Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 293(5): F1539-44, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17715260

ABSTRACT

We have demonstrated that oral contraceptive (OC) users exhibit elevated angiotensin II levels and angiotensin II type 1 receptor expression, indicative of renin-angiotensin system (RAS) activation, yet the renal and systemic consequences are minimal, suggesting that there is increased vasodilatory activity, counteracting the effect of RAS activation. We hypothesized that the nitric oxide (NO) system would be upregulated in OC users and that this would be reflected by a blunted hemodynamic response to l-arginine infusion. All subjects were studied after a 7-day controlled sodium and protein diet. Inulin and para-aminohippurate clearance techniques were used to assess renal function. l-Arginine was infused at 100, 250, and 500 mg/kg, each over 30 min. Skin endothelial NO synthase mRNA expression was assessed by real-time PCR. While OC nonusers exhibited significant increases in effective renal plasma flow (670.8 +/- 35.6 to 816.2 +/- 59.7 ml.min(-1).1.73 m(-2)) and glomerular filtration rate (133.4 +/- 4.3 to 151.0 +/- 5.7 ml.min(-1).1.73 m(-2), P = 0.04) and declines in renal vascular resistance (81.1 +/- 6.1 to 63.5 +/- 6.2 mmHg.ml(-1).min, P = 0.001) at the lower l-arginine infusion rates, the responses in OC users were blunted. While l-arginine reduced mean arterial pressure at the 250 and 500 mg/kg doses in OC nonusers, OC users only exhibited a decrease in mean arterial pressure at the highest infusion rate. In contrast, tissue endothelial NO synthase mRNA levels were higher in the OC users (P = 0.04). In summary, these findings suggest that the NO system is upregulated by OC use in young, healthy women. Increased activity of the NO pathway may modulate the hemodynamic effects of RAS activation in OC users.


Subject(s)
Contraceptives, Oral/pharmacology , Kidney/drug effects , Kidney/physiology , Nitric Oxide/metabolism , Up-Regulation/drug effects , Adult , Arginine/administration & dosage , Arginine/pharmacology , Blood Pressure/drug effects , Female , Glomerular Filtration Rate/drug effects , Hemodynamics/drug effects , Humans , Infusions, Intravenous , Inulin/pharmacokinetics , Nitric Oxide Synthase Type III/genetics , RNA, Messenger/metabolism , Renal Circulation/drug effects , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Skin/enzymology , Vascular Resistance/drug effects , p-Aminohippuric Acid/pharmacokinetics
2.
Kidney Int ; 69(12): 2212-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16672906

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is a homolog of ACE, which is not blocked by ACE inhibitors. High amounts of ACE2 are present in the proximal tubule, and ACE2 catalyzes generation of angiotensin 1-7 (Ang-(1-7)) by this segment. Ang-(1-7) binds to a receptor distinct from the AT1 or AT2 Ang II receptor, identified as the mas receptor. We studied the effects of Ang-(1-7) on Ang II-mediated cell signaling pathways in proximal tubule. In primary cultures of rat proximal tubular cells, activation of mitogen-activated protein kinases (MAPK) was detected by immunoblotting, in the presence or absence of agonists/antagonists. Transforming growth factor-beta1 (TGF-beta1) was measured by enzyme-linked immunosorbent assay. Ang II (5 min, 10(-7) M) stimulated phosphorylation of the three MAPK (p38, extracellular signal-related kinase (ERK 1/2), and c-Jun N-terminal kinase (JNK)). While incubation of proximal tubular cells with Ang-(1-7) alone did not significantly affect MAPK phosphorylation, Ang-(1-7) (10(-7) M) completely inhibited Ang II-stimulated phosphorylation of p38, ERK 1/2, and JNK. This inhibitory effect was reversed by the Ang-(1-7) receptor antagonist, D-Ala7-Ang-(1-7). Ang II significantly increased production of TGF-beta1 in proximal tubular cells, an effect that was partly inhibited by Ang-(1-7). Ang-(1-7) had no significant effect on cyclic 3',5'-adenosine monophosphate production in these cells. In summary, Ang-(1-7) inhibits Ang II-stimulated MAPK phosphorylation in proximal tubular cells. Generation of Ang-(1-7) by proximal tubular ACE2 could thereby serve a protective role by counteracting the effects of locally generated Ang II.


