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.
We recently reported thatarachidonic acid (AA) inhibits L- and N-type Ca2+ currentsat positive test potentials in the presence of the dihydropyridine L-type Ca2+ channel agonist (+)-202-791 in dissociatedneonatal rat superior cervical ganglion neurons [Liu L and RittenhouseAR. J Physiol (Lond) 525: 291-404, 2000]. In thisfirst of two companion papers, we characterized the mechanism ofinhibition by AA at the whole cell level. In the presence of either-conotoxin GVIA or nimodipine, AA decreased current amplitude,confirming that L- and N-type currents, respectively, were inhibited.AA-induced inhibition was concentration dependent and reversible withan albumin-containing wash solution, but appears independent of AAmetabolism and G protein activity. In characterizing inhibition, anAA-induced enhancement of current amplitude was revealed that occurredprimarily at negative test potentials. Cell dialysis with albuminminimized inhibition but had little effect on enhancement, suggestingthat AA has distinct sites of action. We examined AA's actions oncurrent kinetics and found that AA increased holdingpotential-dependent inactivation. AA also enhanced the rate of N-typecurrent activation. These findings indicate that AA causes multiplechanges in sympathetic Ca2+ currents.

  相似文献   

2.
Dietary polyunsaturated fatty acids (PUFAs) have been reported to exhibit antiarrhythmic properties, which have been attributed to their availability to modulate Na+, Ca2+, and several K+ channels. However, their effects on human ether-a-go-go-related gene (HERG) channels are unknown. In this study we have analyzed the effects of arachidonic acid (AA, -6) and docosahexaenoic acid (DHA, -3) on HERG channels stably expressed in Chinese hamster ovary cells by using the whole cell patch-clamp technique. At 10 µM, AA and DHA blocked HERG channels, at the end of 5-s pulses to –10 mV, to a similar extent (37.7 ± 2.4% vs. 50.2 ± 8.1%, n = 7–10, P > 0.05). 5,6,11,14-Eicosatetrayenoic acid, a nonmetabolizable AA analog, induced effects similar to those of AA on HERG current. Both PUFAs shifted the midpoint of activation curves of HERG channels by –5.1 ± 1.8 mV (n = 10, P < 0.05) and –11.2 ± 1.1 mV (n = 7, P < 0.01). Also, AA and DHA shifted the midpoint of inactivation curves by +12.0 ± 3.9 mV (n = 4; P < 0.05) and +15.8 ± 4.3 mV (n = 4; P < 0.05), respectively. DHA and AA accelerated the deactivation kinetics and slowed the inactivation kinetics at potentials positive to +40 mV. Block induced by DHA, but not that produced by AA, was higher when measured after applying a pulse to –120 mV (IO). Finally, both AA and DHA induced a use-dependent inhibition of HERG channels. In summary, block induced by AA and DHA was time, voltage, and use dependent. The results obtained suggest that both PUFAs bind preferentially to the open state of the channel, although an interaction with inactivated HERG channels cannot be ruled out for AA. K+ channel; membrane currents; ion channels; arrhythmia; antiarrhythmics  相似文献   

