Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cardiovasc Pharmacol ; 68(1): 1-10, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26657712

ABSTRACT

Besides the well-known renal effects of aldosterone, the hormone is now known to have direct vascular effects. Clinical observations underline substantial adverse effects of aldosterone on cardiovascular function. The source of systemic circulating aldosterone is the adrenal gland zona glomerulosa cells through stimulus-secretion coupling involving depolarization, opening of L- and T-type calcium channels and aldosterone synthase activation. Local formation and release in peripheral tissues such as perivascular fat is recognized. Where does aldosterone affect the vasculature? Mineralocorticoid receptors (MRs) are present in endothelial and vascular smooth muscle cells, and MR-independent pathways are also involved. The vascular effects of aldosterone are complex, both concentration and temporal and spatial aspects are relevant. The acute response includes vasodilation through endothelial nitric oxide formation and vasoconstrictor effects through endothelial-contracting cyclooxygenase-derived factors and a changed calcium handling. The response to aldosterone can change within the same blood vessels depending on the exposure time and status of the endothelium. Chronic responses involve changed levels of reactive oxygen radicals, endothelial Na-influx and smooth muscle calcium channel expression. Furthermore, perivascular cells for example mast cells have also been suggested to participate in the chronic response. Moreover, the vascular effect of aldosterone depends on the status of the endothelium which is likely the cause of the very different responses to aldosterone and MR treatment observed in human studies going from increased to decreased flow depending on whether the patient had prior cardiovascular disease with endothelial dysfunction or not. A preponderance of constrictor versus dilator responses to aldosterone could therefore be involved in the detrimental vascular actions of the hormone in the setting of endothelial dysfunction and contribute to explain the beneficial action of MR blockers on blood pressure and target organ injury.


Subject(s)
Aldosterone/metabolism , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction , Vasoconstriction , Vasodilation , Animals , Calcium Signaling , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Endothelium, Vascular/physiopathology , Humans , Mice, Transgenic , Mineralocorticoid Receptor Antagonists/therapeutic use , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Signal Transduction/drug effects , Time Factors , Vasoconstriction/drug effects , Vasodilation/drug effects
2.
Pflugers Arch ; 466(12): 2205-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24627154

ABSTRACT

Voltage-gated calcium channels are involved in the vascular excitation-contraction mechanism and regulation of arterial blood pressure. It was hypothesized that T-type channels promote formation of nitric oxide from the endothelium. The present experiments determine the involvement of T-type channels in depolarization-dependent dilatation of mesenteric arteries and blood pressure regulation in Cav3.1 knock-out mice. Nitric oxide-dependent vasodilatation following depolarization-mediated vasoconstriction was reduced significantly in mesenteric arteries from Cav3.1(-/-) compared to wild type mice. Four days of systemic infusion of a nitric oxide (NO)-synthase-inhibitor to conscious wild type elicited a significant increase in mean arterial blood pressure that was absent in Cav3.1(-/-) mice. Immunoprecipitation and immunofluorescence labeling showed co-localization of Cav3.1 and endothelial nitric oxide synthase (eNOS) in arteries from wild type mice. Nitric oxide release measured as DAF fluorescence and cGMP levels were significantly lower in depolarized Cav3.1(-/-) compared to wild type arteries. In summary, the absence of T-type Cav3.1 channels attenuates NO-dependent dilatation in mesenteric arteries in vitro, as well as the hypertension after L-NAME infusion in vivo. Furthermore, Cav3.1 channels cluster with eNOS and promote formation of nitric oxide by the endothelium. The present findings suggest that this mechanism is important for the systemic impact of NO on peripheral resistance.


Subject(s)
Blood Pressure , Calcium Channels, T-Type/metabolism , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Vasodilation , Animals , Calcium Channels, T-Type/genetics , Female , Male , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism
3.
Pflugers Arch ; 465(1): 25-37, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22733355

ABSTRACT

A major rate-limiting step in the renin-angiotensin-aldosterone system is the release of active renin from endocrine cells (juxtaglomerular (JG) cells) in the media layer of the afferent glomerular arterioles. The number and distribution of JG cells vary with age and the physiological level of stimulation; fetal life and chronic stimulation by extracellular volume contraction is associated with recruitment of renin-producing cells. Upon stimulation of renin release, labeled renin granules "disappear;" the number of granules decrease; cell membrane surface area increases in single cells, and release is quantal. Together, this indicates exocytosis as the predominant mode of release. JG cells release few percent of total renin content by physiological stimulation, and recruitment of renin cells is preferred to recruitment of granules during prolonged stimulation. Several endocrine and paracrine agonists, neurotransmitters, and cell swelling converge on the stimulatory cyclic AMP (cAMP) pathway. Renin secretion is attenuated in mice deficient in beta-adrenoceptors, prostaglandin E(2)-EP4 receptors, Gsα protein, and adenylyl cyclases 5 and 6. Phosphodiesterases (PDE) 3 and 4 degrade cAMP in JG cells, and PDE3 is inhibited by cyclic GMP (cGMP) and couples the cGMP pathway to the cAMP pathway. Cyclic AMP enhances K(+)-current in JG cells and is permissive for secretion by stabilizing membrane potential far from threshold that activates L-type voltage-gated calcium channels. Intracellular calcium paradoxically inhibits renin secretion likely through attenuated formation and enhanced degradation of cAMP; by activation of chloride currents and interaction with calcineurin. Connexin 40 is necessary for localization of JG cells in the vascular wall and for pressure- and macula densa-dependent suppression of renin release.


