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.
Although K-Cl cotransporter (KCC1) mRNA is expressed in manytissues, K-Cl cotransport activity has been measured in few cell types,and detection of endogenous KCC1 polypeptide has not yet been reported.We have cloned the mouse erythroid KCC1 (mKCC1) cDNA and its flankinggenomic regions and mapped the mKCC1 gene to chromosome 8. Threeanti-peptide antibodies raised against recombinant mKCC1 function asimmunoblot and immunoprecipitation reagents. The tissue distributionsof mKCC1 mRNA and protein are widespread, and mKCC1 RNA isconstitutively expressed during erythroid differentiation of ES cells.KCC1 polypeptide or related antigen is present in erythrocytes ofmultiple species in which K-Cl cotransport activity has beendocumented. Erythroid KCC1 polypeptide abundance is elevated inproportion to reticulocyte counts in density-fractionated cells, inbleeding-induced reticulocytosis, in mouse models of sickle celldisease and thalassemia, and in the corresponding human disorders.mKCC1-mediated uptake of 86Rb intoXenopus oocytes requires extracellularCl, is blocked by thediureticR(+)-[2-n-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1H-indenyl-5-yl-)oxy]acetic acid, and exhibits an erythroid pattern of acute regulation, with activation by hypotonic swelling,N-ethylmaleimide, and staurosporine and inhibition by calyculin and okadaic acid. These reagents and findings will expedite studies of KCC1 structure-function relationships and of the pathobiology of KCC1-mediated K-Cl cotransport.

  相似文献   

2.
Platelet-derived growth factor (PDGF), apotent serum mitogen for vascular smooth muscle cells (VSMCs), plays animportant role in membrane transport regulation and in atherosclerosis. K-Cl cotransport (K-Cl COT/KCC), the coupled-movement of K and Cl, isinvolved in ion homeostasis. VSMCs possess K-Cl COT activity and theKCC1 and KCC3 isoforms. Here, we report on the effect of PDGF on K-ClCOT activity and mRNA expression in primary cultures of rat VSMCs. K-ClCOT was determined as the Cl-dependent Rb influx and mRNA expression bysemiquantitative RT-PCR. Twenty four-hour serum deprivation inhibitedbasal K-Cl COT activity. Addition of PDGF increased total proteincontent and K-Cl COT activity in a time-dependent manner. PDGFactivated K-Cl COT in a dose-dependent manner, both acutely (10 min)and chronically (12 h). AG-1296, a selective inhibitor of the PDGFreceptor tyrosine kinase, abolished these effects. Actinomycin D andcycloheximide had no effect on the acute PDGF activation of K-Cl COT,suggesting posttranslational regulation by the drug. Furthermore, PDGFincreased KCC1 and decreased KCC3 mRNA expression in a time-dependentmanner. These results indicate that chronic activation of K-Cl COTactivity by PDGF may involve regulation of the two KCC mRNA isoforms,with KCC1 playing a dominant role in the mechanism of PDGF-mediated activation.

  相似文献   

3.
4.
Red blood cells (RBCs) possess the K-Cl cotransport (KCC) isoforms 1, 3, and 4. Mutations within a given isoform may affect overall KCC activity. In a double-blind study, we analyzed, with Rb as a K congener, K fluxes (total flux, ouabain-sensitive Na+/K+ pump, and bumetanide-sensitive Na-K-2Cl cotransport, Cl-dependent, and ouabain- and bumetanide-insensitive KCC with or without stimulation by N-ethylmaleimide (NEM) and staurosporine or Mg removal, and basal channel-mediated fluxes, osmotic fragility, and ions and water in the RBCs of 8 controls, and of 8 patients with hereditary motor and sensory neuropathy with agenesis of corpus callosum (HMSN-ACC) with defined KCC3 mutations (813FsX813 and Phe529FsX532) involving the truncations of 338 and 619 C-terminal amino acids, respectively. Water and ion content and, with one exception, mean osmotic fragility, as well as K fluxes without stimulating agents, were similar in controls and HMSN-ACC RBCs. However, the NEM-stimulated KCC was reduced 5-fold (p < 0.0005) in HMSN-ACC vs control RBCs, as a result of a lower Vmax (p < 0.05) rather than a lower Km (p = 0.109), accompanied by corresponding differences in Cl activation. Low intracellular Mg activated KCC in 6 out of 7 controls vs 1 out of 6 HMSN-ACC RBCs, suggesting that regulation is compromised. The lack of differences in staurosporine-activated KCC indicates different action mechanisms. Thus, in HMSN-ACC patients with KCC3 mutants, RBC KCC activity, although indistinguishable from that of the control group, responded differently to biochemical stressors, such as thiol alkylation or Mg removal, thereby indirectly indicating an important contribution of KCC3 to overall KCC function and regulation.  相似文献   

