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 359 毫秒
1.
In the present study, we examined the mechanisms through which erythropoietin (Epo) activates the calcium-permeable transient receptor potential protein channel (TRPC)2. Erythroblasts were isolated from the spleens of phenylhydrazine-treated mice, and Epo stimulation resulted in a significant and dose-dependent increase in intracellular calcium concentration ([Ca2+]i). This increase in [Ca2+]i was inhibited by pretreatment with the phospholipase C (PLC) inhibitor U-73122 but not by the inactive analog U-73343, demonstrating the requirement for PLC activity in Epo-modulated Ca2+ influx in primary erythroid cells. To determine whether PLC is involved in the activation of TRPC2 by Epo, cell models were used to examine this interaction. Single CHO-S cells that expressed transfected Epo receptor (Epo-R) and TRPC2 were identified, and [Ca2+]i was quantitated. Epo-induced Ca2+ influx through TRPC2 was inhibited by pretreatment with U-73122 or by downregulation of PLC1 by RNA interference. PLC activation results in the production of inositol 1,4,5-trisphosphate (IP3), and TRPC2 has IP3 receptor (IP3R) binding sites. To determine whether IP3R is involved in Epo-R signaling, TRPC2 mutants were prepared with partial or complete deletions of the COOH-terminal IP3R binding domains. In cells expressing TRPC2 IP3R binding mutants and Epo-R, no significant increase in [Ca2+]i was observed after Epo stimulation. TRPC2 coassociated with Epo-R, PLC, and IP3R, and the association between TRPC2 and IP3R was disrupted in these mutants. Our data demonstrate that Epo-R modulates TRPC2 activation through PLC; that interaction of IP3R with TRPC2 is required; and that Epo-R, TRPC2, PLC, and IP3R interact to form a signaling complex. transient receptor potential protein channels; erythropoietin receptor; calcium channels  相似文献   

2.
The effect ofinositol 1,4,5-trisphosphate(IP3) receptor blockade onplatelet-derived growth factor (PDGF), fibroblast growth factor (FGF),endothelin-1 (ET-1), or -thrombin receptor-mediated intracellularCa2+(Ca2+i) release was examined using fura 2 microspectrofluorometry in single Chinese hamster ovary cells andmyoblasts. Blockade of the IP3receptor was achieved by microinjection of heparin or monoclonalantibody (MAb) 18A10 into the IP3type 1 receptor. Heparin completely inhibitedCa2+i release after flash photolysis withcaged IP3 and after exposure toPDGF and FGF. In contrast, heparin failed to blockCa2+i release after -thrombin andET-1. After application of ligand, IP3 levels were five- to sevenfoldhigher for -thrombin than for ET-1 or PDGF.IP3 levels after PDGF and ET-1were comparable. Similar to heparin, MAb 18A10 blockedCa2+i release after PDGF but failed toblock Ca2+i release after ET-1 or-thrombin. These data suggest that the mechanisms of Ca2+i release by tyrosine kinase andcertain 7-transmembrane receptors may differ. Although both receptortypes use the IP3-signaling system, the ET-1 and -thrombin receptors may have a second,alternative mechanism for activatingCa2+i release.

  相似文献   

3.
In smooth muscle of the gut, Gq-coupled receptor agonists activate preferentially PLC-1 to stimulate phosphoinositide (PI) hydrolysis and inositol 1,4,5-trisphosphate (IP3) generation and induce IP3-dependent Ca2+ release. Inhibition of Ca2+ mobilization by cAMP- (PKA) and cGMP-dependent (PKG) protein kinases reflects inhibition of PI hydrolysis by both kinases and PKG-specific inhibitory phosphorylation of IP3 receptor type I. The mechanism of inhibition of PLC-1-dependent PI hydrolysis has not been established. Neither Gq nor PLC-1 was directly phosphorylated by PKA or PKG in gastric smooth muscle cells. However, both kinases 1) phosphorylated regulator of G protein signaling 4 (RGS4) and induced its translocation from cytosol to plasma membrane, 2) enhanced ACh-stimulated association of RGS4 and Gq·GTP and intrinsic Gq·GTPase activity, and 3) inhibited ACh-stimulated PI hydrolysis. RGS4 phosphorylation and inhibition of PI hydrolysis were blocked by selective PKA and PKG inhibitors. Expression of RGS4(S52A), which lacks a PKA/PKG phosphorylation site, blocked the increase in GTPase activity and the decrease in PI hydrolysis induced by PKA and PKG. Blockade of PKA-dependent effects was only partial. Selective phosphorylation of G protein-coupled receptor kinase 2 (GRK2), which contains a RGS domain, by PKA augmented ACh-stimulated GRK2:Gq·GTP association; both effects were blocked in cells expressing GRK2(S685A), which lacks a PKA phosphorylation site. Inhibition of PI hydrolysis induced by PKA was partly blocked in cells expressing GRK2(S685A) and completely blocked in cells coexpressing GRK2(S685A) and RGS4(S52A) or Gq(G188S), a Gq mutant that binds GRK2 but not RGS4. The results demonstrate that inhibition of PLC-1-dependent PI hydrolysis by PKA is mediated via stimulatory phosphorylation of RGS4 and GRK2, leading to rapid inactivation of Gq·GTP. PKG acts only via phosphorylation of RGS4. regulators of G protein signaling; G protein-coupled receptor kinase 2; phospholipase C; cAMP-dependent protein kinase; cGMP-dependent protein kinase  相似文献   

