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.
The phorbol ester phorbol12-myristate 13-acetate (PMA) inhibits Cl secretion(short-circuit current, Isc) and decreasesbarrier function (transepithelial resistance, TER) in T84 epithelia. To elucidate the role of specific protein kinase C (PKC) isoenzymes inthis response, we compared PMA with two non-phorbol activators of PKC(bryostatin-1 and carbachol) and utilized three PKC inhibitors (Gö-6850, Gö-6976, and rottlerin) with different isozymeselectivity profiles. PMA sequentially inhibited cAMP-stimulatedIsc and decreased TER, as measured byvoltage-current clamp. By subcellular fractionation and Western blot,PMA (100 nM) induced sequential membrane translocation of the novelPKC followed by the conventional PKC and activated both isozymesby in vitro kinase assay. PKC was activated by PMA but did nottranslocate. By immunofluorescence, PKC redistributed to thebasolateral domain in response to PMA, whereas PKC moved apically.Inhibition of Isc by PMA was prevented by theconventional and novel PKC inhibitor Gö-6850 (5 µM) but not theconventional isoform inhibitor Gö-6976 (5 µM) or the PKCinhibitor rottlerin (10 µM), implicating PKC in inhibition ofCl secretion. In contrast, both Gö-6976 andGö-6850 prevented the decline of TER, suggesting involvement ofPKC. Bryostatin-1 (100 nM) translocated PKC and PKC andinhibited cAMP-elicited Isc. However, unlikePMA, bryostatin-1 downregulated PKC protein, and the decrease in TERwas only transient. Carbachol (100 µM) translocated only PKC andinhibited Isc with no effect on TER. Gö-6850 but not Gö-6976 or rottlerin blocked bryostatin-1and carbachol inhibition of Isc. We concludethat basolateral translocation of PKC inhibits Clsecretion, while apical translocation of PKC decreases TER. Thesedata suggest that epithelial transport and barrier function can bemodulated by distinct PKC isoforms.

  相似文献   

2.
The protein kinaseC (PKC) family represents an important group of enzymes whoseactivation is associated with their translocation from the cytosol todifferent cellular membranes. In this study, the spatial distributionof PKC-, - and - in rat liver epithelial (WB) cells has beenexamined by Western blot analysis after subcellular fractionation.Cytosolic, membrane, nuclear, and cytoskeletal fractions were obtainedfrom cells stimulated with phorbol 12-myristate 13-acetate (PMA),angiotensin II (ANG II), or epidermal growth factor (EGF). PMA causedmost of the PKC-, - and - initially present in the cytosol tobe transported to the membrane and nuclear fractions. In contrast, bothANG II and EGF induced only a minor translocation of PKC- to themembrane fraction but caused a statistically significantmembrane-directed movement of PKC- and -. Translocation ofPKC- and - to the nucleus induced by ANG II and EGF was transient and quantitatively smaller than that induced by PMA. PKC- and -were present in the cytoskeleton of resting cells, but although PMA,ANG II, and EGF caused some changes in their content, these werevariable, suggesting that the cytoskeleton fraction was heterogeneous. PKC depletion inhibited ANG II-induced mitogenesis and the sustained activation of Raf-1 and extracellular regulated protein kinase (ERK).However, although PKC depletion inhibited EGF-induced mitogenesis, themaximum EGF-induced activation of the ERK pathway was only slightlyretarded. We hypothesize that PKC- and - are involved inmitogenesis via both ERK-dependent and ERK-independent mechanisms. These results support the notion that specific PKC isozymes exert spatially defined effects by virtue of their directed translocation todistinct intracellular sites.