Subject(s)
Juxtaglomerular Apparatus/metabolism , Renin-Angiotensin System , Renin/metabolism , Animals , Cell Differentiation , Humans , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/physiology , Membrane Potentials , Secretory Pathway , Signal Transduction
4.
Am J Physiol Heart Circ Physiol ; 304(8): H1094-102, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23396453

ABSTRACT

In arterioles, aldosterone counteracts the rapid dilatation (recovery) following depolarization-induced contraction. The hypothesis was tested that this effect of aldosterone depends on cyclooxygenase (COX)-derived products and/or nitric oxide (NO) synthase (NOS) inhibition. Recovery of the response to high K(+) was observed in mesenteric arteries of wild-type and COX-2(-/-) mice but it was significantly diminished in preparations from endothelial NOS (eNOS)(-/-) mice. Aldosterone pretreatment inhibited recovery from wild-type and COX-2(-/-) mice. The NO donor sodium nitroprusside (SNP) restored recovery in arteries from eNOS(-/-) mice, and this was inhibited by aldosterone. Actinomycin-D abolished the effect of aldosterone, indicating a genomic effect. The effect was blocked by indomethacin and by the COX-1 inhibitor valeryl salicylate but not by NS-398 (10(-6) mol/l) or the TP-receptor antagonist S18886 (10(-7) mol/l). The effect of aldosterone on recovery in arteries from wild-type mice and the SNP-mediated dilatation in arteries from eNOS(-/-) mice was inhibited by the histamine H2 receptor antagonist cimetidine. RT-PCR showed expression of mast cell markers in mouse mesenteric arteries. The adventitia displayed granular cells positive for toluidine blue vital stain. Confocal microscopy of live mast cells showed loss of quinacrine fluorescence and swelling after aldosterone treatment, indicating degranulation. RT-PCR showed expression of mineralocorticoid receptors in mesenteric arteries and in isolated mast cells. These findings suggest that aldosterone inhibits recovery by stimulation of histamine release from mast cells along mesenteric arteries. The resulting activation of H2 receptors decreases the sensitivity to NO of vascular smooth muscle cells. Aldosterone may chronically affect vascular function through paracrine release of histamine.


Subject(s)
Aldosterone/pharmacology , Endothelium, Vascular/drug effects , Mast Cells/drug effects , Mesenteric Arteries/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Aldosterone/physiology , Animals , Arterioles/drug effects , Arterioles/physiology , Calcium/metabolism , Cimetidine/pharmacology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/physiology , Endothelium, Vascular/physiology , Female , Histamine/pharmacology , Histamine/physiology , Histamine H2 Antagonists/pharmacology , Male , Mast Cells/physiology , Membrane Proteins/physiology , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/drug effects , Microcirculation/physiology , Microscopy, Confocal , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/physiology , Receptors, Mineralocorticoid/physiology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Vasoconstriction/physiology , Vasodilation/physiology
5.
Clin Exp Pharmacol Physiol ; 39(1): 117-24, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21466573

ABSTRACT

1. Sodium retention in disease states characterized by proteinuria, such as nephrotic syndrome, pre-eclampsia and diabetic nephropathy, occurs through poorly understood mechanism(s). 2. In nephrotic syndrome, data from experimental and clinical studies show that the sodium retention originates in the renal cortical collecting duct and involves hyperactivity of the epithelial sodium channel (ENaC). 3. The stimulus for the increased ENaC activity does not appear to involve any of the classical sodium retaining mechanisms, such as the renin-angiotensin-aldosterone system, arginine vasopressin or the sympathetic nervous system. 4. Proteolytic processing of the extracellular domain of γENaC subunit has been shown to stimulate ENaC activity. 5. The serine protease plasmin was recently identified as an ENaC-activating protease in urine from human nephrotic patients and from the puromycin aminonucleoside (PAN) rat model of nephrotic syndrome. 6. This finding suggests that a defective glomerular filtration barrier allows filtration into the tubular fluid of substances that activate ENaC and enhance sodium reabsorption. This concept might be expanded to other disease states, such as pre-eclampsia and diabetic nephropathy, which are also characterized by proteinuria and sodium retention. 7. In this review, we will examine the evidence for a role of urinary serine protease activity in the development of sodium and water retention in diseases characterised by proteinuria with a focus on nephrotic syndrome.