4.
Activation of PLC-delta1 by Gi/o-coupled receptor agonists   总被引:1,自引:0,他引:1  
The mechanism of phospholipase (PLC)- activation by G protein-coupled receptor agonists was examined in rabbit gastric smooth muscle. Ca2+ stimulated an eightfold increase in PLC-1 activity in permeabilized muscle cells. Treatment of dispersed or cultured muscle cells with three Gi/o-coupled receptor agonists (somatostatin, -opioid agonist [D-Pen2,D-Pen5]enkephalin, and A1 agonist cyclopentyl adenosine) caused delayed increase in phosphoinositide (PI) hydrolysis (8- to 10-fold) that was strongly inhibited by overexpression of dominant-negative PLC-1(E341R/D343R; 65–76%) or constitutively active RhoA(G14V). The response coincided with capacitative Ca2+ influx and was not observed in the absence of extracellular Ca2+, but was partly inhibited by nifedipine (16–30%) and strongly inhibited by SKF-96365, a blocker of store-operated Ca2+ channels. Treatment of the cells with a Gq/13-coupled receptor agonist, CCK-8, caused only transient, PLC-1-mediated PI hydrolysis. Unlike Gi/o-coupled receptor agonists, CCK-8 activated RhoA and stimulated RhoA:PLC-1 association. Inhibition of RhoA activity with C3 exoenzyme or by overexpression of dominant-negative RhoA(T19N) or G13 minigene unmasked a delayed increase in PI hydrolysis that was strongly inhibited by coexpression of PLC-1(E341R/D343R) or by SKF-96365. Agonist-independent capacitative Ca2+ influx induced by thapsigargin stimulated PI hydrolysis (8-fold), which was partly inhibited by nifedipine (25%) and strongly inhibited by SKF-96365 (75%) and in cells expressing PLC-1(E341R/D343R). Agonist-independent Ca2+ release or Ca2+ influx via voltage-gated Ca2+ channels stimulated only moderate PI hydrolysis (2- to 3-fold), which was abolished by PLC-1 antibody or nifedipine. We conclude that PLC-1 is activated by Gi/o-coupled receptor agonists that do not activate RhoA. The activation is preferentially mediated by Ca2+ influx via store-operated Ca2+ channels. phospholipase C; G protein  相似文献   

5.
Ca+-induced Ca2+ release (CICR) in the heart involves local Ca2+ signaling between sarcolemmal L-type Ca2+ channels (dihydropyridine receptors, DHPRs) and type 2 ryanodine receptors (RyR2s) in the sarcoplasmic reticulum (SR). We reconstituted cardiac-like CICR by expressing a cardiac dihydropyridine-insensitive (T1066Y/Q1070M) 1-subunit (1CYM) and RyR2 in myotubes derived from RyR1-knockout (dyspedic) mice. Myotubes expressing 1CYM and RyR2 were vesiculated and exhibited spontaneous Ca2+ oscillations that resulted in chaotic and uncontrolled contractions. Coexpression of FKBP12.6 (but not FKBP12.0) with 1CYM and RyR2 eliminated vesiculations and reduced the percentage of myotubes exhibiting uncontrolled global Ca2+ oscillations (63% and 13% of cells exhibited oscillations in the absence and presence of FKBP12.6, respectively). 1CYM/RyR2/FKBP12.6-expressing myotubes exhibited robust and rapid electrically evoked Ca2+ transients that required extracellular Ca2+. Depolarization-induced Ca2+ release in 1CYM/RyR2/FKBP12.6-expressing myotubes exhibited a bell-shaped voltage dependence that was fourfold larger than that of myotubes expressing 1CYM alone (maximal fluorescence change was 2.10 ± 0.39 and 0.54 ± 0.07, respectively), despite similar Ca2+ current densities. In addition, the gain of CICR in 1CYM/RyR2/FKBP12.6-expressing myotubes exhibited a nonlinear voltage dependence, being considerably larger at threshold potentials. We used this molecular model of local 1C-RyR2 signaling to assess the ability of FKBP12.6 to inhibit spontaneous Ca2+ release via a phosphomimetic mutation in RyR2 (S2808D). Electrically evoked Ca2+ release and the incidence of spontaneous Ca2+ oscillations did not differ in wild-type RyR2- and S2808D-expressing myotubes over a wide range of FKBP12.6 expression. Thus a negative charge at S2808 does not alter in situ regulation of RyR2 by FKBP12.6. heart failure; dihydropyridine receptor; excitation-contraction coupling  相似文献   

