Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-26185735

ABSTRACT

KKidney disease could result from hypertension and ischemia/hypoxia. Key mediators of cellular adaptation to hypoxia are oxygen-sensitive hypoxia inducible factor (HIF)s which are regulated by prolyl-4-hydroxylase domain (PHD)-containing dioxygenases. However, HIF activation can be protective as in ischemic death or promote renal fibrosis in chronic conditions. This study tested the hypothesis that increased HIF-1α consequent to reduced PHD expression contributes to the attendant hypertension and target organ damage in deoxycorticosterone acetate (DOCA)/salt hypertension and that PHD inhibition ameliorates this effect. In rats made hypertensive by DOCA/salt treatment (DOCA 50 mg/kg s/c; 1% NaCl orally), PHD inhibition with dimethyl oxallyl glycine (DMOG) markedly attenuated hypertension (P<0.05), proteinuria (P<0.05) and attendant tubular interstitial changes and glomerular damage (P<0.05). Accompanying these changes, DMOG blunted the increased expression of kidney injury molecule (KIM)-1 (P<0.05), a marker of tubular injury and reversed the decreased expression of nephrin (P<0.05), a marker of glomerular injury. DMOG also decreased collagen I staining (P<0.05), increased serum nitrite (P<0.05) and decreased serum 8-isopostane (P<0.05). However, the increased HIF-1α expression (P<0.01) and decreased PHD2 expression (P<0.05) in DOCA/salt hypertensive rats was not affected by DMOG. These data suggest that reduced PHD2 expression with consequent increase in HIF-1α expression probably results from hypoxia induced by DOCA/salt treatment with the continued hypoxia and reduced PHD2 expression evoking hypertensive renal injury and collagen deposition at later stages. Moreover, a PHD inhibitor exerted a protective effect in DOCA/salt hypertension by mechanisms involving increased nitric oxide production and reduced production of reactive oxygen species.

2.
J Hypertens ; 31(10): 2043-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811999

ABSTRACT

BACKGROUND: Nitric oxide just as prolyl hydroxylase domain-containing protein (PHD) is a regulator of hypoxia inducible factor-1 α (HIF-1α), a transcription factor complex that controls the expression of most genes involved in hypoxia and cardiovascular diseases. In the absence of nitric oxide, it is not clear how HIF-1α and PHD are regulated and to what extent they contribute to the ensuing disorder. METHOD: Using the nitric oxide withdrawal/high salt diet model of hypertensive renal injury, this study tested the hypothesis that removal of the inhibition by nitric oxide on PHD predisposes to increased PHD but reduced HIF-1α expression, hypertension and renal injury. RESULTS: In animals treated with N-nitro-L-arginine (L-NNA; 250 mg/l in drinking water for 14 days) and high salt diet (4% NaCl), there was hypertension (41±5%, P<0.05), proteinuria (three-fold, P<0.05), kidney (22±3%, P<0.05) and heart enlargement (24±3%, P<0.05), as well as increased renal osteopontin (21±3%, P<0.05) and collagen IV (24±4%, P<0.05) expression. Accompanying these effects were increased expression of PHD1 (24±4%, P<0.05) and PHD2 (36±4%, P<0.05) but reduced HIF-1α (35±6%, P < 0.05) expression. Dimethyloxallyl glycine (5mg/kg), a PHD inhibitor, paradoxically exacerbated hypertension (46±7%, P<0.05), proteinuria (two-fold, P <0.05), and increased osteopontin (15±2%, P<0.05) and HIF-1α (31±5%, P<0.05) expression with no change in PHD1/2 expression or kidney and heart enlargement. CONCLUSION: These data suggest that the protective effect of physiological levels of nitric oxide may be by virtue of inhibition of PHD or increased HIF-1α expression, hence, the pathological changes produced following its withdrawal was accompanied by increased PHD or decreased HIF-1α expression. Exacerbation of hypertension and renal injury following PHD inhibition suggests a deleterious effect in the chronic setting and challenges the dogma that inhibition of PHD is useful in cardiovascular diseases.


Subject(s)
Hypertension/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Nitric Oxide/physiology , Sodium Chloride, Dietary/adverse effects , Animals , Blood Pressure , Collagen Type IV/metabolism , Female , Gene Expression Regulation , Hypertension/complications , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Kidney Diseases/complications , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Male , Myocardium/pathology , Nitric Oxide Synthase/antagonists & inhibitors , Osteopontin/metabolism , Oxidative Stress , Protein Structure, Tertiary , Proteinuria/complications , Rats , Rats, Sprague-Dawley
3.
J Exp Pharmacol ; 5: 1-13, 2013.
Article in English | MEDLINE | ID: mdl-27186132

ABSTRACT

Cross-talk between insulin and the renin angiotensin system signaling system shows that angiotensin 2 (A2) negatively modulates insulin signaling by stimulating multiple serine phosphorylation events in the early stages of the insulin-signaling cascade; however, the biological actions of A2 on insulin sensitivity remain controversial. Preservation of glucose transporter 4 (GLUT4) expression during hypertension has been shown to prevent the increased vascular reactivity associated with hypertension. This study tested the hypothesis that GLUT4 contributes to the renal actions of A2. In the euvolemic anesthetized rat, acute infusion of the GLUT4 antagonist, indinavir (1 mg/kg/minute), enhanced an A2-induced increase in mean arterial blood pressure (MABP) (P < 0.01), but attenuated an A2-induced increase in medullary blood flow (MBF) and glomerular filtration rate (P < 0.01). Insulin, a GLUT4 activator (20 mU/kg/minute and 40 mU/kg/minute), decreased basal MABP and urine volume (P < 0.05), but it increased MBF, and these effects were reversed and blunted by indinavir. Subchronic indinavir treatment (80 mg/kg/day orally for 15 days) did not affect A2-induced changes in MABP, cortical blood flow, and MBF, but significantly decreased basal MBF (P < 0.01) and global kidney perfusion (P < 0.05). We concluded that acute but not subchronic inhibition of GLUT4 alters A2-induced changes in systemic and renal hemodynamics by attenuating A2-induced increase in MBF and glomerular filtration rate.

