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 We have measured Ca2+ uptake and Ca2+ release in isolated permeabilized pancreatic acinar cells and in isolated membrane vesicles of endoplasmic reticulum prepared from these cells. Ca2+ uptake into cells was monitored with a Ca2+ electrode, whereas Ca2+ uptake into membrane vesicles was measured with45Ca2+. Using inhibitors of known action, such as the H+ ATPase inhibitors NBD-Cl and NEM, the Ca2+ ATPase inhibitor vanadate as well as the second messenger inositol 1,4,5-trisphosphate (IP3) and its analog inositol 1,4,5-trisphosphorothioate (IPS3), we could functionally differentiate two non-mitochondrial Ca2+ pools. Ca2+ uptake into the IP3-sensitive Ca2+ pool (IsCaP) occurs by a MgATP-dependent Ca2+ uptake mechanism that exchanges Ca2+ for H+ ions. In the absence of ATP Ca2+ uptake can occur to some extent at the expense of an H+ gradient that is established by a vacuolar-type MgATP-dependent H+ pump present in the same organelle. The other Ca2+ pool takes up Ca2+ by a vanadate-sensitive Ca2+ ATPase and is insensitive to IP3 (IisCaP). The IsCaP is filled at higher Ca2+ concentrations (10–6 mol/liter) which may occur during stimulation. The low steady-state [Ca2+] of 10–7 mol/liter is adjusted by the IisCaP.It is speculated that both Ca2+ pools can communicate with each other, the possible mechanism of which, however, is at present unknown.  相似文献   

2.
Inositol 1,4,5-trisphosphate (IP3) was found to release Ca2+ from presynaptic nerve endings (synaptosomes) made permeable with saponin. ATP-dependent Ca2+ uptake was carried out until equilibrium was reached. Addition of IP3 produced a rapid release of Ca2+, which was complete within 60 sec, followed by Ca2+ reaccumulation to the original level in 5–7 min. Cholinergic receptor stimulation with muscarine also produced a similar Ca2+ release from synaptic endoplasmic reticulum. Ca2+ release by IP3 was not detectable in the absence of the mitochondrial inhibitors oligomycin or sodium azide. Reaccumulation of Ca2+ was prevented by the presence of vanadate, a potent inhibitor of Ca2+/Mg2+ ATPase. Half maximal and near complete release of Ca2+ took place at 0.4 M and 3 M IP3 concentrations, respectively. These studies demonstrate for the first time IP3 mobilization of Ca2+ from endoplasmic reticulum within synaptic plasma membranes.  相似文献   

3.
We have examined the effect of the Ca2+ (Mg2+)-ATPase inhibitors thapsigargin (TG) and vanadate on ATP-dependent 45Ca2+ uptake into IP3-sensitive Ca2+ pools in isolated microsomes from rat pancreatic acinar cells. The inhibitory effect of TG was biphasic. About 40–50% of total Ca2+ uptake was inhibited by TG up to 10 nm (apparent Ki4.2 nm, Ca2+ pool I). An additional increase of inhibition up to 85–90% of total Ca2+ uptake could be achieved at 15 to 20 nm of TG (apparent Ki12.1 nm, Ca2+ pool II). The rest was due to TG-insensitive contaminating plasma membranes and could be inhibited by vanadate (apparent Ki10 m). In the absence of TG, increasing concentrations of vanadate also showed two phases of inhibition of microsomal Ca2+ uptake. About 30–40% of total Ca2+ uptake was inhibited by 100 m of vanadate (apparent Ki18 m, Ca2+ pool II). The remaining 60–70% could be inhibited either by vanadate at concentrations up to 1 mm (apparent Ki300 m) or by TG up to 10 nm (Ca2+ pool I). The amount of IP3-induced Ca2+ release was constant at 25% over a wide range of Ca2+ filling. About 10–20% remained unreleasable by IP3. Reduction of IP3 releasable Ca2+ in the presence of inhibitors showed similar dose-response curves as Ca2+ uptake (apparent Ki 3.0 nm for IP3-induced Ca2+ release as compared to 4.2 nm for Ca2+ uptake at TG up to 10 nm) indicating that the highly TG-sensitive Ca2+ pump fills the IP3-sensitive Ca2+ pool I. At TG concentrations >10 nm which blocked Ca2+ pool II the apparent Ki values were 11.3 and 12.1 nm, respectively. For inhibition by vanadate up to 100 m the apparent Ki values were 18 m for Ca2+ uptake and 7 m for Ca2+ release (Ca2+ pool II). At vanadate concentrations up to 1 mm the apparent Ki values were 300 and 200 m, respectively (Ca2+ pool I). Both Ca2+ pools I and II also showed different sensitivities to IP3. Dose-response curves for IP3 in the absence of inhibitors (control) showed an apparent Km value for IP3 at 0.6 m. In the presence of TG (inhibition of Ca2+ pool I) the curve was shifted to the left with an apparent Km for IP3 at 0.08 m. In the presence of vanadate (inhibition of Ca2+ pool II), the apparent Km for IP3 was 2.1 m. These data allow the conclusion that there are at least three different Ca2+ uptake mechanisms present in pancreatic acinar cells: TG- and IP3 insensitive but highly vanadate-sensitive Ca2+ uptake occurs into membrane vesicles derived from plasma membranes. Two Ca2+ pools with different TG-, vanadate- and IP3-sensitivities are most likely located in the endoplasmic reticulum at different cell sites, which could have functional implications for hormonal stimulation of pancreatic acinar cells.This work was supported by the Deutsche Forschungsgemeinschaft, Sonderforschungsbereich 246. The authors wish to thank Dr. KlausDieter Preuß for valuable discussions and Mrs. Gabriele Mörschbächer for excellent secretarial help.  相似文献   