Subject(s)
Fibrinolysin/metabolism , Natriuresis , Proteinuria/metabolism , Water-Electrolyte Imbalance/etiology , Animals , Epithelial Sodium Channel Agonists , Epithelial Sodium Channels/metabolism , Fibrinolysin/urine , Humans , Hypertension/etiology , Hypertension/physiopathology , Kidney Tubules/metabolism , Kidney Tubules/physiopathology , Nephrotic Syndrome/physiopathology , Plasminogen/metabolism , Proteinuria/etiology , Proteinuria/physiopathology , Proteinuria/urine , Sodium Chloride, Dietary/adverse effects , Urokinase-Type Plasminogen Activator/metabolism
6.
Kidney Int ; 79(4): 443-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21068717

ABSTRACT

Voltage-gated calcium channels are important for the regulation of renal blood flow and the glomerular filtration rate. Excitation-contraction coupling in afferent arterioles is known to require activation of these channels and we studied their role in the regulation of cortical efferent arteriolar tone. We used microdissected perfused mouse efferent arterioles and found a transient vasoconstriction in response to depolarization with potassium; an effect abolished by removal of extracellular calcium. The T-type voltage-gated calcium channel antagonists mibefradil and nickel blocked this potassium-induced constriction. Further, constriction by the thromboxane analogue U46619 was significantly inhibited by mibefradil at a concentration specific for T-type channels. Using PCR, we found that two channel subtypes, Ca(v)3.1 and Ca(v)3.2, were expressed in microdissected efferent arterioles. Ca(v)3.1 was found by immunocytochemistry to be located in mouse efferent arterioles, human pre- and postglomerular vasculature, and Ca(v)3.2 in rat glomerular arterioles. Inhibition of endothelial nitric oxide synthase by L-NAME or its deletion by gene knockout changed the potassium-elicited transient constriction to a sustained response. Low concentrations of nickel, an agent that blocks Ca(v)3.2, had a similar effect. Thus, T-type voltage-gated calcium channels are functionally important for depolarization-induced vasoconstriction and subsequent dilatation in mouse cortical efferent arterioles.


Subject(s)
Arterioles/physiology , Calcium Channels, T-Type/physiology , Kidney Glomerulus/blood supply , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Acetylcholine/pharmacology , Animals , Arterioles/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , Female , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mibefradil/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nickel/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Rats , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
7.
Curr Opin Nephrol Hypertens ; 20(5): 529-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670672

ABSTRACT

PURPOSE OF REVIEW: Activation of epithelial sodium channel (ENaC) by proteolysis appears to be relevant for day-to-day physiological regulation of channel activity in kidney and other epithelial tissues. Pathophysiogical, proteolytic activation of ENaC in kidney has been demonstrated in proteinuric disease. RECENT FINDINGS: A variation in sodium and potassium intake or plasma aldosterone changes the number of cleaved α and γ-ENaC subunits and is associated with changes in ENaC currents. The protease furin mediates intracellular cleavage, whereas the channel-activating protease prostasin (CAP-1), which is glycophosphatidylinositol-anchored to the apical cell surface, mediates important extracellular cleavage. Soluble protease activity is very low in urine under physiological conditions but rises in proteinuria. In nephrotic syndrome, the dominant soluble protease activity is plasmin, which is formed from filtered plasminogen via urokinase-type plasminogen activator. Plasmin activates ENaC directly at high concentrations and through prostasin at lower concentrations. SUMMARY: The discovery of serine protease-mediated activation of renal ENaC in physiological and pathophysiological conditions opens the way for new understanding of the pathogenesis of proteinuric sodium retention, which may involve plasmin and present several potential new drug targets.


Subject(s)
Epithelial Cells/enzymology , Epithelial Sodium Channels/metabolism , Ion Channel Gating , Kidney Tubules/enzymology , Peptide Hydrolases/metabolism , Aldosterone/metabolism , Animals , Humans , Ion Transport , Kidney Tubules/physiopathology , Potassium/metabolism , Proteinuria/enzymology , Proteinuria/physiopathology , Sodium/metabolism
8.
Kidney Int ; 77(2): 110-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19907416

ABSTRACT

To examine the role of the calcium/calmodulin-dependent phosphatase calcineurin in regulation of renin release, we assayed exocytosis using whole-cell patch clamp of single juxtaglomerular cells in culture. The calcineurin inhibitor, cyclosporine A (CsA), significantly increased juxtaglomerular cell membrane capacitance, an index of cell surface area and an established measure of exocytosis in single-cell assays. This effect was mimicked by intracellular delivery of a calcineurin inhibitory peptide, the calcium chelator ethylene glycol tetraacetic acid (EGTA), or the calmodulin inhibitor W-13. Simultaneous exposure to EGTA and CsA had no additive effect. The protein kinase A (PKA) blocker RpcAMPs had no effect on the CsA-induced increase in membrane capacitance. Intra- and extracellular application of tacrolimus did not alter membrane capacitance. A calmodulin antagonist (calmidazolium) and CsA, but not tacrolimus, significantly stimulated renin release from cultured juxtaglomerular cells. Juxtaglomerular cells expressed the calcineurin isoforms A-beta and A-gamma but not A-alpha. Plasma renin concentrations (PRCs) were not different in wild-type, calcineurin A-alpha, or A-beta knockout mice but increased after CsA treatment of the A-alpha knockout, while renin mRNA was suppressed. We conclude that calcineurin and calcium/calmodulin suppress exocytosis of renin from juxtaglomerular cells independent of PKA.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , Exocytosis , Juxtaglomerular Apparatus/metabolism , Renin/metabolism , Animals , Calcineurin/genetics , Calcium/antagonists & inhibitors , Calmodulin/antagonists & inhibitors , Cells, Cultured , Chelating Agents , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclosporine , Egtazic Acid , Electric Capacitance , Immunosuppressive Agents , Male , Mice , Patch-Clamp Techniques , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Renin/blood , Sulfonamides
9.
J Hepatol ; 52(5): 705-11, 2010 May.
Article in English | MEDLINE | ID: mdl-20347173