Subject(s)
Angiotensin II/pharmacology , Angiotensin I/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Peptide Fragments/pharmacology , Angiotensin I/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , JNK Mitogen-Activated Protein Kinases/analysis , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Tubules, Proximal/enzymology , Male , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/analysis , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/analysis , Peptidyl-Dipeptidase A/physiology , Phosphorylation/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Physiol Renal Physiol ; 281(2): F300-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457722

ABSTRACT

Angiotensin II (ANG II) subtype 2 (AT(2)) receptors are expressed in the adult kidney, but the effects of AT(2) receptor activation are unclear. The proximal tubule cell line LLC-PK(1) was transfected with a plasmid containing cDNA for the rat AT(2) receptor. In transfected cells, specific binding of (125)I-labeled ANG II was detected (dissociation constant = 0.81 nM), with inhibition by the AT(2) antagonist PD-123319, and no effect of the AT(1) antagonist losartan. ANG II (10(-7) M) significantly inhibited mitogen-activated protein kinase (MAPK) activity in transfected cells, associated with decreased phosphorylation of the extracellular signal-related kinases ERK1 and ERK2. ANG II stimulated phosphotyrosine phosphatase activity within 5 min, an effect blocked by PD-123319 and the phosphatase inhibitor vanadate. In transfected cells, ANG II inhibited epidermal growth factor-stimulated [(3)H]thymidine incorporation, an effect reversed by vanadate. In contrast, vanadate did not block ANG II-stimulated apoptosis of transfected cells. In summary, AT(2) receptors in proximal tubule cells inhibit MAPK activity and stimulate phosphotyrosine phosphatase. AT(2) receptor-induced inhibition of mitogenesis is mediated by phosphatase activation, whereas effects on apoptosis are insensitive to phosphatase inhibition. The data suggest that AT(2) receptors inhibit cell growth via distinct signaling pathways in the proximal tubule.


Subject(s)
Angiotensin II/pharmacology , Kidney Tubules, Proximal/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Angiotensin/metabolism , Animals , Antihypertensive Agents/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Division/drug effects , Cell Line , DNA/biosynthesis , Epidermal Growth Factor/metabolism , Imidazoles/pharmacology , Immunoblotting , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Losartan/pharmacology , Mitogen-Activated Protein Kinase 3 , Protein Tyrosine Phosphatases/metabolism , Pyridines/pharmacology , Radioligand Assay , Receptors, Angiotensin/genetics , Transfection , Vanadates/pharmacology
4.
Am J Physiol Renal Physiol ; 280(6): F980-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352837

ABSTRACT

Angiotensin IV (ANG IV), the COOH-terminal hexapeptide fragment of angiotensin II (ANG II), binds to specific sites in the kidney, distinct from type 1 (AT(1)) and type 2 (AT(2)) receptors and designated type 4 (AT(4)) receptors. We determined signaling pathways for ANG IV in a proximal tubular cell line, LLC-PK(1)/Cl(4). In these cells, we found no specific binding of [(125)I]-ANG II. In contrast, ANG IV dose dependently competed for [(125)I]-labeled ANG IV binding, with no displacement by either ANG II, the AT(1) receptor antagonist losartan, or the AT(2) antagonist PD-123319. Saturation binding indicated the presence of AT(4) receptors of high affinity [dissociation constant (K(d)) = 1.4 nM]. ANG IV did not affect cAMP or cGMP production and did not increase cytosolic calcium concentration in these cells. In contrast, immunoprecipitation and immunoblotting studies revealed that ANG IV caused dose-dependent tyrosine phosphorylation of p125-focal adhesion kinase (p125-FAK) and p68-paxillin within 2 min, with maximal stimulation at 30 min. ANG IV-stimulated tyrosine phosphorylation of p125-FAK and paxillin was not affected by pretreatment with either losartan or PD-123319, and ANG II (10(-7) M) did not induce protein tyrosine phosphorylation. Our results indicate that LLC-PK(1)/Cl(4) cells express ANG IV receptors, which we demonstrate for the first time are linked to tyrosine phosphorylation of focal adhesion-associated proteins. This suggests that ANG IV, a product of ANG II metabolism, may regulate function of the focal adhesion complex in proximal tubule cells.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Cytoskeletal Proteins/metabolism , Kidney Tubules, Proximal/enzymology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Binding, Competitive/physiology , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Focal Adhesion Protein-Tyrosine Kinases , Iodine Radioisotopes , Kidney Tubules, Proximal/cytology , LLC-PK1 Cells , Paxillin , Phosphorylation , Renin-Angiotensin System/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Swine , Thymidine/pharmacokinetics , Tritium , Tyrosine/metabolism
5.
Am J Physiol Renal Physiol ; 280(2): F254-65, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208601