6.
The role of glycolytically generated ATP in Ca2+/calmodulin-dependent kinase II (CaMKII)-mediated regulation of intracellular Ca2+ signaling was examined in cultured calf pulmonary artery endothelial (CPAE) cells. Exposure of cells (extracellular Ca2+ concentration = 2 mM) to glycolytic inhibitors 2-deoxy-D-glucose (2-DG), pyruvate (pyr) + -hydroxybutyrate (-HB), or iodoacetic acid (IAA) caused an increase of intracellular Ca2+ concentration ([Ca2+]i). CaMKII inhibitors (KN-93, W-7) triggered a similar increase of [Ca2+]i. The rise of [Ca2+]i was characterized by a transient spike followed by a small sustained plateau of elevated [Ca2+]i. In the absence of extracellular Ca2+ 2-DG caused an increase in [Ca2+]i, suggesting that inhibition of glycolysis directly triggered release of Ca2+ from intracellular endoplasmic reticulum (ER) Ca2+ stores. The inositol-1,4,5-trisphosphate receptor (IP3R) inhibitor 2-aminoethoxydiphenyl borate abolished the KN-93- and 2-DG-induced Ca2+ response. Ca2+ release was initiated in peripheral cytoplasmic processes from which activation propagated as a [Ca2+]i wave toward the central region of the cell. Focal application of 2-DG resulted in spatially confined elevations of [Ca2+]i. Propagating [Ca2+]i waves were preceded by [Ca2+]i oscillations and small, highly localized elevations of [Ca2+]i (Ca2+ puffs). Inhibition of glycolysis with 2-DG reduced the KN-93-induced Ca2+ response, and vice versa during inhibition of CaMKII 2-DG-induced Ca2+ release was attenuated. Similar results were obtained with pyr + -HB and W-7. Furthermore, 2-DG and IAA caused a rapid increase of intracellular Mg2+ concentration, indicating a concomitant drop of cellular ATP levels. In conclusion, CaMKII exerts a profound inhibition of ER Ca2+ release in CPAE cells, which is mediated by glycolytically generated ATP, possibly through ATP-dependent phosphorylation of the IP3R. Ca2+/calmodulin-dependent kinase II; glycolysis; calcium regulation  相似文献   

7.
Calcineurin mediates repression of plasma membrane Ca2+-ATPase-4 (PMCA4) expression in neurons, whereas c-Myb is known to repress PMCA1 expression in vascular smooth muscle cells (VSMC). Here, we describe a novel mouse VSMC line (MOVAS) in which 45Ca efflux rates decreased 50%, fura 2-AM-based intracellular Ca2+ concentrations ([Ca2+]i) increased twofold, and real-time RT-PCR and Western blot revealed a 40% decrease in PMCA4 expression levels from G0 to G1/S in the cell cycle, where PMCA4 constituted 20% of total PMCA protein. Although calcineurin activity increased fivefold as MOVAS progressed from G0 to G1/S, inhibition of this increase with either BAPTA or retroviral transduction with peptide inhibitors of calcineurin (CAIN), or its downstream target nuclear factor of activated T cells (NFAT) (VIVIT), had no effect on the repression of PMCA4 mRNA expression at G1/S. By contrast, Ca2+-independent activity of the calmodulin-dependent protein kinase-II (CaMK-II) increased eightfold as MOVAS progressed from G0 to G1/S, and treatment with an inhibitor of CaMK-II (KN-93) or transduction of a c-Myb-neutralizing antibody significantly alleviated the G1/S-associated repression of PMCA4. These data show that G1/S-specific PMCA4 repression in proliferating VSMC is brought about by c-Myb and CaMK-II and that calcineurin may regulate cell cycle-associated [Ca2+]i through alternate targets. calcineurin; c-Myb; plasma membrane Ca2+-ATPase-4; cell cycle  相似文献   