  相似文献   

3.
Investigation of the role ofindividual protein kinase C (PKC) isozymes in the regulation ofNa+ channels has been largely limited by the lack ofisozyme-selective modulators. Here we used a novel peptide-specificactivator (V1-7) of PKC and other peptide isozyme-specificinhibitors in addition to the general PKC activator phorbol12-myristate 13-acetate (PMA) to dissect the role of individual PKCs inthe regulation of the human cardiac Na+ channel hH1,heterologously expressed in Xenopus oocytes. Peptides wereinjected individually or in combination into the oocyte. Whole cellNa+ current (INa) was recorded usingtwo-electrode voltage clamp. V1-7 (100 nM) and PMA (100 nM)inhibited INa by 31 ± 5% and 44 ± 8% (at 20 mV), respectively. These effects were not seen with thescrambled peptide for V1-7 (100 nM) or the PMA analog4-phorbol 12,13-didecanoate (100 nM). However, V1-7-and PMA-induced INa inhibition was abolished byV1-2, a peptide-specific antagonist of PKC. Furthermore,PMA-induced INa inhibition was not altered by100 nM peptide-specific inhibitors for -, -, -, or PKC. PMAand V1-7 induced translocation of PKC from soluble toparticulate fraction in Xenopus oocytes. This translocationwas antagonized by V1-2. In native rat ventricular myocytes,PMA and V1-7 also inhibited INa; thisinhibition was antagonized by V1-2. In conclusion, the resultsprovide evidence for selective regulation of cardiac Na+channels by PKC isozyme.

  相似文献   

4.
Protein kinase C (PKC) is known to regulate epithelial barrier function. However, the effect of specific PKC isozymes, and their mechanism of action, are largely unknown. We determined that the nonphorbol ester PKC agonist bryostatin-1 increased transepithelial electrical resistance (TER), a marker of barrier function, in confluent T84 epithelia. Bryostatin-1, which has been shown to selectively activate PKC-, -, and - (34), was associated with a shift in the subcellular distribution of the tight junction proteins claudin-1 and ZO-2 from a detergent-soluble fraction into a detergent-insoluble fraction. Bryostatin-1 also led to the appearance of a higher-molecular-weight form of occludin previously shown to correspond to protein phosphorylation. These changes were attenuated by the conventional and novel PKC inhibitor Gö-6850 but not the conventional PKC inhibitor Gö-6976 or the PKC- inhibitor röttlerin, implicating a novel isozyme, likely PKC-. The results suggest that enhanced epithelial barrier function induced by bryostatin-1 involves a PKC--dependent signaling pathway leading to recruitment of claudin-1 and ZO-2, and phosphorylation of occludin, into the tight junctional complex. protein kinase C; epithelial barrier function  相似文献   

5.
Protein kinase C (PKC) plays animportant role in activating store-operated Ca2+ channels(SOC) in human mesangial cells (MC). The present study was performed todetermine the specific isoform(s) of conventional PKC involved inactivating SOC in MC. Fura 2 fluorescence ratiometry showed that thethapsigargin-induced Ca2+ entry (equivalent to SOC) wassignificantly inhibited by 1 µM Gö-6976 (a specific PKC andI inhibitor) and PKC antisense treatment (2.5 nM for 24-48h). However, LY-379196 (PKC inhibitor) and2,2',3,3',4,4'-hexahydroxy-1,1'-biphenyl-6,6'-dimethanoldimethyl ether(HBDDE; PKC and  inhibitor) failed to affect thapsigargin-evoked activation of SOC. Single-channel analysis in the cell-attached configuration revealed that Gö-6976 and PKC antisensesignificantly depressed thapsigargin-induced activation of SOC.However, LY-379196 and HBDDE did not affect the SOC responses. Ininside-out patches, application of purified PKC or I, but notII or , significantly rescued SOC from postexcision rundown.Western blot analysis revealed that thapsigargin evoked a decrease incytosolic expression with a corresponding increase in membraneexpression of PKC and . However, the translocation from cytosolto membranes was not detected for PKCI or II. These resultssuggest that PKC participates in the intracellular signaling pathwayfor activating SOC upon release of intracellular stores ofCa2+.