4.
Mitochondria modulate cellular Ca2+ signals by accumulating the ion via a uniporter and releasing it via Na+- or H+-exchange. In smooth muscle, inhibition of mitochondrial Ca2+ uptake inhibits Ca2+ release from the sarcoplasmic reticulum (SR) via inositol-1,4,5-trisphosphate-sensitive receptors (IP3R). At least two mechanisms may explain this effect. First, localised uptake of Ca2+ by mitochondria may prevent negative feedback by cytosolic Ca2+ on IP3R activity, or secondly localised provision of Ca2+ by mitochondrial efflux may maintain IP3R function or SR Ca2+ content. To distinguish between these possibilities the role of mitochondrial Ca2+ efflux on IP3R function was examined. IP3 was liberated in freshly isolated single colonic smooth muscle cells and mitochondrial Na+–Ca2+ exchanger inhibited with CGP-37157 (10 μM). Mitochondria accumulated Ca2+ during IP3-evoked [Ca2+]c rises and released the ion back to the cytosol (within 15 s) when mitochondrial Ca2+ efflux was active. When mitochondrial Ca2+ efflux was inhibited by CGP-37157, an extensive and sustained loading of mitochondria with Ca2+ occurred after IP3-evoked Ca2+ release. IP3-evoked [Ca2+]c rises were initially unaffected, then only slowly inhibited by CGP-37157. IP3R activity was required for inhibition to occur; incubation with CGP-37157 for the same duration without IP3 release did not inhibit IP3R. CGP-37157 directly inhibited voltage-gated Ca2+ channel activity, however SR Ca2+ content was unaltered by the drug. Thus, the gradual decline of IP3R function that followed mitochondrial Na+–Ca2+ exchanger inhibition resulted from a gradual overload of mitochondria with Ca2+, leading to a reduced capacity for Ca2+ uptake. Localised uptake of Ca2+ by mitochondria, rather than mitochondrial Ca2+ efflux, appears critical for maintaining IP3R activity.  相似文献   

5.
A monoclonal antibody (PL/IM 430), previously found to inhibit the uptake of Ca2+ into highly purified platelet intracellular membrane vesicles (Hack, N., Wilkinson, J. M. and Crawford, N. 1988,Biochem. J. 250, 355–361) has been introduced into saponin-permeabilised platelets. At a saponin concentration (20–25 g/ml) commensurate with total LDH release, sequestration of Ca2+ into intracellular non-mitochondrial stores is inhibited by the antibody (50% inhibition at 20 g/ml IgG). At higher saponin concentrations when intracellular binding of125I-labelled mAb is maximum, inhibition of Ca2+ sequestration approaches 70%. The inhibition is specific, control studies with non-platelet directed mouse IgG and mAbs which immunoblot platelet antigens other than the 100 kDa protein did not affect the Ca2+ sequestration.No effect of the antibody were observed against IP3-induced release of prestored Ca2+, either in permeabilised platelets or with isolated intracellular membrane vesicles. The mAb PL/IM 430 appears to bind only to the Ca2+ translocating channel protein associated with the intracellular membrane (Ca2++Mg2+) ATPase and not to Ca2+ channels responsive to IP3.Abbreviations mAb monoclonal antibody - PBS phosphate buffered saline - LDH lactate dehydrogenase  相似文献   