3.
Enterochromaffin-like (ECL) cells are histamine-containingendocrine cells in the gastric mucosa that maintain a negative membranepotential of about 50 mV, largely due to voltage-gated K+ currents [D. F. Loo, G. Sachs, and C. Prinz. Am. J. Physiol. 270 (Gastrointest Liver Physiol. 33):G739-G745, 1996]. The current study investigated thepresence of voltage-gated Ca2+channels in single ECL cells. ECL cells were isolated from rat fundicmucosa by elutriation, density gradient centrifugation, and primaryculture to a purity >90%. Voltage-gatedCa2+ currents were measured insingle ECL cells using the whole cell configuration of the patch-clamptechnique. Depolarization-activated currents were recorded in thepresence of Na+ orK+ blocking solutions and additionof 20 mM extracellular Ca2+. ECLcells showed inward currents in response to voltage steps that wereactivated at a test potential of around 20 mV with maximalinward currents observed at +20 mV and 20 mM extracellular Ca2+. The inactivation rate of thecurrent decreased with increasingly negative holding potentials and wastotally abolished at a holding potential of 30 mV. Addition ofextracellular 20 mM Ba2+ insteadof 20 mM Ca2+ increased thedepolarization-induced current and decreased the inactivation rate. Theinward current was fully inhibited by the specific L-typeCa2+ channel inhibitor verapamil(0.2 mM) and was augmented by the L-typeCa2+ channel activator BAY K 8644 (0.07 mM). We conclude that depolarization activateshigh-voltage-activated Ca2+channels in ECL cells. Activation characteristics,Ba2+ effects, and pharmacologicalresults imply the presence of L-type Ca2+ channels, whereasinactivation kinetics suggest the presence of additional N-typechannels in rat gastric ECL cells.

  相似文献   

4.
The effects ofcyanide (CN) on whole cell current measured with the perforated-patchmethod were studied in adrenal medullary cells. Application of CNproduced initially inward and then outward currents at 52 mV ormore negative. As the membrane potential was hyperpolarized, amplitudeand latency of the outward current (Io) by CNbecame small and long, respectively. A decrease in the externalNa+ concentration did not affectthe latency for CN-inducedIo but enhancedthe amplitude markedly. The CNIo reversedpolarity at 85 mV, close to the Nernst potential forK+, and was suppressed by theK+ channel blockers curare andapamin but not by glibenclamide, suggesting thatIo is due to theactivation of Ca2+-dependentK+ channels. Consistent with thisnotion, the Ca2+-mobilizingagents, muscarine and caffeine, also producedIo. Exposure toCN in a Ca2+-deficient medium for4 min abolished caffeine- or muscarine-induced Io withoutdevelopment ofIo, and additionof Ca2+ to the CN-containingsolution inducedIo. We concludethat exposure to CN producesCa2+-dependentK+ currents in an externalCa2+-dependent manner, probablyvia facilitation of Ca2+ influx.

  相似文献   

5.
This study investigated the interaction between L-type Ca2+ current (ICaL) and Ca2+ release from the sarcoplasmic reticulum (SRCR) in whole cell voltage-clamped guinea pig ventricular myocytes. Quasiphysiological cation solutions (Nao+:KI+) were used for most experiments. In control conditions, there was no obvious interaction between ICaL and SRCR. In isoproterenol, activation of ICaL from voltages between -70 and -50 mV reduced the amplitude and accelerated the decay of the current. Short (50 ms), small-amplitude voltage steps applied 60 or 510 ms before stimulating ICaL inhibited and facilitated the current, respectively. These changes were blocked by ryanodine. Low-voltage activated currents such as T-type Ca2+ current, TTX-sensitive ICa (ICaTTX), or slip mode Ca2+ conductance via INa+ were not responsible for low-voltage SRCR. However, L-type Ca2+ currents could be distinguished at voltages as negative as -45 mV. It is concluded that in the presence of isoproterenol, Ca2+ release from the SR at negative potentials is due to activation of L-type Ca2+ channels. heart; calcium current; low-voltage activation  相似文献   