4.
Exp Biol Med (Maywood) ; 235(11): 1365-74, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20943934

ABSTRACT

The production of 20-hydroxyeicosatetraenoic acid (20-HETE) in the kidney is thought to be involved in the control of renal vascular tone and tubular sodium and chloride reabsorption. Cytochrome (Cyp) P-450 enzymes of the Cyp4a family in the mouse, namely 4a10, -12 and 14, are involved in 20-HETE synthesis. Recent advances in the molecular genetics of the mouse have produced mice in which Cyp4a isoforms have been disrupted and the consequence of such an approach is examined. This study evaluated the effect of deletion of the Cyp4a14 gene on blood pressure, renal vascular responses and tubular function. When compared with the wild-type (WT) litter mates, systolic blood pressure was greater in Cyp4a14 null (KO) mice as were renal vascular responses to angiotensin II or phenyephrine, G protein-coupled receptor (GPCR) agonists, but not KCl, a non-GPCR agonist. Renal vascular responses to guanosine 5'-O-(gamma-thio)triphosphate, a non-hydrolyzable GTP analog, or NaF(4), an activator of G-proteins, were also enhanced. However, vasodilation to bradykinin or apocynin but not sodium nitroprusside was blunted in Cyp4a14 null (KO) kidneys. These changes in KO mice were accompanied by increased 20-HETE synthesis, reduced renal production of nitric oxide (NO), increased lipid hydroperoxides and increased apocynin-inhibitable vascular NADPH oxidase activity that was prevented by administration of NO synthase (NOS) inhibitor, suggesting endothelial nitric oxide synthase (eNOS) uncoupling. Cyp4a14 KO mice also exhibited a diminished capacity to excrete an acute sodium load (0.9% NaCl, 2.5 mL/kg). These data suggest that deletion of the Cyp4a gene conferred a prohypertensive status via mechanisms involving increased 20-HETE synthesis and eNOS uncoupling leading to increased oxidative stress, enhanced vasoconstriction but diminished vasodilation as well as a defect in the renal excretory capacity in Cyp4a14 KO mice. These mechanisms suggest that the Cyp4a14-deficient mouse may be a useful model for evaluation of NO/20-HETE interactions.


Subject(s)
Blood Pressure/genetics , Cytochrome P-450 Enzyme System/genetics , Kidney/physiology , Renal Circulation/genetics , Animals , Cytochrome P-450 Enzyme System/physiology , Cytochrome P450 Family 4 , Enzyme Inhibitors/pharmacology , Female , Gene Deletion , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/genetics , Hydroxyeicosatetraenoic Acids/physiology , Hypertension/genetics , In Vitro Techniques , Kidney/metabolism , Lipid Peroxides/metabolism , Male , Mice , Mice, Knockout , Microsomes/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitrites/urine , Sodium Chloride/metabolism , Vasoconstriction/genetics
5.
Bratisl Lek Listy ; 111(5): 258-64, 2010.
Article in English | MEDLINE | ID: mdl-20568414

ABSTRACT

BACKGROUND: Peroxisome proliferator activator receptor alpha (PPAR alpha), a member of the nuclear receptor superfamily, is known to increase nitric oxide (NO) production and the mechanisms by which PPAR alpha activation alleviates vascular dysfunction may predicate its activation and possible expression. OBJECTIVES: We have evaluated the effects of acute clofibrate, a PPAR alpha ligand and the role of PKC on PPAR alpha expression and NO production in cultured cerebral microvascular endothelial cell (CMVEC). METHODS: Confluent CMVEC derived from pig brain were cultured and the role of PKC in acute clofibrate-induced PPAR alpha expression and NO production was determined in the presence or absence of PKC activator phorbol myristate acetate (PMA) or inhibitor (calphostin C). RESULTS: Incubation of CMVEC with clofibrate or PMA increased NO production by 40% or 27%, respectively, whereas co-incubation of cells with PMA and clofibrate had no effect on NO production. Incubation of cells with Calphostin C blunted PMA but not clofibrate-induced increase in NO production. L-NAME (0.1 mM), an inhibitor of NO synthase, reduced basal (47%; p<0.01) and abolished clofibrate-induced increase in NO production. Clofibrate increased PPAR alpha expression (26%; p<0.05) while PMA with or without clofibrate reduced PPAR alpha expression (p<0.01). On the other hand, calphostin C reduced basal (69%, ap<0.01) as well as clofibrate-induced increase (59%, p<0.01) in PPAR expression, and further reduced PMA-induced down regulation of PPAR expression. eNOS expression was not significantly affected by either clofibrate or PMA, alone or in combination. CONCLUSION: These results show that in the brain microvascular endothelial cell, PPAR alpha activation increases NO production-independent of eNOS and PKC signaling pathways, a regulates PPAR alpha expression through a complex PKC signaling mechanism(s) as both PKC activation and inhibition reduced clofibrate-induced activation of PPAR expression (Fig. 4, Ref. 32). Full Text (Free, PDF) www.bmj.sk.