8.
Stimulatory concentrations of glucose induce two patterns of cytosolic Ca2+ concentration ([Ca2+]c) oscillations in mouse islets: simple or mixed. In the mixed pattern, rapid oscillations are superimposed on slow ones. In the present study, we examined the role of the membrane potential in the mixed pattern and the impact of this pattern on insulin release. Simultaneous measurement of [Ca2+]c and insulin release from single islets revealed that mixed [Ca2+]c oscillations triggered synchronous oscillations of insulin secretion. Simultaneous recordings of membrane potential in a single -cell within an islet and of [Ca2+]c in the whole islet demonstrated that the mixed pattern resulted from compound bursting (i.e., clusters of membrane potential oscillations separated by prolonged silent intervals) that was synchronized in most -cells of the islet. Each slow [Ca2+]c increase during mixed oscillations was due to a progressive summation of rapid oscillations. Digital image analysis confirmed the good synchrony between subregions of an islet. By contrast, islets from sarco(endo)plasmic reticulum Ca2+-ATPase isoform 3 (SERCA3)-knockout mice did not display typical mixed [Ca2+]c oscillations in response to glucose. This results from a lack of progressive summation of rapid oscillations and from altered spontaneous electrical activity, i.e., lack of compound bursting, and membrane potential oscillations characterized by lower-frequency but larger-depolarization phases than observed in SERCA3+/+ -cells. We conclude that glucose-induced mixed [Ca2+]c oscillations result from compound bursting in all -cells of the islet. Disruption of SERCA3 abolishes mixed [Ca2+]c oscillations and augments -cell depolarization. This latter observation indicates that the endoplasmic reticulum participates in the control of the -cell membrane potential during glucose stimulation. electrical activity; insulin-secreting cell; thapsigargin  相似文献   

9.
We demonstrated recently that norepinephrine activates Ca2+-permeable nonselective cation channels (NSCCs) in Chinese hamster ovary cells stably expressing 1A-adrenergic receptors (CHO-1A). Moreover, extracellular Ca2+ through NSCCs plays essential roles in norepinephrine-induced arachidonic acid release. The purpose of the present study was to identify the G proteins involved in the activation of NSCCs and arachidonic acid release by norepinephrine. For these purposes, we used U73122, an inhibitor of phospholipase C (PLC), and dominant negative mutants of G12 and G13 (G12G228A and G13G225A, respectively). U73122 failed to inhibit NSCCs activation by norepinephrine. The magnitudes of norepinephrine-induced extracellular Ca2+ influx in CHO-1A microinjected with G13G225A were smaller than those in CHO-1A. In contrast, the magnitudes of norepinephrine-induced extracellular Ca2+ influx in CHO-1A microinjected with G12G228A were similar to those in CHO-1A. In addition, neither a Rho-associated kinase (ROCK) inhibitor nor a phosphoinositide 3-kinase inhibitor affected norepinephrine-induced extracellular Ca2+ influx. G13G225A, but not G12G228A, also inhibited arachidonic acid release partially. These results demonstrate that 1) the Gq/PLC-pathway is not involved in NSCCs activation by norepinephrine, 2) G13 couples with CHO-1A and plays important roles for norepinephrine-induced NSCCs activation, 3) neither ROCK- nor PI3K-dependent cascade is involved in NSCCs activation, and 4) G13 is involved in norepinephrine-induced arachidonic acid release in CHO-1A. norepinephrine; 1A-adrenergic receptor; nonselective cation channel; G13 protein; arachidonic acid release  相似文献   