ABSTRACT

The interaction of ANG II with intrarenal AT1 receptors has been implicated in the progression of diabetic nephropathy, but the role of intrarenal AT2 receptors is unknown. The present studies determined the effect of early diabetes on components of the glomerular renin-angiotensin system and on expression of kidney AT2 receptors. Three groups of rats were studied after 2 wk: 1) control (C), 2) streptozotocin (STZ)-induced diabetic (D), and 3) STZ-induced diabetic with insulin implant (D+I), to maintain normoglycemia. By competitive RT-PCR, early diabetes had no significant effect on glomerular mRNA expression for renin, angiotensinogen, or angiotensin-converting enzyme (ACE). In isolated glomeruli, nonglycosylated (41-kDa) AT1 receptor protein expression (AT1A and AT1B) was increased in D rats, with no change in glycosylated (53-kDa) AT1 receptor protein or in AT1 receptor mRNA. By contrast, STZ diabetes caused a significant decrease in glomerular AT2 receptor protein expression (47.0 +/- 6.5% of C; P < 0.001; n = 6), with partial reversal in D+I rats. In normal rat kidney, AT2 receptor immunostaining was localized to glomerular endothelial cells and tubular epithelial cells in the cortex, interstitial, and tubular cells in the outer medulla, and inner medullary collecting duct cells. STZ diabetes caused a significant decrease in AT2 receptor immunostaining in all kidney regions, an effect partially reversed in D+I rats. In summary, early diabetes has no effect on glomerular mRNA expression for renin, angiotensinogen, or ACE. AT2 receptors are present in glomeruli and are downregulated in early diabetes, as are all kidney AT2 receptors. Our data suggest that alterations in the balance of kidney AT1 and AT2 receptor expression may contribute to ANG II-mediated glomerular injury in progressive diabetic nephropathy.


Subject(s)
Angiotensins/metabolism , Diabetes Mellitus, Experimental/metabolism , Down-Regulation/physiology , Kidney Glomerulus/metabolism , Receptors, Angiotensin/metabolism , Renin/metabolism , Animals , Anti-Bacterial Agents , Diabetes Mellitus, Experimental/chemically induced , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Male , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Streptozocin
6.
Am J Physiol Renal Physiol ; 280(2): F266-77, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208602

ABSTRACT

The prostacyclin (IP) message was detected by RT-PCR in the renal cortex, outer (OM) and inner medulla (IM), and in freshly isolated (IMCD-f) and cultured inner medullary collecting duct (IMCD-c), and also the E-prostanoid (EP)1,3,4 receptor subtypes, but not EP2. Digoxigenin in situ hybridization localized IP mRNA in the tubules of the OM and IM, and the vasculature, and also in the glomeruli, arteries, and tubules of the cortex. IP splice variants or subtypes could not be detected by RT-PCR followed by TA cloning, though several nonfunctional point mutations or single base pair deletions were observed. Iloprost (ILP), cicaprost (CCP), PGE2, and arginine vasopressin (AVP) stimulated cAMP in both IMCD preparations. In addition, AVP-stimulated cAMP in IMCD-f was inhibited by all three prostanoids, but not in IMCD-c. Calcium experiments were performed on IMCD-c or microdissected IMCD (IMCD-m). CCP, ILP, and PGE2 did not alter intracellular calcium concentration ([Ca2+]i) in IMCD-c. However, on IMCD-m, both PGE2 and ILP increased [Ca2+]i levels equipotently and CCP had no effect. Pretreatment with the EP1 antagonist AH-6809 indicates that the response to ILP and PGE2 is mediated via EP1. These results suggest that IP receptors in the rat IMCD mediate the cAMP but not calcium signaling linked to PGI2; to date no subtypes or splice variants have been identified.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Kidney/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin/metabolism , Animals , Arginine Vasopressin/pharmacology , Kidney/drug effects , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Male , Prostaglandins/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Epoprostenol , Receptors, Prostaglandin/drug effects , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E, EP1 Subtype , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Kidney Int ; 58(6): 2320-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115066

