Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 87: 180-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26283571

ABSTRACT

Interendothelial junctions play an important role in the maintenance of endothelial integrity and the regulation of vascular functions. We report here that cationic amino acid transporter-1 (CAT-1) is a novel interendothelial cell adhesion molecule (CAM). We identified that CAT-1 protein localized at cell-cell adhesive junctions, similar to the classic CAM of VE-cadherin, and knockdown of CAT-1 with siRNA led to an increase in endothelial permeability. In addition, CAT-1 formed a cis-homo-dimer and showed Ca(2+)-dependent trans-homo-interaction to cause homophilic cell-cell adhesion. Co-immunoprecipitation assays showed that CAT-1 can associate with ß-catenin. Furthermore, we found that the sub-cellular localization and function of CAT-1 are associated with cell confluency, in sub-confluent ECs CAT-1 proteins distribute on the entire surface and function as L-Arg transporters, but most of the CAT-1 in the confluent ECs are localized at interendothelial junctions and serve as CAMs. Further functional characterization has disclosed that extracellular L-Arg exposure stabilizes endothelial integrity via abating the cell junction disassembly of CAT-1 and blocking the cellular membrane CAT-1 internalization, which provides the new mechanisms for L-Arg paradox and trans-stimulation of cationic amino acid transport system (CAAT). These results suggest that CAT-1 is a novel CAM that directly regulates endothelial integrity and mediates the protective actions of L-Arg to endothelium via a NO-independent mechanism.


Subject(s)
Capillary Permeability/genetics , Cationic Amino Acid Transporter 1/biosynthesis , Cationic Amino Acid Transporter 1/metabolism , Cell Adhesion/genetics , Animals , Arginine/metabolism , Cationic Amino Acid Transporter 1/genetics , Gap Junctions/genetics , Gap Junctions/pathology , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Nitric Oxide/metabolism , Swine , beta Catenin/metabolism
2.
Mol Cell Biochem ; 360(1-2): 309-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21948261

ABSTRACT

We previously reported that the vasoactive peptide 1 (P1, "SSWRRKRKESS") modulates the tension of pulmonary artery vessels through caveolar endothelial nitric oxide synthase (eNOS) activation in intact lung endothelial cells (ECs). Since PKC-α is a caveolae resident protein and caveolae play a critical role in the peptide internalization process, we determined whether modulation of caveolae and/or caveolar PKC-α phosphorylation regulates internalization of P1 in lung ECs. Cell monolayers were incubated in culture medium containing Rhodamine red-labeled P1 (100 µM) for 0-120 min. Confocal examinations indicate that P1 internalization is time-dependent and reaches a plateau at 60 min. Caveolae disruption by methyl-ß-cyclodextrin (CD) and filipin (FIL) inhibited the internalization of P1 in ECs suggesting that P1 internalizes via caveolae. P1-stimulation also enhances phosphorylation of caveolar PKC-α and increases intracellular calcium (Ca(2+)) release in intact cells suggesting that P1 internalization is regulated by PKC-α in ECs. To confirm the roles of increased phosphorylation of PKC-α and Ca(2+) release in internalization of P1, PKC-α modulation by phorbol ester (PMA), PKC-α knockdown, and Ca(2+) scavenger BAPTA-AM model systems were used. PMA-stimulated phosphorylation of caveolar PKC-α is associated with significant reduction in P1 internalization. In contrast, PKC-α deficiency and reduced phosphorylation of PKC-α enhanced P1 internalization. P1-mediated increased phosphorylation of PKC-α appears to be associated with increased intracellular calcium (Ca(2+)) release since the Ca(2+) scavenger BAPTA-AM enhanced P1 internalization. These data indicate that caveolar integrity and P1-mediated increased phosphorylation of caveolar PKC-α play crucial roles in the regulation of P1 internalization in lung ECs.