10.
Differentiated primary myotubes isolated from wild-type mice exhibit ryanodine-sensitive, spontaneous global Ca2+ oscillations as well as spontaneous depolarizations in the plasma membrane. Immunolabeling of these myotubes showed expression of both 1S dihydropyridine receptors (DHPRs) and ryanodine-sensitive Ca2+-release channel 1 (RyR1), the two key proteins in skeletal excitation-contraction (E-C) coupling. Spontaneous global Ca2+ oscillations could be inhibited by addition of 0.1 mM CdCl2/0.5 mM LaCl3 or 5 µM nifedipine to the extracellular bathing solution. After either treatment, Ca2+ oscillations could be restored upon extensive washing. Although exposure to DHPR antagonists completely blocked Ca2+ oscillations, normal orthograde signaling between DHPRs and RyRs, such as that elicited by 80 mM KCl depolarization, was still observed. In addition, we showed that spontaneous Ca2+ oscillations were never present in cultured mdg myotubes, which lack the expression of 1SDHPRs. These results suggest that under physiological conditions in conjunction with the mechanical coupling between the 1SDHPRs and RyR1, the initiation of Ca2+ oscillations in myotubes may be facilitated, in part, by the Ca2+ influx through the 1s-subunit of the DHPR. calcium-induced calcium release; dihydropyridine receptors; excitation-contraction coupling; ryanodine receptors; skeletal muscle  相似文献   

11.
Sympathetic efferent nerves are present in tendons, but their function within tendon is unknown. 1-Adrenoceptors are expressed by a variety of cell types. In the presence of norepinephrine (NE), adrenoceptors activate Gq/11 signaling pathways that subsequently increase intracellular Ca2+ concentration ([Ca2+]ic). It was hypothesized that avian tendon cells express functional adrenoceptors that respond to NE by increasing [Ca2+]ic. Avian tendon cells were analyzed for mRNA expression of 1-adrenoceptors by RT-PCR. Avian tendons expressed the 1A- and 1B-adrenoceptor subtypes. Furthermore, both tendon surface epitenon cells and internal fibroblasts infused with a Ca2+-sensitive dye, fura 2, and stimulated with NE responded by increasing [Ca2+]ic. KMD-3213, an 1A-adrenoceptor antagonist, significantly reduced the Ca2+ response. Other adrenoceptor antagonists had no effect on the Ca2+ response. The absence of extracellular Ca2+ also significantly reduced the response to NE, indicating that Ca2+ influx contributed to the rise in [Ca2+]ic. This study provides the first evidence that tendon cells express adrenoceptors and that the NE-induced Ca2+ response is coupled to the 1A-adrenoceptor subtype. -adrenoceptors; fibroblasts; catecholamines; tenocytes  相似文献   

12.
The effect of sphingosine-1-phosphate (S1P) on large-conductance Ca2+-activated K+ (BKCa) channels was examined in primary cultured human umbilical vein endothelial cells by measuring intracellular Ca2+ concentration ([Ca2+]i), whole cell membrane currents, and single-channel activity. In nystatin-perforated current-clamped cells, S1P hyperpolarized the membrane and simultaneously increased [Ca2+]i. [Ca2+]i and membrane potentials were strongly correlated. In whole cell clamped cells, BKCa currents were activated by increasing [Ca2+]i via cell dialysis with pipette solution, and the activated BKCa currents were further enhanced by S1P. When [Ca2+]i was buffered at 1 µM, the S1P concentration required to evoke half-maximal activation was 403 ± 13 nM. In inside-out patches, when S1P was included in the bath solution, S1P enhanced BKCa channel activity in a reversible manner and shifted the relationship between Ca2+ concentration in the bath solution and the mean open probability to the left. In whole cell clamped cells or inside-out patches loaded with guanosine 5'-O-(2-thiodiphosphate) (GDPS; 1 mM) using a patch pipette, GDPS application or pretreatment of cells with pertussis toxin (100 ng/ml) for 15 h did not affect S1P-induced BKCa current and channel activation. These results suggest that S1P enhances BKCa channel activity by increasing Ca2+ sensitivity. This channel activation hyperpolarizes the membrane and thereby increases Ca2+ influx through Ca2+ entry channels. Inasmuch as S1P activates BKCa channels via a mechanism independent of G protein-coupled receptors, S1P may be a component of the intracellular second messenger that is involved in Ca2+ mobilization in human endothelial cells. sphingolipid metabolites; intracellular second messenger; Ca2+ mobilization  相似文献   