ABSTRACT

BACKGROUND & AIMS: Suppression of the hypothalamic-pituitary-adrenal axis occurs in cirrhosis and cholestasis and is associated with increased concentrations of bile acids. We investigated whether this was mediated through bile acids acting to impair steroid clearance by inhibiting glucocorticoid metabolism by 5beta-reductase. METHODS: The effect of bile acids on glucocorticoid metabolism was studied in vitro in hepatic subcellular fractions and hepatoma cells, allowing quantitation of the kinetics and transcript abundance of 5beta-reductase. Metabolism was subsequently examined in vivo in rats following dietary manipulation or bile duct ligation. Finally, glucocorticoid metabolism was assessed in humans with obstructive jaundice. RESULTS: In rat hepatic cytosol, chenodeoxycholic acid competitively inhibited 5beta-reductase (K(i) 9.19+/-0.40 microM) and reduced its transcript abundance (in H4iiE cells) and promoter activity (reporter system, HepG2 cells). In Wistar rats, dietary chenodeoxycholic acid (1% w/w chow) inhibited hepatic 5beta-reductase activity, reduced urinary excretion of 3alpha,5beta-tetrahydrocorticosterone and reduced adrenal weight. Conversely, a fat-free diet suppressed bile acid levels and increased hepatic 5beta-reductase activity, supplementation of the fat-free diet with CDCA reduced 5beta-reductase activity, and urinary 3alpha,5beta-reduced corticosterone. Cholestasis in rats suppressed hepatic 5beta-reductase activity and transcript abundance. In eight women with obstructive jaundice, relative urinary excretion of 3alpha,5beta-tetrahydrocortisol was significantly lower than in healthy controls. CONCLUSION: These data suggest a novel role for bile acids in inhibiting hepatic glucocorticoid clearance, of sufficient magnitude to suppress hypothalamic-pituitary-adrenal axis activity. Elevated hepatic bile acids may account for adrenal insufficiency in liver disease.


Subject(s)
Bile Acids and Salts/pharmacology , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/physiology , Jaundice, Obstructive/drug therapy , Pituitary-Adrenal System/physiology , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Base Sequence , Bile Acids and Salts/therapeutic use , Bile Ducts/physiology , Cytosol/enzymology , Female , Humans , Hydrocortisone/metabolism , Hypothalamo-Hypophyseal System/drug effects , Jaundice, Obstructive/metabolism , Jaundice, Obstructive/urine , Kinetics , Ligation , Liver/enzymology , Male , Pituitary-Adrenal System/drug effects , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Tetrahydrocortisol/urine , Transcription, Genetic/drug effects
10.
J Am Soc Nephrol ; 20(2): 299-310, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19073825

ABSTRACT

Proteinuria and increased renal reabsorption of NaCl characterize the nephrotic syndrome. Here, we show that protein-rich urine from nephrotic rats and from patients with nephrotic syndrome activate the epithelial sodium channel (ENaC) in cultured M-1 mouse collecting duct cells and in Xenopus laevis oocytes heterologously expressing ENaC. The activation depended on urinary serine protease activity. We identified plasmin as a urinary serine protease by matrix-assisted laser desorption/ionization time of-flight mass spectrometry. Purified plasmin activated ENaC currents, and inhibitors of plasmin abolished urinary protease activity and the ability to activate ENaC. In nephrotic syndrome, tubular urokinase-type plasminogen activator likely converts filtered plasminogen to plasmin. Consistent with this, the combined application of urokinase-type plasminogen activator and plasminogen stimulated amiloride-sensitive transepithelial sodium transport in M-1 cells and increased amiloride-sensitive whole-cell currents in Xenopus laevis oocytes heterologously expressing ENaC. Activation of ENaC by plasmin involved cleavage and release of an inhibitory peptide from the ENaC gamma subunit ectodomain. These data suggest that a defective glomerular filtration barrier allows passage of proteolytic enzymes that have the ability to activate ENaC.


Subject(s)
Epithelial Sodium Channels/metabolism , Fibrinolysin/urine , Nephrosis/urine , Amiloride/pharmacology , Animals , Humans , Kidney/metabolism , Mice , Oocytes/metabolism , Patch-Clamp Techniques , Peptide Hydrolases/metabolism , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Xenopus laevis
11.
J Am Soc Nephrol ; 20(10): 2154-61, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628672

ABSTRACT

The mechanism by which extracellular hypotonicity stimulates release of renin from juxtaglomerular (JG) cells is unknown. We hypothesized that osmotically induced renin release depends on water movement through aquaporin-1 (AQP1) water channels and subsequent prostanoid formation. We recorded membrane capacitance (C(m)) by whole-cell patch clamp in single JG cells as an index of exocytosis. Hypotonicity increased C(m) significantly and enhanced outward current. Indomethacin, PLA(2) inhibition, and an antagonist of prostaglandin transport impaired the C(m) and current responses to hypotonicity. Hypotonicity also increased exocytosis as determined by a decrease in single JG cell quinacrine fluorescence in an indomethacin-sensitive manner. In single JG cells from COX-2(-/ -) and AQP1(-/ -) mice, hypotonicity increased neither C(m) nor outward current, but 0.1-muM PGE(2) increased both in these cells. A reduction in osmolality enhanced cAMP accumulation in JG cells but not in renin-producing As4.1 cells; only the former had detectable AQP1 expression. Inhibition of protein kinase A blocked the hypotonicity-induced C(m) and current response in JG cells. Taken together, our results show that a 5 to 7% decrease in extracellular tonicity leads to AQP1-mediated water influx in JG cells, PLA(2)/COX-2-mediated prostaglandin-dependent formation of cAMP, and activation of PKA, which promotes exocytosis of renin.