6.
The effect of -adrenergic stimulation on cardiac Na+/Ca2+ exchange has been controversial. To clarify the effect, we measured Na+/Ca2+ exchange current (INCX) in voltage-clamped guinea pig, mouse, and rat ventricular cells. When INCX was defined as a 5 mM Ni2+-sensitive current in guinea pig ventricular myocytes, 1 µM isoproterenol apparently augmented INCX by 32%. However, this increase was probably due to contamination of the cAMP-dependent Cl current (CFTR-Cl current, ICFTR-Cl), because Ni2+ inhibited the activation of ICFTR-Cl by 1 µM isoproterenol with a half-maximum concentration of 0.5 mM under conditions where INCX was suppressed. Five or ten millimolar Ni2+ did not inhibit ICFTR-Cl activated by 10 µM forskolin, an activator of adenylate cyclase, suggesting that Ni2+ acted upstream of adenylate cyclase in the -adrenergic signaling pathway. Furthermore, in a low-extracellular Cl bath solution, 1 µM isoproterenol did not significantly alter the amplitude of Ni2+-sensitive INCX at +50 mV, which is close to the reversal potential of ICFTR-Cl. No change in INCX amplitude was induced by 10 µM forskolin. When INCX was activated by extracellular Ca2+, it was not significantly affected by 1 µM isoproterenol in guinea pig, mouse, or rat ventricular cells. We concluded that -adrenergic stimulation does not have significant effects on INCX in guinea pig, mouse, or rat ventricular myocytes. cystic fibrosis transmembrane conductance regulator; nickel ion  相似文献   

7.
Ca2+-activatedCl currents (ICl,Ca) wereexamined using fluorescence confocal microscopy to monitorintracellular Ca2+ liberation evoked by flash photolysis ofcaged inositol 1,4,5-trisphosphate (InsP3) involtage-clamped Xenopus oocytes. Currents at +40 mV exhibited asteep dependence on InsP3 concentration([InsP3]), whereas currents at140 mV exhibited a higher threshold and more graded relationshipwith [InsP3]. Ca2+ levelsrequired to half-maximally activate ICl,Ca wereabout 50% larger at 140 mV than at +40 mV, and currents evokedby small Ca2+ elevations were reduced >25-fold. Thehalf-decay time of Ca2+ signals shortened at increasinglypositive potentials, whereas the decay of ICl,Calengthened. The steady-state current-voltage (I-V) relationshipfor ICl,Ca exhibited outward rectification withweak photolysis flashes but became more linear with stronger stimuli.Instantaneous I-V relationships were linear with both strongand weak stimuli. Current relaxations following voltage steps duringactivation of ICl,Ca decayed with half-times that shortened from about 100 ms at +10 mV to 20 ms at 160 mV. We conclude that InsP3-mediated Ca2+liberation activates a single population of Clchannels, which exhibit voltage-dependent Ca2+ activationand voltage-independent instantaneous conductance.

  相似文献   

8.
We studied the K+-selective conductances in primary cultures of rat renal inner medullary collecting duct (IMCD) using perforated-patch and conventional whole cell techniques. Depolarizations above –20 mV induced a time-dependent outward K+ current (Ivto) similar to a delayed rectifier. Ivto showed a half-maximal activation around 5.6 mV with a slope factor of 6.8 mV. Its K+/Na+ selectivity ratio was 11.7. It was inhibited by tetraethylammonium, quinidine, 4-aminopyridine, and Ba2+ and was not Ca2+ dependent. The delayed rectifying characteristics of Ivto prompted us to screen the expression of Kv1 and Kv3 families by RT-PCR. Analysis of RNA isolated from cell cultures revealed the presence of three Kv -subunits (Kv1.1, Kv1.3, and Kv1.6). Western blot analysis with Kv -subunit antibodies for Kv1.1 and Kv1.3 showed labeling of 70-kDa proteins from inner medulla plasmatic and microsome membranes. Immunocytochemical analysis of cell culture and kidney inner medulla showed that Kv1.3 is colocalized with the Na+-K+-ATPase at the basolateral membrane, although it is also in the cytoplasm. This is the first evidence of recording, protein expression, and localization of a voltage-gated Kv1 in the kidney IMCD cells. kidney; Kv1.3; potassium channel; potassium transport; whole cell clamp; immunocytochemistry; confocal microscopy  相似文献   