5.
K-Cl cotransport, theelectroneutral-coupled movement of K and Cl ions, plays an importantrole in regulatory volume decrease. We recently reported that nitrite,a nitric oxide derivative possessing potent vasodilation properties,stimulates K-Cl cotransport in low-K sheep red blood cells (LK SRBCs).We hypothesized that activation of vascular smooth muscle (VSM) K-Clcotransport by vasodilators decreases VSM tension. Here we tested thishypothesis by comparing the effects of commonly used vasodilators,hydralazine (HYZ), sodium nitroprusside, isosorbide mononitrate, andpentaerythritol, on K-Cl cotransport in LK SRBCs and in primarycultures of rat VSM cells (VSMCs) and of HYZ-induced K-Clcotransport activation on relaxation of isolated porcine coronaryrings. K-Cl cotransport was measured as the Cl-dependent K efflux or Rbinflux in the presence and absence of inhibitors for other K/Rbtransport pathways. All vasodilators activated K-Cl cotransport in LKSRBCs and HYZ in VSMCs, and this activation was inhibited by calyculinand genistein, two inhibitors of K-Cl cotransport. KT-5823, a specificinhibitor of protein kinase G, abolished the sodiumnitroprusside-stimulated K-Cl cotransport in LK SRBCs, suggestinginvolvement of the cGMP pathway in K-Cl cotransport activation.Hydralazine, in a dose-dependent manner, and sodium nitroprussiderelaxed (independently of the endothelium) precontractedarteries when only K-Cl cotransport was operating and other pathwaysfor K/Rb transport, including the Ca-activated K channel, wereinhibited. Our findings suggest that K-Cl cotransport may be involvedin vasodilation.

  相似文献   

6.
K-Cl cotransport plays a crucial role in regulatory volume decrease of erythrocytes. K-Cl cotransport activities in dog erythrocytes with an inherited high Na-K pump activity (HK) and normal erythrocytes (LK) were compared. Nitrite (NO(2)) stimulated K-Cl cotransport activity in HK cells around 14-fold at 2.4 mM, and it also increased the Km value of this cotransporter. Real-time PCR and western blot analysis revealed that K-Cl cotransporter 1 was dominant, and that the quantity of K-Cl cotransporter 1 protein was comparable between HK and LK erythrocytes. These results suggest that the difference in cotransport activity was not caused by the amount of K-Cl cotransport protein but by a difference in the regulation system, which is susceptible to oxidant.  相似文献   

7.
8.
9.
Active potassium absorption in the rat distal colon is electroneutral, Na(+)-independent, partially chloride-dependent, and energized by an apical membrane H,K-ATPase. Both dietary sodium and dietary potassium depletion substantially increase active potassium absorption. We have recently reported that sodium depletion up-regulates H,K-ATPase alpha-subunit mRNA and protein expression, whereas potassium depletion up-regulates H,K-ATPase beta-subunit mRNA and protein expression. Because overall potassium absorption is non-conductive, K-Cl cotransport (KCC) at the basolateral membrane may also be involved in potassium absorption. Although KCC1 has not been cloned from the colon, we established, in Northern blot analysis with mRNA from the rat distal colon using rabbit kidney KCC1 cDNA as a probe, the presence of an expected size mRNA in the rat colon. This KCC1 mRNA is substantially increased by potassium depletion but only minimally by sodium depletion. KCC1-specific antibody identified a 155-kDa protein in rat colonic basolateral membrane. Potassium depletion but not sodium depletion resulted in an increase in KCC1 protein expression in basolateral membrane. The increase of colonic KCC1 mRNA abundance and KCC1 protein expression in potassium depletion of the rat colonic basolateral membrane suggests that K-Cl cotransporter: 1) is involved in transepithelial potassium absorption and 2) regulates the increase in potassium absorption induced by dietary potassium depletion. We conclude that active potassium absorption in the rat distal colon involves the coordinated regulation of both apical membrane H,K-ATPase and basolateral membrane KCC1 protein.  相似文献   

10.
Swelling-activated [K-Cl] cotransport and shrinkage-activated Na/H exchange were studied in dog red cells with altered internal Mg or Li content. The two pathways responded in a coordinated fashion. When cells were depleted of Mg, [K-Cl] cotransport was stimulated and Na/H exchange was inhibited. Raising internal Mg had the opposite effect: [K-Cl] cotransport was inhibited and Na/H exchange was stimulated. Li loading, previously shown to stimulate Na/H exchange, inhibited [K-Cl] cotransport. From these reciprocal effects and from other evidence, we surmise that the regulation of Na/H exchange and [K-Cl] cotransport is conducted and coordinated by a discrete mechanism that responds to changes in cell volume and is sensitive to cytoplasmic Mg and Li concentrations.  相似文献   