13.
The P2Y1 ADP receptor activates Gq and causes increases in intracellular Ca2+ concentration through stimulation of PLC. In this study, we investigated the role of the amino acid residues in the COOH terminus of the human P2Y1 receptor in Gq activation. Stimulation of Chinese hamster ovary (CHO-K1) cells stably expressing the wild-type human P2Y1 receptor (P2Y1-WT cells), P2Y1-R340-L373, or P2Y1-D356-L373 with 2-methylthio-ADP (2-MeSADP) caused inositol phosphate production. In contrast, cells expressing P2Y1-T330-L373, a mutant lacking the entire COOH terminus, completely lost their response to 2-MeSADP. Similar data were obtained by using these cell lines and measuring Ca2+ mobilization upon stimulation with 2-MeSADP, indicating that the 10 amino acids (330TFRRRLSRAT339) in the COOH terminus of the human P2Y1 receptor are essential for Gq coupling. Radioligand binding demonstrated that both the P2Y1-WT and P2Y1-T330-L373-expressing cells have almost equal binding of [3H]MRS2279, a P2Y1 receptor antagonist, indicating that COOH-terminal truncation did not drastically affect the conformation of the receptor. CHO-K1 cells expressing a chimeric P2Y12 receptor with the P2Y1 COOH terminus failed to elicit Gq functional responses, indicating that the P2Y1 COOH terminus is essential but not sufficient for Gq activation. Finally, cells expressing a double-mutant P2Y1 receptor (R333A/R334A) in the conserved BBXXB region of the COOH terminus of the Gq-activating P2Y receptors completely lost their functional ability to activate Gq. We conclude that the two arginine residues (R333R334) in the COOH terminus of the human P2Y1 receptor are essential for Gq coupling. carboxyl terminus; adenosine diphosphate; truncation; inositol phosphate  相似文献   

14.
Previous studies have shown that inhibition of L-type Ca2+ current (ICa) by cytosolic free Mg2+ concentration ([Mg2+]i) is profoundly affected by activation of cAMP-dependent protein kinase pathways. To investigate the mechanism underlying this counterregulation of ICa, rat cardiac myocytes and tsA201 cells expressing L-type Ca2+ channels were whole cell voltage-clamped with patch pipettes in which [Mg2+] ([Mg2+]p) was buffered by citrate and ATP. In tsA201 cells expressing wild-type Ca2+ channels (1C/2A/2), increasing [Mg2+]p from 0.2 mM to 1.8 mM decreased peak ICa by 76 ± 4.5% (n = 7). Mg2+-dependent modulation of ICa was also observed in cells loaded with ATP--S. With 0.2 mM [Mg2+]p, manipulating phosphorylation conditions by pipette application of protein kinase A (PKA) or phosphatase 2A (PP2A) produced large changes in ICa amplitude; however, with 1.8 mM [Mg2+]p, these same manipulations had no significant effect on ICa. With mutant channels lacking principal PKA phosphorylation sites (1C/S1928A/2A/S478A/S479A/2), increasing [Mg2+]p had only small effects on ICa. However, when channel open probability was increased by 1C-subunit truncation (1C1905/2A/S478A/S479A/2), increasing [Mg2+]p greatly reduced peak ICa. Correspondingly, in myocytes voltage-clamped with pipette PP2A to minimize channel phosphorylation, increasing [Mg2+]p produced a much larger reduction in ICa when channel opening was promoted with BAY K8644. These data suggest that, around its physiological concentration range, cytosolic Mg2+ modulates the extent to which channel phosphorylation regulates ICa. This modulation does not necessarily involve changes in channel phosphorylation per se, but more generally appears to depend on the kinetics of gating induced by channel phosphorylation. voltage-gated Ca2+ channel; cardiac myocytes; human embryonic kidney cells; protein kinase A; protein phosphatase 2A  相似文献   

15.
The Ca2+-sensing receptor (CaR) couples to multiple G proteins involved in distinct signaling pathways: Gi to inhibit the activity of adenylyl cyclase and activate ERK, Gq to stimulate phospholipase C and phospholipase A2, and G to stimulate phosphatidylinositol 3-kinase. To determine whether the receptor also couples to G12/13, we investigated the signaling pathway by which the CaR regulates phospholipase D (PLD), a known G12/13 target. We established Madin-Darby canine kidney (MDCK) cell lines that stably overexpress the wild-type CaR (CaRWT) or the nonfunctional mutant CaRR796W as a negative control, prelabeled these cells with [3H]palmitic acid, and measured CaR-stimulated PLD activity as the formation of [3H]phosphatidylethanol (PEt). The formation of [3H]PEt increased in a time-dependent manner in the cells that overexpress the CaRWT but not the CaRR796W. Treatment of the cells with C3 exoenzyme inhibited PLD activity, which indicates that the CaR activates the Rho family of small G proteins, targets of G12/13. To determine which G protein(s) the CaR couples to in order to activate Rho and PLD, we pretreated the cells with pertussis toxin to inactivate Gi or coexpressed regulators of G protein-signaling (RGS) proteins to attenuate G protein signaling (RGS4 for Gi and Gq, and a p115RhoGEF construct containing the RGS domain for G12/13). Overexpression of p115RhoGEF-RGS in the MDCK cells that overexpress CaRWT inhibited extracellular Ca2+-stimulated PLD activity, but pretreatment of cells with pertussis toxin and overexpression of RGS4 were without effect. The involvement of other signaling components such as protein kinase C, ADP-ribosylation factor, and phosphatidylinositol biphosphate was excluded. These findings demonstrate that the CaR couples to G12/13 to regulate PLD via a Rho-dependent mechanism and does so independently of Gi and Gq. This suggests that the CaR may regulate cytoskeleton via G12/13, Rho, and PLD. calcium-sensing receptor; G proteins; RGS proteins  相似文献   