Subject(s)
Caveolae/enzymology , Cell-Penetrating Peptides/metabolism , Endothelial Cells/metabolism , Lung/cytology , Peptides/metabolism , Protein Kinase C-alpha/metabolism , Animals , Calcium Signaling , Caveolae/drug effects , Caveolin 1/metabolism , Cells, Cultured , Endocytosis/drug effects , Endothelial Cells/enzymology , Enzyme Activation , Enzyme Activators/pharmacology , Filipin/pharmacology , Phosphorylation , Protein Kinase C-alpha/genetics , Swine , Tetradecanoylphorbol Acetate/pharmacology , beta-Cyclodextrins/pharmacology
3.
Biochim Biophys Acta ; 1800(9): 906-11, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20594946

ABSTRACT

BACKGROUND: The H,K-ATPase, consisting of α and ß subunits, belongs to the P-type ATPase family. There are two isoforms of the α subunit, HKα1 and HKα2 encoded by different genes. The ouabain-resistant gastric HKα1-H,K-ATPase is Sch28080-sensitive. However, the colonic HKα2-H,K-ATPase from different species shows poor primary structure conservation of the HKα2 subunit between species and diverse pharmacological sensitivity to ouabain and Sch28080. This study sought to determine the contribution of each gene to functional activity and its pharmacological profile using mouse models with targeted disruption of HKα1, HKα2, or HKbeta genes. METHODS: Membrane vesicles from gastric mucosa and distal colon in wild-type (WT), HKα1, HKα2, or HKß knockout (KO) mice were extracted. K-ATPase activity and pharmacological profiles were examined. RESULTS: The colonic H,K-ATPase demonstrated slightly greater affinity for K(+) than the gastric H,K-ATPase. This K-ATPase activity was not detected in the colon of HKα2 KO but was observed in HKß KO with properties indistinguishable from WT. Neither ouabain nor Sch28080 had a significant effect on the WT colonic K-ATPase activity, but orthovanadate abolished this activity. Amiloride and its analogs benzamil and 5-N-ethyl-N-isopropylamiloride inhibited K-ATPase activity of HKα1-containing H,K-ATPase; the dose dependence of inhibition was similar for all three inhibitors. In contrast, the colonic HKα2-H,K-ATPase was not inhibited by these compounds. CONCLUSIONS: These data demonstrate that the mouse colonic H,K-ATPase exhibits a ouabain- and Sch28080-insensitive, orthovanadate-sensitive K-ATPase activity. Interestingly, pharmacological studies suggested that the mouse gastric H,K-ATPase is sensitive to amiloride. GENERAL SIGNIFICANCE: Characterization of the pharmacological profiles of the H,K-ATPases is important for understanding the relevant knockout animals and for considering the specificity of the inhibitors.


Subject(s)
Colon/enzymology , Drug Resistance/physiology , Enzyme Inhibitors/pharmacology , Gastric Mucosa/enzymology , H(+)-K(+)-Exchanging ATPase/metabolism , Proton Pump Inhibitors , Amiloride/pharmacology , Animals , Catalytic Domain/genetics , Drug Resistance/drug effects , H(+)-K(+)-Exchanging ATPase/genetics , Imidazoles/pharmacology , Mice , Mice, Knockout , Organ Specificity/drug effects , Organ Specificity/physiology , Ouabain/pharmacology , Sodium Channel Blockers/pharmacology , Vanadates/pharmacology
4.
Am J Physiol Renal Physiol ; 298(2): F408-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923411

ABSTRACT

In the collecting duct (CD), H-K-ATPases function in cation reabsorption and H secretion. This study evaluated H-K-ATPase-mediated H secretion along the mouse CD, measured as EIPA- and luminal bafilomycin A(1)-insensitive intracellular pH (pH(i)) recovery from acute H loading (NH(4)) using BCECF. pH(i) recovery was measured in 1) microperfused cortical, outer medullary, and inner medullary CDs (CCD, OMCD, and IMCD) from C57BL/6J mice fed a normal diet and 2) common murine CD cell lines. H-K-ATPase activity along the native, microperfused CD was greatest in the CCD, less in the OMCD, and least in the IMCD (0.10 +/- 0.02, 0.04 +/- 0.01, and 0.01 +/- 0.002 U/min, respectively). H-K-ATPase activity was 0.30 +/- 0.03 and 0.26 +/- 0.03 in A- and B-type ICs, respectively, and was sensitive to Sch-28080 or ouabain. pH(i) recovery was greatest in the OMCD(1) cell line (0.25 +/- 0.01) and less in mpkCCD(c14) (0.17 +/- 0.01), mIMCD-K2 (0.12 +/- 0.01), and mIMCD-3 (0.05 +/- 0.01) cells. EIPA inhibited the majority of pH(i) recovery in these cells (100%, 64%, 75%, and 80% in mpkCCD(c14), OMCD(1), mIMCD-K2, and mIMCD-3, respectively). In OMCD(1) cells, where EIPA-insensitive pH(i) recovery was greatest, H-K-ATPase activity was 0.10 +/- 0.01 and was significantly inhibited (80%) by Sch-28080. We conclude that 1) H-K-ATPase-mediated H secretion in the native mouse CD is greatest in the ICs of the CCD, 2) A- and B-type ICs possess HKalpha(1) and HKalpha(2) H-K-ATPase activity, and 3) the OMCD(1) cell line best exhibits H-K-ATPase.


Subject(s)
Acids/metabolism , H(+)-K(+)-Exchanging ATPase/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Female , H(+)-K(+)-Exchanging ATPase/genetics , Hydrogen-Ion Concentration , Imidazoles/pharmacology , In Vitro Techniques , Intracellular Membranes/metabolism , Isoenzymes/genetics , Kidney Cortex/cytology , Kidney Medulla/cytology , Kidney Tubules, Collecting/cytology , Macrolides/pharmacology , Mice , Mice, Inbred C57BL , Ouabain/pharmacology , Perfusion/methods , Proton Pump Inhibitors , RNA, Messenger/metabolism , Tissue Distribution
5.
Biophys J ; 97(6): 1578-85, 2009 Sep 16.
Article in English | MEDLINE | ID: mdl-19751662

ABSTRACT

Tension generation in endothelial cells of the aorta, spleen, and eye occurs in actin stress fibers, and is necessary for normal cell function. Sarcomeres are the tension-generating units of actin stress fibers in endothelial cells. How sarcomeres generate and maintain tension in stress fibers is not well understood. Using femtosecond laser ablation, we severed living stress fibers and measured sarcomere contraction under zero tension. The length of the sarcomere decreased in two phases: an instantaneous initial response, followed by a slower change in length attributed to myosin activity. The latter phase ceased abruptly after a minimum sarcomere length was reached, suggesting a rigid resistance that prevents further contraction. Furthermore, severed, contracted stress fibers did not relax when treated with myosin inhibitors, indicating that contracted stress fibers do not store elastic potential energy. These novel measurements combined with modeling suggest that myosin-generated forces in adjacent sarcomeres are directly in balance, and argue against sarcomere models with springlike elements in parallel with myosin contractile elements. We propose a new model for tension generation in the sarcomere, which provides a mechanistic interpretation for our observations and previous observations of inhomogeneous sarcomere contraction and apparent stress fiber viscoelastic behavior.


Subject(s)
Endothelial Cells/cytology , Sarcomeres/metabolism , Animals , Biomechanical Phenomena , Cattle , Elasticity , Endothelial Cells/metabolism , Laser Therapy , Linear Models , Models, Biological , Myosins/metabolism , Stress Fibers/metabolism , Time Factors
6.
Mediators Inflamm ; 2009: 535348, 2009.
Article in English | MEDLINE | ID: mdl-19325914

ABSTRACT

Macrophage migration inhibitory factor (MIF), an inflammatory cytokine, and its receptor CD74 are upregulated by bladder inflammation. MIF-mediated signal transduction involves binding to cell-surface CD74, this study documents, in vivo, MIF-CD74 interactions at the urothelial cell surface. N-hydroxysulfosuccinimide biotin ester-labeled surface urothelial proteins in rats treated either with saline or substance P (SP, 40 microg/kg). The bladder was examined by histology and confocal microscopy. Biotinylated proteins were purified by avidin agarose, immunoprecipitated with anti-MIF or anti-CD74 antibodies, and detected with strepavidin-HRP. Only superficial urothelial cells were biotinylated. These cells contained a biotinylated MIF/CD74 cell-surface complex that was increased in SP-treated animals. SP treatment increased MIF and CD74 mRNA in urothelial cells. Our data indicate that intraluminal MIF, released from urothelial cells as a consequence of SP treatment, interacts with urothelial cell-surface CD74. These results document that our previously described MIF-CD74 interaction occurs at the urothelial cell surface.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Histocompatibility Antigens Class II/metabolism , Membrane Proteins/metabolism , Substance P/metabolism , Urothelium/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Biotinylation , Histocompatibility Antigens Class II/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Male , Membrane Proteins/genetics , Rats , Rats, Sprague-Dawley , Urothelium/cytology
7.
Am J Pathol ; 172(3): 839-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18276778