11.
K-Cl cotransport, KCC, is activated by swelling in many cells types, and promotes volume regulation by a KCl efflux osmotically coupled to water efflux. KCC is probably activated by swelling-inhibition of a kinase, permitting dephosphorylation, and activation of the cotransporter by a phosphatase. The myosin light chain kinase (MLCK) inhibitor ML-7 inhibits transporters activated by shrinkage. In red blood cells from three mammalian species, ML-7 stimulated KCC in a volume-dependent manner. Relative stimulation was greatest in more shrunken cells. Stimulation was reduced by moderate cell swelling and abolished by further swelling. The half-maximal stimulation is at ∼20 μm ML-7, 50-fold greater than the IC50 for inhibition of MLCK in vitro. Stimulation of KCC by ML-7 did not require cell Ca, while MLCK does. Therefore the target of ML-7 in stimulating KCC in red cells is probably not MLCK. The evidence favors stimulation of KCC by ML-7 by inhibiting the volume-sensitive kinase. Qualitatively similar effects of ML-7 on KCC in red cells from three mammalian species suggest a general mechanism. Received: 17 March 2000/Revised: 28 July 2000  相似文献   

12.
Zhang J  Lauf PK  Adragna NC 《Life sciences》2005,77(9):953-965
K-Cl cotransport (K-Cl COT, KCC) is an electroneutrally coupled movement of K and Cl present in most cells. In this work, we studied the pathways of regulation of K-Cl COT by platelet-derived growth factor (PDGF) in primary cultures of vascular smooth muscle cells (VSMCs). Wortmannin and LY 294002 blocked the PDGF-induced K-Cl COT activation, indicating that the phosphoinositide 3-kinase (PI 3-K) pathway is involved. However, PD 98059 had no effect on K-Cl COT activation by PDGF, suggesting that the mitogen-activated protein kinase pathway is not involved under the experimental conditions tested. Involvement of phosphatases was also examined. Sodium orthovanadate, cyclosporin A and okadaic acid had no effect on PDGF-stimulated K-Cl COT. Calyculin A blocked the PDGF-stimulated K-Cl COT by 60%, suggesting that protein phosphatase-1 (PP-1) is a mediator in the PDGF signaling pathway/s. In conclusion, our results indicate that the PDGF-mediated pathways of K-Cl COT regulation involve the signaling molecules PI 3-K and PP-1.  相似文献   

13.
K-Cl cotransporter expression in the human kidney   总被引:2,自引:0,他引:2  
The K-Cl cotransporter protein KCC1 is a membrane transportprotein that mediates the coupled, electroneutral transport of K and Clacross plasma membranes. The precise cell type(s) in the kidney thatexpress the K-Cl cotransporter have remained unknown. The aim of thepresent investigation was to define the distribution of KCC1 mRNA inthe human kidney. We used in situ hybridization with a nonradioactivedigoxigenin-labeled riboprobe. We identified abundant KCC1 mRNAexpression in the epithelial cells throughout the distal and proximalrenal tubular epithelium. The transporter was also expressed inglomerular mesangial cells and endothelial cells of the renal vessels.These findings suggest that the K-Cl cotransporter may have animportant role in transepithelial K and Cl reabsorption.

  相似文献   

14.
Volume-sensitive K-Cl cotransport occurs in red blood cells of many species. In intact cells, activation of K-Cl cotransport by swelling requires dephosphorylation of some cell protein, but maximal activity requires the presence of intracellular ATP. We have examined the relation between K-Cl cotransport activity and ATP in ghosts prepared from human red blood cells. K-Cl cotransport activity in swollen ghosts increased by ATP, and the increase requires Mg so that it almost certainly results from the phosphorylation of some membrane component. However, even in ATP-free ghosts residual volume-sensitive K-Cl cotransport can be demonstrated. This residual cotransport in ATP-free ghosts is greater in the presence of vanadate, a tyrosyl phosphatase inhibitor, and in ghosts that contain ATP cotransport is reduced by genistein, a tyrosyl kinase inhibitor. Okadaic acid, an inhibitor of serine and threonine phosphatases, inhibits K-Cl cotransport in ghosts as it does in intact cells. Experiments in which ghosts were preexposed to okadaic acid showed that the protein dephosphorylation that permits K-Cl cotransport can proceed to completion before the ghosts are swollen and K transport measured and therefore dephosphorylation is not a response to ghost swelling. In experiments with ATP-free ghosts we found that phosphorylation is not necessary to increase the cotransport rate when shrunken ghosts are swollen, nor is rephosphorylation necessary to decrease the cotransport rate when swollen ghosts are shrunken. Cotransport is greater in swollen than in shrunken ghosts even when the swollen and shrunken ghosts have the same concentration of cytoplasmic solutes. We conclude that, although phosphorylation and dephosphorylation modify the activity of the cotransporter in swollen and in shrunken ghosts, neither of these processes nor any other known messenger is involved in signal transduction between the cell volume sensor and the cotransporter as originally proposed by Jennings and Al- Rohil (Jennings, M. L., and N. Al-Rohil. 1990. Journal of General Physiology. 95: 1021-1040).  相似文献   

