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.
Exposure of renal proximal tubule cells to oxalate may play an important role in cell proliferation, but the signaling pathways involved in this effect have not been elucidated. Thus the present study was performed to examine the effect of oxalate on 3H-labeled thymidine incorporation and its related signal pathway in primary cultured rabbit renal proximal tubule cells (PTCs). The effects of oxalate on [3H]thymidine incorporation, lactate dehydrogenase (LDH) release, Trypan blue exclusion, H2O2 release, activation of mitogen-activated protein kinases (MAPKs), and 3H-labeled arachidonic acid (AA) release were examined in primary cultured renal PTCs. Oxalate inhibited [3H]thymidine incorporation in a time- and dose-dependent manner. However, its analogs did not affect [3H]thymidine incorporation. Oxalate (1 mM) significantly increased H2O2 release, which was blocked by N-acetyl-L-cysteine (NAC) and catalase (antioxidants). Oxalate significantly increased p38 MAPK and stress-activated protein kinase (SAPK)/c-Jun NH2-terminal kinase (JNK) activity, not p44/42 MAPK. Oxalate stimulated [3H]AA release and translocation of cytosolic phospholipase A2 (cPLA2) from the cytosolic fraction to the membrane fraction. Indeed, oxalate significantly increased prostaglandin E2 (PGE2) production compared with control. Oxalate-induced inhibition of [3H]thymidine incorporation and increase of [3H]AA release were prevented by antioxidants (NAC), a p38 MAPK inhibitor (SB-203580), a SAPK/JNK inhibitor (SP-600125), or PLA2 inhibitors [mepacrine and arachidonyl trifluoromethyl ketone (AACOCF3)], but not by a p44/42 MAPK inhibitor (PD-98059). These findings suggest that oxalate inhibits renal PTC proliferation via oxidative stress, p38 MAPK/JNK, and cPLA2 signaling pathways. kidney; mitogen-activated protein kinase; phospholipase A2  相似文献   

2.
The accumulation of radiolabeled arachidonicacid (AA), immunoblot analysis of subcellular fractions, andimmunofluorescence tagging of proteins in intact cells were used toexamine the coupling of ANG II receptors with the activity and locationof a cytosolic phospholipase A2(cPLA2) in vascular smoothmuscle cells (VSMC). ANG II induced the accumulation of AA, whichpeaked by 10 min and was downregulated by 20 min. A large proportion ofthe AA released in response to ANG II was due to the activation of a Ca2+-dependent lipase coupled toan AT1 receptor. However,regulation of Ca2+ availabilityfailed to completely block AA release, and a small but significantreduction in ANG II-mediated AA release was observed in the presence ofan AT2 antagonist. These findings,coupled with a 25% reduction in the ANG II-induced AA release by aninhibitor specific for aCa2+-independentPLA2, are consistent with thepresence and activation of aCa2+-independentPLA2. In contrast, immunoblotanalysis and immunofluorescence detection showed that the ANGII-mediated translocation of cPLA2 to a membrane fraction was exclusivelyAT1 dependent and regulated byCa2+ availability. Furthermore,the nucleus was the membrane target. We conclude that ANG II regulatesthe Ca2+-dependent activation andtranslocation of cPLA2 through anAT1 receptor and that this eventis targeted at the nucleus in VSMC.

  相似文献   

3.
To test thehypothesis that intracellular Ca2+activation of large-conductanceCa2+-activatedK+ (BK) channels involves thecytosolic form of phospholipase A2 (cPLA2), we first inhibited theexpression of cPLA2 by treating GH3 cells with antisenseoligonucleotides directed at the two possible translation start siteson cPLA2. Western blot analysis and a biochemical assay of cPLA2activity showed marked inhibition of the expression ofcPLA2 in antisense-treated cells.We then examined the effects of intracellularCa2+ concentration([Ca2+]i)on single BK channels from these cells. Open channel probability (Po) for thecells exposed to cPLA2 antisenseoligonucleotides in 0.1 µM intracellularCa2+ was significantly lower thanin untreated or sense oligonucleotide-treated cells, but the voltagesensitivity did not change (measured as the slope of thePo-voltagerelationship). In fact, a 1,000-fold increase in[Ca2+]ifrom 0.1 to 100 µM did not significantly increasePoin these cells, whereas BK channels from cells in the other treatmentgroups showed a normalPo-[Ca2+]iresponse. Finally, we examined the effect of exogenous arachidonic acidon thePoof BK channels from antisense-treated cells. Although arachidonic aciddid significantly increasePo,it did so without restoring the[Ca2+]isensitivity observed in untreated cells. We conclude that although [Ca2+]idoes impart some basal activity to BK channels inGH3 cells, the steepPo-[Ca2+]irelationship that is characteristic of these channels involves cPLA2.