6.
《Insect Biochemistry》1990,20(1):83-89
Isolated tick salivary glands, permeabilized with digitonin in the presence of the Ca2+ uptake inhibitors, sodium azide and vanadate, released Ca2+ in response to 20 μM inositol-1,4,5-trisphosphate (IP3). Inositol-1-phosphate (IP1) and inositol-1,4-bisphosphate (IP2) appeared to stimulate an uptake of Ca2+ into whole glands. Inositol-1,4,5-trisphosphate caused release of Ca2+ from a 100,000 g microsome enriched pellet; however, IP1 and IP2 were ineffective in stimulating an uptake or efflux of Ca2+. The combined 900 and 11,500 g pellets showed no significant release of Ca2+ in response to addition of IP3. Inositol-1,4,5-trisphosphate concentrations as low as 1 μM are capable of stimulating a significant release of Ca2+ from microsomes. Results suggest that intracellular Ca2+ is mobilized from microsomal intracellular stores in response to agonists which increase cytosolic IP3 in tick salivary glands. Results also suggest a possible role for IP1 and IP2 or both in stimulating an uptake of Ca2+ into vanadate and azide-insensitive intracellular pools.  相似文献   

7.
We examined the effect of tricyclic antidepressants on intracellular Ca2+ signalling in cultured cells of neuronal and glial origin. High concentrations of amitriptyline and desipramine increased the intracellular Ca2+ in PC-12 and U-87 MG cells. In PC-12 cells amitriptyline induced a biphasic rise in intracellular Ca2+. A rapid and transient increase due to release of Ca2+ from intracellular pools was followed by sustained elevation of [Ca2+]i due to influx from the extracellular medium. Desipramine evoked the Ca2+ release from intracellular pools but the influx of Ca2+ was not elicited. In U-87 MG cells both the drugs induced Ca2+ release from intracellular pools, however amitriptyline also induced a transient influx of Ca2+. To delineate the mechanisms involved in mobilization of Ca2+ by the drugs pharmacological agents that inhibit IP3 formation in cells and Ca2+ channel blockers were used and changes in [Ca2+]i and membrane potential were monitored. The results show that both the drugs release Ca2+ from IP3 sensitive pools by activation of phospholipase C and amitriptyline in addition activates a non specific cation channel in the plasma membrane of cells. Paradoxically at relatively lower concentrations (< 50 M) amitriptyline and desipramine inhibited the Ca2+ signal induced by adenosine triphosphate in both the cell types. Our data demonstrate that tricyclic antidepressants at different doses may have inhibitory or stimulatory effects on cellular Ca2+ signalling.  相似文献   

8.
It is known that inositol (1, 4, 5)-trisphosphate (IP3) stimulates Ca2+ release from sarcoplasmic reticulum (SR) in several tissues, but in cardiac myocytes this phenomenon has not been confirmed. The purpose of the present study was to confirm the effect of (1, 4, 5)-IP3 on Ca2+ release from SR in cardiac myocytes. The effect of IP3 on Ca2+ release from SR in hypertrophic cardiac cells was also determined.We examined the effects of IP3 on Ca2+ release from cardiac myocyte SR by the bigital-image method in a single cell. We also determined the effect of IP3 on calcium release from isolated SR. SR was prepared from spontaneous hypertensive rat hearts and Wistar kyoto rat hearts. The SR was prelabeled with45Ca2+, and then incubated with the indicated concentrations of IP3 for 1 min at 37°C. In cardiac myocytes treated with saponin, Ca2+ release stimulated by 10 M (1, 4, 5)-IP3 was detected by fura-2. In45Ca2+ prelabeled SR, the maximal Ca2+ release was achieved at 10 M IP3 incubated for 1 min. The release of Ca2+ was higher in Sr of SHR than in the SR of WKY. IP3 stimulates Ca2+ release from cardiac SR, and this release is greater in SHR than in WKY. However, it is uncertain whether this phenomenon plays a role in cardiac hypertrophy.  相似文献   