15.
Mg is an importantdeterminant of erythrocyte cation transport system(s) activity. Weinvestigated cation transport in erythrocytes from mice bred for high(MGH) and low (MGL) Mg levels in erythrocytes and plasma. We found thatK-Cl cotransport activity was higher in MGL than in MGH erythrocytes,and this could explain their higher mean corpuscular hemoglobinconcentration, median density, and reduced cell K content. Althoughmouse KCC1 protein abundance was comparable in MGL and MGHerythrocytes, activities of Src family tyrosine kinases were higher inMGH than in MGL erythrocytes. In contrast, protein phosphatase (PP)isoform 1 (PP1) enzymatic activity, which has been suggested toplay a positive regulatory role in K-Cl cotransport, was lower in MGHthan in MGL erythrocytes. Additionally, we found that the Src familykinase c-Fgr tyrosine phosphorylates PP1 in vitro. These findingssuggest that in vivo downregulation of K-Cl cotransport activity by Mgis mediated by enhanced Src family kinase activity, leading toinhibition of the K-Cl cotransport stimulator PP1.

  相似文献   

16.
K-Cl co-transport (COT) is significantly higher in low K (LK), L-antigen (L) positive, than in high K (HK), M-antigen (M) positive, sheep red blood cells (SRBCs) and is inhibited by sheep allo-anti-L1 antibody. To answer the question of whether this difference in K-Cl co-transport activity resides at the level of the transporter or its regulation, a combined immunocytochemical and functional approach was taken. At least four isoforms of K-Cl COT encoded by different KCC genes are known, with 12 transmembrane domains and cytoplasmic C- and N-terminal domains (Ctd and Ntd, respectively). Polyclonal anti-rat (rt)KCC1 antibodies against a fusion peptide with 77 amino acids from the Ctd of rtKCC1 and anti-human (h)KCC3 against an 18-aa peptide from the Ntd of hKCC3, were prepared in rabbits (rb). Two distinctly separate protein bands of 180 and 145 kDa molecular mass were detected in hemoglobin-free ghosts from RBCs of two LK (one homozygous LL and one heterozygous LM) and one HK (homozygous MM) sheep by Western blots with rb anti-rtKCC1 and rb anti-hKCC3. Confocal microscopy showed specific immunostaining of KCC1 with rb anti-rtKCC1, and of KCC3 with rb anti-hKCC3, in white ghosts from both LK and HK SRBCs. To test the functional heterogeneity of K-Cl COT, the effect of the anti-L1 antibody was assessed on K-Cl COT activated by the kinase inhibitor staurosporine. Incubation of LK SRBCs with anti-L1 serum inhibited by 30% staurosporine-stimulated K-Cl COT suggesting that approximately two-thirds of the transport activity is independent of the L1 antigen. That staurosporine altered the L1 antigen/antibody reaction is unlikely since the action of another antibody, anti-Lp, stimulating the Na/K pump flux, was not modified. The present results, in conjunction with earlier work, lead to the hypothesis that the partial anti-L1 inhibition of K-Cl COT may be related to the molecular KCC dimorphism, seen in these cells with anti-KCC1 and anti-KCC3 antibodies.  相似文献   

17.
We isolated and characterized a novelK-Cl cotransporter, KCC3, from human placenta. The deduced proteincontains 1,150 amino acids. KCC3 shares 75-76% identity at theamino acid level with human, pig, rat, and rabbit KCC1 and 67%identity with rat KCC2. KCC3 is 40 and 33% identical to twoCaenorhabditis elegans K-Cl cotransporters and ~20%identical to other members of the cation-chloride cotransporter family(CCC), two Na-K-Cl cotransporters (NKCC1, NKCC2), and the Na-Clcotransporter (NCC). Hydropathy analysis indicates a typical KCCtopology with 12 transmembrane domains, a large extracellular loopbetween transmembrane domains 5 and 6 (unique to KCCs), and largeNH2 and COOH termini. KCC3 is predominantly expressed inkidney, heart, and brain, and is also expressed in skeletal muscle,placenta, lung, liver, and pancreas. KCC3 was localized to chromosome15. KCC3 transiently expressed in human embryonic kidney (HEK)-293cells fulfilled three criteria for increased expression of K-Clcotransport: stimulation of cotransport by swelling, treatment withN-ethylmaleimide, or treatment with staurosporine.