9.
Arterial smooth muscle cell large-conductance Ca2+-activated potassium (KCa) channels have been implicated in modulating hypoxic dilation of systemic arteries, although this is controversial. KCa channel activity in arterial smooth muscle cells is controlled by localized intracellular Ca2+ transients, termed Ca2+ sparks, but hypoxic regulation of Ca2+ sparks and KCa channel activation by Ca2+ sparks has not been investigated. We report here that in voltage-clamped (–40 mV) cerebral artery smooth muscle cells, a reduction in dissolved O2 partial pressure from 150 to 15 mmHg reversibly decreased Ca2+ spark-induced transient KCa current frequency and amplitude to 61% and 76% of control, respectively. In contrast, hypoxia did not alter Ca2+ spark frequency, amplitude, global intracellular Ca2+ concentration, or sarcoplasmic reticulum Ca2+ load. Hypoxia reduced transient KCa current frequency by decreasing the percentage of Ca2+ sparks that activated a transient KCa current from 89% to 63%. Hypoxia reduced transient KCa current amplitude by attenuating the amplitude relationship between Ca2+ sparks that remained coupled and the evoked transient KCa currents. Consistent with these data, in inside-out patches at –40 mV hypoxia reduced KCa channel apparent Ca2+ sensitivity and increased the Kd for Ca2+ from 17 to 32 µM, but did not alter single-channel amplitude. In summary, data indicate that hypoxia reduces KCa channel apparent Ca2+ sensitivity via a mechanism that is independent of cytosolic signaling messengers, and this leads to uncoupling of KCa channels from Ca2+ sparks. Transient KCa current inhibition due to uncoupling would oppose hypoxic cerebrovascular dilation. transient calcium-activated potassium current  相似文献   

10.
The effects of niflumic acid on ryanodinereceptors (RyRs) of frog skeletal muscle were studied by incorporatingsarcoplasmic reticulum (SR) vesicles into planar lipid bilayers. Frogmuscle had two distinct types of RyRs in the SR: one showed abell-shaped channel activation curve against cytoplasmicCa2+ or niflumic acid, and its mean open probability(Po) was increased by perchlorate at 20-30mM (termed "-like" RyR); the other showed a sigmoidalactivation curve against Ca2+ or niflumic acid, with noeffect on perchlorate (termed "-like" RyR). The unitaryconductance and reversal potential of both channel types wereunaffected after exposure to niflumic acid when clamped at 0 mV. Whenclamped at more positive potentials, the -like RyRchannel rectified this, increasing the unitary current. Treatment withniflumic acid did not inhibit the response of both channels toCa2+ release channel modulators such as caffeine,ryanodine, and ruthenium red. The different effects of niflumic acid onPo and the unitary current amplitude in both typesof channels may be attributable to the lack or the presence ofinactivation sites and/or distinct responses to agonists.

  相似文献   

11.
Modulation of the L-type current by sarcoplasmicreticulum (SR) Ca2+ release hasbeen examined in patch-clamped mouse myotubes. Inhibition of SRCa2+ release by inclusion ofryanodine in the internal solution shifted the half-activating voltage(V0.5) of theL-type current from 1.1 ± 2.1 to 7.7 ± 1.7 mV. Rutheniumred in the internal solution shiftedV0.5 from 5.4 ± 1.9 to 3.2 ± 4.1 mV. Chelation of myoplasmic Ca2+ with1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraaceticacid perfusion shiftedV0.5 from 4.4 ± 1.7 to 3.5 ± 3.3 mV and increased the peak current.Extracellular caffeine (1 mM), which should enhance SRCa2+ release, significantlydecreased the peak Ca2+ current.In low (0.1 mM) internal EGTA, myotube contraction was abolished byinternal perfusion with ryanodine or ruthenium red, whereas addition ofcaffeine to the extracellular solution lowered the contractilethreshold, indicating that these modulators of SRCa2+ release had the expectedeffects on contraction. Therefore, SR Ca2+ release appears to modulatethe sarcolemmal L-type current, suggesting a retrograde communicationfrom the SR to the sarcolemmal L-type channels inexcitation-contraction coupling.