Subject(s)
Aquaporin 1/physiology , Cyclooxygenase 2/physiology , Exocytosis , Juxtaglomerular Apparatus/metabolism , Renin/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Hypotonic Solutions , Juxtaglomerular Apparatus/cytology , Male , Mice , Mice, Inbred C57BL , Osmolar Concentration , Phospholipases A2/physiology , Rats , Rats, Sprague-Dawley
12.
Kidney Int ; 75(8): 793-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19165174

ABSTRACT

Adenosine can induce vasodilatation and vasoconstriction of the renal afferent arteriole of the mouse. We determined here its direct effect on efferent arterioles of mouse kidneys. Using isolated-perfused cortical efferent arterioles, we measured changes in luminal diameter in response to adenosine. Extraluminal application of adenosine and cyclohexyladenosine had no effect on the luminal diameter. When the vessels were constricted by the thromboxane mimetic U46619, application of adenosine and 5'-N-ethylcarboxamido-adenosine dilated the efferent arterioles in a dose-dependent manner. We also found that the adenosine-induced vasodilatation was inhibited by the A(2)-specific receptor blocker 3,7-dimethyl-1-propargylxanthine. In the presence of this inhibitor, adenosine failed to alter the basal vessel diameter of quiescent efferent arterioles. Using primer-specific polymerase chain reaction we found that the adenosine A(1), A(2a), A(2b), and A(3) receptors were expressed in microdissected mouse efferent arterioles. We conclude that adenosine dilates the efferent arteriole using the A(2) receptor subtype at concentrations compatible with activation of the A(2b) receptor.


Subject(s)
Kidney Cortex/blood supply , Receptors, Adenosine A2/physiology , Vasodilation , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Arterioles/chemistry , Dose-Response Relationship, Drug , In Vitro Techniques , Mice , Polymerase Chain Reaction , Receptor, Adenosine A1/analysis , Receptor, Adenosine A1/genetics , Receptor, Adenosine A2A/analysis , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2B/analysis , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A3/analysis , Receptor, Adenosine A3/genetics , Receptors, Adenosine A2/metabolism , Vasodilation/drug effects
13.
J Hypertens ; 27(1): 41-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19145766

ABSTRACT

OBJECTIVES: High-salt diet likely elevates blood pressure (BP), thus increasing the risk of cardiovascular events. We hypothesized that a high-salt diet plays a critical role in subjects whose renin-angiotensin systems cannot adjust to variable salt intake, rendering them more susceptible to atherosclerosis. METHODS: Apolipoprotein E-deficient (ApoE-/-) mice received standard or high-salt diet (8%) alone or in combination with fixed angiotensin II (Ang II) infusion (0.5 microg/kg per min). BP was measured using telemetry, and plaque burden was assessed in the thoracic aorta and innominate artery. We used urinary isoprostane as a marker for oxidative stress. RESULTS: Although high-salt diet per se did not affect plaque extension, high salt combined with Ang II increased plaque area significantly in both the aorta and the innominate artery as compared with Ang II or salt alone (P < 0.05 and P < 0.01, respectively). High-salt diet did not affect BP or isoprostane levels, whereas Ang II infusion increased both BP and isoprostane levels (P < 0.05 and P < 0.01, respectively). Although high-salt diet combined with Ang II did not amplify BP, salt in combination with Ang II increased isoprostane levels further (P < 0.001 vs. Ang II alone). Ang II increases macrophage content in lesions (P < 0.05), whereas salt likely increases collagen content. CONCLUSION: High-salt diet per se does not influence BP in ApoE-/- mice and is only moderately atherogenic. Possibly mediated via increased oxidative stress, a high-salt diet combined with fixed high Ang II levels accelerates atherogenesis synergistically, beyond the effect of BP.