ABSTRACT

Intimal hyperplasia of autologous vein grafts is a critical problem affecting the long-term patency of many types of vascular reconstruction. Within intimal hyperplasia lesions, smooth muscle cells are a major component, playing an essential role in the pathological process. Given that bone marrow-derived cells may differentiate into smooth muscle cells in the neointima of injured arteries, we hypothesized that the bone marrow may serve as a source for some of the smooth muscle cells within intimal hyperplasia lesions of vein grafts. To test this hypothesis, we used an established mouse model for intimal hyperplasia in wild-type mice that had been transplanted with bone marrow from a green fluorescent protein (GFP+/+) transgenic mouse. High-resolution confocal microscopy analysis performed 2 and 8 weeks after grafting demonstrated expression of GFP in 5.4 +/- 0.8% and 11.9 +/- 2.3%, respectively, of smooth muscle cells within intimal hyperplasia lesions. By 16 weeks, GFP expression in smooth muscle cells was not detected by immunohistochemistry; however, real-time PCR revealed that 20.2 +/- 1.7% of the smooth muscle cells captured from the neointima lesion by laser capture microdissection at 16 weeks contained GFP DNA. Our results suggest that bone marrow-derived cells differentiated into smooth muscle cells within the intimal lesion and may provide a novel clinical approach for decreasing intimal hyperplasia in vein grafts.


Subject(s)
Bone Marrow Transplantation/physiology , Tunica Intima/pathology , Veins/transplantation , Animals , Cell Proliferation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyperplasia/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdissection , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Time Factors , Transplantation, Autologous , Treatment Outcome
8.
Am J Physiol Renal Physiol ; 294(3): F621-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18057185

ABSTRACT

Two classes of H pumps, H-K-ATPase and H-ATPase, contribute to luminal acidification and HCO(3) transport in the collecting duct (CD). At least two H-K-ATPase alpha-subunits are expressed in the CD: HKalpha(1) and HKalpha(2). Both exhibit K dependence but have different inhibitor sensitivities. The HKalpha(1) H-K-ATPase is Sch-28080 sensitive, whereas the pharmacological profile of the HKalpha(2) H-K-ATPase is not completely understood. The present study used a nonpharmacological, genetic approach to determine the contribution of HKalpha(1) and HKalpha(2) to cortical CD (CCD) intercalated cell (IC) proton transport in mice fed a normal diet. Intracellular pH (pH(i)) recovery was determined in ICs using in vitro microperfusion of CCD after an acute intracellular acid load in wild-type mice and mice of the same strain lacking expression of HKalpha(1), HKalpha(2), or both H-K-ATPases (HKalpha(1,2)). A-type and B-type ICs were differentiated by luminal loading with BCECF-AM and peritubular chloride removal from CO(2)/HCO(3)-buffered solutions to identify the membrane locations of Cl/HCO(3) exchange activity. H-ATPase- and Na/H exchange-mediated H transport were inhibited with bafilomycin A(1) (100 nM) and EIPA (10 microM), respectively. Here, we report 1) initial pH(i) and buffering capacity were not significantly altered in the ICs of HKalpha-deficient mice, 2) either HKalpha(1) or HKalpha(2) deficiency resulted in slower acid extrusion, and 3) A-type ICs from HKalpha(1,2)-deficient mice had significantly slower acid extrusion compared with A-type ICs from HKalpha(1)-deficient mice alone. These studies are the first nonpharmacological demonstration that both HKalpha(1) and HKalpha(2) contribute to H secretion in both A-type and B-type ICs in animals fed a normal diet.


Subject(s)
Acids/metabolism , H(+)-K(+)-Exchanging ATPase/deficiency , Kidney Tubules, Collecting/metabolism , Protons , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Diet , Female , Genotype , H(+)-K(+)-Exchanging ATPase/genetics , Hydrogen-Ion Concentration/drug effects , Kidney Tubules, Collecting/cytology , Macrolides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/metabolism , Proton-Translocating ATPases/antagonists & inhibitors
9.
Am J Physiol Renal Physiol ; 293(5): F1751-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17686958

