Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Metallomics ; 7(10): 1407-19, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26106847

ABSTRACT

The potentially lethal human pathogen Bacillus anthracis expresses a putative metal import system, MntBCA, which belongs to the large family of ABC transporters. MntBCA is essential for virulence of Bacillus anthracis: deletion of MntA, the system's substrate binding protein, yields a completely non-virulent strain. Here we determined the metal binding spectrum of MntA. In contrast to what can be inferred from growth complementation studies we find no evidence that MntA binds Fe(2+) or Fe(3+). Rather, MntA binds a variety of other metal ions, including Mn(2+), Zn(2+), Cd(2+), Co(2+), and Ni(2+) with affinities ranging from 10(-6) to 10(-8) M. Binding of Zn(2+) and Co(2+) have a pronounced thermo-stabilizing effect on MntA, with Mn(2+) having a milder effect. The thermodynamic stability of MntA, competition experiments, and metal binding and release experiments all suggest that Mn(2+) is the metal that is likely transported by MntBCA and is therefore the limiting factor for virulence of Bacillus anthracis. A homology-model of MntA shows a single, highly conserved metal binding site, with four residues that participate in metal coordination: two histidines, a glutamate, and an aspartate. The metals bind to this site in a mutually exclusive manner, yet surprisingly, mutational analysis shows that for proper coordination each metal requires a different subset of these four residues. ConSurf evolutionary analysis and structural comparison of MntA and its homologues suggest that substrate binding proteins (SBPs) of metal ions use a pair of highly conserved prolines to interact with their cognate ABC transporters. This proline pair is found exclusively in ABC import systems of metal ions.


Subject(s)
Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Proteins/metabolism , Metals/metabolism , Bacterial Proteins/chemistry , Cadmium/metabolism , Cobalt/metabolism , Manganese/metabolism , Nickel/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Virulence , Zinc/metabolism
2.
Proc Natl Acad Sci U S A ; 110(14): 5440-5, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23513215

ABSTRACT

In all kingdoms of life, ATP Binding Cassette (ABC) transporters participate in many physiological and pathological processes. Despite the diversity of their functions, they have been considered to operate by a largely conserved mechanism. One deviant is the vitamin B12 transporter BtuCD that has been shown to operate by a distinct mechanism. However, it is unknown if this deviation is an exotic example, perhaps arising from the nature of the transported moiety. Here we compared two ABC importers of identical substrate specificity (molybdate/tungstate), and find that their interactions with their substrate binding proteins are utterly different. One system forms a high-affinity, slow-dissociating complex that is destabilized by nucleotide and substrate binding. The other forms a low-affinity, transient complex that is stabilized by ligands. The results highlight significant mechanistic divergence among ABC transporters, even when they share the same substrate specificity. We propose that these differences are correlated with the different folds of the transmembrane domains of ABC transporters.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Escherichia coli Proteins/metabolism , Models, Molecular , Molybdenum/metabolism , Multiprotein Complexes/metabolism , Tungsten Compounds/metabolism , ATP-Binding Cassette Transporters/chemistry , Archaeoglobus fulgidus , Chromatography, Gel , Escherichia coli Proteins/chemistry , Haemophilus influenzae , Kinetics , Liposomes/metabolism , Multiprotein Complexes/chemistry , Periplasmic Binding Proteins/metabolism , Protein Folding , Protein Structure, Tertiary , Species Specificity , Substrate Specificity
3.
Proc Natl Acad Sci U S A ; 110(14): 5434-9, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23513227

ABSTRACT

In all kingdoms of life, ATP binding cassette (ABC) transporters are essential to many cellular functions. In this large superfamily of proteins, two catalytic sites hydrolyze ATP to power uphill substrate translocation. A central question in the field concerns the relationship between the two ATPase catalytic sites: Are the sites independent of one another? Are both needed for function? Do they function cooperatively? These issues have been resolved for type I ABC transporters but never for a type II ABC transporter. The many mechanistic differences between type I and type II ABC transporters raise the question whether in respect to ATP hydrolysis the two subtypes are similar or different. We have addressed this question by studying the Escherichia coli vitamin B12 type II ABC transporter BtuCD. We have constructed and purified a series of BtuCD variants where both, one, or none of the ATPase sites were rendered inactive by mutation. We find that, in a membrane environment, the ATPase sites of BtuCD are highly cooperative with a Hill coefficient of 2. We also find that, when one of the ATPase sites is inactive, ATP hydrolysis and vitamin B12 transport by BtuCD is reduced by 95%. These exact features are also shared by the archetypical type I maltose ABC transporter. Remarkably, mutants that have lost 95% of their ATPase and transport capabilities still retain the ability to fully use vitamin B12 in vivo. The results demonstrate that, despite the many differences between type I and type II ABC transporters, the fundamental mechanism of ATP hydrolysis remains conserved.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Escherichia coli Proteins/metabolism , Multiprotein Complexes/metabolism , ATP-Binding Cassette Transporters/genetics , Biological Transport, Active/genetics , Biological Transport, Active/physiology , Catalytic Domain/genetics , Chromatography, Gel , Escherichia coli , Escherichia coli Proteins/genetics , Hydrolysis , Multiprotein Complexes/genetics , Mutation/genetics , Vitamin B 12/metabolism
4.
Physiol Rep ; 1(6): e00155, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24400157