16.
Spontaneous Ca2+ sparks were observed in fluo 4-loaded myocytes from guinea pig vas deferens with line-scan confocal imaging. They were abolished by ryanodine (100 µM), but the inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) blockers 2-aminoethoxydiphenyl borate (2-APB; 100 µM) and intracellular heparin (5 mg/ml) increased spark frequency, rise time, duration, and spread. Very prolonged Ca2+ release events were also observed in 20% of cells treated with IP3R blockers but not under control conditions. 2-APB and heparin abolished norepinephrine (10 µM; 0 Ca2+)-evoked Ca2+ transients but increased caffeine (10 mM; 0 Ca2+) transients in fura 2-loaded myocytes. Transients evoked by ionomycin (25 µM; 0 Ca2+) were also enhanced by 2-APB. Ca2+ sparks and transients evoked by norepinephrine and caffeine were abolished by thimerosal (100 µM), which sensitizes the IP3R to IP3. In cells voltage clamped at –40 mV, spontaneous transient outward currents (STOCs) were increased in frequency, amplitude, and duration in the presence of 2-APB. These data are consistent with a model in which the Ca2+ store content in smooth muscle is limited by tonic release of Ca2+ via an IP3-dependent pathway. Blockade of IP3Rs elevates sarcoplasmic reticulum store content, promoting Ca2+ sparks and STOC activity. calcium ion release; calcium ion transients; smooth muscle  相似文献   

17.
The role of the thyroid hormone agonist 3,3',5 L-tri-iodothyronine (T3) on cholangiocytes is unknown. We evaluated the in vivo and in vitro effects of T3 on cholangiocyte proliferation of bile duct-ligated (BDL) rats. We assessed the expression of 1-, 2-, 1-, and 2-thyroid hormone receptors (THRs) by immunohistochemistry in liver sections from normal and BDL rats. BDL rats were treated with T3 (38.4 µg/day) or vehicle for 1 wk. We evaluated 1) biliary mass and apoptosis in liver sections and 2) proliferation in cholangiocytes. Serum-free T3 levels were measured by chemiluminescence. Purified BDL cholangiocytes were treated with 0.2% BSA or T3 (1 µM) in the absence/presence of U-73122 (PLC inhibitor) or BAPTA/AM (intracellular Ca2+ chelator) before measurement of PCNA protein expression by immunoblots. The in vitro effects of T3 (1 µM) on 1) cAMP, IP3, and Ca2+ levels and 2) the phosphorylation of Src Tyr139 and Tyr530 (that, together, regulate Src activity) and ERK1/2 of BDL cholangiocytes were also evaluated. 1-, 2-, 1-, and 2-THRs were expressed by bile ducts of normal and BDL rats. In vivo, T3 decreased cholangiocyte proliferation of BDL rats. In vitro, T3 inhibition of PCNA protein expression was blocked by U-73122 and BAPTA/AM. Furthermore, T3 1) increased IP3 and Ca2+ levels and 2) decreased Src and ERK1/2 phosphorylation of BDL cholangiocytes. T3 inhibits cholangiocyte proliferation of BDL rats by PLC/IP3/Ca2+-dependent decreased phosphorylation of Src/ERK1/2. Activation of the intracellular signals triggered by T3 may modulate the excess of cholangiocyte proliferation in liver diseases. cholestasis; cholangiopathies; hyperplasia; intrahepatic biliary epithelium; mitosis  相似文献   