Subject(s)
Cerebral Cortex/blood supply , Endothelium, Vascular/metabolism , Hypolipidemic Agents/pharmacology , PPAR alpha/metabolism , Animals , Cells, Cultured , Clofibrate/pharmacology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Microvessels/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Naphthalenes/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/pharmacology , Sus scrofa , Tetradecanoylphorbol Acetate/pharmacology
6.
Blood Press ; 17(1): 55-63, 2008.
Article in English | MEDLINE | ID: mdl-18568693

ABSTRACT

This study evaluated the role of PPARalpha in renal function and whether PPARalpha knockout (KO) mice are hypertensive or salt-sensitive. We hypothesize that PPARalpha modulation of ion transport defines the capacity for sodium excretion (U(Na)V). PPARalpha KO and wild-type (WT) mice were placed on a normal salt (NS, 0.5% NaCl) or high salt (8% NaCl, HS) diet for 28 days and mean arterial blood pressure (MABP) and heart rate (HR) determined. In a group of anesthetized animals on NS diet, pressure natriuresis (P/N) was determined and in another group, acute sodium load (0.9% NaCl) was administered and U(Na)V compared in mice pretreated with amiloride (200 microg/kg) or hydrochlorothiazide (3 mg/kg), in vivo measurements of sodium hydrogen exchanger or Na-Cl-cotransporter activity, respectively. MABP and HR were similar in PPARalpha KO and WT mice placed on a NS diet (116+/-6 mmHg, 587+/-40 beats/min, KO; 116+/-4 mmHg, 551+/-20 beats/min, WT). HS diet increased MABP to a greater extent in KO mice (Delta = 29+/-3 vs 14+/-3 mmHg, p<0.05) as did proteinuria (8- vs 2.5-fold, p<0.05). P/N was blunted in untreated KO mice. In response to an acute NaCl-load, U(Na)V was faster in PPARalpha KO mice (4.31+/-1.11 vs 0.77+/-0.31 micromol, p<0.05). However, U(Na)V was unchanged in hydrochlorothiazide-treated KO mice but increased 6.9-fold in WT mice. Similarly, U(Na)V was less in amiloride-treated KO mice (3.4- vs 15.5-fold). These data suggest that PPARalpha participates in pressure natriuresis and affects Na transport via amiloride- and thiazide-sensitive mechanisms. Thus, despite defective fatty acid oxidation, PPARalpha null mice are not hypertensive but develop salt-sensitive hypertension.


Subject(s)
Amiloride/therapeutic use , Hypertension, Renal/physiopathology , Natriuresis/drug effects , PPAR alpha/deficiency , Thiazides/therapeutic use , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Body Weight/drug effects , Diuretics/therapeutic use , Drug Resistance/genetics , Hypertension, Renal/drug therapy , Hypertension, Renal/genetics , Kidney Function Tests/methods , Male , Mice , Mice, Knockout , Natriuresis/genetics , Organ Size/drug effects , PPAR alpha/genetics , Sodium Chloride/administration & dosage , Sodium Chloride/urine , Sodium Chloride, Dietary
7.
Acta Pharmacol Sin ; 28(10): 1566-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17883941

ABSTRACT

AIM: We tested the hypothesis that bradykinin (BK)-induced relaxation of phenylephrine (PE) and endothelin-1 (ET-1) contractions can be differentially modulated by reactive oxygen species (ROS). METHODS: Aortic rings isolated from Sprague-Dawley rats were used for the study. The contribution of ROS to PE (1 x 10(-9)-1 x 10(-5) mol/L)- and ET-1 (1 x 10(-10)-1 x 10(-8) mol/L)-induced contractions and the influence of ROS in BK (1 x 10(-9)-1 x 10(-5) mol/L) relaxation of PE (1 x 10(-7) mol/L) or ET-1 (1 x 10(-9) mol/L)-induced tension was evaluated in the aorta in the presence or absence of the following antioxidants: catalase (CAT, 300 U/mL), superoxide dismutase (SOD, 300 U/mL), and vitamin C (1 x 10(-4) mol/L). RESULTS: Tension generated by ET-1 (1 x 10(-9) mol/L) or PE (1 x 10(-7) mol/L) was differentially relaxed by BK (1 x 10(-5) mol/L), producing a maximal relaxation of 75%+/-5% and 35+/-4%, respectively. The BK (1 x 10(-5) mol/L)-induced relaxation of PE (1 x 10(-7) mol/L) tension was significantly enhanced from 35%+/-4% (control) to 56%+/-9%, 60%+/-5%, and 49%+/-6% by SOD, CAT, and vitamin C, respectively (P<0.05, n=8). However, the relaxation of ET-1 (1 x 10(-9) mol/L) tension was significantly attenuated from 75%+/-5% (control) to 37%+/-9%, 63%+/-4%, and 39%+/-7% by SOD, CAT, and vitamin C, respectively (P<0.05, n=8). On the other hand, CAT had no effect on PE-induced tension, while SOD enhanced PE-induced tension (36%, P<0.05, n=10) and vitamin C attenuated (66%, P<0.05, n=8) the tension induced by PE. By contrast, SOD or vitamin C had no effect, but CAT attenuated (44%, P<0.05, n=9) the tension induced by ET-1. CONCLUSION: We have demonstrated that O2(-) and H2O2 differentially modulate BK relaxation in an agonist-specific manner. O2(-) attenuates BK-induced relaxation of PE contraction, but contributes to the relaxation of ET-1 contraction. O2(-) seems to inhibit PE contraction, while H2O2 contributes to ET-1-induced contraction. Thus, ROS differentially modulate vascular tone depending on the vasoactive agent that is used to generate the tone.