ABSTRACT

This study determined whether nucleotides that bind to purinergic receptors (P2R) regulate the expression or function of serum- and glucocorticoid-inducible kinase-1 (SGK1) in mouse renal inner medullar collecting duct cells (mIMCD-3). The SGK1 protein was detected by Western blotting. A significant reduction of cytosolic SGK1 expression was observed in the cells pretreated with P2R agonist adenosine 5'-O-(3-thiotriphosphate) (ATPgammaS), and the reduction could be reversed by P2R antagonists. This reduction was also observed in cells that were pretreated with agonists for P2R subtypes. Using ELISA, we observed a reduced SGK1 kinase activity in ATPgammaS-pretreated cells. This effect was reversed by P2R antagonists. Furthermore, an increase of SGK1 kinase activity in aldosterone-pretreated cells was suppressed by ATPgammaS. These studies demonstrate for the first time that SGK1 can be downregulated by nucleotides in renal collecting duct epithelial cells, likely via the activation of P2R, and suggest that activation of renal purinergic signaling regulates a SGK1-dependent pathway that is known to modulate ion transport in the renal collecting duct.


Subject(s)
Down-Regulation/physiology , Immediate-Early Proteins/metabolism , Kidney Tubules, Collecting/metabolism , Nucleotides/physiology , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Aldosterone/pharmacology , Animals , Cell Line , Cytosol/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Medulla , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Mice , Nucleotides/pharmacology , Purinergic Antagonists , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Sulfonic Acids/pharmacology , Suramin/pharmacology
10.
Biochem Biophys Res Commun ; 359(3): 438-44, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17560948

ABSTRACT

We examined P2X receptor expression and distribution in the mouse collecting duct (CD) and their functional role in Ca(2+) signaling. Both P2X(1) and P2X(4) were detected by RT-PCR and Western blot. Immunohistochemistry demonstrated apical P2X(1) and P2X(4) immunoreactivity in principal cells in the outer medullary CD (OMCD) and inner medullary CD (IMCD). Luminal ATP induced an increase in Ca(2+) signaling in native medullary CD (MCD) as measured by fluorescence imaging. ATP also induced an increase in Ca(2+) signaling in MCD cells grown in primary culture but not in the presence of P2XR antagonist PPNDS. Short circuit current (I(sc)) measurement with mouse IMCD cells showed that P2XR agonist BzATP induced a larger I(sc) than did P2YR agonist UTP in the apical membrane. Our data reveal for the first time that P2X(1) and P2X(4) are cell-specific with prominent immunoreactivity in the apical area of MCD cells. The finding that P2XR blockade inhibits ATP-induced Ca(2+) signaling suggests that activation of P2XR is a key step in Ca(2+)-dependent purinergic signaling. The result that activation of P2XR produces large I(sc) indicates the necessity of P2XR in renal CD ion transport.


Subject(s)
Calcium Signaling , Kidney Tubules, Collecting/metabolism , Receptors, Purinergic P2/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation , Immunohistochemistry , Male , Mice , RNA, Messenger/genetics , Receptors, Purinergic P2/classification , Receptors, Purinergic P2/genetics
11.
Br J Pharmacol ; 148(5): 732-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16715118

ABSTRACT

1. Myristoylated pseudosubstrate of PKCzeta (mPS) - a synthetic myristoylated peptide with a sequence (13 amino acids) mimicking the endogenous PKCzeta pseudosubstrate region -- is considered a selective cell-permeable inhibitor of PKCzeta. We present strong evidence that in endothelial cells the action of mPS is not limited to inhibition of PKC activity and that myristoylation of certain peptides can activate eNOS (endothelial nitric oxide synthase) through Akt phosphorylation. 2. mPS at micromolar concentrations (1-10 microM) induced profound phosphorylation of eNOS, Akt, ERK 1/2, and p38 MAPK in cultured pulmonary artery endothelial cells (PAEC). The same changes were observed after treatment of PAEC with a myristoylated scrambled version of mPS (mScr), whereas a cell-permeable version of PKCzeta pseudosubstrate fused to the HIV-TAT membrane-translocating peptide did not induce analogous changes, suggesting that myristoylation confers new properties on the peptides consisting of activation of different signaling pathways in endothelial cells. 3. In addition to mPS and mScr, a number of other myristoylated peptides induced phosphorylation of eNOS suggesting that myristoylation of peptides can activate eNOS by mechanisms unrelated to inhibition of PKC. All active myristoylated peptides contained basic amino acids motif and were longer than six amino acids. 4. Activation of eNOS by myristoylated peptides was dependent on the PI3K/Akt pathway and the rise of intracellular calcium and was associated with an elevation of cGMP levels in PAEC and with relaxation of precontracted isolated pulmonary artery segments. 5. Myristoylated peptides can be considered a new class of activators of NO production in endothelial cells and that using mPS as a specific inhibitor of PKC should be done with caution, especially in endothelial cells.