Subject(s)
Angiotensin II/toxicity , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Sodium Chloride, Dietary/adverse effects , Animals , Aorta, Thoracic/pathology , Brachiocephalic Trunk/pathology , Female , Lipids/blood , Mice , Oxidative Stress , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 1/physiology
14.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1733-41, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19793956

ABSTRACT

Several pathophysiological conditions, including nephrotic syndrome, are characterized by increased renal activity of the epithelial Na(+) channel (ENaC). We recently identified plasmin in nephrotic urine as a stimulator of ENaC activity and undertook this study to investigate the mechanism by which plasmin stimulates ENaC activity. Cy3-labeled plasmin was found to bind to the surface of the mouse cortical collecting duct cell line, M-1. Binding depended on a glycosylphosphatidylinositol (GPI)-anchored protein. Biotin-label transfer showed that plasmin interacted with the GPI-anchored protein prostasin on M-1 cells and that plasmin cleaved prostasin. Prostasin activates ENaC by cleavage of the gamma-subunit, which releases an inhibitory peptide from the extracellular domain. Removal of GPI-anchored proteins from the M-1 cells with phosphatidylinositol-specific phospholipase C (PI-PLC) inhibited plasmin-stimulated ENaC current in monolayers of M-1 cells at low plasmin concentration (1-4 microg/ml). At a high plasmin concentration of 30 microg/ml, there was no difference between cell layers treated with or without PI-PLC. Knockdown of prostasin attenuated binding of plasmin to M1 cells and blocked plasmin-stimulated ENaC current in single M-1 cells, as measured by whole-cell patch clamp. In M-1 cells expressing heterologous FLAG-tagged prostasin, gammaENaC and prostasin were colocalized. A monoclonal antibody directed against the inhibitory peptide of gammaENaC produced specific immunofluorescence labeling of M-1 cells. Pretreatment with plasmin abolished labeling of M-1 cells in a prostasin-dependent way. We conclude that, at low concentrations, plasmin interacts with GPI-anchored prostasin, which leads to cleavage of the gamma-subunit and activation of ENaC, while at higher concentrations, plasmin directly activates ENaC.


Subject(s)
Epithelial Sodium Channels/metabolism , Fibrinolysin/metabolism , Ion Channel Gating , Kidney Tubules, Collecting/metabolism , Serine Endopeptidases/metabolism , Animals , Biotinylation , Carbocyanines/metabolism , Cell Line , Disease Models, Animal , Fluorescent Antibody Technique , Fluorescent Dyes/metabolism , Kidney Tubules, Collecting/cytology , Membrane Potentials , Mice , Nephrotic Syndrome/urine , Oligopeptides , Patch-Clamp Techniques , Peptides/metabolism , Phosphoinositide Phospholipase C/metabolism , Protein Binding , RNA Interference , Rats , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Time Factors
15.
Cardiovasc Res ; 78(1): 130-8, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18093985

ABSTRACT

AIMS: The present study examined the hypothesis that prostaglandin E2 (PGE2) through activation of prostaglandin E (EP) receptor contributes to endothelium-dependent contractions. METHODS AND RESULTS: Western blotting revealed that the protein expression of EP1 receptor was significantly down-regulated in the aorta of the spontaneously hypertensive rat (SHR), but there was no significant difference in the expression of EP2, EP4, and total EP3 receptors between preparations of Wistar Kyoto rats (WKY) and SHR. Isometric tension studies showed that low concentrations of PGE2 caused endothelium-dependent relaxations in WKY but not in aortas of the SHR. High concentrations of PGE2 evoked contractions predominately through the activation of thromboxane-prostanoid (TP) receptors in the WKY, but involves the dual activation EP and TP receptors in the SHR. SQ29,548, BAYu3405 and Terutroban (TP receptor antagonists), and AH6809 (non-selective EP receptor antagonist) abolished, while SC19220 (preferential EP1 receptor antagonist) did not inhibit endothelium-dependent contractions. Both SC19220 and AH6809 significantly inhibited contractions to U46619 (TP receptor agonist). CONCLUSION: The present study demonstrates that the contraction caused by PGE2 in the SHR aorta is dependent on the activation of EP1 and TP receptors, but that endothelium-dependent contractions do not require the former. Thus, PGE2 is unlikely to be an endothelium-derived contracting factor in this artery. The ability of AH6809 to inhibit endothelium-dependent contractions can be attributed to its partial antagonism at TP receptors. Nevertheless, the impairment of PGE2-mediated relaxation may contribute to endothelial dysfunction in the aorta of the SHR.


Subject(s)
Aorta, Thoracic/metabolism , Dinoprostone/metabolism , Endothelium, Vascular/metabolism , Hypertension/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Thromboxane/metabolism , Vasoconstriction , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic , Carbazoles/pharmacology , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Fatty Acids, Unsaturated , Hydrazines/pharmacology , Hypertension/physiopathology , Immunohistochemistry , Naphthalenes/pharmacology , Phenylephrine/pharmacology , Potassium Chloride/pharmacology , Propionates/pharmacology , Prostaglandin Antagonists/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Thromboxane/antagonists & inhibitors , Sulfonamides/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation , Xanthones/pharmacology
16.
Am J Hypertens ; 21(1): 111-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18091753