Subject(s)
Antioxidants/pharmacology , Bradykinin/pharmacology , Endothelin-1/pharmacology , Phenylephrine/pharmacology , Vasodilation/drug effects , Animals , Aorta/drug effects , Aorta/physiology , Ascorbic Acid/pharmacology , Catalase/pharmacology , In Vitro Techniques , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/pharmacology , Vasodilator Agents/pharmacology
8.
J Pharmacol Exp Ther ; 320(2): 774-81, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17105827

ABSTRACT

Elevated endothelin (ET)-1 has been implicated in cerebrovascular complications following brain trauma characterized by dysregulation of endothelial nitric oxide synthase (eNOS), protein kinase C (PKC), and cerebral function. Recently, vascular expression of PPARalpha has been observed and suggested to improve vascular dysfunction. We speculate that activation of PPARalpha in cerebral microvessels can improve cerebral dysfunction following trauma, and we tested the hypothesis that activation of cerebral endothelial peroxisome proliferator-activated receptor (PPAR)alpha will attenuate ET-1 production via a mechanism involving nitric oxide (NO) and PKC. Phorbol 12-myristate 13-acetate (PMA) (1 microM), bradykinin (BK, 1 microM), angiotensin II (AII, 1 microM), or hemoglobin (Hem, 10 microM) increased ET-1 levels by 24-, 11.4-, 3.6-, or 1.3-fold increasing ET-1 levels from 0.36 +/- 0.08 to 8.6 +/- 0.8, 4.1 +/- 0.7, 1.30 +/- 0.1, or 0.47 +/- 0.03 fmol/microg protein (p < 0.05), respectively. Clofibrate (10 microM) reduced basal ET-1 from 0.36 +/- 0.08 (control) to 0.03 +/- 0.01 and blunted vasoactive agent-induced increase to 0.12 +/- 0.07 (PMA), 0.6 +/- 0.04 (BK), 0.25 +/- 0.03 (AII), or 0.12 +/- 0.03 (Hem) fM/microg protein (p < 0.05). L-arginine methyl ester (100 microM) inhibited clofibrate-induced reduction in basal ET-1 production. Clofibrate increased PPARalpha expression, accompanied by increased NO production and eNOS expression. PKC inhibition by calphostin C (10 microM) blocked these effects, whereas activation by PMA reduced basal PPARalpha expression. Thus, PPARalpha activation attenuated ET-1 production by agents that mediate brain injury through mechanisms that probably result from PPARalpha-induced increase in eNOS expression/NO production and complex PKC signaling pathways. Therefore, PPARalpha activators can be appropriate therapeutic agents to alleviate cerebrovascular dysfunction following cerebral vasospasm.


Subject(s)
Brain/blood supply , Endothelial Cells/metabolism , Endothelin-1/biosynthesis , Nitric Oxide/physiology , PPAR alpha/physiology , Protein Kinase C/physiology , Signal Transduction/physiology , Animals , Clofibrate/pharmacology , Microcirculation , Nitric Oxide Synthase Type III/analysis , PPAR alpha/analysis , Swine
9.
Kidney Int ; 68(5): 2154-63, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16221214

ABSTRACT

BACKGROUND: Aging has considerable structural and functional effects on the vascular system of the kidney. One such effect is an alteration in vascular tone which potentially will initiate renal damage. Vascular tone is determined by the balance between vasoconstrictors and vasodilators. Therefore, we hypothesized that aging attenuates vasodilatory responses in the kidney. These changes may be mediated by a loss of nitric oxide and endothelial-derived hyperpolarizing factor (EDHF). METHODS: The systemic and renal responses of nitric oxide and EDHF were investigated in aging (18 months old) and young (3 months old) Sprague-Dawley rats. RESULTS: We demonstrated a general loss of vasodilatory responses in the aging kidney. In addition, nitric oxide levels were reduced in the serum and kidney cortex of aging versus young animals, although this was not accompanied with a loss of endothelial nitric oxide synthase (eNOS) protein in the kidney cortex. Aging animals also exhibited a loss in EDHF-mediated vasodilation following stimulation with either acetylcholine or bradykinin in the isolated perfused kidney. CONCLUSION: These findings indicate that not only a defect in the nitric oxide pathway, but also a loss of EDHF-mediated responses may be responsible for impaired vasodilation in the aging kidney. This may result in enhanced vasoconstrictive responses in aging which potentially will cause renal damage and ultimately a loss in glomerular filtration rate (GFR).


Subject(s)
Aging/metabolism , Biological Factors/metabolism , Kidney/physiology , Nitric Oxide/metabolism , Renal Circulation/physiology , Acetylcholine/pharmacology , Animals , Glomerular Filtration Rate , Kidney/blood supply , Male , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
10.
J Pharmacol Exp Ther ; 313(3): 1289-95, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15769866