  相似文献   

4.
Ceramide and the metabolites including ceramide-1-phosphate (C1P) and sphingosine are reported to regulate the release of arachidonic acid (AA) and/or phospholipase A2 (PLA2) activity in many cell types including lymphocytes. Recent studies established that C1P, a product of ceramide kinase, interacts directly with Ca2+ binding regions in the C2 domain of α type cytosolic PLA2 (cPLA2α), leading to translocation of the enzyme from the cytosol to the perinuclear region in cells. However, a precise mechanism for C1P-induced activation of cPLA2α has not been well elucidated; such as the phosphorylation signal caused by the extracellular signal-regulated kinases (ERK1/2) pathway, a downstream of the protein kinase C activation with 4β-phorbol myristate acetate (PMA), is required or not. In the present study, we showed that the increase in intracellular ceramide levels (exogenously added cell permeable ceramides and an inhibition of ceramidase by (1S,2R)-D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol and the increase in C1P formation by transfection with the vector for human ceramide kinase significantly enhanced the Ca2+ ionophore (A23187) -induced release of AA via cPLA2α's activation in CHO cells. Ceramides did not show additional effects on the release from the cells treated with the inhibitor of ceramidase. Ceramides and C2-C1P neither had effect on the intracellular mobilization of Ca2+ nor the phosphorylation of cPLA2α in cells. A23187/PMA-induced release of AA was enhanced by ceramides and C2-C1P and by expression of ceramide kinase. Our findings suggest that C1P is a stimulatory factor on cPLA2α that is independent of the Ca2+ signal and the PKC-ERK-mediated phosphorylation signal.  相似文献   

5.
We have examined the mechanisms regulatingprostacyclin (PGI2) synthesis after acute exposure of humanumbilical vein endothelial cells (HUVEC) to interleukin-1 (IL-1).IL-1 evoked an early (30 min) release of PGI2 and[3H]arachidonate that was blocked by the cytosolicphospholipase A2 (cPLA2) inhibitorarachidonyl trifluoromethyl ketone. IL-1-mediated activationof extracellular signal-regulated kinase 1/2 (ERK1/2; p42/p44mapk) coincided temporally with phosphorylation ofcPLA2 and with the onset of PGI2synthesis. The mitogen-activated protein kinase (MAPK) kinase (MEK)inhibitors, PD-98059 and U-0126, blocked IL-1-induced ERKactivation and partially attenuated cPLA2phosphorylation and PGI2 release, suggesting thatERK-dependent and -independent pathways regulate cPLA2phosphorylation. SB-203580 treatment enhanced IL-1-induced MEK,p42/44mapk, and cPLA2 phosphorylation butreduced thrombin-stimulated MEK and p42/44mapk activation.IL-1, but not thrombin, activated Raf-1 as assessed byimmune-complex kinase assay, as did SB-203580 alone. These results showthat IL-1 causes an acute upregulation of PGI2generation in HUVEC, establish a role for theMEK/ERK/cPLA2 pathway in this early release, and provideevidence for an agonist-specific cross talk between p38mapkand p42/44mapk that may reflect receptor-specificdifferences in the signaling elements proximal to MAPK activation.