  相似文献   

6.
Bufalin, anNa+-K+-ATPase inhibitor, simultaneouslyinduced cell differentiation and apoptosis in human monocyticleukemia THP-1 cells. In this study, we investigated the regulatoryrole of protein kinase C (PKC) isozymes in bufalin-induced celldifferentiation and apoptosis. A PKC-specific butisozyme-nonselective inhibitor, Ro-31-8220, and a cPKC selectiveinhibitor, Gö-6976, caused significant attenuation ofbufalin-induced interleukin-1 (IL-1) gene expression, a maturemonocytic marker, indicating that cPKC participates in thebufalin-induced cell differentiation. On the other hand, cPKC- andnPKC-defective THP-1/TPA cells displayed strong resistance tothe bufalin-induced DNA ladder formation. Rottlerin, annPKC-specific inhibitor, partially attenuated preapoptotic effectsof bufalin, such as the limited proteolysis of nPKC andpoly(ADP-ribose) polymerase and the cell staining by terminaldeoxynucleotidyl transferase-mediated dUTP nick end labeling,suggesting that nPKC is involved, at least in part, inbufalin-induced apoptosis. In contrast, Gö-6976 androttlerin significantly augmented bufalin-induced apoptosis anddifferentiation, respectively. The findings suggest thatbufalin-induced cell differentiation and apoptosis areinterlinked and that distinct PKC isozymes are involved in the fate ofthe cell.

  相似文献   

7.
Protein kinase C(PKC) regulates cystic fibrosis transmembrane conductance regulator(CFTR) channel activity but the PKC signaling mechanism is not yetknown. The goal of these studies was to identify PKC isotype(s)required for control of CFTR function. CFTR activity was measured as36Cl efflux in a Chinese hamsterovary cell line stably expressing wild-type CFTR (CHO-wtCFTR) and in aCalu-3 cell line. Chelerythrine, a PKC inhibitor, delayed increasedCFTR activity induced with phorbol 12-myristate 13-acetate or with thecAMP-generating agents ()-epinephrine or forskolin plus8-(4-chlorophenylthio)adenosine 3',5'- cyclicmonophosphate. Immunoblot analysis of Calu-3 cells revealed thatPKC-, -II, -, -, and- were expressed in confluent cell cultures. Pretreatment of cellmonolayers with Lipofectin plus antisense oligonucleotide to PKC-for 48 h prevented stimulation of CFTR with ()-epinephrine,reduced PKC- activity in unstimulated cells by 52.1%, and decreasedPKC- mass by 76.1% but did not affect hormone-activated proteinkinase A activity. Sense oligonucleotide to PKC- and antisenseoligonucleotide to PKC- and - did not alter()-epinephrine-stimulated CFTR activity. These results demonstrate the selective regulation of CFTR function by constitutively active PKC-.

  相似文献   

8.
Short-term regulation of sodiummetabolism is dependent on the modulation of the activity of sodiumtransporters by first and second messengers. In understanding diseasesassociated with sodium retention, it is necessary to identify thecoupling between these messengers. We have examined whether dopamine,an important first messenger in tubular cells, activates andtranslocates various protein kinase C (PKC) isoforms. We used aproximal tubular-like cell line, LLCPK-1 cells, in which dopamine wasfound to inhibit Na+-K+-ATPase in aPKC-dependent manner. Translocation of PKC isoforms was studied withboth subcellular fractionation and confocal microscopy. Both techniquesrevealed a dopamine-induced translocation from cytosol to plasmamembrane of PKC- and -, but not of PKC-, -, and -. Theprocess of subcellular fractionation resulted in partial translocationof PKC-. This artifact was eliminated in confocal studies. Confocalimaging permitted detection of translocation within 20 s.Translocation was abolished by a phospholipase C inhibitor and by anantagonist against the dopamine 1 subtype (D1) but not the2 subtype of receptor (D2). In conclusion, this studyvisualizes in renal epithelial cells a very rapid activation of thePKC- and - isoforms by the D1 receptor subtype.