ABSTRACT

BACKGROUND: Nitric oxide deficiency is involved in the development of hypertension, but the mechanisms are currently unclear. This study was conducted to further elucidate the role of neuronal nitric oxide synthase (nNOS) in blood pressure regulation and renin release in relation to different sodium loads. METHODS: Blood pressure and heart rate were measured telemetrically and assessed during periods of physical activity and inactivity. Urinary solute excretion was measured by metabolism cages and plasma renin concentration (PRC) was determined by radioimmunoassay; all in nNOS knockout (nNOS(-/-)) and wild-type (nNOS(+/+)) mice after 10 days of low (0.01% NaCl) and high (4% NaCl) sodium diets. RESULTS: The resting heart rate was reduced in nNOS(-/-) mice, but the two genotypes had similar blood pressure during the low (nNOS(+/+) 104 +/- 2 mm Hg; nNOS(-/-) 103 +/- 2 mm Hg) and high (nNOS(+/+) 107 +/- 3 mm Hg; nNOS(-/-) 108 +/- 2 mm Hg) sodium diets. During the high sodium diet, PRC did not differ between the genotypes (nNOS(+/+) 743 +/- 115 10(-5) Goldblatt units; nNOS(-/-) 822 +/- 63 10(-5) Goldblatt units), but during the low sodium diet, nNOS(-/-) mice failed to increase PRC (nNOS(+/+) 2164 +/- 220 10(-5) Goldblatt units; nNOS(-/-) 907 +/- 101 10(-5) Goldblatt units) and renal renin mRNA. On the low sodium diet, nNOS(-/-) mice also showed increased urine flow rate and osmolar excretion, observations not made during a high sodium diet. CONCLUSIONS: Our results show that nNOS is necessary for stimulation of renin in response to sodium restriction. Furthermore, nNOS(-/-) mice are normotensive, and their blood pressure responds normally to an increased dietary sodium intake, indicating that nNOS deficiency does not cause salt-sensitive hypertension.


Subject(s)
Blood Pressure/drug effects , Kidney/drug effects , Nitric Oxide Synthase Type I/metabolism , Renin-Angiotensin System/drug effects , Renin/metabolism , Sodium Chloride, Dietary/administration & dosage , Aldosterone/blood , Animals , Dose-Response Relationship, Drug , Female , Genotype , Heart Rate/drug effects , Kidney/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/genetics , Phenotype , Potassium/blood , Potassium/urine , RNA, Messenger/metabolism , Renin/blood , Renin/genetics , Telemetry , Urination/drug effects
17.
Nephrol Dial Transplant ; 23(9): 2777-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18424460

ABSTRACT

BACKGROUND: Recent data indicate that aldosterone aggravates cyclosporin A (CsA)-induced nephrotoxicity. We examined whether the mineralocorticoid receptor (MR) blocker eplerenone (EPL) antagonized early deterioration of renal function and blood pressure (BP) increase in CsA-treated rats. METHODS: Male Sprague-Dawley rats received CsA (15 mg/kg/day i.p.) and/or EPL (100 mg/kg/day p.o.) for 21 days. After 2 weeks, arterial, venous and urinary bladder catheters were implanted and the rats were trained to accept a restraining device allowing arterial blood sampling and direct measurement of BP and renal function. BP was measured on-line in conscious rats. RESULTS: CsA significantly increased systolic BP: 139 +/- 4 versus 134 +/- 2 mmHg, reduced body weight gain: -5 +/- 6 versus 36 +/- 7 g, glomerular filtration rate (GFR): 1.02 +/- 0.16 versus 2.64 +/- 0.27 ml/min, renal blood flow (RBF): 5.3 +/- 2.4 versus 13.5 +/- 2.1 ml/min and lithium clearance (C(Li+)): 0.16 +/- 0.04 versus 0.26 +/- 0.07 ml/min compared to controls. These changes were prevented by simultaneous EPL treatment: systolic BP, 130 +/- 4 mmHg; weight gain, 53 +/- 7 g; GFR, 1.67 +/- 0.26 ml/min; RBF, 12.3 +/- 2.1 ml/min and C(Li+), 0.27 +/- 0.03 ml/min. Analysis of kidney morphology after the CsA treatment showed hyaline vacuolization in tubules and vascular depositions in arterioles; these changes were less pronounced after combination therapy. No significant changes were seen regarding haemoglobin, haematocrit, plasma renin and vasopressin, plasma and urinary sodium, potassium, or osmolality. CONCLUSIONS: MR blockade by EPL prevented short-term alterations in GFR, RBF and hypertension associated with CsA nephrotoxicity. We conclude that the aldosterone-MR pathway contributes markedly to the renal toxicity induced by this calcineurin inhibitor.


Subject(s)
Cyclosporine/adverse effects , Kidney/drug effects , Mineralocorticoid Receptor Antagonists/pharmacology , Receptors, Mineralocorticoid/drug effects , Spironolactone/analogs & derivatives , Animals , Blood Pressure/drug effects , Cyclosporine/blood , Disease Models, Animal , Eplerenone , Glomerular Filtration Rate/drug effects , Kidney/physiology , Kidney Function Tests , Male , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Spironolactone/pharmacology
18.
Pharmacol Ther ; 111(2): 495-507, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16413609

ABSTRACT

The mineralocorticoid receptor (MR) and the enzyme 11betahydroxysteroid dehydrogenase type 2, which confers aldosterone specificity to the MR, are present in endothelium and vascular smooth muscle. In several pathological conditions aldosterone promotes vascular damage by formation of reactive oxygen species. The effect of aldosterone on vascular function, however, is far from clear. By rapid non-genomic mechanisms aldosterone may cause calcium mobilization and vasoconstriction, or may stimulate nitric oxide formation through the PI-3 kinase/Akt pathway and thereby counteract vasoconstriction. Vasoconstrictor, vasodilator or no effects of aldosterone have been reported from studies on human forearm blood flow. Inhibition of MR with spironolactone improves endothelial function in patients with heart failure but worsens endothelial function in type 2 diabetic patients. The aim of the present review is to reconcile some of the apparently conflicting data. A key observation is that reactive oxygen and nitrogen species serve as physiological signaling molecules at low concentrations, while they initiate pathological processes at higher concentrations. The net effect of aldosterone, which stimulates ROS production, therefore depends on the ambient level of oxidative stress. Thus, in situations with low levels of oxidative stress aldosterone may promote vasodilatation, while at higher oxidative stress (high NaCl intake, pre-existing vascular pathological conditions, high oxygen tension in vitro) aldosterone is likely to be associated with vasoconstriction and oxidative damage, and in this setting inhibition of the MR is likely to be beneficial.