ABSTRACT

Xanthurenic acid 8-o-ß-d-glucoside is an endogenous derivative of tryptophan metabolism, isolated from urine of patients with chronic renal disease. This compound was suggested previously to act as a natriuretic hormone based on its ability to block short circuit currents in a frog skin assay and to induce a sustained natriuresis when injected into rats (C. D. Cain et al., Proc. Natl. Acad. Sci. USA 2007: 17873-17878). The present communication describes the effects of the compound on renal clearance and hemodynamic parameters in male Sprague-Dawley rats maintained on a normal salt (0.4-0.5%) diet. Intravenous administration of synthetic xanthurenic acid 8-o-ß-d-glucoside in two consecutive incremental doses (6.3 and 31.5 nmol) resulted in a significant increase (P < 0.05), in urine flow (43.91 ± 6.31 µL/min vs. 10.54 ± 2.21 µL/min), absolute rate of sodium excretion (3.99 ± 0.95 µEq/min vs. 1.15 ± µEq/min), and percentage sodium excretion (1.63 ± 0.46% vs. 0.37 ± 0.12%, peak response vs. baseline, respectively). The natriuretic/diuretic effect was associated also with a significant increase in potassium excretion. These effects were not related to changes in renal hemodynamics or in arterial blood pressure. Pretreatment with the sodium channel blocker, amiloride, completely abolished the natriuretic and kaluretic actions of the compound. Administration of the xanthurenic acid derivative caused a dose-related increase in urinary nitrite/nitrate excretion. Moreover, under chronic nitric oxide blockade by l-NG-Nitro-Arginine-Methyl-Esther (l-NAME) sodium excretion was similar in rats treated or untreated with the compound. Our data demonstrate that xanthurenic acid 8-o-ß-d-glucoside has significant diuretic/natriuretic and kaluretic properties. An intact amiloride-sensitive sodium channel is required for the renal effects of the compound. The data further suggest that the natriuretic effect is mediated in part by a nitric oxide-dependent mechanism.

5.
PLoS One ; 7(11): e47890, 2012.
Article in English | MEDLINE | ID: mdl-23166584

ABSTRACT

TVP1022, the S-enantiomer of rasagiline (Azilect®) (N-propargyl-1R-aminoindan), exerts cyto/cardio-protective effects in a variety of experimental cardiac and neuronal models. Previous studies have demonstrated that the protective activity of TVP1022 and other propargyl derivatives involve the activation of p42/44 mitogen-activated protein kinase (MAPK) signaling pathway. In the current study, we further investigated the molecular mechanism of action and signaling pathways of TVP1022 which may account for the cyto/cardio-protective efficacy of the drug. Using specific receptor binding and enzyme assays, we demonstrated that the imidazoline 1 and 2 binding sites (I(1) & I(2)) are potential targets for TVP1022 (IC(50) =9.5E-08 M and IC(50) =1.4E-07 M, respectively). Western blotting analysis showed that TVP1022 (1-20 µM) dose-dependently increased the immunoreactivity of phosphorylated p42 and p44 MAPK in rat pheochromocytoma PC12 cells and in neonatal rat ventricular myocytes (NRVM). This effect of TVP1022 was significantly attenuated by efaroxan, a selective I(1) imidazoline receptor antagonist. In addition, the cytoprotective effect of TVP1022 demonstrated in NRVM against serum deprivation-induced toxicity was markedly inhibited by efaroxan, thus suggesting the importance of I(1)imidazoline receptor in mediating the cardioprotective activity of the drug. Our findings suggest that the I(1)imidazoline receptor represents a novel site of action for the cyto/cardio-protective efficacy of TVP1022.