Subject(s)
Endothelial Cells/drug effects , Fatty Acids, Monounsaturated/chemistry , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , 3T3-L1 Cells , Animals , Calcium/physiology , Cells, Cultured , Cyclic GMP/biosynthesis , Humans , Isoenzymes/chemistry , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Organ Specificity/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation/drug effects , Placebos/pharmacology , Protein Kinase C/chemistry , Protein Kinase C/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Swine , Vasodilation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Vascul Pharmacol ; 43(5): 336-45, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16257270

ABSTRACT

In addition to its role as a vasoconstrictor, angiotensin II also acts as a potent growth factor by activating several tyrosine kinases, including Jak2. Interestingly, Jak2 has been linked to similar cardiovascular pathologies as have been previously linked to the renin-angiotensin system. Identifying the downstream targets of Jak2 via the AT(1) receptor may therefore elucidate its role in the progression of various pathologies. Previously, microarray analysis from our laboratory identified the Type 1 inositol 1,4,5 trisphosphate (IP(3)) receptor as a potential target of Jak2 following chronic stimulation by angiotensin II. Therefore, we hypothesized that Jak2 regulates IP(3) receptor expression in response to angiotensin II. To test this hypothesis, rat aortic smooth muscle (RASM) cells over-expressing a dominant negative (DN) Jak2 protein were used. The Jak2-dependent signaling in these cells is reduced approximately 90% when compared to RASM control cells. Analysis of protein expression showed that the IP(3) receptor was degraded approximately 2-fold (P<0.05) in cells lacking functional Jak2 within 1 h of treatment by angiotensin II. Notably, degradation of the IP(3) receptor was reversible since protein levels were restored to normal following 2 h of recovery from angiotensin II. To eliminate the possibility of clonal artifact in the DN cells, wild type RASM cells were treated with the Jak2 pharmacological inhibitor, AG490. We found that angiotensin II treatment degraded IP(3) receptor in AG490-treated cells, but not in the vehicle controls. Treatment with lactacystin, a proteasome inhibitor, completely blocked angiotensin II-mediated degradation of IP(3) receptor, thereby suggesting that the degradation occurs through a proteasome-dependent mechanism. Moreover, the degradation of IP(3) receptor in DN cells correlated with a significant loss of intracellular calcium mobilization when treated with angiotensin II (DN 27.4+/-1.1% vs. WT 42.2+/-4.7%; n=5, P=0.002). We next examined through what mechanism Jak2 regulates the IP(3) receptor. When wild type RASM cells were treated with PP2, an Src-family inhibitor, IP(3) receptor expression was markedly reduced. Since previous data show that Fyn, a downstream target of Jak2, is able to phosphorylate the IP(3) receptor at Tyr 353, we believe our data suggest that Jak2 prevents the angiotensin II-mediated IP(3) receptor degradation through the activation of Fyn. In conclusion, these data suggest that Jak2 has a protective role in maintaining IP(3) receptor expression, potentially through activation of Fyn and subsequent phosphorylation of the IP(3) receptor.


Subject(s)
Angiotensin II/antagonists & inhibitors , Calcium Channels/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Blotting, Western , Calcium/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Inositol 1,4,5-Trisphosphate Receptors , Janus Kinase 2 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Tyrphostins/pharmacology
13.
Genomics ; 85(3): 352-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15718102

ABSTRACT

We have cloned and characterized the gene encoding the porcine cationic amino acid transporter, member 1 (CAT-1) (HGMW-approved gene symbol SLC7A1) from porcine pulmonary artery endothelial cells. The porcine SLC7A1 encodes 629 deduced amino acid residues showing a higher degree of sequence similarity with the human counterpart (91.1%) than with the rat (87.3%) and mouse (87.6%) counterparts. Confocal microscopic examination of porcine CAT-1-GFP-expressing HEK293 cells revealed that porcine CAT-1 localizes on the plasma membrane. Amino acid uptake studies in Xenopus oocytes injected with cRNA encoding this protein demonstrated transport properties consistent with system y(+). Radiation hybrid mapping data indicate that the porcine SLC7A1 maps to the distal end of the short arm of pig chromosome 11 (SSC11). This map location is consistent with the known conservation of genome organization between human and pig and provides further confirmation that we have characterized the porcine orthologue of the human SLC7A1.