Subject(s)
Aldosterone , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Aldosterone/metabolism , Aldosterone/pharmacology , Animals , Arteries/drug effects , Arteries/physiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Humans , Muscle, Smooth, Vascular/physiology , Oxidation-Reduction , Oxidative Stress , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Sodium Chloride/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
19.
Cardiovasc Res ; 71(3): 557-65, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16831408

ABSTRACT

OBJECTIVE: Extracellular retention of PDGF-B has been proposed to play an important role in PDGF-B signalling. We used the PDGF-B retention motif knockout mouse (RetKO) to study the effects of retention motif deletion on development of micro- and macrovascular structure and function. METHODS: Passive and active properties of conduit vessels were studied using myograph techniques and histological examination. Capillary structure and function was studied using measurements of capillary density in skeletal muscle and by assessing aerobic physical performance in a treadmill setup. Cardiac function was assessed using echocardiography. RESULTS: Myograph experiments revealed an increased diameter and stiffness of the aorta in RetKO. Histological examination showed increased media collagen content and a decreased number of aortic wall layers, however with a similar number of vascular smooth muscle cells. This outward eutrophic remodelling of the aorta was accompanied by endothelial dysfunction. RetKO showed decreased capillary density in skeletal muscle and signs of a defective delivery of capillary oxygen to skeletal muscle, as shown by a decreased physical performance. In RetKO mice, echocardiography revealed an adaptive eccentric cardiac hypertrophy. CONCLUSION: We conclude that retention of PDGF-B during development is essential for a normal conduit vessel function in the adult mouse. Furthermore, PDGF-B retention is also necessary for the development of an adequate capillary density, and thereby for a normal oxygen delivery to skeletal muscle. The lack of primary effects on cardiac function supports the redundant role of PDGF-B in cardiac development.


Subject(s)
Aorta, Thoracic/physiopathology , Muscle, Skeletal/blood supply , Proto-Oncogene Proteins c-sis/physiology , Animals , Aorta, Thoracic/pathology , Blood Pressure , Capillaries/pathology , Capillaries/physiopathology , Cardiomegaly/diagnosis , Cardiomegaly/genetics , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Male , Mice , Mice, Knockout , Motor Activity , Myography , Oxygen Consumption , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/genetics , Renin/blood , Tunica Media/metabolism
20.
J Hypertens ; 24(10): 2001-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16957560

ABSTRACT

OBJECTIVE: Using an aortic constriction model in mice, we studied whether the increase in pressure or the activation of the renin-angiotensin system (RAS) and its main receptors is the main driving force for plaque progression. METHODS: Male ApoE mice underwent sham surgery or placement of a suprarenal silver clip around the aorta (AoC). Half the group was treated with the selective AT1 receptor antagonist losartan (30 mg/kg per day) for 4 weeks. RESULTS: Anesthetized mean arterial pressure (MAP) was increased in AoC mice compared to sham (106 +/- 3 versus 90 +/- 1 mmHg, P < 0.001). Losartan reduced MAP in sham mice (78 +/- 2 mmHg, P < 0.01) but not in AoC (AoC losartan 104 +/- 2 mmHg). Plasma renin concentration (PRC) was increased in AoC mice compared to sham [1.6 +/- 0.3 versus 0.8 +/- 0.2 milliGoldblatt units (mGU)/ml, P < 0.001]. Losartan treatment augmented this difference (18.7 +/- 3.7 versus 4.6 +/- 1.7 mGU/ml, P < 0.01). AT2 receptor mRNA expression was increased 5.8-fold by aortic constriction in thoracic aorta (P < 0.05) and the major site for expression of the AT2 receptor protein was within the plaques. The plaque area was increased in AoC mice compared to sham (0.61 +/- 0.09 versus 0.07 +/- 0.01%, P < 0.001); however, losartan did not alter plaque area. CONCLUSIONS: Our data do not support a role for the AT1 receptor in the progression of atherosclerosis in this model, since blockade with losartan did not alter plaque distribution. Furthermore, we found no support for the counteraction of atherogenesis by increased activity of the RAS acting on the AT2 receptor. Our data suggest that increased pressure is the main driving force for atherosclerosis in this model.


Subject(s)
Aortic Valve Stenosis/physiopathology , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Blood Pressure/physiology , Receptor, Angiotensin, Type 1/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aortic Valve Stenosis/metabolism , Atherosclerosis/metabolism , Blood Pressure/drug effects , Disease Models, Animal , Losartan/pharmacology , Male , Mice , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL