a sudden but quickly reversible drop inI sc.G andI sc decreased continuously with increasing Ba2+ concentration, (Ba2+) o . The apparent Michaelis constant of the inhibition by Ba2+ lies within the range 40–80 m. The apical membrane seems to remain permselective for K+ up to 500 m (Ba2+) o . Higher (Ba2+) o , however, appears to induce a shunt (PD falls,G increases). This finding made an accurate determination of the nature of the inhibition difficult but our results tend to suggest a K+-channel block by K+–Ba2+ competition. In the presence of Ba2+, the power spectrum of the K+ current shows a second Lorentzian component in the low-frequency range, in addition to the high-frequency Lorentzian caused by spontaneous K+-channel fluctuations (Van Driessche & Zeiske, 1980). Both Lorentzian components are only present with mucosal K+ and can be depressed by addition of Cs+ ions, thus indicating that Ba2+ ions induce K+-channel fluctuations. The dependence of the parameters of the induced Lorentzian on (Ba2+) o , shows a rise in the plateau values to a maximum around 60 m (Ba2+) o , followed by a sharp and progressive decrease to very low values. The corner frequency which reflects the rate of the Ba2+-induced fluctuations, however, increases quasi-linearly up to 1mm (Ba2+) o with a tendency to saturate at higher (Ba2+) o . Based on a three-state model for the K+ channel (having one open state, one closed by the spontaneous fluctuation and one blocked by Ba2+) computer calculations compared favorably with our results. The effect of Ba2+ could be explained by assuming reversible binding at the outer side of the apical K+ channel, thereby blocking the open channel in competition with K+. The association-dissociation of Ba2+ at its receptor site is thought to cause a chopping of the K+ current, resulting in modulated current fluctuations.  相似文献   