ABSTRACT

We recently demonstrated that endothelin-1-induced medullary vasodilation despite a potent cortical vasoconstriction in the rat kidney may be accounted for by 20-hydroxyeicosatetraenoic acid (20-HETE) production. This study characterized the effects of 20-HETE and its metabolites, 20-hydroxy prostaglandin E(2) (20-OH PGE(2)) and 20-hydroxy prostaglandin F(2alpha) (20-OH PGF(2alpha)), and the contribution of nitric oxide (NO) and prostanoids to the changes evoked in cortical blood flow (CBF) and medullary blood flow (MBF). We tested the hypothesis that 20-HETE produces qualitatively different regional hemodynamic effects in the kidney with 20-OH PGF(2alpha) or 20-OH PGE(2), accounting for the vasoconstriction or vasodilation, respectively, in the cortex and medulla. Renal intra-arterial infusion of 1, 2.5, 5, and 10 ng/min 20-HETE decreased CBF by 10 +/- 3, 24 +/- 4, 40 +/- 7, and 58 +/- 9 perfusion units (PU), respectively, but increased MBF by 4 +/- 2, 16 +/- 4, 27 +/- 3, and 41 +/- 10 PU, respectively. 20-OH PGF(2alpha) mimics the effects of 20-HETE, as did PGF(2alpha). However, 20-OH PGE(2) increased both CBF and MBF, as did PGE(2). Indomethacin (5 mg/kg) blunted the effects of 20-HETE but not that of 20-OH PGE(2) and 20-OH PGF(2alpha). However, SQ29548 ([1S-[1alpha,2alpha(Z),3alpha,4alpha]]-7-[3[[2-[(phenylamino)carbonyl[hydrazino]methyl]-7-oxabicyclo]2.2.1]hept-2-yl]-5-heptenoic acid) (0.1 mg/kg), a prostaglandin H(2)/thromboxane A(2) receptor antagonist, blunted the cortical and medullary hemodynamic effects elicited by 20-HETE, 20-OH PGE(2), 20-OH PGF(2alpha), and PGF(2alpha) but not PGE(2). N(omega)-L-nitro arginine methyl ester (5 mg/kg), the inhibitor of NO synthase, exacerbated the cortical constrictor effects of 20-HETE and 20-OH PGF(2alpha) without affecting the medullary perfusion produced by 20-HETE or its metabolites. These findings suggest that 20-HETE, through its hydroxyl metabolites, produced differential effects in the kidney. The medullary perfusion appears to be independent of NO.


Subject(s)
Dinoprostone/analogs & derivatives , Hydroxyeicosatetraenoic Acids/pharmacology , Kidney Medulla/blood supply , Renal Circulation/drug effects , Animals , Bridged Bicyclo Compounds, Heterocyclic , Cerebrovascular Circulation/drug effects , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated , Hydrazines/pharmacology , Hydroxyeicosatetraenoic Acids/metabolism , Indomethacin/pharmacology , Male , Nitric Oxide/physiology , Rats , Rats, Sprague-Dawley , Vasoconstriction/drug effects
11.
Life Sci ; 75(24): 2921-32, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15454343

ABSTRACT

Development of vascular complications in diabetes has been linked to the quality of glucose regulation and characterized by endothelial dysfunction. The exact mechanism behind vascular complications in diabetes is poorly understood. However, alteration of nitric oxide (NO) biosynthesis or bioactivity is strongly implicated and the mechanism behind such alterations is still a subject for research investigations. In the present study, we tested the hypothesis that glucose-induced attenuation of vascular relaxation involves protein kinase C (PKC)-linked generation of free radicals. Vascular relaxation to acetylcholine (ACh; 10(-9)-10(-5) M), isoproterenol (10(-9)-10(-5) M), or NO donor, sodium nitropruside (SNP; 10(-9)-10(-6) M) was determined in phenylephrine (PE, 10(-7) M) pre-constricted aortic rings from Sprague-Dawley rats in the presence or absence of 30 mM glucose (30 min), L-nitro-arginine methyl ester (L-NAME; 10(-4) M for 15 min), a NO synthase inhibitor, or xanthine (10(-5) M), a free radical generator. ACh dose-dependently caused relaxation that was attenuated by L-NAME, glucose, or xanthine. Pre-incubation (15 min) of the rings with vitamin C (10(-4) M), an antioxidant or calphostin C (10(-6) M), a PKC inhibitor, restored the ACh responses. However, high glucose had no significant effects on SNP or isoproterenol-induced relaxation. ACh-induced NO production by aortic ring was significantly reduced by glucose or xanthine. The reduced NO production was restored by pretreatment with vitamin C or calphostin C in the presence of glucose, but not xanthine. These data demonstrate that oxidants or PKC contribute to glucose-induced attenuation of vasorelaxation which could be mediated via impaired endothelial NO production and bioavailability. Thus, pathogenesis of glucose-induced vasculopathy involves PKC-coupled generation of oxygen free radicals which inhibit NO production and selectively inhibit NO-dependent relaxation.


Subject(s)
Acetylcholine/pharmacology , Aorta/drug effects , Free Radicals/metabolism , Protein Kinase C/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Analysis of Variance , Animals , Ascorbic Acid/metabolism , Dose-Response Relationship, Drug , Glucose/metabolism , Isoproterenol/pharmacology , Male , NG-Nitroarginine Methyl Ester/metabolism , Naphthalenes/metabolism , Nitric Oxide/biosynthesis , Nitroprusside/pharmacology , Phenylephrine , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Xanthine/metabolism
12.
BMC Pharmacol ; 4: 1, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-15018640