  相似文献   

9.
Inhibitory actions of ceramide upon PKC-epsilon/ERK interactions   总被引:4,自引:0,他引:4  
We have previously shown that interleukin-1receptor-generated ceramide induces growth arrest in smooth musclepericytes by inhibiting an upstream kinase in the extracellularsignal-regulated kinase (ERK) cascade. Here, we now report themechanism by which ceramide inhibits ERK activity. Ceramide renders thehuman embryonic kidney 293 cells (HEK 293) resistant to the mitogenicactions of growth factors and activators of protein kinase C (PKC). A role for PKC to mediate ceramide inhibition of growth factor-induced ERK activity and mitogenesis is suggested, as exogenous ceramide directly inhibits both immunoprecipitated and recombinant PKC- activities. To confirm that PKC- is necessary for ceramide-inhibited ERK activity, HEK 293 cells were transfected with a dominant-negative mutant of PKC- (PKC-). These transfected cells respond toinsulin-like growth factor I (IGF-I) with a significantly decreased ERKactivity that is not further reduced by ceramide treatment.Coimmunoprecipitation studies reveal that the treatment with IGF-Iinduces the association of ERK with PKC- but not with PKC-.Ceramide treatment significantly inhibits the IGF-I-induced PKC-interaction with bioactive phosphorylated ERK. Ceramide also inhibitsIGF-I-induced PKC- association with Raf-1, an upstream kinase ofERK. Together, these studies demonstrate that ceramide exertsanti-mitogenic actions by limiting the ability of PKC- to form asignaling complex with Raf-1 and ERK.

  相似文献   

10.
It has long been known thatNa+ channels in electrically tight epithelia are regulatedby homeostatic mechanisms that maintain a steady state and allow newlevels of transport to be sustained in hormonally challenged cells.Little is known about the potential pathways involved in theseprocesses. In addition to short-term effect, recent evidence alsoindicates the involvement of PKC in the long-term regulation of theepithelial Na+ channel (ENaC) at the protein level(40). To determine whether stimulation of ENaC involvesfeedback regulation of PKC levels, we utilized Western blot analysis todetermine the distribution of PKC isoforms in polarized A6 epithelia.We found the presence of PKC isoforms in the conventional ( and), novel (, , and ), and atypical (, , and) groups. Steady-state stimulation of Na+ transport withaldosterone was accompanied by a specific decrease of PKC proteinlevels in both the cytoplasmic and membrane fractions. Similarly,overnight treatment with an uncharged amiloride analog (CDPC), aprocedure that through feedback regulation causes a stimulation ofNa+ transport, also decreased PKC levels. These effectswere additive, indicating separate mechanisms that converge at thelevel of PKC. These effects were not accompanied by changes ofPKC mRNA levels as determined by Northern blot analysis. We proposethat this may represent a novel regulatory feedback mechanism necessary for sustaining an increase of Na+ transport.

  相似文献   

11.
Work from this and other laboratories has identified a role forprotein tyrosine kinases in interleukin-1 (IL-1)- and tumor necrosis factor- (TNF-)-induced responses in endothelial cells. In this study, we show that activation of human umbilical vein endothelial cells (HUVEC) by IL-1 leads to increased tyrosine phosphorylation of several proteins including one with a molecular massof ~42 kDa. This protein was identified asp42mapk by Western blot analysis.Tyrosine phosphorylation and catalytic activation ofp42mapk by IL-1 was transient,reaching maximal levels after 30 min and returning to basal levels by120-300 min. Activation ofp42mapk in HUVEC was also observedin response to TNF- or to the protein kinase C (PKC)-activatingphorbol ester phorbol 12-myristate 13-acetate (PMA). Pretreatment ofHUVEC with IL-1 or TNF- prevented reactivation ofp42mapk by either cytokine but didnot affect subsequent activation in response to PMA. Activation ofp42mapk by PMA was significantlyreduced by the PKC inhibitor Ro-31-8220 and completely inhibited by theprotein tyrosine kinase inhibitor genistein. Genistein, but notRo-31-8220, attenuated IL-1- and TNF--inducedp42mapk activation. Takentogether, the results of this study demonstrate 1) thatp42mapk is transiently activatedin HUVEC by IL-1 and TNF-, 2)that this activation is PKC independent, and3) that a genistein-inhibitable tyrosine kinase may be an upstream regulator of cytokine-induced p42mapk activation in humanendothelium.