10.
Potassium channels are tetrameric proteins that mediate K+-selective transmembrane diffusion. For KcsA, tetramer stability depends on interactions between permeant ions and the channel pore. We have examined the role of pore blockers on the tetramer stability of KirBac1.1. In 150 mm KCl, purified KirBac1.1 protein migrates as a monomer (∼40 kDa) on SDS-PAGE. Addition of Ba2+ (K1/2 ∼ 50 μm) prior to loading results in an additional tetramer band (∼160 kDa). Mutation A109C, at a residue located near the expected Ba2+-binding site, decreased tetramer stabilization by Ba2+ (K1/2 ∼ 300 μm), whereas I131C, located nearby, stabilized tetramers in the absence of Ba2+. Neither mutation affected Ba2+ block of channel activity (using 86Rb+ flux assay). In contrast to Ba2+, Mg2+ had no effect on tetramer stability (even though Mg2+ was a potent blocker). Many studies have shown Cd2+ block of K+ channels as a result of cysteine substitution of cavity-lining M2 (S6) residues, with the implicit interpretation that coordination of a single ion by cysteine side chains along the central axis effectively blocks the pore. We examined blocking and tetramer-stabilizing effects of Cd2+ on KirBac1.1 with cysteine substitutions in M2. Cd2+ block potency followed an α-helical pattern consistent with the crystal structure. Significantly, Cd2+ strongly stabilized tetramers of I138C, located in the center of the inner cavity. This stabilization was additive with the effect of Ba2+, consistent with both ions simultaneously occupying the channel: Ba2+ at the selectivity filter entrance and Cd2+ coordinated by I138C side chains in the inner cavity.Potassium channels are expressed in many cell types and are key players in a wide range of physiological processes. One subset of potassium channels, the inward-rectifying potassium (Kir) channels, are functionally blocked by cytosolic cations such as Mg2+ and polyamines and contribute to the regulation of membrane excitability, cardiac rhythm, vascular tone, insulin release, and salt flow across epithelia (13). There are seven subfamilies of eukaryotic Kir channel genes. Among them, Kir1 encodes weak rectifiers, whereas Kir2 and Kir5 encode strong rectifiers; Kir3 encodes G-protein-regulated channels; and Kir6 encodes ATP-sensitive channels (4). Recently, a related bacterial family of genes (KirBac) has been identified (5, 6), and in 2003, the first member (KirBac1.1) was crystallized (7), providing a structural model for eukaryotic channels.The crystal structure of KirBac1.1 revealed a tetrameric pore structure similar to that seen in KcsA and a novel cytoplasmic domain (7, 8). The selectivity filter of both KirBac1.1 and KcsA consists of an extremely conserved pore loop followed by a central cavity, forming a transmembrane ion-selective permeation pore (7, 8). The linear arrangement of five oxygen rings (four from carbonyl oxygens and one from a Thr side chain) in the selectivity filter coordinates with ions, compensating for the energy barrier caused by K+ dehydration, thereby facilitating the rapid diffusion of K+ across the membrane (812). Two-thirds of the KirBac1.1 amino acid residues constitute the cytosolic domain that is highly conserved among the Kir subfamilies and form the cytosolic vestibule (1316), which, together with the transmembrane pore, generates an 88-Å-long ion conduction pore (7).The prototypic potassium channel KcsA exists very stably as a tetramer, even in the harsh conditions of SDS-PAGE (17). In addition to protein-protein interaction between monomers, protein-lipid and protein-ion interactions play important roles in stabilizing the KcsA tetramer (1720). The selectivity filter of KcsA, coordinated with K+ ions, can serve as a bridge between the four monomers to maintain the structure of the selectivity filter and the tetrameric architecture of the channel as a whole (11, 21). Blocking ions, such as Ba2+, also act as strong stabilizers (17). In the crystal structure of KcsA, Ba2+ occupies a site equivalent to the S4 K+-binding site within the selectivity filter (22). Other permeant ions (Rb+, Cs+, Tl+, and NH+4) and strong blockers (Sr2+) can also contribute to the thermostability of the KcsA tetramer in SDS-PAGE (17). In contrast, impermeant ions such as Na+ and Li+ or weak blockers such as Mg2+ tend to destabilize the KcsA tetramer (17, 19).Like KcsA, KirBac1.1 purified using decylmaltoside or tridecylmaltoside is active and presumably stable as a tetramer in mild detergent solutions. However, in SDS-PAGE, KirBac1.1 migrates exclusively as a monomer (23). Because KcsA and KirBac1.1 are structurally similar in the transmembrane region of the pore, we hypothesized that permeant and blocking ions would also affect KirBac1.1 tetramer stability in SDS-PAGE. In the present work, the effects of blocking ions such as Ba2+ and Mg2+ on KirBac1.1 tetramer stability were examined to provide insight to the physical nature of their interaction with KirBac1.1, particularly in the selectivity filter and TM2 cavity. The data reveal important differences in the nature of the interaction of Mg2+ and Ba2+ with the channel as well as provide previously unavailable evidence for the nature of Cd2+ coordination within the channel.  相似文献   

11.
Members of the eukaryotic PIEZO family (the human orthologs are noted hPIEZO1 and hPIEZO2) form cation-selective mechanically-gated channels. We characterized the selectivity of human PIEZO1 (hPIEZO1) for alkali ions: K+, Na+, Cs+ and Li+; organic cations: TMA and TEA, and divalents: Ba2+, Ca2+, Mg2+ and Mn2+. All monovalent ions permeated the channel. At a membrane potential of -100 mV, Cs+, Na+ and K+ had chord conductances in the range of 35–55 pS with the exception of Li+, which had a significantly lower conductance of ~ 23 pS. The divalents decreased the single-channel permeability of K+, presumably because the divalents permeated slowly and occupied the open channel for a significant fraction of the time. In cell-attached mode, 90 mM extracellular divalents had a conductance for inward currents carried by the divalents of: 25 pS for Ba2+ and 15 pS for Ca2+ at -80 mV and 10 pS for Mg2+ at -50 mV. The organic cations, TMA and TEA, permeated slowly and attenuated K+ currents much like the divalents. As expected, the channel K+ conductance increased with K+ concentration saturating at ~ 45 pS and the KD of K+ for the channel was 32 mM. Pure divalent ion currents were of lower amplitude than those with alkali ions and the channel opening rate was lower in the presence of divalents than in the presence of monovalents. Exposing cells to the actin disrupting reagent cytochalasin D increased the frequency of openings in cell-attached patches probably by reducing mechanoprotection.  相似文献   

12.
L-type Ca2+ channels select for Ca2+ over sodium Na+ by an affinity-based mechanism. The prevailing model of Ca2+ channel permeation describes a multi-ion pore that requires pore occupancy by at least two Ca2+ ions to generate a Ca2+ current. At [Ca2+] < 1 μM, Ca2+ channels conduct Na+. Due to the high affinity of the intrapore binding sites for Ca2+ relative to Na+, addition of μM concentrations of Ca2+ block Na+ conductance through the channel. There is little information, however, about the potential for interaction between Na+ and Ca2+ for the second binding site in a Ca2+ channel already occupied by one Ca2+. The two simplest possibilities, (a) that Na+ and Ca2+ compete for the second binding site or (b) that full time occupancy by one Ca2+ excludes Na+ from the pore altogether, would imply considerably different mechanisms of channel permeation. We are studying permeation mechanisms in N-type Ca2+ channels. Similar to L-type Ca2+ channels, N-type channels conduct Na+ well in the absence of external Ca2+. Addition of 10 μM Ca2+ inhibited Na+ conductance by 95%, and addition of 1 mM Mg2+ inhibited Na+ conductance by 80%. At divalent ion concentrations of 2 mM, 120 mM Na+ blocked both Ca2+ and Ba2+ currents. With 2 mM Ba2+, the IC50 for block of Ba2+ currents by Na+ was 119 mM. External Li+ also blocked Ba2+ currents in a concentration-dependent manner, with an IC50 of 97 mM. Na+ block of Ba2+ currents was dependent on [Ba2+]; increasing [Ba2+] progressively reduced block with an IC50 of 2 mM. External Na+ had no effect on voltage-dependent activation or inactivation of the channel. These data suggest that at physiological concentrations, Na+ and Ca2+ compete for occupancy in a pore already occupied by a single Ca2+. Occupancy of the pore by Na+ reduced Ca2+ channel conductance, such that in physiological solutions, Ca2+ channel currents are between 50 and 70% of maximal.  相似文献   

13.
Potassium (K+) channels are specialized membrane proteins that are able to facilitate and regulate the conduction of K+ through cell membranes. Comprising five specific cation binding sites (S0-S4) formed by the backbone carbonyl groups of conserved residues common to all K+ channels, the narrow selectivity filter allows fast conduction of K+ while being highly selective for K+ over Na+. To extend our knowledge of the microscopic mechanism underlying selectivity in K+ channels, we characterize the free energy landscapes governing the entry and translocation of a Na+ or a K+ from the extracellular side into the selectivity filter of KcsA. The entry process of an extracellular ion is examined in the presence of two additional K+ in the pore, and the three-ion potential of mean force is computed using extensive all-atom umbrella sampling molecular dynamics simulations. A comparison of the potentials of mean force yields a number of important results. First, the free energy minima corresponding to configurations with extracellular K+ or Na+ in binding site S0 or S1 are similar in depth, suggesting that the thermodynamic selectivity governed by the free energy minima for those two binding sites is insignificant. Second, the free energy barriers between stable multi-ion configurations are generally higher for Na+ than for K+, implying that the kinetics of ion conduction is slower when a Na+ enters the pore. Third, the region corresponding to binding site S2 near the center of the narrow pore emerges as the most selective for K+ over Na+. In particular, while there is a stable minimum for K+ in site S2, Na+ faces a steep free energy increase with no local free energy well in this region. Lastly, analysis shows that selectivity is not correlated with the overall coordination number of the ion entering the pore, but is predominantly affected by changes in the type of coordinating ligands (carbonyls versus water molecules). These results further highlight the importance of the central region near binding site S2 in the selectivity filter of K+ channels.  相似文献   