ABSTRACT

BACKGROUND: Enhanced intrarenal angiotensin II (Ang II) activity may contribute to diabetic nephropathy. The proximal tubule is a proposed site of significant intrarenal Ang II production. We determined the effect of early diabetes on mRNA expression of components of the proximal tubule renin-angiotensin system. METHODS: Three groups of male Sprague-Dawley rats were studied after two weeks: (1) control (C), (2) streptozotocin-induced diabetes (STZ), and (3) STZ-induced diabetes, with normoglycemia maintained by insulin implants (STZ-I). Competitive reverse transcription-polymerase chain reaction was used to assay mRNA for renin, angiotensinogen, and angiotensin-converting enzyme in suspensions of proximal tubules; plasma and kidney levels of Ang II were measured by radioimmunoassay, and Western analysis of Ang II subtype 1 (AT1) receptors was performed. RESULTS: STZ rats tended to have increased plasma and intrarenal levels of Ang II compared with C and STZ-I rats. In proximal tubules, mRNA for renin was significantly increased in STZ rats, with reversal to control values in STZ-I rats (C, 2432 +/- 437 vs. STZ, 5688 +/- 890 fg/0.25 microg RNA, P < 0.05 vs. C, N = 9, vs. STZ-I, 1676 +/- 376 fg/0.25 microg RNA, P = NS vs. C). In STZ rats, the AT1 receptor antagonist losartan caused a further fivefold increase in proximal tubule renin mRNA, associated with proximal tubular renin immunostaining. STZ had no significant effect on mRNA expression for angiotensinogen or angiotensin-converting enzyme in proximal tubules. By Western blot analysis, cortical and proximal tubule AT1 receptor protein expression was significantly decreased in STZ rats. CONCLUSIONS: These data suggest activation of the proximal tubule renin-angiotensin system in early STZ diabetes, mediated at least partly by enhanced expression of renin mRNA. Increased local production of Ang II could contribute to tubulointerstitial injury in this model.


Subject(s)
Diabetic Nephropathies/physiopathology , Kidney Tubules, Proximal/physiopathology , Renin/genetics , Angiotensin II/analysis , Angiotensin II/blood , Angiotensin Receptor Antagonists , Angiotensinogen/genetics , Animals , Antihypertensive Agents/pharmacology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Gene Expression/physiology , Hypertrophy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/pathology , Losartan/pharmacology , Male , Nephritis, Interstitial/genetics , Nephritis, Interstitial/pathology , Nephritis, Interstitial/physiopathology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Renin/analysis , Renin-Angiotensin System/physiology
8.
Am J Physiol ; 275(1): F46-54, 1998 07.
Article in English | MEDLINE | ID: mdl-9689003

ABSTRACT

Nitric oxide (NO) derived from neuronal NO synthase (nNOS) in the kidney inner medulla has been implicated in the regulation of arterial blood pressure. The purpose of the present study was to determine the effect of high dietary NaCl on the expression of nNOS in the rat inner medullary collecting duct (IMCD). After 3 days or 3 wk of high (4.0%)-NaCl diet in rats, urinary NO-2/NO-3 excretion significantly increased. In freshly microdissected IMCD, nNOS was readily detected by immunofluorescence with polyclonal antibody, an effect that was completely blocked by neutralization of antibody with immunizing antigen. In rats fed a 4.0% NaCl diet for 3 days, IMCD nNOS mRNA, detected by RT-PCR, did not change from control values (0.3% NaCl, 19.84 +/- 1.57 x 10(3), vs. 4.0% NaCl, 20.44 +/- 3.14 x 10(3) cpm; P = not significant, n = 3). By Western blotting however, nNOS protein expression significantly increased (0.3% NaCl, 0.51 +/- 0.12, vs. 4.0% NaCl, 0.92 +/- 0.14 arbitrary units; P < 0. 05, n = 5). After 3 wk of 4.0% dietary NaCl, expression of nNOS mRNA and protein in IMCD did not differ significantly from control values. In contrast to these data, renal cortical expression of nNOS mRNA and protein was significantly decreased after 4.0% NaCl diet for 3 days. High dietary NaCl had no significant effect on expression of mRNA for inducible NO synthase (iNOS) in IMCD after either 3 days or 3 wk. In summary, our data indicate that nNOS mRNA and protein are expressed in IMCD and that high dietary NaCl differentially regulates nNOS expression in IMCD and cortex. The early increase in nNOS protein in IMCD may contribute to enhanced local production of NO and thereby represent an adaptive response to salt intake.