  相似文献   

12.
We reported previously that Ro-318220 blocked expression ofmitogen-activated protein kinase phosphatase-1 (MKP-1) induced by tumornecrosis factor- (TNF-) and subsequently caused apopotosis inmesangial cells (Y.-L. Guo, B. Kang, and J. R. Williamson. J. Biol. Chem. 273: 10362-10366,1998). These data support our hypothesis that a TNF--induciblephosphatase may be responsible for preventing sustained activation ofc-Jun NH2-terminal protein kinase(JNK) and consequent cell death in these cells (Y.-L. Guo, K. Baysal,B. Kang, L.-J. Yang, and J. R. Williamson. J. Biol. Chem. 273: 4027-4034, 1998). In this study, weinvestigated the involvement of protein kinase C (PKC) in regulation ofMKP-1 expression in mesangial cells together with effects on viability.Although originally characterized as a PKC inhibitor, Ro-318220inhibited TNF--induced MKP-1 expression through a mechanism otherthan blocking the PKC pathway. Furthermore, inhibition of the PKCpathway neither significantly affected TNF--induced MKP-1 expression nor made cells susceptible to toxic effect of TNF-. Thus PKC activation is not essential for cells to achieve the resistance toTNF- cytotoxicity displayed by normal mesangial cells. However, activation of PKC by phorbol 12-myristate 13-acetate (PMA) dramatically increased cellular resistance to the apoptotic effect of TNF-. Coincidentally, PMA stimulated MKP-1 expression and suppressed JNKactivation. Therefore, PMA-induced MKP-1 expression may contribute tothe protective effect of PMA. These results provide a mechanistic explanation for previous documentation that PKC activation can rescuesome cells from apopotosis.  相似文献   

13.
LLC-PK1, an epithelial cellline derived from the kidney proximal tubule, was used to study theability of the G protein -subunit, Gq, to regulate celldifferentiation. A constitutively active mutant protein,qQ209L, was expressed using theLacSwitch-inducible mammalian expression system. Induction ofqQ209L expression with isopropyl--D-thiogalactopyranoside(IPTG) enhanced phospholipase C activity maximally by 6- to 7.5-fold.Increasing concentrations of IPTG progressively inhibited the activityof two differentiation markers,Na+-dependent hexose transport andalkaline phosphatase activity. Induction ofqQ209L expression also caused achange from an epithelial to a spindle-shaped morphology. The effectsof qQ209L expression on celldifferentiation were similar to those observed with12-O-tetradecanoylphorbol 13-acetate(TPA) treatment. However, protein kinase C (PKC) levels weredownregulated in TPA-treated cells but not inqQ209L-expressing cells,suggesting that the regulation of PKC byGq may be different fromregulation by TPA. Interestingly, the PKC inhibitor GF-109203X did notinhibit the effect of IPTG on the development ofNa+-dependent hexose transport inqQ209L-expressing cells. These data implicate PKC and PKC in the pathway used byGq to block the development ofNa+-dependent hexose transport inIPTG-treated cells.

  相似文献   

14.
The involvement of PKC, the isoforms of which are categorized into three subtypes: conventional (, I, II, and ), novel [, , , and µ (also known as PKD),], and atypical ( and /), in the regulation of endothelial monolayer integrity is well documented. However, isoform activity varies among different cell types. Our goal was to reveal isoform-specific PKC activity in the microvascular endothelium in response to phorbol 12-myristate 13-acetate (PMA) and diacylglycerol (DAG). Isoform activity was demonstrated by cytosol-to-membrane translocation after PMA treatment and phosphorylation of the myristoylated alanine-rich C kinase substrate (MARCKS) protein after PMA and DAG treatment. Specific isoforms were inhibited by using both antisense oligonucleotides and pharmacological agents. The data showed partial cytosol-to-membrane translocation of isoforms , I, and and complete translocation of PKC and PKD in response to PMA. Furthermore, antisense treatment and pharmacological studies indicated that the novel isoform PKC and PKD are both required for PMA- and DAG-induced MARCKS phosphorylation and hyperpermeability in pulmonary microvascular endothelial cells, whereas isoforms , I, and were dispensable with regard to these same phenomena. signal transduction; permeability; myristolated alanine-rich C kinase substrate; microvasculature; pulmonary endothelium  相似文献   