Subject(s)
Cationic Amino Acid Transporter 1/genetics , Chromosome Mapping , Swine/genetics , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , DNA Primers , Humans , Molecular Sequence Data , Nitric Oxide/biosynthesis , Xenopus laevis
14.
Am J Physiol Renal Physiol ; 287(2): F204-14, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15068972

ABSTRACT

Kidney tubules are targets for the activation of locally released nucleotides through multiple P2 receptor types. Activation of these P2 receptors modulates cellular Ca(2+) signaling and downstream cellular function. The purpose of this study was to determine whether P2 receptors were present in mIMCD-3 cells, a mouse inner medullary collecting duct cell line, and if so, to examine their link with intracellular Ca(2+) homeostasis. To monitor intracellular Ca(2+) concentration ([Ca(2+)](i)), experiments were conducted using the fluorescent dye fura 2. ATP (0.1-100 microM) produced a dose-dependent increase in [Ca(2+)](i) in a physiological Ca(2+)-containing solution, with an EC(50) of 2.5 microM. The P2-receptor antagonist PPADS reduced the effect of ATP on [Ca(2+)](i), and the P1-receptor agonist adenosine caused only a small increase in [Ca(2+)](i). Preincubation of cells with the phospholipase C antagonist U-73122 blocked the ATP-induced increase in [Ca(2+)](i), indicating P2Y receptors were involved in this process. In a Ca(2+)-free bath solution, thapsigargin and ATP induced intracellular Ca(2+) release from an identical pool. Nucleotides caused an increase in [Ca(2+)](i) in the potency order of UTP = ATP > ATP gamma S > ADP > UDP that is best fitted with the P2Y(2) subtype profile. Although the P2Y agonist UTP induced a similar large transient increase in [Ca(2+)](i) as did ATP, a small but sustained increase in [Ca(2+)](i) occurred only in ATP-stimulated cells, suggesting the role of P2X receptors in Ca(2+) influx. The sustained increase in [Ca(2+)](i) could be blocked by either nonselective cation channel blockers Gd(3+) or P2X antagonists PPADS and PPNDS. Furthermore, when either Gd(3+) or PPNDS was applied to the bath solution before ATP application, the ATP-induced increase in [Ca(2+)](i) was significantly reduced. Both RT-PCR and Western blotting corroborated the presence of P2X(1) and P2Y(2) receptors. These studies demonstrate that mIMCD-3 cells have both P2X and P2Y subtype receptors and that the activation of both P2X and P2Y receptors by extracellular ATP appears to be required to regulate intracellular Ca(2+) signaling.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Signaling , Extracellular Space/metabolism , Kidney Tubules, Collecting/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/pharmacology , Animals , Base Sequence , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Intracellular Membranes/metabolism , Kidney Medulla , Kidney Tubules, Collecting/cytology , Mice , Molecular Sequence Data , Osmolar Concentration , RNA, Messenger/metabolism , Receptors, Purinergic P2/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Biochemistry ; 41(41): 12457-66, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12369836

ABSTRACT

We have characterized single and double mutations in the M1-M2 segment of an inwardly rectifying K(+) channel, Kir2.1, using the cell-attached configuration of the patch-clamp technique. These mutations generated novel N-glycosylation sites at positions 128, 140, 143, and 147. Previously, we showed that these mutants were glycosylated, functional, and at the cell surface, which indicated that the putative pore-forming segment, including the invariant G(Y/F)G sequence of K(+) channels, was extracellular [Schwalbe, R. A., Rudin, A., Xia, S.-L., and Wingo, C. S. (2002) J. Biol. Chem. 277, 24382-24389]. In this study, three conductance states, corresponding to the main open state and two subconductance states, were identified in WT Kir2.1 channels expressed in infected Sf9 cells. Kir2.1 channels with mutations in the M1-M2 linker had at least one distinguishable conductance state of WT channels. In addition, these mutations altered the transitions, duration, and frequency of the defined populations of permeating and nonpermeating states. Of note, S128N had permeation rates similar to those of WT Kir2.1, but the total duration of the lower subconductance state was 3-5 times longer. Mutations in the signature sequence, I143N/Y145T, produced channels with permeation rates similar to those of the main open state and lower subconductance state of WT Kir2.1; however, the frequencies of these states were substantially different. These results demonstrate a novel functional role of the M1-M2 segment in regulating the transitions of the Kir2.1 channel and therefore suggest that this segment is a critical structural determinant in adjustments of pore conformations. Additionally, our results indicate that these mutants are correctly folded and thus further substantiate that the M1-M2 segment, including the G(Y/F)G sequence, is topologically extracellular.