Subject(s)
Diet, Sodium-Restricted , Gene Expression Regulation, Enzymologic/physiology , Kidney Medulla/enzymology , Kidney Tubules, Collecting/enzymology , Nitric Oxide Synthase/biosynthesis , Sodium, Dietary/pharmacology , Animals , Cell Survival , DNA Primers , Gene Expression Regulation, Enzymologic/drug effects , Kidney Medulla/drug effects , Kidney Medulla/physiology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiology , Male , Nitrates/urine , Nitric Oxide Synthase Type I , Nitrites/urine , Polymerase Chain Reaction , Protein Biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Time Factors , Transcription, Genetic
9.
J Clin Invest ; 90(4): 1458-66, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1328300

ABSTRACT

Renal tubule solute and water transport is subject to regulation by numerous factors. To characterize direct effects of the recently discovered peptide endothelin (ET) on renal tubule transport, we determined signaling mechanisms for ET effects on vasopressin (AVP)-stimulated water permeability (PF) in rat terminal inner medullary collecting duct (IMCD) perfused in vitro. ET caused a rapid, dose-dependent, and reversible fall in AVP- but not cyclic AMP-stimulated PF, suggesting that its effect on PF is by inhibition of cyclic AMP accumulation. Indomethacin did not block ET actions, ruling out a role for prostaglandins in its effect. The protein kinase C (PKC) inhibitor calphostin, or pretreatment of perfused tubules with pertussis toxin, blocked ET-mediated inhibition of AVP-stimulated PF. ET caused a transient increase in intracellular calcium ([Ca2+]i) in perfused tubules, an effect unchanged in zero calcium bath or by PT pretreatment. ET effects on PF and [Ca2+]i desensitized rapidly. Inhibition of PF was transient and largely abolished by 20 min ET preexposure, and repeat exposure to ET did not alter [Ca2+]i. In contrast, PGE2-mediated inhibition of AVP-stimulated PF and increase of [Ca2+]i were sustained and unaltered by prior exposure of IMCD to ET. Thus desensitization to ET is homologous. We conclude that ET is a potent inhibitor of AVP-stimulated water permeability in rat terminal IMCD. Signaling pathways for its effects involve both an inhibitory guanine nucleotide-binding protein and phospholipase-mediated activation of PKC. Since ET is synthesized by IMCD cells, this peptide may be an important autocrine modulator of renal epithelial transport.


Subject(s)
Arginine Vasopressin/pharmacology , Body Water/metabolism , Endothelins/pharmacology , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Calcium/metabolism , Cyclic AMP/physiology , GTP-Binding Proteins/physiology , Kidney Tubules, Collecting/drug effects , Male , Permeability , Pertussis Toxin , Prostaglandins/physiology , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Virulence Factors, Bordetella/pharmacology
10.
Am J Physiol ; 262(2 Pt 2): F229-35, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1311524

ABSTRACT

To assess sites and mechanism of action of prostaglandin E2 (PGE2) on water permeability (PF), we determined PGE2 effects on antidiuretic hormone (ADH)- and adenosine 3',5'-cyclic monophosphate (cAMP)-stimulated PF in rat terminal inner medullary collecting ducts perfused in vitro. PGE2 (10(-7) M) reversibly inhibited ADH-stimulated PF (1.131 +/- 192 to 532 +/- 208 microns/s). In contrast to that observed in rabbit, PGE2 also inhibited an established PF response to the exogenous cAMP analogue 8-p-(chlorophenylthio)-cAMP (696 +/- 107 to 399 +/- 99 microns/s). PGE2 alone had no effect on PF. The protein kinase C inhibitor staurosporine (10(-8) M) blocked PGE2-mediated inhibition of cAMP-stimulated PF. PGE2 caused a rapid spikelike increase in intracellular calcium [( Ca2+]i) followed by a stable elevation above basal values. Only the latter effect was abolished in a zero calcium bath. Neither staurosporine nor cAMP altered the [Ca2+]i response. These studies are the first to demonstrate PGE2-mediated inhibition of an established PF response to cAMP independent of changes in intracellular cAMP. The pattern of [Ca2+]i release and sensitivity to staurosporine suggests that this effect is mediated via signaling through phospholipase C. The results underscore the importance of species differences, axial heterogeneity, and/or in vivo conditioning for functional expression of cellular signaling pathways.


Subject(s)
Cyclic AMP/metabolism , Dinoprostone/pharmacology , Kidney Tubules, Collecting/metabolism , Water/metabolism , Alkaloids/pharmacology , Animals , Calcium/metabolism , Cyclic AMP/pharmacology , Intracellular Membranes/metabolism , Kidney Medulla , Male , Permeability/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Inbred Strains , Staurosporine , Vasopressins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...