9.
A rise in the intracellular concentration of ionized calcium ([Ca2+]i) is a primary signal for contraction in all types of muscles. Recent progress in the development of imaging techniques, with special accent on fluorescence confocal microscopy, and new achievements in the synthesis of organelle- and ion-specific fluorochromes provide an experimental basis for studying the relationship between the structural organization of living smooth muscle cells (SMCs) and features of calcium signaling at the subcellular level. Applying fluorescent confocal imaging, patch-clamp recording, immunostaining, and flash photolysis techniques to freshly isolated SMCs, we have demonstrated that: (i) Ca2+ sparks are mediated by spontaneous clustered opening of ryanodine receptors (RyRs) and occur at the highest rate at preferred sites (frequent discharge sites, FDSs), the number of which depends on SMC type; (ii) FDSs are associated with sub-plasmalemmal sarcoplasmic reticulum (SR) elements, but not with polarized mitochondria; (iii) Ca2+ spark frequency increases with membrane depolarization in voltage-clamped SMCs or following neurotransmitter application to SMCs, in which the membrane potential was not controlled, leading to spark summation and resulting in a cell-wide increase in [Ca2+]i and myocyte contraction; (iv) cross-talk between RyRs and inositol trisphosphate receptors (IP3Rs) is an important determinant of the [Ca2+]i dynamics and recruits neighboring Ca2+-release sites to generate [Ca2+]i waves; (v) [Ca2+]i waves induced by depolarization of the plasma membrane or by noradrenaline or caffeine, but not by carbachol (CCh), originate at FDSs; (vi) Ca2+-dependent K+ and Cl- channels sense the local changes in [Ca2+]i during a Ca2+ spark and thereby may couple changes in [Ca2+]i within a microdomain to changes in the membrane potential, thus affecting the cell excitability; (vii) the muscarinic cation current (mI cat) does not mirror changes in [Ca2+]i, thus reflecting the complexity of [Ca2+]i — muscarinic cationic channel coupling; (viii) RyR-mediated Ca2+ release, either spontaneous or caffeine-induced, does not augment mI cat; (ix) intracellular flash release of Ca2+ is less effective in augmentation of mI cat than flash release of IP3, suggesting that IP3 may sensitize muscarinic cationic channels to Ca2+; (x) intracellular flash release of IP3 fails to augment mI cat in SMCs, in which [Ca2+]i was strongly buffered, suggesting that IP3 exerts no direct effect on muscarinic cationic channel gating, and that these channels sense an increase in [Ca2+]i rather than depletion of the IP3-dependent Ca2+ store; and (xi) predominant expression of IP3R type 1 in the peripheral SR provides a structural basis for a tight functional coupling between IP3R-mediated Ca2+ release and muscarinic cationic channel opening.Neirofiziologiya/Neurophysiology, Vol. 36, Nos. 5/6, pp. 455–465, September–December, 2004.This revised version was published online in April 2005 with a corrected cover date and copyright year.  相似文献   

10.
We investigated the direct effect of inositol 1,4,5-trisphosphate (IP3) and ryanodine receptor agonists on Ca2+ release from vesicles of a rat liver Golgi apparatus (GA) enriched fraction, which were actively loaded with 45Ca2+. Results in GA were compared with those obtained in a rat liver endoplasmic reticulum (ER) enriched fraction. The addition of IP3 at concentrations ranging from 100 nm to 100 μm, in the presence of thapsigargin, a specific inhibitor of sarcoplasmic/endoplasmic reticulum Ca2+-ATPases, promoted a rapid decrease in the Ca2+ content of GA vesicles. The amount of Ca2+ released from the vesicles was a function of IP3 concentration, reaching about 60% in both GA and ER fractions at 100 μm IP3. Calcium release was inhibited by heparin, an antagonist of IP3 receptors. Calcium exhibited a bell-shaped effect on IP3-dependent Ca2+ released from GA vesicles: it activated Ca2+ release at concentrations up to 1 μm, and inhibited it at higher concentrations. In contrast to that found in the endoplasmic reticulum fraction, none of the ryanodine receptor agonists tested (cyclic ADP-ribose, caffeine and ryanodine) significantly induced Ca2+ release from GA fraction vesicles in the presence of thapsigargin. Our results indicate the presence of an IP3-sensitive Ca2+ release mechanism in the Golgi apparatus membrane analogous to that of the ER. However, a Ca2+ release mechanism sensitive to ryanodine receptor agonists like that of ER is not evident in the GA membrane. Received: 13 March 2000/Revised: 13 July 2000  相似文献   

11.
The expression of protein kinase C (PKC) isoforms and the modulation of Ca2+ mobilization by PKC were investigated in the human submandibular duct cell line A253. Three new PKC (nPKC) isoforms (, , and ) and one atypical PKC (aPKC) isoform () are expressed in this cell line. No classical PKC (cPKC) isoforms were present. The effects of the PKC activator phorbol 12-myristate-13-acetate (PMA) and of the PKC inhibitors calphostin C (CC) and bisindolymaleimide I (BSM) on inositol 1,4,5-trisphosphate (IP3) and Ca2+ responses to ATP and to thapsigargin (TG) were investigated. Pre-exposure to PMA inhibited IP3 formation, Ca2+ release and Ca2+ influx in response to ATP. Pre-exposure to CC or BSM slightly enhanced IP3 formation but inhibited the Ca2+ release and the Ca2+ influx induced by ATP. In contrast, pre-exposure to PMA did not modify the Ca2+ release induced by TG, but reduced the influx of Ca2+ seen in the presence of this Ca2+-ATPase inhibitor. These results suggest that PKC modulates elements of the IP3/Ca2+ signal transduction pathway in A253 cells by (1) inhibiting phosphatidylinositol turnover and altering the sensitivity of the Ca2+ channels to IP3, (2) altering the activity, the sensitivity to inhibitors, or the distribution of the TG-sensitive Ca2+ ATPase, and (3) modulating Ca2+ entry pathways.  相似文献   