14.
Summary The outer membranes of plant cells contain channels which are highly selective for K+. In the giant-celled green algaChara corallina, K+ currents in the plasmalemma were measured during the action potential and when the cell was depolarized to the K+ equilibrium potential in high external K+ concentrations. Currents in both conditions were reduced by externally added tetraethylammonium (TEA+), Ba2+, Na+ and Cs+. In contrast to inhibition by TEA+, the latter three ions inhibited inward K+ current in a voltage-dependent manner, and reduced inward current more than outward. Ba2+ and Na+ also appeared to inhibit outward current in a strongly voltage-dependent manner. The blockade by Cs+ is studied in more detail in the following paper. TEA+ inhibited both inward and outward currents in a largely voltage-independent manner, with an apparentK D of about 0.7 to 1.1mm, increasing with increasing external K+. All inhibitors reduced current towards a similar linear leak, suggesting an insensitivity of the background leak inChara to these various K+ channel inhibitors. The selectivity of the channel to various monovalent cations varied depending on the method of measurement, suggesting that ion movement through the K+-selective channel may not be independent.  相似文献   

15.
The Na+/K+-ATPase mediates electrogenic transport by exporting three Na+ ions in exchange for two K+ ions across the cell membrane per adenosine triphosphate molecule. The location of two Rb+ ions in the crystal structures of the Na+/K+-ATPase has defined two “common” cation binding sites, I and II, which accommodate Na+ or K+ ions during transport. The configuration of site III is still unknown, but the crystal structure has suggested a critical role of the carboxy-terminal KETYY motif for the formation of this “unique” Na+ binding site. Our two-electrode voltage clamp experiments on Xenopus oocytes show that deletion of two tyrosines at the carboxy terminus of the human Na+/K+-ATPase α2 subunit decreases the affinity for extracellular and intracellular Na+, in agreement with previous biochemical studies. Apparently, the ΔYY deletion changes Na+ affinity at site III but leaves the common sites unaffected, whereas the more extensive ΔKETYY deletion affects the unique site and the common sites as well. In the absence of extracellular K+, the ΔYY construct mediated ouabain-sensitive, hyperpolarization-activated inward currents, which were Na+ dependent and increased with acidification. Furthermore, the voltage dependence of rate constants from transient currents under Na+/Na+ exchange conditions was reversed, and the amounts of charge transported upon voltage pulses from a certain holding potential to hyperpolarizing potentials and back were unequal. These findings are incompatible with a reversible and exclusively extracellular Na+ release/binding mechanism. In analogy to the mechanism proposed for the H+ leak currents of the wild-type Na+/K+-ATPase, we suggest that the ΔYY deletion lowers the energy barrier for the intracellular Na+ occlusion reaction, thus destabilizing the Na+-occluded state and enabling inward leak currents. The leakage currents are prevented by aromatic amino acids at the carboxy terminus. Thus, the carboxy terminus of the Na+/K+-ATPase α subunit represents a structural and functional relay between Na+ binding site III and the intracellular cation occlusion gate.  相似文献   

16.
HL‐1 is a line of immortalized cells of cardiomyocyte origin that are a useful complement to native cardiomyocytes in studies of cardiac gene regulation. Several types of ion channel have been identified in these cells, but not the physiologically important inward rectifier K+ channels. Our aim was to identify and characterize inward rectifier K+ channels in HL‐1 cells. External Ba2+ (100 µM) inhibited 44 ± 0.05% (mean ± s.e.m., n = 11) of inward current in whole‐cell patch‐clamp recordings. The reversal potential of the Ba2+‐sensitive current shifted with external [K+] as expected for K+‐selective channels. The slope conductance of the inward Ba2+‐sensitive current increased with external [K+]. The apparent Kd for Ba2+ was voltage dependent, ranging from 15 µM at ?150 mV to 148 µM at ?75 mV in 120 mM external K+. This current was insensitive to 10 µM glybenclamide. A component of whole‐cell current was sensitive to 150 µM 4,4′‐diisothiocyanatostilbene‐2,2′‐disulfonic acid (DIDS), although it did not correspond to the Ba2+‐sensitive component. The effect of external 1 mM Cs+ was similar to that of Ba2+. Polymerase chain reaction using HL‐1 cDNA as template and primers specific for the cardiac inward rectifier Kir2.1 produced a fragment of the expected size that was confirmed to be Kir2.1 by DNA sequencing. In conclusion, HL‐1 cells express a current that is characteristic of cardiac inward rectifier K+ channels, and express Kir2.1 mRNA. This cell line may have use as a system for studying inward rectifier gene regulation in a cardiomyocyte phenotype. J. Cell. Physiol. 225: 751–756, 2010. © 2010 Wiley‐Liss, Inc.  相似文献   