15.
Protein kinase C (PKC) has been reported tobe associated with the activation of extracellular signal-regulatedkinase (ERK) by hyperosmolality. However, it is unclear whetherhyperosmolality induces PKC activation and which PKC isoforms areinvolved in ERK activation. In this study, we demonstrate that NaClincreases total PKC activity and induces PKC, PKC, andPKC translocation from the cytosol to the membrane inNIH/3T3 cells, suggesting that hyperosmotic stress activatesconventional PKC (cPKC) and novel PKC (nPKC). Further studies show thatNaCl-inducible ERK1 and ERK2 (ERK1/2) activation is a consequence ofcPKC and nPKC activation, because either downregulation with12-O-tetradecanoylphorbol 13-acetateor selective inhibition of cPKC and nPKC by GF-109203X and rottlerinlargely inhibited the stimulation of ERK1/2 phosphorylation by NaCl. Inaddition, we show that NaCl increases diacylglycerol (DAG) levels andthat a phospholipase C (PLC) inhibitor, U-73122, inhibits NaCl-inducedERK1/2 phosphorylation. These results, together, suggest that ahyperosmotic NaCl-induced signaling pathway that leads to activation ofERK1/2 may sequentially involve PLC activation, DAG release, and cPKCand nPKC activation.

  相似文献   

16.
Tumor necrosis factor-(TNF-) triggers degranulation and oxygen radical release in adherentneutrophils. The p60TNF receptor (p60TNFR) is responsible forproinflammatory signaling, and protein kinase C (PKC) is a candidatefor the regulation of p60TNFR. Both TNF- and the PKC-activatorphorbol 12-myristate 13-acetate triggered phosphorylation of p60TNFR.Receptor phosphorylation was on both serine and threonine but not ontyrosine residues. The PKC- isotype is a candidate enzyme for serinephosphorylation of p60TNFR. Staurosporine and the PKC- inhibitorrottlerin inhibited TNF--triggered serine but not threoninephosphorylation. Serine phosphorylation was associated withreceptor desensitization, as inhibition of PKC resulted in enhanceddegranulation (elastase release). After neutrophil activation, PKC-was the only PKC isotype that associated with p60TNFR within thecorrect time frame for receptor phosphorylation. In vitro, onlyPKC-, but not the -, I-, II-, or -isotypes, wascompetent to phosphorylate the receptor, indicating that p60TNFR is adirect substrate for PKC-. These findings suggest a selective rolefor PKC- in negative regulation of the p60TNFR and ofTNF--induced signaling.

  相似文献   

17.
This study examined the ability of protein kinase C (PKC) toinduce heterologous desensitization by targeting specific G proteinsand limiting their ability to transduce signals in smooth muscle.Activation of PKC by pretreatment of intestinal smooth muscle cellswith phorbol 12-myristate 13-acetate, cholecystokinin octapeptide, orthe phosphatase 1 and phosphatase 2A inhibitor, calyculin A,selectively phosphorylated Gi-1 and Gi-2,but not Gi-3 or Go, and blockedinhibition of adenylyl cyclase mediated by somatostatin receptorscoupled to Gi-1 and opioid receptors coupled toGi-2, but not by muscarinic M2 and adenosineA1 receptors coupled to Gi-3. Phosphorylationof Gi-1 and Gi-2 and blockade of cyclaseinhibition were reversed by calphostin C and bisindolylmaleimide, andadditively by selective inhibitors of PKC and PKC. Blockade ofinhibition was prevented by downregulation of PKC. Phosphorylation ofG-subunits by PKC also affected responses mediated by-subunits. Pretreatment of muscle cells withcANP-(4-23), a selective agonist of the natriureticpeptide clearance receptor, NPR-C, which activates phospholipase C(PLC)-3 via the -subunits of Gi-1 andGi-2, inhibited the PLC- response to somatostatin and[D-Pen2,5]enkephalin. The inhibition waspartly reversed by calphostin C. Short-term activation of PKC had noeffect on receptor binding or effector enzyme (adenylyl cyclase orPLC-) activity. We conclude that selective phosphorylation ofGi-1 and Gi-2 by PKC partly accounts forheterologous desensitization of responses mediated by the - and-subunits of both G proteins. The desensitization reflects adecrease in reassociation and thus availability of heterotrimeric G proteins.