12.
Ryanodine receptors (RyR) are Ca2+ channels that mediate Ca2+ release from intracellular stores in response to diverse intracellular signals. In RINm5F insulinoma cells, caffeine, and 4-chloro-m-cresol (4CmC), agonists of RyR, stimulated Ca2+ entry that was independent of store-operated Ca2+ entry, and blocked by prior incubation with a concentration of ryanodine that inactivates RyR. Patch-clamp recording identified small numbers of large-conductance (γK = 169 pS) cation channels that were activated by caffeine, 4CmC or low concentrations of ryanodine. Similar channels were detected in rat pancreatic β-cells. In RINm5F cells, the channels were blocked by cytosolic, but not extracellular, ruthenium red. Subcellular fractionation showed that type 3 IP3 receptors (IP3R3) were expressed predominantly in endoplasmic reticulum, whereas RyR2 were present also in plasma membrane fractions. Using RNAi selectively to reduce expression of RyR1, RyR2, or IP3R3, we showed that RyR2 mediates both the Ca2+ entry and the plasma membrane currents evoked by agonists of RyR. We conclude that small numbers of RyR2 are selectively expressed in the plasma membrane of RINm5F pancreatic β-cells, where they mediate Ca2+ entry.Ryanodine receptors (RyR)3 and inositol 1,4,5-trisphosphate receptors (IP3R) (1, 2) are the archetypal intracellular Ca2+ channels. Both are widely expressed, although RyR are more restricted in their expression than IP3R (3, 4). In common with many cells, pancreatic β-cells and insulin-secreting cell lines express both IP3R (predominantly IP3R3) (5, 6) and RyR (predominantly RyR2) (7). Both RyR and IP3R are expressed mostly within membranes of the endoplasmic (ER), where they mediate release of Ca2+. Functional RyR are also expressed in the secretory vesicles (8, 9) or, and perhaps more likely, in the endosomes of β-cells (10). Despite earlier suggestions (11), IP3R are probably not present in the secretory vesicles of β-cells (8, 12, 13).All three subtypes of IP3R are stimulated by IP3 with Ca2+ (1), and the three subtypes of RyR are each directly regulated by Ca2+. However, RyR differ in whether their most important physiological stimulus is depolarization of the plasma membrane (RyR1), Ca2+ (RyR2) or additional intracellular messengers like cyclic ADP-ribose. The latter stimulates both Ca2+ release and insulin secretion in β-cells (8, 14). The activities of both families of intracellular Ca2+ channels are also modulated by many additional signals that act directly or via phosphorylation (15, 16). Although they commonly mediate release of Ca2+ from the ER, both IP3R and RyR select rather poorly between Ca2+ and other cations (permeability ratio, PCa/PK ∼7) (1, 17). This may allow electrogenic Ca2+ release from the ER to be rapidly compensated by uptake of K+ (18), and where RyR or IP3R are expressed in other membranes it may allow them to affect membrane potential.Both Ca2+ entry and release of Ca2+ from intracellular stores contribute to the oscillatory increases in cytosolic Ca2+ concentration ([Ca2+]i) that stimulate exocytosis of insulin-containing vesicles in pancreatic β-cells (7). Glucose rapidly equilibrates across the plasma membrane (PM) of β-cells and its oxidative metabolism by mitochondria increases the cytosolic ATP/ADP ratio, causing KATP channels to close (19). This allows an unidentified leak current to depolarize the PM (20) and activate voltage-gated Ca2+ channels, predominantly L-type Ca2+ channels (21). The resulting Ca2+ entry is amplified by Ca2+-induced Ca2+ release from intracellular stores (7), triggering exocytotic release of insulin-containing dense-core vesicles (22). The importance of this sequence is clear from the widespread use of sulfonylurea drugs, which close KATP channels, in the treatment of type 2 diabetes. Ca2+ uptake by mitochondria beneath the PM further stimulates ATP production, amplifying the initial response to glucose and perhaps thereby contributing to the sustained phase of insulin release (23). However, neither the increase in [Ca2+]i nor the insulin release evoked by glucose or other nutrients is entirely dependent on Ca2+ entry (7, 24) or closure of KATP channels (25). This suggests that glucose metabolism may also more directly activate RyR (7, 26) and/or IP3R (27) to cause release of Ca2+ from intracellular stores. A change in the ATP/ADP ratio is one means whereby nutrient metabolism may be linked to opening of intracellular Ca2+ channels because both RyR (28) and IP3R (1) are stimulated by ATP.The other major physiological regulators of insulin release are the incretins: glucagon-like peptide-1 and glucose-dependent insulinotropic hormone (29). These hormones, released by cells in the small intestine, stimulate synthesis of cAMP in β-cells and thereby potentiate glucose-evoked insulin release (30). These pathways are also targets of drugs used successfully to treat type 2 diabetes (29). The responses of β-cells to cAMP involve both cAMP-dependent protein kinase and epacs (exchange factors activated by cAMP) (31, 32). The effects of the latter are, at least partly, due to release of Ca2+ from intracellular stores via RyR (3335) and perhaps also via IP3R (36). The interplays between Ca2+ and cAMP signaling generate oscillatory changes in the concentrations of both messengers (37). RyR and IP3R are thus implicated in mediating responses to each of the major physiological regulators of insulin secretion: glucose and incretins.Here we report that in addition to expression in intracellular stores, which probably include both the ER and secretory vesicles and/or endosomes, functional RyR2 are also expressed in small numbers in the PM of RINm5F insulinoma cells and rat pancreatic β-cells.  相似文献   