  相似文献   

12.
Phospholamban(PLB) ablation is associated with enhanced sarcoplasmic reticulum (SR)Ca2+ uptake and attenuation of thecardiac contractile responses to -adrenergic agonists. In thepresent study, we compared the effects of isoproterenol (Iso) on theCa2+ currents(ICa) ofventricular myocytes isolated from wild-type (WT) and PLB knockout(PLB-KO) mice. Current density and voltage dependence ofICa were similarbetween WT and PLB-KO cells. However, ICa recorded fromPLB-KO myocytes had significantly faster decay kinetics. Iso increasedICa amplitude inboth groups in a dose-dependent manner (50% effective concentration,57.1 nM). Iso did not alter the rate ofICa inactivationin WT cells but significantly prolonged the rate of inactivation inPLB-KO cells. When Ba2+ was usedas the charge carrier, Iso slowed the decay of the current in both WTand PLB-KO cells. Depletion of SRCa2+ by ryanodine also slowed therate of inactivation ofICa, and subsequent application of Iso further reduced the inactivation rate ofboth groups. These results suggest that enhancedCa2+ release from the SR offsetsthe slowing effects of -adrenergic receptor stimulation on the rateof inactivation ofICa.

  相似文献   

13.
Opposite effects of Ni2+ on Xenopus and rat ENaCs expressed in Xenopus oocytes. Am J Physiol Cell Physiol 289: C946–C958, 2005. First published June 8, 2005; .—The epithelial Na+ channel (ENaC) is modulated by various extracellular factors, including Na+, organic or inorganic cations, and serine proteases. To identify the effect of the divalent Ni2+ cation on ENaCs, we compared the Na+ permeability and amiloride kinetics of Xenopus ENaCs (xENaCs) and rat ENaCs (rENaCs) heterologously expressed in Xenopus oocytes. We found that the channel cloned from the kidney of the clawed toad Xenopus laevis [wild-type (WT) xENaC] was stimulated by external Ni2+, whereas the divalent cation inhibited the channel cloned from the rat colon (WT rENaC). The kinetics of amiloride binding were determined using noise analysis of blocker-induced fluctuation in current adapted for the transoocyte voltage-clamp method, and Na+ conductance was assessed using the dual electrode voltage-clamp (TEVC) technique. The inhibitory effect of Ni2+ on amiloride binding is not species dependent, because Ni2+ decreased the affinity (mainly reducing the association rate constant) of the blocker in both species in competition with Na+. Importantly, using the TEVC method, we found a prominent difference in channel conductance at hyperpolarizing voltage pulses. In WT xENaCs, the initial ohmic current response was stimulated by Ni2+, whereas the secondary voltage-activated current component remained unaffected. In WT rENaCs, only a voltage-dependent block by Ni2+ was obtained. To further study the origin of the xENaC stimulation by Ni2+, and based on the rationale of the well-known high affinity of Ni2+ for histidine residues, we designed -subunit mutants of xENaCs by substituting histidines that were expressed in oocytes, together with WT - and -subunits. Changing His215 to Asp in one putative amiloride-binding domain (WYRFHY) in the extracellular loop between Na+ channel membrane segments M1 and M2 had no influence on the stimulatory effect of Ni2+, and neither did complete deletion of this segment. Next, we mutated His416 flanked by His411 and Cys417, a unique site for possible heavy metal ion chelation, and, with this quality, most proximal (100 amino acids upstream of the second putative amiloride binding site at the pore entrance), was found localized at M2. Replacing His416 with arginine, aspartate, tyrosine, and alanine clearly affected amiloride binding in all cases, as well as Na+ conductance, as expressed in the xENaC current-voltage relationship, especially with regard to aspartate and tyrosine. However, similarly to those obtained with the WYRFHY stretch, none of these mutations could either abolish the stimulating effect of Ni2+ or reverse it to an inhibitory type. epithelia; divalent cations; amiloride; Na+; voltage clamp  相似文献   