  相似文献   

6.
Mechanisms of MAPK activation by bradykinin in vascular smooth muscle cells   总被引:4,自引:0,他引:4  
Vascular smooth muscle cell (VSMC) proliferation is a prominentfeature of the atherosclerotic process occurring after endothelial injury. A vascular wall kallikrein-kinin system has been described. Thecontribution of this system to vascular disease is undefined. In thepresent study we characterized the signal transduction pathway leadingto mitogen-activated protein kinase (MAPK) activation in response tobradykinin (BK) in VSMC. Addition of1010-107M BK to VSMC resulted in a rapid and concentration-dependent increasein tyrosine phosphorylation of several 144- to 40-kDa proteins. Thiseffect of BK was abolished by theB2-kinin receptor antagonistHOE-140, but not by the B1-kininreceptor antagonist des-Arg9-Leu8-BK.Immunoprecipitation with anti-phosphotyrosine antibodies followed byimmunoblot revealed that109 M BK induced tyrosinephosphorylation of focal adhesion kinase (p125FAK). BK(108 M) promoted theassociation of p60src with theadapter protein growth factor receptor binding protein-2 and alsoinduced a significant increase in MAPK activity. Pertussis and choleratoxins did not inhibit BK-induced MAPK tyrosine phosphorylation. Protein kinase C downregulation by phorbol 12-myristate 13-acetate and/or inhibitors to protein kinase C,p60src kinase, and MAPK kinaseinhibited BK-induced MAPK tyrosine phosphorylation. These findingsprovide evidence that activation of theB2-kinin receptor in VSMC leads togeneration of multiple second messengers that converge to activateMAPK. The activation of this crucial kinase by BK provides a strongrationale to investigate the mitogenic actions of BK on VSMCproliferation in disease states of vascular injury.

  相似文献   

7.
In cultured porcine aortic smooth muscle cells,sphingosylphosphorylcholine (SPC), ATP, or bradykinin (BK) induced arapid dose-dependent increase in the cytosolicCa2+ concentration([Ca2+]i)and also stimulated inositol 1,4,5-trisphosphate(IP3) generation. Pretreatmentof cells with pertussis toxin blocked the SPC-induced IP3 generation and[Ca2+]iincrease but had no effect on the action of ATP or BK. In addition, SPCstimulated the mitogen-activated protein kinase (MAPK) and increasedDNA synthesis, whereas neither ATP nor BK produced such effects. Boththe SPC-induced MAPK activation and DNA synthesis were pertussis toxinsensitive. SPC-induced MAPK activation was blocked by treatment ofcells with the phospholipase C inhibitor, U-73122, or the intracellularCa2+-ATPase inhibitor,thapsigargin, but not by removal of extracellular Ca2+. Lysophosphatidic acidinduced cellular responses similar to SPC in a pertussistoxin-sensitive manner in terms of[Ca2+]iincrease, IP3 generation, MAPKactivation, and DNA synthesis. Platelet-derived growth factor (PDGF)also induced a[Ca2+]iincrease, MAPK activation, and DNA synthesis in the same cells; however, the PDGF-induced MAPK activation was not sensitive to pertussis toxin and changes in[Ca2+]i.SPC-induced MAPK activation was inhibited by pretreatment of cells withstaurosporine, W-7, or calmidazolium. Our results suggest that, inporcine aortic smooth muscle cells, MAPK is not activated by theincrease in[Ca2+]iunless a pertussis toxin-sensitive G protein is simultaneously stimulated, indicating the role ofCa2+ in pertussis toxin-sensitiveG protein-mediated MAPK activation.

  相似文献   

8.
Little is known about the mechanism and signal transduction by LPS-mediated immunomodulation of murine peritoneal macrophages. It is found that the signal molecules of the down-stream of Ras, Raf-1, MAPK p44, and MAPK p42 are phosphorylated, and cPLA2 is activated with a significant increase of the release of [ H3 ] AA by macrophages in response to LPS and PMA. Compared with the very recent finding that LPS and PMA trigger the activation and translocation of PKC-α and PKC-ε, these findings suggest that there is a connection between PKC signaling pathway and the Raf-1/MAPK pathway and that the activation of these main signaling events may be closely related to the secretion of IL-12 during LPS-induced modulation of macrophages.  相似文献   

9.
LocalCa2+ transients("Ca2+ sparks") caused bythe opening of one or the coordinated opening of a number of tightlyclustered ryanodine-sensitiveCa2+-release (RyR) channels in thesarcoplasmic reticulum (SR) activate nearbyCa2+-dependentK+(KCa) channels to cause anoutward current [referred to as a "spontaneous transientoutward current" (STOC)]. TheseKCa currents cause membranepotential hyperpolarization of arterial myocytes, which would lead tovasodilation through decreasingCa2+ entry throughvoltage-dependent Ca2+ channels.Therefore, modulation of Ca2+spark frequency should be a means to regulation ofKCa channel currents and hencemembrane potential. We examined the frequency modulation ofCa2+ sparks and STOCs byactivation of protein kinase C (PKC). The PKC activators, phorbol12-myristate 13-acetate (PMA; 10 nM) and 1,2-dioctanoyl-sn-glycerol (1 µM),decreased Ca2+ spark frequency by72% and 60%, respectively, and PMA reduced STOC frequency by 83%.PMA also decreased STOC amplitude by 22%, which could be explained byan observed reduction (29%) inKCa channel open probability inthe absence of Ca2+ sparks. Thereduction in STOC frequency occurred in the presence of an inorganicblocker (Cd2+) ofvoltage-dependent Ca2+ channels.The reduction in Ca2+ sparkfrequency did not result from SRCa2+ depletion, sincecaffeine-induced Ca2+ transientsdid not decrease in the presence of PMA. These results suggest thatactivators of PKC can modulate the frequency ofCa2+ sparks, through an effect onthe RyR channel, which would decrease STOC frequency (i.e.,KCa channel activity).

  相似文献   

10.
In inflammatory cells, agonist-stimulated arachidonic acid (AA) release is thought to be induced by activation of group IV Ca(2+)-dependent cytosolic phospholipase A(2) (cPLA(2)) through mitogen-activated protein kinase (MAP kinase)- and/or protein kinase C (PKC)-mediated phosphorylation and Ca(2+)-dependent translocation of the enzyme to the membrane. Here we investigated the role of phospholipases in N-formylmethionyl-l-leucyl-l-phenylalanine (fMLP; 1 nM-10 microM)-induced AA release from neutrophil-like db-cAMP-differentiated HL-60 cells. U 73122 (1 microM), an inhibitor of phosphatidyl-inositol-4,5-biphosphate-specific phospholipase C, or the membrane-permeant Ca(2+)-chelator 1, 2-bis?2-aminophenoxy?thane-N,N,N',N'-tetraacetic acid (10 microM) abolished fMLP-mediated Ca(2+) signaling, but had no effect on fMLP-induced AA release. The protein kinase C-inhibitor Ro 318220 (5 microM) or the inhibitor of cPLA(2) arachidonyl trifluoromethyl ketone (AACOCF(3); 10-30 microM) did not inhibit fMLP-induced AA release. In contrast, AA release was stimulated by the Ca(2+) ionophore A23187 (10 microM) plus the PKC activator phorbol myristate acetate (PMA) (0.2 microM). This effect was inhibited by either Ro 318220 or AACOCF(3). Accordingly, a translocation of cPLA(2) from the cytosol to the membrane fraction was observed with A23187 + PMA, but not with fMLP. fMLP-mediated AA release therefore appeared to be independent of Ca(2+) signaling and PKC and MAP kinase activation. However, fMLP-mediated AA release was reduced by approximately 45% by Clostridium difficile toxin B (10 ng/ml) or by 1-butanol; both block phospholipase D (PLD) activity. The inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC), D609 (100 microM), decreased fMLP-mediated AA release by approximately 35%. The effect of D609 + 1-butanol on fMLP-induced AA release was additive and of a magnitude similar to that of propranolol (0.2 mM), an inhibitor of phosphatidic acid phosphohydrolase. This suggests that the bulk of AA generated by fMLP stimulation of db-cAMP-differentiated HL-60 cells is independent of the cPLA(2) pathway, but may originate from activation of PC-PLC and PLD.  相似文献   