17.
K+ channels exhibit strong selectivity for K+ ions over Na+ ions based on electrophysiology experiments that measure ions competing for passage through the channel. During this conduction process, multiple ions interact within the region of the channel called the selectivity filter. Ion selectivity may arise from an equilibrium preference for K+ ions within the selectivity filter or from a kinetic mechanism whereby Na+ ions are precluded from entering the selectivity filter. Here, we measure the equilibrium affinity and selectivity of K+ and Na+ ions binding to two different K+ channels, KcsA and MthK, using isothermal titration calorimetry. Both channels exhibit a large preference for K+ over Na+ ions at equilibrium, in line with electrophysiology recordings of reversal potentials and Ba2+ block experiments used to measure the selectivity of the external-most ion-binding sites. These results suggest that the high selectivity observed during ion conduction can originate from a strong equilibrium preference for K+ ions in the selectivity filter, and that K+ selectivity is an intrinsic property of the filter. We hypothesize that the equilibrium preference for K+ ions originates in part through the optimal spacing between sites to accommodate multiple K+ ions within the selectivity filter.  相似文献   

18.
KcsA 通道对Na+、K+及Rb+离子选择性的统计热力学研究   总被引:2,自引:0,他引:2  
钾离子的通透率至少比钠离子的通透率大10000倍,这个问题至今没有很好地解决.为了在分子水平阐释钾离子通道的选择性机制,以KcsA钾通道X射线衍射结构为基础,采用密度泛函理论计算了不同离子在离子通道中的位能.计算结果表明,Rb+离子具有与K+离子相类似的位能曲线,但是其在通透过程遇到的位垒要比K+离子的位垒高,因而所对应的通透率也就小于钾离子的通透率,而钠离子的的通透率仅仅是钾离子通透率的0.0067%.文中所涉及的系统仅仅包含269个原子,而用分子动力学虽然也可以得到相近的结果,但是它的系统大小为41 000个原子.  相似文献   

19.
The interactions of divalent cations with the adenosine triphosphatase (ATPase) and para-nitrophenyl phosphatase (pNPPase) activity of the purified dog kidney Na pump and the fluorescence of fluorescein isothiocyanate (FITC)-labeled pump were determined. Sr2+ and Ba2+ did not compete with K+ for ATPase (an extracellular K+ effect). Sr2+ and Ba2+ did compete with Na+ for ATPase (an intracellular Na+ effect) and with K+ for pNPPase (an intracellular K+ effect). These results suggest that Ba2+ or Sr2+ can bind to the intracellular transport site, yet neither Ba2+ nor Sr2+ was able to activate pNPPase activity; we confirmed that Ca2+ and Mn2+ did activate. As another measure of cation binding, we observed that Ca2+ and Mn2+, but not Ba2+, decreased the fluorescence of the FITC-labeled pump; we confirmed that K+ substantially decreased the fluorescence. Interestingly, Ba2+ did shift the K+ dose-response curve. Ethane diamine inhibited Mn2+ stimulation of pNPPase (as well as K+ and Mg2+ stimulation) but did not shift the 50% inhibitory concentration (IC50) for the Mn2+-induced fluorescence change of FITC, though it did shift the IC50 for the K+-induced change. These results suggest that the Mn2+-induced fluorescence change is not due to Mn2+ binding at the transport site. The drawbacks of models in which Mn2+ stimulates pNPPase by binding solely to the catalytic site vs. those in which Mn2+ stimulates by binding to both the catalytic and transport sites are presented. Our results provide new insights into the pNPPase kinetic mechanism as well as how divalent cations interact with the Na pump.  相似文献   

20.
Patch clamping whole-cell recording techniques were applied to study the inward K+ -channels inArabidopsis root cortex cells. The inward K+ -channels in the plasma membranes of the root cortex cell protoplasts were activated by hyperpolarized membrane potentials. The channels were highly selective for K+ ions over Na+ ions. The channel activity was significantly inhibited by the external TEA+ or Ba2+. The changes in cytoplasmic Ca2+ concentrations did not affect the whole-cell inward K+ -currents. The possible association between the channel selectivity to K+ and Na+ ions and plant salt-tolerance was also discussed.  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