13.
Regulation of bi-directional communication between intracellular Ca2+ pools and surface Ca2+ channels remains incompletely characterized. We report Ca2+ release mediated by inositol 1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RyR) pathways is diminished under actin cytoskeleton disruption in NG115-401L (401L) neuronal cells, yet despite truncated Ca2+ release, Ca2+ influx was not significantly altered in these experiments. However, disruption of cortical actin networks completely abolished IP3R induced Ca2+ release, whereas RyR-mediated Ca2+ release was preserved, albeit attenuated. Moreover, cortical actin disruption completely abolished IP3R and RyR linked Ca2+ influx even though Ca2+ pool sensitivities were different. These findings suggest discrete Ca2+ store/Ca2+ channel coupling mechanisms in the IP3R and RyR pathways as revealed by the differential sensitivity to actin perturbation.  相似文献   

14.
To investigate the presence and the size of different non-mitochondria) Ca2+ pools of Ehrlich ascites tumor cells (EATCs) , digitonin-permeabilized cells were allowed to accumulate Ca2+ in the presence of mitochondrial inhibitors and treated with the reticular Ca2+-ATPase inhibitor thapsigargin, IP3 and the Ca2+ ionophore A23187. Emptying of thapsigargin-sensitive Ca2+ stores prevented any Ca2+ release by IP3, and, after IP3 addition, little or no Ca2+ was released by thapsigargin. In both instances, a further Ca2+ release was accomplished by A23187. The IP3-thapsigargin-sensitive pool and the residual A23187-sensitive one corresponded to approximately 60 and 37% of non-mitochondria) stored Ca2+, respectively. In intact EATCs, IP3-dependent agonists and thapsigargin discharged Ca 2+ pools almost completely overlapping, and A32187 released a minor residual Ca2+ pool. The IP3-insensitive pool appeared to have a relatively low affinity for Ca2+ (below 600 nM). The high affinity, IP3-sensitive Ca2+ pool was discharged in a ‘quantal’ manner following step additions of sub maximal [IP3], and the IP3-induced fractional Ca2+ release was more marked at higher concentrations of stored (luminal) Ca2+, The IP3-sensitive Ca2+ pool appeared to be devoid of the Ca2+-activated Ca2+ release channel since caffeine did not released any Ca2+ in intact and permeabilized EATCs, and Western blot analyses of EATC microsomal membranes failed to detect any known ryanodine receptor isoform.  相似文献   