11.
The L-type Ca2+ channel is the primary voltage-dependent Ca2+-influx pathway in many excitable and secretory cells, and direct phosphorylation by different kinases is one of the mechanisms involved in the regulation of its activity. The aim of this study was to evaluate the participation of Ser/Thr kinases and tyrosine kinases (TKs) in depolarization-induced Ca2+ influx in the endocrine somatomammotrope cell line GH3. Intracellular Ca2+ concentration ([Ca2+]i) was measured using a spectrofluorometric method with fura 2-AM, and 12.5 mM KCl (K+) was used as a depolarization stimulus. K+ induced an abrupt spike (peak) in [Ca2+]i that was abolished in the presence of nifedipine, showing that K+ enhances [Ca2+]i, preferably activating L-type Ca2+ channels. H89, a selective PKA inhibitor, significantly reduced depolarization-induced Ca2+ mobilization in a concentration-related manner when it was applied before or after K+, and okadaic acid, an inhibitor of Ser/Thr phosphatases, which has been shown to regulate PKA-stimulated L-type Ca2+ channels, increased K+-induced Ca2+ entry. When PKC was activated by PMA, the K+-evoked peak in [Ca2+]i, as well as the plateau phase, was significantly reduced, and chelerythrine (a PKC inhibitor) potentiated the K+-induced increase in [Ca2+]i, indicating an inhibitory role of PKC in voltage-dependent Ca2+ channel (VDCC) activity. Genistein, a TK inhibitor, reduced the K+-evoked increase in [Ca2+]i, but, unexpectedly, the tyrosine phosphatase inhibitor orthovanadate reduced not only basal Ca2+ levels but, also, Ca2+ influx during the plateau phase. Both results suggest that different TKs may act differentially on VDCC activation. Activation of receptor TKs with epidermal growth factor (EGF) or vascular endothelial growth factor potentiated K+-induced Ca2+ influx, and AG-1478 (an EGF receptor inhibitor) decreased it. However, inhibition of the non-receptor TK pp60 c-Src enhanced K+-induced Ca2+ influx. The present study strongly demonstrates that a complex equilibrium among different kinases and phosphatases regulates VDCC activity in the pituitary cell line GH3: PKA and receptor TKs, such as vascular endothelial growth factor receptor and EGF receptor, enhance depolarization-induced Ca2+ influx, whereas PKC and c-Src have an inhibitory effect. These kinases modulate membrane depolarization and may therefore participate in the regulation of a plethora of intracellular processes, such as hormone secretion, gene expression, protein synthesis, and cell proliferation, in pituitary cells. phosphatases; protein kinase A; protein kinase C; epidermal growth factor  相似文献   

12.
Large-conductance Ca2+-activated K+ (BK) channels are reported to be essential for NADPH oxidase-dependent microbial killing and innate immunity in leukocytes. Using human peripheral blood and mouse bone marrow neutrophils, pharmacological targeting, and BK channel gene-deficient (BK–/–) mice, we stimulated NADPH oxidase activity with 12-O-tetradecanoylphorbol-13-acetate (PMA) and performed patch-clamp recordings on isolated neutrophils. Although PMA stimulated NADPH oxidase activity as assessed by O2 and H2O2 production, our patch-clamp experiments failed to show PMA-activated BK channel currents in neutrophils. In our studies, PMA induced slowly activating currents, which were insensitive to the BK channel inhibitor iberiotoxin. Instead, the currents were blocked by Zn2+, which indicates activation of proton channel currents. BK channels are gated by elevated intracellular Ca2+ and membrane depolarization. We did not observe BK channel currents, even during extreme depolarization to +140 mV and after elevation of intracellular Ca2+ by N-formyl-L-methionyl-L-leucyl-phenylalanine. As a control, we examined BK channel currents in cerebral and tibial artery smooth muscle cells, which showed characteristic BK channel current pharmacology. Iberiotoxin did not block killing of Staphylococcus aureus or Candida albicans. Moreover, we addressed the role of BK channels in a systemic S. aureus and Yersinia enterocolitica mouse infection model. After 3 and 5 days of infection, we found no differences in the number of bacteria in spleen and kidney between BK–/– and BK+/+ mice. In conclusion, our experiments failed to identify functional BK channels in neutrophils. We therefore conclude that BK channels are not essential for innate immunity. killing assay; reactive oxygen species; BK-deficient mice; mice infection  相似文献   