ABSTRACT

BACKGROUND: Nitric oxide (NO) regulates renal proximal tubular (PT) Na+ handling through modulation of Na+-K+ ATPase. Peroxisome Proliferator Activated Receptor alpha (PPARalpha), a nuclear transcription factor, is expressed in PTs and has been reported to influence NO generation/activity in renal tissues. This study tested the hypothesis that PPARalpha interacts with NO and thereby affects renal tubular Na+ transport. Urinary excretion of nitrite (UNOXV) and Na+ (UNaV) and PT Na+ transport (Na+-K+ ATPase activity) were determined in rats treated with clofibrate (250 mg/kg i.p) or WY14643 (45 mg/kg; i.p.), a PPARalpha ligand, 2% NaCl (orally), clofibrate/NaCl, L-NAME, an inhibitor of NO production (100 mg/kg; orally), L-NAME/Clofibrate. RESULTS: Clofibrate or WY14643 increased PPARalpha expression by 106 +/- 7% (p < 0.05) and 113 +/- 8% (p < 0.05), respectively. Similarly, clofibrate and WY14643 increased expression of MCAD, a downstream target protein of PPARalpha by 123 +/- 8% (p < 0.05) and 143 +/- 8% (p < 0.05), respectively. L-NAME attenuated clofibrate-induced increase in PPARalpha expression by 27 +/- 2% (p < 0.05) but did not affect MCAD expression. UNOXV excretion increased 3-4 fold in rats treated with clofibrate, WY14643 or NaCl from 44 +/- 7 to 170 +/- 15, 144 +/- 18 or 132 +/- 11 nmol/24 hr, respectively (p < 0.05). Similarly, clofibrate, WY14643 or NaCl elicited a 2-5 fold increase in UNaV. L-NAME significantly reduced basal UNOXV and UNaV and abolished the clofibrate-induced increase. Clofibrate, WY14643, NaCl or clofibrate + NaCl treatment reduced Na+-K+-ATPase activity in the PT by 89 +/- 23, 62 +/- 10, 43 +/- 9 and 82 +/- 15% (p < 0.05), respectively. On the contrary, L-NAME or ODQ, inhibitor of sGC, abolished the inhibition of Na+-K+-ATPase activity by clofibrate (p < 0.05). Clofibrate either alone or with NaCl elicited approximately 2-fold increase in the expression of the alpha1 subunit of Na+-K+ ATPase in the PT while L-NAME abolished clofibrate-induced increase in Na+-K+ ATPase expression. CONCLUSION: These data suggest that PPARalpha activation, through increased NO generation promotes renal excretion of Na+ through reduced Na+-K+ ATPase activity in the PT probably via post translational modification of Na+-K+-ATPase.


Subject(s)
Kidney Tubules, Proximal/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sodium/metabolism , Transcription Factors/metabolism , Acyl-CoA Dehydrogenase/metabolism , Animals , Biological Transport , Clofibrate/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/urine , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/metabolism
13.
J Pharmacol Exp Ther ; 308(2): 636-43, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14600247

ABSTRACT

Carbon monoxide (CO), a product of heme metabolism by heme-oxygenase (HO), has biological actions similar to those of nitric oxide (NO). The role of CO in decreasing vascular responses to constrictor agents produced by experimental cirrhosis induced by carbon tetrachloride was evaluated before and after inhibition of HO with tin-mesoporphyrin (SnMP) in the perfused superior mesenteric vasculature (SMV) of cirrhotic and normal rats and in normal rats transfected with the human HO-1 (HHO-1) gene. Perfusion pressure and vasoconstrictor responses of the SMV to KCl, phenylephrine (PE), and endothelin-1 (ET-1) were decreased in cirrhotic rats. SnMP increased SMV perfusion pressure and restored the constrictor responses of the SMV to KCl, PE, and ET-1 in cirrhotic rats. The relative roles of NO and CO in producing hyporeactivity of the SMV to PE in cirrhotic rats were examined. Vasoconstrictor responses to PE were successively augmented by stepwise inhibition of CO and NO production, suggesting a complementary role for these gases in the regulation of reactivity of the SMV. Expression of constitutive but not of inducible HO (HO-1) was increased in the SMV of cirrhotic rats as was HO activity. Administration of adenovirus containing HHO-1 gene produced detection of HHO-1 RNA and increased HO activity in the SMV within 7 days. Rats transfected with HO-1 demonstrated reduction in both perfusion pressure and vasoconstrictor responses to PE in the SMV. We propose that HO is an essential component in mechanisms that modulate reactivity of the mesenteric circulation in experimental hepatic cirrhosis in rats.


Subject(s)
Heme Oxygenase (Decyclizing)/physiology , Liver Cirrhosis/physiopathology , Mesenteric Arteries/pathology , Splanchnic Circulation , Animals , Blood Pressure , Heme Oxygenase (Decyclizing)/metabolism , Liver Cirrhosis/enzymology , Male , Rats , Rats, Sprague-Dawley
14.
J Physiol ; 551(Pt 3): 971-9, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12857783