15.
Summary Plasma membrane vesicles, which are mostly right side-out, were isolated from corn leaves by aqueous two-phase partitioning method. Characteristics of Ca2+ transport were investigated after preparing inside-out vesicles by Triton X-100 treatment.45Ca2+ transport was assayed by membrane filtration technique. Results showed that Ca2+ transport into the plasma membrane vesicles was Mg-ATP dependent. The active Ca2+ transport system had a high affinity for Ca2+(K m (Ca2+)=0.4 m) and ATP(K m (ATP)=3.9 m), and showed pH optimum at 7.5. ATP-dependent Ca2+ uptake in the plasma membrane vesicles was stimulated in the presence of Cl or NO 3 . Quenching of quinacrine fluorescence showed that these anions also induced H+ transport into the vesicles. The Ca2+ uptake stimulated by Cl was dependent on the activity of H+ transport into the vesicles. However, carbonylcyanidem-chlorophenylhydrazone (CCCP) and VO 4 3– which is known to inhibit the H+ pump associated with the plasma membrane, canceled almost all of the Cl-stimulated Ca2+ uptake. Furthermore, artificially imposed pH gradient (acid inside) caused Ca2+ uptake into the vesicles. These results suggest that the Cl-stimulated Ca2+ uptake is caused by the efflux of H+ from the vesicles by the operation of Ca2+/H+ antiport system in the plasma membrane. In Cl-free medium, H+ transport into the vesicles scarcely occurred and the addition of CCCP caused only a slight inhibition of the active Ca2+ uptake into the vesicles. These results suggest that two Ca2+ transport systems are operating in the plasma membrane from corn leaves, i.e., one is an ATP-dependent active Ca2+ transport system (Ca2+ pump) and the other is a Ca2+/H+ antiport system. Little difference in characteristics of Ca2+ transport was observed between the plasma membranes isolated from etiolated and green corn leaves.  相似文献   

16.
Summary The presence of a coupled Na+/Ca2+ exchange system has been demonstrated in plasma membrane vesicles from rat pancreatic acinar cells. Na+/Ca2+ exchange was investigated by measuring45Ca2+ uptake and45Ca2+ efflux in the presence of sodium gradients and at different electrical potential differences across the membrane (=) in the presence of sodium. Plasma membranes were prepared by a MgCl2 precipitation method and characterized by marker enzyme distribution. When compared to the total homogenate, the typical marker for the plasma membrane, (Na++K+)-ATPase was enriched by 23-fold. Markers for the endoplasmic reticulum, such as RNA and NADPH cytochromec reductase, as well as for mitochondria, the cytochromec oxidase, were reduced by twofold, threefold and 10-fold, respectively. For the Na+/Ca2+ countertransport system, the Ca2+ uptake after 1 min of incubation was half-maximal at 0.62 mol/liter Ca2+ and at 20 mmol/liter Na+ concentration and maximal at 10 mol/liter Ca2+ and 150 mmol/liter Na+ concentration, respecitively. When Na+ was replaced by Li+, maximal Ca2+ uptake was 75% as compared to that in the presence of Na+. Amiloride (10–3 mol/liter) at 200 mmol/liter Na+ did not inhibit Na+/Ca2+ countertransport, whereas at low Na+ concentration (25 mmol/liter) amiloride exhibited dose-dependent inhibition to be 62% at 10–2 mol/liter. CFCCP (10–5 mol/liter) did not influence Na+/Ca2+ countertransport. Monensin inhibited dose dependently; at a concentration of 5×10–6 mol/liter inhibition was 80%. A SCN or K+ diffusion potential (=), being positive at the vesicle inside, stimulated calcium uptake in the presence of sodium suggesting that Na+/Ca2+ countertransport operates electrogenically, i.e. with a stoichiometry higher than 2 Na+ for 1 Ca2+. In the absence of Na+, did not promote Ca2+ uptake. We conclude that in addition to ATP-dependent Ca2+ outward transport as characterized previously (E. Bayerdörffer, L. Eckhardt, W. Haase & 1. Schulz, 1985,J. Membrane Biol. 84:45–60) the Na+/Ca2+ countertransport system, as characterized in this study, represents a second transport system for the extrusion of calcium from the cell. Furthermore, the high affinity for calcium suggests that this system might participate in the regulation of the cytosolic free Ca2+ level.  相似文献   

17.
In this study we have used saponin to permeabilize platelet membranes in order to test directly the involvement of IP3 in regulating internal Ca2+ release, and to measure IP3 binding to its receptor. Our results indicate that platelet vesicles release Ca2+ as early as 3 seconds after IP3 addition. Using [3H]IP3, we have found that platelets contain a single class of high affinity IP3 binding sites with a Kd of ~0.20 (± 0.01) nM. Immuno-blotting shows that platelets contain a 260 kDa polypeptide which shares immunological cross reactivity with brain IP3 receptor. Immunofluorescence staining data indicate that the IP3 receptor is preferentially located at the periphery of the platelet plasma membrane. Most importantly, both IP3 binding and IP3-induced Ca2+ release activities are significantly inhibited by cytochalasin D (a microfilament inhibitor) and colchicine (a microtubule inhibitor). These findings suggest that the cytoskeleton is involved in the regulation of IP3 binding and IP3 receptor-mediated Ca2+ release during platelet activation.  相似文献   