13.
We demonstrated previously that thrombin stimulation of endothelial cells activates a membrane-associated, Ca2+-independent phospholipase A2 (iPLA2) that selectively hydrolyzes arachidonylated plasmalogen phospholipids. We report that incubation of human coronary artery endothelial cells (HCAEC) with phorbol 12-myristate 13-acetate (PMA) to activate protein kinase C (PKC) resulted in hydrolysis of cellular phospholipids similar to that observed with thrombin stimulation (0.05 IU/ml; 10 min). Thrombin stimulation resulted in a decrease in arachidonylated plasmenylcholine (2.7 ± 0.1 vs. 5.3 ± 0.4 nmol PO4/mg of protein) and plasmenylethanolamine (7.5 ± 1.0 vs. 12.0 ± 0.9 nmol PO4/mg of protein). Incubation with PMA resulted in decreases in arachidonylated plasmenylcholine (3.2 ± 0.3 nmol PO4/mg of protein) and plasmenylethanolamine (6.0 ± 1.0 nmol PO4/mg of protein). Incubation of HCAEC with the selective iPLA2 inhibitor bromoenol lactone (5 mM; 10 min) inhibited accelerated plasmalogen phospholipid hydrolysis in response to both PMA and thrombin stimulation. Incubation of HCAEC with PMA (100 nM; 5 min) resulted in increased arachidonic acid release (7.1 ± 0.3 vs. 1.1 ± 0.1%) and increased production of lysoplasmenylcholine (1.4 ± 0.2 vs. 0.6 ± 0.1 nmol PO4/mg of protein), similar to the responses observed with thrombin stimulation. Downregulation of PKC by prolonged exposure to PMA (100 nM; 24 h) completely inhibited thrombin-stimulated increases in arachidonic acid release (7.1 ± 0.6 to 0.5 ± 0.1%) and lysoplasmenylcholine production (2.0 ± 0.1 to 0.2 ± 0.1 nmol PO4/mg of protein). These data suggest that PKC activates iPLA2 in HCAEC, leading to accelerated plasmalogen phospholipid hydrolysis and increased phospholipid metabolite production. lysophospholipids; cell signaling; phospholipid metabolism; arachidonic acid  相似文献   

14.
Localized Ca2+ transients resulting from inositoltrisphosphate (IP3)-dependent Ca2+ releasecouple to spontaneous transient outward currents (STOCs) in murinecolonic myocytes. Confocal microscopy and whole cell patch-clamptechniques were used to investigate coupling between localizedCa2+ transients and STOCs. Colonic myocytes were loadedwith fluo 3. Reduction in external Ca2+([Ca2+]o) reduced localized Ca2+transients but increased STOC amplitude and frequency. Simultaneous recordings of Ca2+ transients and STOCs showed increasedcoupling strength between Ca2+ transients and STOCs when[Ca2+]o was reduced. Gd3+ (10 µM) did not affect Ca2+ transients but increased STOCamplitude and frequency. Similarly, an inhibitor of Ca2+influx,1-2-(4-methoxyphenyl)-2-[3-(4-methoxyphenyl)propoxy]ethyl-1H-imidazole (SKF-96365), increased STOC amplitude and frequency. A protein kinase C(PKC) inhibitor, GF-109203X, also increased the amplitude and frequencyof STOCs but had no effect on Ca2+ transients. Phorbol12-myristate 13-acetate (1 µM) reduced STOC amplitude and frequencybut did not affect Ca2+ transients. 4-Phorbol (1 µM)had no effect on STOCs or Ca2+ transients. Single channelstudies indicated that large-conductance Ca2+-activatedK+ (BK) channels were inhibited by aCa2+-dependent PKC. In summary 1)Ca2+ release from IP3 receptor-operated storesactivates Ca2+-activated K+ channels;2) Ca2+ influx through nonselective cationchannels facilitates activation of PKC; and 3) PKC reducesthe Ca2+ sensitivity of BK channels, reducing the couplingstrength between localized Ca2+ transients and BK channels.

  相似文献   

15.
Binding of LA350, a lymphoblastoid human B cell line, by phorbol myristate acetate (PMA) plus a calcium ionophore, either ionomycin or A23187, produced unique alterations in the release of arachidonic acid (AA) from cellular phospholipids. After equilibrium labeling of cells with radioactive fatty acids, [14C]AA demonstrated a selective enhanced release from the cells in response to the binding of PMA plus calcium ionophore as compared to the release of [14C]stearic acid (STE), [3H]oleic acid (OLE) and [3H]palmitic acid (PAL). The major phospholipid sources of the released [14C]AA were shown to be phosphatidylcholine, phosphatidylethanolamine, and phosphatidylinositol. The participation of protein kinase C (PKC) in the enhanced synergistic release of [14C]AA was demonstrated by the inhibition of the release by the PKC inhibitor, staurosporine. Approximately 2-6% of the labeled AA liberated was converted to 5-hydroxyeicosatetraenoic acid by an endogenous 5-lipoxygenase. Therefore during cell activation the B cell is capable of liberating AA via a PKC-dependent mechanism, implicating AA and/or its metabolites in signal transduction.  相似文献   

16.
Phorbol esters, potent activators of protein kinase C (PKC), greatly enhance the release of arachidonic acid and its metabolites (TXA2, HETES, HHT) by Ca2+ ionophores in human platelets. In this paper, we report the relationship between intracellular Ca2+ mobilization and external calcium influx into platelets and the ability of PMA plus A23187 to promote thromboxane A2 (TXA2) synthesis. The enhanced levels of TXA2 due to the synergistic stimulation of the platelets with A23187 and phorbol esters are not affected significantly by the presence of external Ca2+ or the calcium-chelator EGTA. PKC inhibitors, staurosporine and sphingosine, abolished phorbol myristate acetate (PMA) potentiation of TXA2 production which strongly supports the role of PKC in the synergism. Platelet aggregation is more sensitive to PMA and external calcium than TXA2 formation. PMA increased TXA2 production as much as 4-fold at low ionophore concentrations. The A23187-induced rise in [Ca2+]i was reduced by pretreatment of human platelets with phorbol esters, both in the presence and absence of EGTA, and staurosporine reversed this inhibitory effect. These results indicate that the synergistic stimulation of TXA2 production by A23187 and phorbol esters is promoted by intracellular Ca2+ mobilization and not by external calcium influx. Our data also suggest that PKC is involved in the regulation of Ca2+ mobilization from some specific intracellular stores and that PKC may also stimulate the Ca(2+)-dependent phospholipase A2 at suboptimal Ca2+i concentrations.  相似文献   

17.
We examined the effect of EGF on the proliferation of mouse embryonic stem (ES) cells and their related signal pathways. EGF increased [3H]thymidine and 5-bromo-2'-deoxyuridine incorporation in a time- and dose-dependent manner. EGF stimulated the phosphorylation of EGF receptor (EGFR). Inhibition of EGFR tyrosine kinase with AG-1478 or herbimycin A, inhibition of PLC with neomycin or U-73122, inhibition of PKC with bisindolylmaleimide I or staurosporine, and inhibition of L-type Ca2+ channels with nifedipine or methoxyverapamil prevented EGF-induced [3H]thymidine incorporation. PKC-, -I, -, -, and - were translocated to the membrane and intracellular Ca2+ concentration ([Ca2+]i) was increased in response to EGF. Moreover, inhibition of EGFR tyrosine kinase, PLC, and PKC completely prevented EGF-induced increases in [Ca2+]i. EGF also increased inositol phosphate levels, which were blocked by EGFR tyrosine kinase inhibitors. Furthermore, EGF rapidly increased formation of H2O2, and pretreatment with antioxidant (N-acetyl-L-cysteine) inhibited EGF-induced increase of [Ca2+]i. In addition, we observed that p44/42 MAPK phosphorylation by EGF and inhibition of EGFR tyrosine kinase, PLC, PKC, or Ca2+ channels blocked EGF-induced phosphorylation of p44/42 MAPKs. Inhibition of p44/42 MAPKs with PD-98059 (MEK inhibitor) attenuated EGF-induced increase of [3H]thymidine incorporation. Finally, inhibition of EGFR tyrosine kinase, PKC, Ca2+ channels, or p44/42 MAPKs attenuated EGF-stimulated cyclin D1, cyclin E, cyclin-dependent kinase (CDK)2, and CDK4, respectively. In conclusion, EGF partially stimulates proliferation of mouse ES cells via PLC/PKC, Ca2+ influx, and p44/42 MAPK signal pathways through EGFR tyrosine kinase phosphorylation. calcium; epidermal growth factor; mitogen-activated protein kinases; protein kinase C  相似文献   