Subject(s)
Ion Channel Gating/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Point Mutation , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Asparagine/genetics , Cysteine/genetics , Glycosylation , Isoleucine/genetics , Mice , Molecular Sequence Data , Patch-Clamp Techniques , Peptide Fragments/biosynthesis , Phenylalanine/genetics , Potassium Channels, Inwardly Rectifying/biosynthesis , Protein Conformation , Protein Structure, Tertiary/genetics , Serine/genetics , Spodoptera/genetics , Threonine/genetics , Time Factors , Tyrosine/genetics
17.
Am J Physiol Cell Physiol ; 283(4): C1080-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12225972

ABSTRACT

We investigated whether nitric oxide (NO) upregulates a cyclic nucleotide-gated (CNG) channel and whether this contributes to sustained elevation of intracellular calcium levels ([Ca(2+)](i)) in porcine pulmonary artery endothelial cells (PAEC). Exposure of PAEC to an NO donor, NOC-18 (1 mM), for 18 h increased the protein and mRNA levels of CNGA2 40 and 50%, respectively (P < 0.05). [Ca(2+)](i) in NO-treated cells was increased 50%, and this increase was maintained for up to 12 h after removal of NOC-18 from medium. Extracellular calcium is required for the increase in [Ca(2+)](i) in NO-treated cells. Thapsigargin induced a rapid cytosolic calcium rise, whereas both a CNG and a nonselective cation channel blocker caused a faster decline in [Ca(2+)](i), suggesting that capacitive calcium entry contributes to the elevated calcium levels. Antisense inhibition of CNGA2 expression attenuated the NO-induced increases in CNGA2 expression and [Ca(2+)](i) and in capacitive calcium entry. Our results demonstrate that exogenous NO upregulates CNGA2 expression and that this is associated with elevated [Ca(2+)](i) and capacitive calcium entry in porcine PAEC.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/metabolism , Ion Channels/metabolism , Nitric Oxide/metabolism , Up-Regulation/physiology , Animals , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Green Fluorescent Proteins , Immunoblotting , Intracellular Fluid/metabolism , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Nitric Oxide Donors/pharmacology , Oligonucleotides, Antisense/pharmacology , Pulmonary Artery , RNA, Messenger/antagonists & inhibitors , Swine , Transfection , Up-Regulation/drug effects
18.
J Biol Chem ; 277(27): 24382-9, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-11991952

ABSTRACT

Inwardly rectifying K+ channels or Kirs are a large gene family and have been predicted to have two transmembrane segments, M1 and M2, intracellular N and C termini, and two extracellular loops, E1 and E2, separated by an intramembranous pore-forming segment, H5. H5 contains a stretch of eight residues that are similar in voltage-dependent K+ channels, Kvs, and this stretch is called the signature sequence of K+ channels. Because mutations in this sequence altered selectivity in Kvs, it has been designated as the selectivity filter. Previously, we used N-glycosylation substitution mutants to map the extracellular topology of a weak inwardly rectifying K+ channel, Kir1.1 or ROMK1, and found that the entire H5 segment was extracellular. We now report utilization of introduced N-glycosylation sites, NX(S/T), at positions Ser(128) in E1, and Gln(140), Ileu(143), and Phe(147) in the H5 sequence of a strong inwardly rectifying K+ channel, Kir2.1. Furthermore, we show that biotinylated channel proteins with N-linked oligosaccharides attached at positions 140 and 143 in the signature sequence are located at the cell surface. Mutant channels were functional as detected by whole-cell and single-channel recordings. Unlike Kir1.1, position Lys(117) was not occupied. We conclude that, for yet another K+ channel, the invariant G(Y/F)G sequence is extracellular rather than intramembranous.


Subject(s)
Potassium Channels, Inwardly Rectifying/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Glutamine , Glycosylation , Isoleucine , Molecular Sequence Data , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Phenylalanine , Polymerase Chain Reaction , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Recombinant Proteins/metabolism , Serine , Spodoptera , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...