14.
G protein-coupled receptors (GPCRs) control neuronal functions via ion channel modulation. For voltage-gated ion channels, gating charge movement precedes and underlies channel opening. Therefore, we sought to investigate the effects of G protein activation on gating charge movement. Nonlinear capacitive currents were recorded using the whole cell patch-clamp technique in cultured rat sympathetic neurons. Our results show that gating charge movement depends on voltage with average Boltzmann parameters: maximum charge per unit of linear capacitance (Qmax) = 6.1 ± 0.6 nC/µF, midpoint (Vh) = –29.2 ± 0.5 mV, and measure of steepness (k) = 8.4 ± 0.4 mV. Intracellular dialysis with GTPS produces a nonreversible 34% decrease in Qmax, a 10 mV shift in Vh, and a 63% increase in k with respect to the control. Norepinephrine induces a 7 mV shift in Vh and 40% increase in k. Overexpression of G protein 14 subunits produces a 13% decrease in Qmax, a 9 mV shift in Vh, and a 28% increase in k. We correlate charge movement modulation with the modulated behavior of voltage-gated channels. Concurrently, G protein activation by transmitters and GTPS also inhibit both Na+ and N-type Ca2+ channels. These results reveal an inhibition of gating charge movement by G protein activation that parallels the inhibition of both Na+ and N-type Ca2+ currents. We propose that gating charge movement decrement may precede or accompany some forms of GPCR-mediated channel current inhibition or downregulation. This may be a common step in the GPCR-mediated inhibition of distinct populations of voltage-gated ion channels. ion channel modulation; G protein-coupled receptors; charge movement  相似文献   

15.
These experiments were performed to determine the effects ofreducing Ca2+ influx(Cain) onK+ currents(IK) inmyocytes from rat small mesenteric arteries by1) adding externalCd2+ or2) lowering externalCa2+ to 0.2 mM. When measured froma holding potential (HP) of 20 mV(IK20),decreasing Cain decreasedIK at voltageswhere it was active (>0 mV). When measured from a HP of 60 mV(IK60),decreasing Cain increasedIK at voltagesbetween 30 and +20 mV but decreased IK at voltagesabove +40 mV. Difference currents(IK) weredetermined by digital subtraction of currents recorded under controlconditions from those obtained whenCain was decreased. At testvoltages up to 0 mV,IK60 exhibitedkinetics similar to controlIK60, with rapidactivation to a peak followed by slow inactivation. At 0 mV, peakIK60 averaged75 ± 13 pA (n = 8) withCd2+ and 120 ± 20 pA(n = 9) with lowCa2+ concentration. At testvoltages from 0 to +60 mV,IK60 always had an early positive peak phase, but its apparent "inactivation" increased with voltage and its steady value became negative above +20mV. At +60 mV, the initial peakIK60 averaged115 ± 18 pA with Cd2+ and 187 ± 34 pA with low Ca2+. With 10 mM pipette BAPTA, Cd2+ produced asmall inhibition ofIK20 but stillincreased IK60 between 30 and +10 mV. InCa2+-free external solution,Cd2+ only decreased bothIK20 andIK60. In thepresence of iberiotoxin (100 nM) to inhibitCa2+-activatedK+ channels(KCa),Cd2+ increasedIK60 at allvoltages positive to 30 mV while BAY K 8644 (1 µM) decreasedIK60. Theseresults suggest that Cain, through L-type Ca2+ channels and perhapsother pathways, increases KCa(i.e., IK20) and decreases voltage-dependent K+currents in this tissue. This effect could contribute to membrane depolarization and force maintenance.

  相似文献   

16.
The following is the abstract of the article discussed in thesubsequent letter:

Mitchell, Claire H., Jin Jun Zhang, Liwei Wang, andTim J. C. Jacob. Volume-sensitive chloride current in pigmented ciliary epithelial cells: role of phospholipases. Am. J. Physiol. 272 (Cell Physiol. 41): C212-C222, 1997.Thewhole cell recording technique was used to examine an outwardlyrectifying chloride current activated by hypotonic shock in bovinepigmented ciliary epithelial (PCE) cells. Removal of internal andexternal Ca2+ did not affect the activation of thesecurrents, but they were abolished by the phospholipase C inhibitorneomycin. The current was blocked by5-nitro-2-(3-phenylpropylamino)benzoic acid,4-acetamido-4'-isothiocyanostilbene-2,2'-disulfonic acid, and4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS) in avoltage-dependent manner, but tamoxifen, dideoxyforskolin, andquinidine did not affect it. This blocking profile differs from that ofthe volume-sensitive chloride channel in neighboring nonpigmentedciliary epithelial cells (Wu, J., J. J. Zhang, H. Koppel, and T. J. C. Jacob. J. Physiol. Lond. 491: 743-755, 1996), and thisdifference implies that the volume responses of the two cell types aremediated by different chloride channels (Jacob, T. J. C., and J. J. Zhang. J. Physiol. Lond. In press). Intracellular administration of guanosine 5'-O-(3-thiotriphosphate) (GTPS) to PCE cells induced a transient, time-independent, outwardly rectifying chloride current that closely resembled the current activated by hypotonic shock. DIDS produced a voltage-dependent blockof the GTPS-activated current similar to the block of the hypotonically activated current. Intracellular neomycin completely prevented activation of this current as did incubation of the cells incalphostin C, an inhibitor of protein kinase C (PKC). Removal ofCa2+ did not affect activation of the current by GTPSbut extended the duration of the response. Inhibition of phospholipaseA2 (PLA2) with p-bromophenacyl bromideprevented the activation of the hypotonically induced current and alsoinhibited the current once activated by hypotonic solution. Thefindings imply that the hypotonic response in PCE cells is mediated byboth phospholipase C (PLC) and PLA2. Both phospholipasesgenerate arachidonic acid, and, in addition, the PLC pathway regulatesthe PLA2 pathway via a PKC-dependent phosphorylation ofPLA2.

  相似文献   

17.
A voltage-dependent outwardK+(KV) current in the intermediatecell (melanocyte) of the cochlear stria vascularis was studied usingthe whole cell patch-clamp technique. TheKV current had an activationthreshold voltage of approximately 80 mV, and 50% activationwas observed at 42.6 mV. The time courses of activation andinactivation were well fitted by two exponential functions: the timeconstants at 0 mV were 7.9 and 58.8 ms for activation and 0.6 and 4.3 sfor inactivation. The half-maximal activation time was 13.8 ms at 0 mV.Inactivation of the current was incomplete even after a prolongeddepolarization of 10 s. This current was independent of intracellularCa2+. Quinine, verapamil,Ba2+, and tetraethylammoniuminhibited the current in a dose-dependent manner, but 4-aminopyridinewas ineffective at 50 mM. We conclude that theKV conductance in the intermediatecell may stabilize the membrane potential, which is thought to beclosely related to the endocochlear potential, and may provide anadditional route for K+ secretioninto the intercellular space.