18.
Intracellular Ca2+ mobilization events were assessed in mouse L cells, which contain native prostaglandin E1 receptors and transfected human 2 adrenergic receptors. Both Fura2 (single cell measurements) and Quin 2, (cuvette assays) were used to determine [Ca2+]i levels. Our results demonstrate that in the transfected cells there is a dose-dependent increase in [Ca2+]i in response to isoproterenol (0.1 nM–100 nM), which is inhibited by the -adrenergic antagonist, propranolol, and is a result of intracellular Ca2+ release. [Ca2+]1 in these cells was also increased by prostaglandin E1, 8 bromo cyclic AMP, and aluminum fluoride. Both 8 bromo cAMP and isoproterenol induced a rapid increase in the levels of IP1, IP2, and IP3. The data presented demonstrate that the elevation of intracellular cyclic AMP induces an increase in IP3 production which leads to an elevation in [Ca2+];. We propose that this cyclic AMP dependent activation of the IP3 generating system occurs at a post-receptor site.Abbreviations cAMP Adenosine Cyclic 3-5-Monophosphate - [Ca2+]i intracellular [Ca2+]i - 8 Br cAMP 8 Bromo Adenosine Cyclic 3-5-Monophosphate - DAG Diacylglycerol - EGTA] [Ethylene Bis (oxyethylenenitrilo)] Tetracetic acid - BSA Bovine Serum Albumin - HBSS-H Hanks' Balanced Salt Solution buffered with HEPES to pH 7.4 - HEPES 4-(2-Hydroxyethyl)-1-piperazineethanesulfonic acid - PIP2 Phosphatidylinositol 4,5-bisphosphate - IP2 Inositol 4 Phosphate - IP2 Inositol 4,5 Bisphosphate - IP3 Inositol Trisphosphate - PGE1 Prostaglandin E1 - PBS Phosphate Buffered Saline Solution  相似文献   

19.
Inositol 1,4,5-trisphosphate-induced calcium release from canine aortic smooth muscle sarcoplasmic reticulum vesicles was examined using the calcium indicator antipyrylazo III. Calcium release was initiated by addition of inositol 1,4,5-trisphosphate (IP3) to aortic vesicles 7 min after initiation of ATP-supported calcium uptake. Half-maximal calcium release occurred at 1 μM IP3, with maximal calcium release amounting to 25±2% of the intravesicular calcium (n=12, 9 preparations). Ruthenium red (10–20 μM), which has been reported to block IP3-induced calcium release from skeletal muscle sarcoplasmic reticulum, did not inhibit aortic IP3-induced calcium release. Elevation of Mg2+ concentration from 0.06 to 7.8 mM inhibited aortic IP3-induced calcium release 75%, which contrasts with the Mg2+-insensitive IP3-induced calcium release from platelet reticular membranes. The IP3-dependence of aortic calcium release suggested that Mg2+ acted as a noncompetitive inhibitor. Thus, aortic sarcoplasmic reticulum vesicles contain an IP3-sensitive calcium pathway which is inhibited by millimolar concentrations of Mg2+, but which is not inhibited by Ruthenium red and so differs from the previously described IP3-sensitive calcium pathways in skeletal muscle and platelet reticular membranes.  相似文献   

20.
KRAS-induced actin-interacting protein (KRAP) was originally characterized as a filamentous- actin-interacting protein. We have recently found that KRAP is an associated molecule with inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and is responsible for the proper subcellular localization of IP3R. Since it remains unknown whether KRAP regulates the IP3R-mediated Ca2+ signaling, we herein examined the effects of KRAP on the IP3R-mediated Ca2+ release by Ca2+ imagings in the cultured HEK293 or MCF7 cells. Reduction of KRAP protein by KRAP-specific siRNA diminishes ATP-induced Ca2+ release and the ATP-induced Ca2+ release is completely quenched by the pretreatment with the IP3R inhibitor but not with the ryanodine receptor inhibitor, indicating that KRAP regulates IP3R-mediated Ca2+ release. To further reveal mechanistic insights into the regulation of IP3R-mediated Ca2+ release by KRAP, we examined the effects of the KRAP-knockdown on the releasable Ca2+ content of intracellular Ca2+ stores. Consequently, reduction of KRAP does not affect the amount of ionophore- or Ca2+-ATPase inhibitor-induced Ca2+ release in the HEK293 cells, indicating that releasable Ca2+ content of intracellular Ca2+ stores is not altered by KRAP. Thus, KRAP is involved in the proper regulation of IP3R-mediated Ca2+ release.  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