ABSTRACT

20-Hydroxyeicosatetraenoic acid (20-HETE), a major renal eicosanoid, regulates renal function and contributes to renal responses following withdrawal of nitric oxide (NO). However, the role of 20-HETE-synthesizing isoforms in renal function resulting from NO inhibition is unknown. The present study evaluated the role of cytochrome (CYP)4A1, -4A2 and -4A3 isoforms on renal function in the presence and absence of NO. Antisense oligonucleotides (ASODN) to CYP4A1, -4A2 and -4A3 reduced 20-HETE synthesis and downregulated the expression of CYP4A isoforms in renal microsomes. Nomega-L-nitromethyl arginine ester (L-NAME, 25 mg kg(-1)), an inhibitor of NO production, increased mean arterial blood pressure (MABP, Delta = +18 to 26 mmHg), reduced renal blood flow (RBF, Delta = -1.8 to 2.9 ml min(-1)), increased renal vascular resistance (RVR, Delta = +47 to 54 mmHg ml(-1) min(-1)), reduced glomerular filtration rate (GFR), but increased sodium excretion (UNaV). ASODN to CYP4A1 and -4A2 but not -4A3 reduced basal MABP and RVR and increased basal GFR, while ASODN to CYP4A2 significantly reduced basal UNaV suggesting a differential role for CYP4A isoforms in the regulation of renal function. ASODN to CYP4A2 but not -4A1 or -4A3 blunted the increase in MABP by L-NAME (38 +/- 9 %, P < 0.05). ASODN to CYP4A1, -4A2 and -4A3 attenuated the reduction in RBF and the consequent increase in RVR by L-NAME with a potency order of CYP4A2 = CYP4A1 > CYP4A3. ASODN to CYP4A1 and -4A2 but not -4A3 attenuated L-NAME-induced reduction in GFR, but ASODN to all three CYP4A isoforms blunted the L-NAME-induced increase in UNaV (CYP4A3 > CYP4A1 >> CYP4A2). We conclude from these data that CYP4A isoforms contribute to different extents to basal renal function. Moreover, CYP4A2 contributes greatest to haemodynamic responses while CYP4A3 contributes greatest to tubular responses following NO inhibition. We therefore propose that NO differentially regulates the function of CYP4A1, -4A2, and -4A3 isoforms in the renal vasculature and the nephron.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Kidney Glomerulus/enzymology , Kidney Tubules/enzymology , Nitric Oxide/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cytochrome P-450 CYP4A/metabolism , Cytochrome P450 Family 4 , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Kidney Glomerulus/blood supply , Kidney Tubules/blood supply , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Circulation/physiology
16.
J Pharmacol Exp Ther ; 304(2): 507-12, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538801

ABSTRACT

We recently demonstrated that U46619 (9-11-dideoxy-11 alpha,9a-epoxymethano-prostaglandin F(2a)) evoked a medullary vasodilation and a reduction in blood pressure despite a potent cortical vasoconstriction in the anesthetized rat. The present study tested the hypothesis that nitric oxide (NO) and prostanoids contribute to U46619-induced increase in medullary blood flow (MBF). U46619 at 1, 3, and 5 microg/kg increased MBF (above basal values) by 16 +/- 3, 45 +/- 10, and 58 +/- 8 perfusion units, respectively, and increased NO current in the medulla by 17 +/- 4, 34 +/- 7, and 60 +/- 12 pA, respectively. N(omega)-L-Nitro-arginine methyl ester (5 mg/kg), the inhibitor of NO production, attenuated the increase in MBF (75 +/- 8%, p < 0.05) as did indomethacin (10 mg/kg), the inhibitor of cyclooxygenase (38 +/- 5%, p < 0.05), suggesting the involvement of NO and dilator prostanoids. H-Arg-Lys-Arg-Ala-Arg-Lys-Glu-OH, a synthetic peptide and selective inhibitor of cGMP-dependent protein kinase, attenuated U46619-induced medullary perfusion (52 +/- 6%, p < 0.05), but H-89 ((N-[2-((p-bromocinnamyl)aminoethyl)]-5-isoquinolinesulfonamide hydrochloride), a cell-permeable, selective, and potent inhibitor of cAMP-dependent protein kinase A, was without effect. Glybenclamide, a K(ATP) channel blocker, also blunted the increase by U46619 in MBF (58 +/- 7%, p < 0.05). These data suggest that NO and prostanoids contribute to U46619-induced medullary perfusion and that the effects of these mediators are coupled to activation of protein kinase G and K(ATP) channels but not protein kinase A.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Kidney Medulla/drug effects , Nitric Oxide/biosynthesis , Signal Transduction/drug effects , Vasodilation/drug effects , Animals , Enzyme Inhibitors/pharmacology , Kidney Medulla/blood supply , Kidney Medulla/physiology , Male , Nitric Oxide/antagonists & inhibitors , Prostaglandins/biosynthesis , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Circulation/physiology , Signal Transduction/physiology , Vasodilation/physiology
17.
J Cardiovasc Pharmacol ; 39(6): 834-41, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12021577

ABSTRACT

Administration of glycerol produces acute renal failure (ARF) accompanied by profound vasoconstriction. It was hypothesized that impaired arachidonic acid metabolism may contribute to the vasoconstriction through alteration of renal eicosanoids or endothelin-1 or angiotensin II stimulation of renal oxygenases. Arachidonic acid (5, 10, 25 microg) in the control kidney produced increases in perfusion pressure of 15 +/- 9, 18 +/- 8, and 43 +/- 18 mm Hg, respectively. These responses were increased 1.5-fold in glycerol-induced renal failure (p < 0.01). Indomethacin (10 microM), the cyclooxygenase inhibitor, converted arachidonic acid vasoconstriction to epoxide-mediated vasodilator responses, which were unchanged in ARF. In ARF, 5,8,11,14-eicosatetraynoic acid (10 microM), the all-purpose inhibitor of arachidonic acid metabolism; indomethacin (10 microM), a cyclooxygenase inhibitor; 5,8,11-eicosatriyenoic acid (2.5 microM), the 5- and 12-lipoxygenase inhibitor; or aminobenzotriazole (50 mM), the cytochrome P-450 monooxygenase inhibitor, markedly attenuated arachidonic acid-induced vasoconstriction by 73 +/- 11% (p < 0.01), 89 +/- 1% (p < 0.01), 62 +/- 11% (p < 0.01), and 82 +/- 2% (p < 0.01), respectively. In ARF, angiotensin II-induced vasoconstriction was amplified by 67% (p < 0.01). Eicosatetraynoic acid, eicosatriyenoic acid, and aminobenzotriazole reduced these responses by 33 +/- 6% (p < 0.05), 53 +/- 6% (p < 0.01), and 52 +/- 11% (p < 0.05), respectively. Vasoconstriction by endothelin-1 was unchanged in ARF (24 +/- 17%). However, indomethacin attenuated endothelin-1 vasoconstriction by 41 +/- 11% (p < 0.05), whereas eicosatriyenoic acid and aminobenzotriazole were without effect. These data suggest that the increased renal vascular reactivity in ARF in response to arachidonic acid involves a relatively greater production of cyclooxygenase metabolites than monoxygenase- or lipoxygenase-derived eicosanoid metabolites. Furthermore, increased angiotensin II vasoconstriction is predominantly through lipoxygenase and monoxygenase metabolic pathways, whereas for endothelin-1, increased cyclooxygenase-derived vasoconstrictor metabolites play a significant role in its amplified vasoconstrictor effect in glycerol-induced ARF.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/enzymology , Glycerol/adverse effects , Kidney/enzymology , Oxygenases/metabolism , Animals , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Kidney/drug effects , Male , Rats , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Vasoconstriction/physiology
18.
Am J Physiol Renal Physiol ; 282(1): F144-50, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11739122