  相似文献   

18.
The effects ofMg2+ andBa2+ on single-channel propertiesof the inositol 1,4,5-trisphosphate receptor(IP3R) were studied by patch clampof isolated nuclei from Xenopusoocytes. In 140 mM K+ theIP3R channel kinetics and presenceof conductance substates were similar over a range (0-9.5 mM) offree Mg2+. In 0 mMMg2+ the channel current-voltage(I-V) relation was linear withconductance of ~320 pS. Conductance varied slowly and continuouslyover a wide range (SD  60 pS) and sometimes fluctuated during single openings. The presence of Mg2+ oneither or both sides of the channel reduced the current (blocking constant ~0.6 mM in symmetricalMg2+), as well as the range ofconductances observed, and made the I-V relation nonlinear (slopeconductance ~120 pS near 0 mV and ~360 pS at ±70 mV insymmetrical 2.5 mM Mg2+).Ba2+ exhibited similar effects onchannel conductance. Mg2+ andBa2+ permeated the channel with aratio of permeability of Ba2+ toMg2+ toK+ of 3.5:2.6:1. These resultsindicate that divalent cations induce nonlinearity in theI-V relation and reduce current by amechanism involving permeation block of theIP3R due to strong binding to site(s) in the conduction pathway. Furthermore, stabilization ofconductance by divalent cations reveals a novel interaction between thecations and the IP3R.

  相似文献   

19.
We usedsingle-channel recording techniques to identify and characterize alarge-conductance,Ca2+-independentK+ channel in the colonicsecretory cell line T84. In symmetric potassium gluconate, this channelhad a linear current-voltage relationship with a single-channelconductance of 161 pS. Channel open probability(Po) wasincreased at depolarizing potentials. Partial substitution of bathK+ withNa+ indicated a permeability ratioof K+ toNa+ of 25:1. ChannelPo was reduced byextracellular Ba2+. Event-durationanalysis suggested a linear kinetic model for channel gating having asingle open state and three closed states: C3C2C1O.Arachidonic acid (AA) increased thePo of thechannel, with an apparent stimulatory constant(Ks)of 1.39 µM. Neither channel open time (O) nor the fast closed time(C1) was affected by AA. Incontrast, AA dramatically reduced mean closed time by decreasing bothC3 andC2. Thecis-unsaturated fatty acid linoleate increased Poalso, whereas the saturated fatty acid myristate and thetrans-unsaturated fatty acid elaidatedid not affectPo. This channelis activated also by negative pressure applied to the pipette duringinside-out recording. Thus we determined the effect of thestretch-activated channel blockers amiloride and Gd3+ on theK+ channel after activation by AA.Amiloride (2 mM) on the extracellular side reduced single-channelamplitude in a voltage-dependent manner, whereasGd3+ (100 µM) had no effect onchannel activity. Activation of this K+ channel may be important duringstimulation of Cl secretionby agonists that use AA as a second messenger (e.g., vasoactiveintestinal polypeptide, adenosine) or during the volume regulatoryresponse to cell swelling.

  相似文献   

20.
Voltage-clamp studies offreshly isolated smooth muscle cells from rabbit portal veinrevealed the existence of a time-dependent cation current evoked bymembrane hyperpolarization (termed Ih). Both therate of activation and the amplitude of Ih wereenhanced by membrane hyperpolarization. Half-maximal activation ofIh was about 105 mV with conventional wholecell and 80 mV when the perforated patch technique was used. Incurrent clamp, injection of hyperpolarizing current produced a markeddepolarizing "sag" followed by rebound depolarization. Activationof Ih was augmented by an increase in theextracellular K+ concentration and was blocked rapidly byexternally applied Cs+ (1-5 mM). The bradycardic agentZD-7288 (10 µM), a selective inhibitor of Ih,produced a characteristically slow inhibition of the portal veinIh. The depolarizing sag recorded in current clamp was also abolished by application of 5 mM Cs+.Cs+ significantly decreased the frequency of spontaneouscontractions in both whole rat portal vein and rabbit portal veinsegments. Multiplex RT-PCR of rabbit portal vein myocytes using primers derived from existing genes for hyperpolarization-activated cation channels (HCN1-4) revealed the existence of cDNA clonescorresponding to HCN2, 3, and 4. The present study shows that portalvein myocytes contain genes shown to encode forhyperpolarization-activated channels and exhibit an endogenous currentwith characteristics similar to Ih in other celltypes. This conductance appears to determine, in part, the rhythmicityof this vessel.

  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