18.
We examined expression of sphingosine 1-phosphate (S1P) receptors and sphingosine kinase (SPK) in gastric smooth muscle cells and characterized signaling pathways mediating S1P-induced 20-kDa myosin light chain (MLC20) phosphorylation and contraction. RT-PCR demonstrated expression of SPK1 and SPK2 and S1P1 and S1P2 receptors. S1P activated Gq, G13, and all Gi isoforms and stimulated PLC-1, PLC-3, and Rho kinase activities. PLC- activity was partially inhibited by pertussis toxin (PTX), G or Gq antibody, PLC-1 or PLC-3 antibody, and by expression of Gq or Gi minigene, and was abolished by a combination of antibodies or minigenes. S1P-stimulated Rho kinase activity was partially inhibited by expression of G13 or Gq minigene and abolished by expression of both. S1P stimulated Ca2+ release that was inhibited by U-73122 and heparin and induced concentration-dependent contraction of smooth muscle cells (EC50 1 nM). Initial contraction and MLC20 phosphorylation were abolished by U-73122 and MLC kinase (MLCK) inhibitor ML-9. Initial contraction was also partially inhibited by PTX and Gq or G antibody and abolished by a combination of both antibodies. In contrast, sustained contraction and MLC20 phosphorylation were partially inhibited by a PKC or Rho kinase inhibitor (bisindolylmaleimide and Y-27632) and abolished by a combination of both inhibitors but not affected by U-73122 or ML-9. These results indicate that S1P induces 1) initial contraction mediated by S1P2 and S1P1 involving concurrent activation of PLC-1 and PLC-3 via Gq and Gi, respectively, resulting in inositol 1,4,5-trisphosphate-dependent Ca2+ release and MLCK-mediated MLC20 phosphorylation, and 2) sustained contraction exclusively mediated by S1P2 involving activation of RhoA via Gq and G13, resulting in Rho kinase- and PKC-dependent MLC20 phosphorylation. muscle contraction; signal transduction  相似文献   

19.
We recentlydemonstrated expression of a novel, glioma-specificCl current in glial-derivedtumor cells (gliomas), including stable cell lines such as STTG1,derived from a human anaplastic astrocytoma. We used STTG1 cells tostudy whether glioma Clchannel (GCC) activity is regulated during cell cycle progression. Cells were arrested in defined stages of cell cycle(G0,G1,G1/S, S, and M phases) using serumstarvation, mevastatin, hydroxyurea, demecolcine, and cytosine-D-arabinofuranoside. Cellcycle arrest was confirmed by measuring[3H]thymidineincorporation and by DNA flow cytometry. Using whole cell patch-clamprecordings, we demonstrate differential changes in GCC activity aftercell proliferation and cell cycle progression was selectively altered;specifically, channel expression was low in serum-starved,G0-arrested cells, increasedsignificantly in early G1,decreased during S phase, and increased after arrest in M phase.Although the link between the cell cycle and GCC activity is not yetclear, we speculate that GCCs are linked to the cytoskeleton and thatcytoskeletal rearrangements associated with cell division lead to theobserved changes in channel activity. Consistent with this hypothesis,we demonstrate the activation of GCC by disruption of F-actin usingcytochalasin D or osmotic cell swelling.

  相似文献   

20.
Inisolated rat pancreatic -cells, the nitric oxide (NO) donor NOC-7 at1 µM reduced the amplitude of the oscillations of cytosolicCa2+ concentration ([Ca2+]c)induced by 11.1 mM glucose, and at 10 µM terminated them. In thepresence of NG-nitro-L-arginine(L-NNA), however, NOC-7 at 0.5 and 1 µM increased theamplitude of the [Ca2+]c oscillations,although the NO donor at 10 µM still suppressed them. Aqueous NOsolution also had a dual effect on the[Ca2+]c oscillations. The soluble guanylatecyclase inhibitor LY-83583 and the cGMP-dependent protein kinaseinhibitor KT5823 inhibited the stimulatory effect of NO, and8-bromo-cGMP increased the amplitude of the[Ca2+]c oscillations. Patch-clamp analyses inthe perforated configuration showed that 8-bromo-cGMP inhibited wholecell ATP-sensitive K+ currents in the isolated ratpancreatic -cells, suggesting that the inhibition by cGMP ofATP-sensitive K+ channels is, at least in part, responsiblefor the stimulatory effect of NO on the[Ca2+]c oscillations. In the presence ofL-NNA, the glucose-induced insulin secretion from isolatedislets was facilitated by 0.5 µM NOC-7, whereas it was suppressed by10 µM NOC-7. These results suggest that NO facilitatesglucose-induced [Ca2+]c oscillations of-cells and insulin secretion at low concentrations, which effectsare mediated by cGMP, whereas NO inhibits them in a cGMP-independentmanner at high concentrations.

  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