ABSTRACT

We examined the rat proximal tubule (PT) response to endothelin-1 (ET-1) in terms of 20-hydroxyeicosatetraenoic acid (HETE) dependency. Arachidonic acid (AA) (1 microM) decreased ouabain-sensitive (86)Rb uptake from 2.1 +/- 0.1 to 0.3 +/- 0.08 ng Rb. 10 microg protein(-1). 2 min(-1) (P < 0.05); 20-HETE (1 microM) had similar effects. Dibromododecenoic acid (DBDD) (2 microM), an inhibitor of omega-hydroxylase, abolished the inhibitory action of AA on (86)Rb uptake whereas the PT response to 20-HETE was unaffected. ET-1 at 0.1, 1, 10, and 100 nM reduced (86)Rb uptake from 2.8 +/- 0.3 in control PTs to 2.4 +/- 0.2, 1.7 +/- 0.1, 0.67 +/- 0.08, and 0.1 +/- 0.03 ng Rb. 10 microg protein(-1). 2 min(-1), respectively. DBDD (2 microM) abolished the inhibitory effect of ET-1 on (86)Rb uptake as did BMS182874 (1 microM), an ET(A)-selective receptor antagonist. ET-1 (100 nM) significantly increased PT 20-HETE release by approximately 50%, an effect prevented by DBDD. N(omega)-nitro-L-arginine-methyl ester (L-NAME), given for 4 days to inhibit nitric oxide synthase (NOS), increased arterial pressure from 92 +/- 12 to 140 +/- 8 mmHg and increased endogenous release of 20-HETE from isolated PTs (measured by gas chromatography/mass spectrometry). In L-NAME-treated PTs, but not in control PTs, 0.1 microM AA inhibited ouabain-sensitive (86)Rb uptake by > 40%; the response to AA was attenuated by DBDD. We conclude that, in the PTs, 1) 20-HETE is a second messenger for ET-1 and 2) conversion of AA to 20-HETE is augmented when NOS is inhibited.


Subject(s)
Arachidonic Acid/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Endothelin-1/metabolism , Kidney Tubules, Proximal/metabolism , Animals , Biological Transport/physiology , Endothelin Receptor Antagonists , Enzyme Inhibitors/pharmacology , Hydroxyeicosatetraenoic Acids/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptors, Endothelin/metabolism , Rubidium Radioisotopes/pharmacokinetics , Signal Transduction/drug effects , Signal Transduction/physiology
19.
European journal of pharmacology ; 277(2-3): 123-132, April 24, 1995. graf
Article in English | MedCarib | ID: med-17382

ABSTRACT

The isolated perfused kidney of the rat was used to examine the hypothesis that lipopolysaccharide-induced nitric oxide (NO) production inhibits cytochrome P450-dependent vasodilation. The vasodilator responses to arachidonic acid and bradykinin were examined as the response to arachidonic acid is wholly dependent, and that to bradykinin partly dependent on cytochrome P450 metabolism. In endotoxin-treated rats, the vasodilator response to arachidonic acid was inhibited, and those to bradykinin and acetylcholine were enhanced. Following treatment with phenobarbitone, the inducer of certain isoforms of cytochrome P450 enzymes, the vasodilator effects of all three agonists,especially that of arachidonic acid were amplified. Lipopolysaccharide inhibited the effect of phenobarbitone on the vasodilator effect of arachidonic acid and bradykinin but enhanced that of acetylcholine. The effect of lipopolysaccharide was antagonized by haemoglobin, a NO antagonist, and N (omega)- nitro-L-arginine, an inhibitor of NO synthase, suggesting that the inhibitory effect of lipopolysaccharide on arachidonic acid- and bradykinin-induced vasodilation was mediated by NO/NO synthase. N(omega)-Nitro-L-arginine enhanced vasodilation induced by arachidonic acid while that induced by bradykinin or acetylcholine was reduced, implying that endogenous NO inhibits vasodilator cytochrome P450 metabolites in the rat kidney. Pretreatment with dexamethasone, an inhibitor of inducible NO synthase, resulted in inhibition of the lipopolysaccharide modulation of arachidoni acid-induced vasodilation, suggesting that the inducible NO synthase is the target of the inhibitory effect of lipopolysaccharide. The inhibitory effect of lipopolysaccharide was mimicked by nitroprusside, the L-arginine-independent NO donor, and by L-arginine, the biosynthetic precusor of NO ...


Subject(s)
Rats , Lipopolysaccharides , Cytochrome P-450 Enzyme System , Arachidonic Acid , Nitric Oxide , Trinidad and Tobago
SELECTION OF CITATIONS
SEARCH DETAIL
...