18.
The presentstudy was performed to characterize thrombin-stimulated phospholipaseA2(PLA2) activity and theresultant release of lysophospholipids from endothelial cells. Themajority of PLA2 activity inendothelial cells was membrane associated,Ca2+ independent, and arachidonateselective. Incubation with thrombin increased membrane-associatedPLA2 activity using bothplasmenylcholine and alkylacyl glycerophosphocholine substrates in theabsence of Ca2+, with no increasein activity observed with phosphatidylcholine substrate. The increasedPLA2 activity was accompanied byarachidonic acid and lysoplasmenylcholine (LPlasC) release fromendothelial cells into the surrounding medium. Thrombin-induced changeswere duplicated by stimulation with the thrombin-receptor-directed peptide SFLLRNPNDKYEPF. Pretreatment with theCa2+-independentPLA2 inhibitor bromoenol lactoneblocked thrombin-stimulated increases inPLA2 activity, arachidonic acid,and LPlasC release. Stimulation of protein kinase C (PKC) increasedbasal PLA2 activity and LPlasCproduction. Thrombin-stimulatedPLA2 activity and LPlasC production were enhanced with PKC activation and completely prevented with PKC downregulation. Thus thrombin treatment of endothelial cellsactivates a PKC-activated, membrane-associated,Ca2+-independentPLA2 that selectively hydrolyzesarachidonylated, ether-linked phospholipid substrates, resulting inLPlasC and arachidonic acid release.

  相似文献   

19.
Nigericin decreases intracellular pH(pHi) and stimulates prostanoid(PG) synthesis in endothelial cells from cerebral microvessels ofnewborn pigs. Nigericin-induced PG production was abolished by proteintyrosine kinase (PTK) inhibitors and amplified by phorbol 12-myristate13-acetate (PMA) or protein tyrosine phosphatase (PTP) inhibitors.Nigericin-induced PG production in PMA-primed cells was potentiated byPTP inhibitors and abrogated by PTK inhibitors. PhospholipaseA2(PLA2) activity was stimulatedby nigericin in a phosphorylation-dependent manner. Nigericin'seffects on PG production and PLA2activity were reproduced by ionomycin, which activates cytosolicPLA2(cPLA2).cPLA2 was immunodetected in endothelial cell lysates. We found no evidence that nigericin's effects are mediated via mitogen-activated protein (MAP) kinase [extracellularly regulated kinase 1 (ERK1) and ERK2]activation: although nigericin stimulateddetergent-soluble MAP kinase, its effects were not amplified by PMA orPTP inhibitors. Phosphorylation-dependent stimulation of PG synthesiswas also observed when pHi wasdecreased by sodium propionate or a high level ofCO2. Altogether, our data indicatethat nigericin and decreased pHistimulate PG synthesis by a protein phosphorylation-dependent mechanisminvolving cross talk between pathways mediated by PTK and PTP and byprotein kinase C; cPLA2 appears tobe a key enzyme affected by nigericin and decreasedpHi.

  相似文献   

20.
Early activation of p160ROCK by pressure overload in rat heart   总被引:1,自引:0,他引:1  
We investigated the mechanisms underlying regulation of contraction with measurements of isometric force and intracellular Ca2+ concentration ([Ca2+]i) in NIH 3T3 fibroblast reconstituted into fibers with the use of a collagen matrix. Treatment with the major phospholipids, neurotransmitters, and growth factors had little effect on baseline isometric force. However, U-46619, a thromboxane A2 (TxA2) analog, increased force and [Ca2+]i; EC50 values were 11.0 and 10.0 nM, respectively. The time courses were similar to those induced by calf serum (CS), and the maximal force was 65% of a CS-mediated contraction. The selective TxA2 receptor antagonist SQ-29548 abolished the U-46619-induced responses. CS-induced contractions are dependent on an intracellular Ca2+ store function; however, the U-46619 response depended not only on intracellular Ca2+ stores, but also on Ca2+ influx from the extracellular medium. Inhibition of Rho kinase suppressed U-46619- and CS-induced responses; in contrast, inhibition of C kinase (PKC) reduced only the U-46619 response. Moreover, addition of U-46619 to a CS contracture enhanced force and [Ca2+]i responses. These results indicate that U-46619-induced responses involve PKC and Rho kinase pathways, in contrast to activation by CS. Thus TxA2 may have a role in not only the initial step of wound repair as an activator of blood coagulation, but also in fibroblast contractility in later stages. collagen matrix; signal transduction; wound repair  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