  相似文献   

18.
In the estrogen-treated rat myometrium, carbachol increased thegeneration of inositol phosphates by stimulating the muscarinic receptor-Gq/G11-phospholipaseC-3 (PLC-3) cascade. Exposure to carbachol resulted in a rapidand specific (homologous) attenuation of the subsequent muscarinicresponses in terms of inositol phosphate production, PLC-3translocation to membrane, and contraction. Refractoriness wasaccompanied by a reduction of membrane muscarinic binding sites and anuncoupled state of residual receptors. Protein kinase C (PKC) alteredthe functionality of muscarinic receptors and contributed to theinitial period of desensitization. A delayed phase of the muscarinicrefractoriness was PKC independent and was associated with adownregulation ofGq/G11.Atropine failed to induce desensitization as well asGq/G11downregulation, indicating that both events involve active occupancy ofthe receptor. Prolonged exposure toAlF4 reduced subsequent AlF4 as well as carbachol-mediatedinositol phosphate responses and similarly induced downregulation ofGq/G11. Data suggest that a decrease in the level ofGq/G11is subsequent to its activation and may account forheterologous desensitization.

  相似文献   

19.
-Opioid receptor (-OR)stimulation with U50,488H, a selective -OR agonist, or activation ofprotein kinase C (PKC) with 4-phorbol 12-myristate 13-acetate (PMA), anactivator of PKC, decreased the electrically induced intracellularCa2+ ([Ca2+]i) transient andincreased the intracellular pH (pHi) in single ventricularmyocytes of rats subjected to 10% oxygen for 4 wk. The effects ofU50,488H were abolished by nor-binaltorphimine, a selective -ORantagonist, and calphostin C, a specific inhibitor of PKC, while theeffects of PMA were abolished by calphostin C andethylisopropylamiloride (EIPA), a potent Na+/H+exchange blocker. In both right hypertrophied and leftnonhypertrophied ventricles of chronically hypoxic rats, the effects ofU50,488H or PMA on [Ca2+]i transient andpHi were significantly attenuated and completely abolished,respectively. Results are first evidence that the[Ca2+]i and pHi responses to-OR stimulation are attenuated in the chronically hypoxic rat heart,which may be due to reduced responses to PKC activation. Responses toall treatments were the same for right and left ventricles, indicatingthat the functional impairment is independent of hypertrophy. -ORmRNA expression was the same in right and left ventricles of bothnormoxic and hypoxic rats, indicating no regional specificity.

  相似文献   

20.
A role for protein kinase C (PKC)- and -isotypes in 1-adrenergicregulation of human tracheal epithelial Na-K-2Cl cotransport wasstudied with the use of isotype-specific PKC inhibitors and antisenseoligodeoxynucleotides to PKC- or - mRNA. Rottlerin, a PKC-inhibitor, blocked 72% of basolateral-to-apical, bumetanide-sensitive 36Cl flux innystatin-permeabilized cell monolayers stimulated with methoxamine, an1-adrenergic agonist, with a50% inhibitory concentration of 2.3 µM. Methoxamine increased PKCactivity in cytosol and a particulate fraction; the response wasinsensitive to PKC- and -IIisotype-specific inhibitors, but was blocked by general PKC inhibitorsand rottlerin. Rottlerin also inhibited methoxamine-induced PKCactivity in immune complexes of PKC-, but not PKC-. At the subcellular level, methoxamine selectively elevated cytosolic PKC-activity and particulate PKC- activity. Pretreatment of cellmonolayers with antisense oligodeoxynucleotide to PKC- for 48 hreduced the amount of whole cell and cytosolic PKC-, diminished whole cell and cytosolic PKC- activity, and blockedmethoxamine-stimulated Na-K-2Cl cotransport. Sense oligodeoxynucleotideto PKC- and antisense oligodeoxynucleotide to PKC- did not altermethoxamine-induced cotransport activity. These results demonstrate theselective activation of Na-K-2Cl cotransport by cytosolic PKC-.

  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