  相似文献   

18.
We have studiedthe regulation of the K-Cl cotransporter KCC1 and its functionalinteraction with the Na-K-Cl cotransporter. K-Cl cotransporter activitywas substantially activated in HEK-293 cells overexpressing KCC1(KCC1-HEK) by hypotonic cell swelling, 50 mM external K, andpretreatment with N-ethylmaleimide(NEM). Bumetanide inhibited 86Rbefflux in KCC1-HEK cells after cell swelling [inhibition constant (Ki) ~190µM] and pretreatment with NEM(Ki ~60 µM).Thus regulation of KCC1 is consistent with properties of the red cellK-Cl cotransporter. To investigate functional interactions between K-Cland Na-K-Cl cotransporters, we studied the relationship between Na-K-Clcotransporter activation and intracellular Cl concentration([Cl]i). Without stimulation, KCC1-HEK cells had greater Na-K-Cl cotransporter activitythan controls. Endogenous Na-K-Cl cotransporter of KCC1-HEK cells wasactivated <2-fold by low-Cl hypotonic prestimulation, compared with10-fold activation in HEK-293 cells and >20-fold activation in cellsoverexpressing the Na-K-Cl cotransporter (NKCC1-HEK). KCC1-HEK cellshad lower resting[Cl]i than HEK-293cells; cell volume was not different among cell lines. We found a steeprelationship between[Cl]i and Na-K-Clcotransport activity within the physiological range, supporting aprimary role for [Cl]iin activation of Na-K-Cl cotransport and in apical-basolateral crosstalk in ion-transporting epithelia.  相似文献   

19.
Cervical cancer is a major world health problem for women, but the pathophysiology of this disease has received scant attention. Here we show that the growth and invasion of cervical cancer cells are strongly linked the expression and activity of the KCl cotransporter (KCC), an important regulator of the ionic and cellular osmotic homeostasis. Functional assays of KCl cotransport activation by osmotic swelling, staurosporine, and N-ethylmaleimide indicate that removal of the N-terminal 117 amino acids from KCC1 produces a dominant-negative loss-of-function phenotype for KCl cotransport in human cervical cancer cells. The capability for regulatory volume decrease is much attenuated in the loss-of-function KCC mutant cervical cancer cells. The loss-of-function KCC mutant cervical cancer cells exhibit inhibited cell growth accompanied by decreased activity of the cell cycle gene products retinoblastoma and cdc2 kinase. Reduced cellular invasiveness is in parallel by reduced expression of alpha v beta 3 and alpha 6 beta 4 integrins, accompanied by decreased activity of matrix metalloproteinase 2 and 9. Inhibition of tumor growth in SCID mice confirms the crucial role of KCC in promoting cervical cancer growth and invasion. Thus, blockade of KCl cotransport may be a useful therapeutic adjunctive strategy to retard or prevent cervical cancer invasion.  相似文献   

20.
Activation of K-Cl cotransport is associated with activation ofmembrane-bound serine/threonine protein phosphatases (S/T-PPases). Wecharacterize red blood cell S/T-PPases and K-Clcotransport activity regarding protein phosphatase inhibitors andresponse to changes in ionic strength and cell size. Proteinphosphatase type 1 (PP1) activity is highly sensitive to calyculin A(CalA) but not to okadaic acid (OA). PP2A activity is highly sensitive to CalA and OA. CalA completely inhibits K-Cl cotransport activity, whereas OA partially inhibits K-Cl cotransport. Membrane PP1 and membrane PP2A activities are elevated in cells suspended in hypotonic solutions, where K-Cl cotransport is elevated. Increases in membrane PP1 activity (62 ± 10% per 100 meq/l) result from decreases in intracellular ionic strength and correlate with increases in K-Cl cotransport activity (54 ± 10% per 100 meq/l). Increasesin membrane PP2A activity (270 ± 77% per 100 mosM) result fromvolume increases and also correlate with increases in K-Cl cotransportactivity (420 ± 47% per 100 mosM). The characteristics ofmembrane-associated PP1 and PP2A are consistent with a role for bothphosphatases in K-Cl cotransport activation in human erythrocytes.

  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