Subject(s)
Cardiotonic Agents/pharmacology , Imidazoline Receptors/metabolism , Indans/pharmacology , Signal Transduction/drug effects , Animals , Benzofurans/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , Blotting, Western , Cardiotonic Agents/chemistry , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Imidazoline Receptors/antagonists & inhibitors , In Vitro Techniques , Indans/chemistry , Inhibitory Concentration 50 , Mitogen-Activated Protein Kinase 1/metabolism , Myocytes, Cardiac/metabolism , PC12 Cells , Rats
6.
Life Sci ; 91(13-14): 669-75, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22365956

ABSTRACT

AIMS: Ischemic acute kidney injury (iAKI) in experimental diabetes mellitus (DM) is associated with a rapid kidney dysfunction more than in non-diabetic rats. We hypothesize that this vulnerability is due to excessive endothelin-1 (ET-1) expression along with dysregulation of nitric oxide synthase (NOS) isoforms. The aim of the present study was to assess the impact of ischemia on renal function in diabetic rats as compared with non-diabetic rats, and to investigate the involvement of ET-1 and NO systems in the susceptibility of diabetic kidney to ischemic damage. MAIN METHODS: DM was induced by Streptozotocin. iAKI was induced by clamping of left renal artery for 30 min. Right intact kidney served as control. 48 h following ischemia, clearance protocols were applied to assess glomerular filtration rate (GFR), urinary flow (V) and sodium excretion (U(Na)V) in both kidneys. The renal effects of ABT-627, ET(A) antagonist; A192621.1, ET(B) antagonist; L-NAME, NOS non-selective inhibitor; 1400 W, inducible NOS (iNOS) inhibitor; and NPLA, neuronal NOS (nNOS) inhibitor, were assessed following ischemic renal injury in diabetic rats. KEY FINDINGS: Induction of iAKI in diabetic and non-diabetic rats caused significant reductions in GFR, V, and U(Na)V, which were greater in diabetic than non-diabetic rats. While, treatment with ABT-627 decreased V and U(Na)V, and increased GFR, A192621.1 decreased all these parameters. L-NAME, 1400 W, and NPLA improved GFR in the ischemic diabetic kidney. SIGNIFICANCE: Excessive vasoconstrictive effects of ET-1 via ET(A) and upregulation of iNOS, are partly responsible for the impaired recovery of renal function following ischemia in diabetic rats.


Subject(s)
Acute Kidney Injury/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Endothelin-1/metabolism , Nitric Oxide Synthase Type II/metabolism , Receptor, Endothelin A/metabolism , Acute Kidney Injury/etiology , Animals , Diabetes Mellitus, Experimental/complications , Glomerular Filtration Rate , Male , Nitric Oxide , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Streptozocin , Up-Regulation , Vasoconstriction
7.
Circ Heart Fail ; 4(4): 463-73, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558446

ABSTRACT

BACKGROUND: Despite the availability of many pharmacological and mechanical therapies, the mortality rate among patients with congestive heart failure (CHF) remains high. We tested the hypothesis that TVP1022 (the S-isomer of rasagiline; Azilect), a neuroprotective and cytoprotective molecule, is also cardioprotective in the settings of experimental CHF in rats. METHODS AND RESULTS: In rats with volume overload-induced CHF, we investigated the therapeutic efficacy of TVP1022 (7.5 mg/kg) on cardiac function, structure, biomarkers, and kidney function. Treatment with TVP1022 for 7 days before CHF induction prevented the increase in left ventricular end-diastolic area and end-systolic area, and the decrease in fractional shortening measured 14 days after CHF induction. Additionally, TVP1022 pretreatment attenuated CHF-induced cardiomyocyte hypertrophy, fibrosis, plasma and ventricular B-type natriuretic peptide levels, and reactive oxygen species expression. Further, in CHF rats, TVP1022 decreased cytochrome c and caspase 3 expression, thereby contributing to the cardioprotective efficacy of the drug. TVP1022 also enhanced the urinary Na(+) excretion and improved the glomerular filtration rate. Similar cardioprotective effects were obtained when TVP1022 was given to rats after CHF induction. CONCLUSIONS: TVP1022 attenuated the adverse functional, structural, and molecular alterations in CHF, rendering this drug a promising candidate for improving cardiac and renal function in this disease state.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Failure/physiopathology , Indans/pharmacology , Kidney/drug effects , Kidney/physiopathology , Neuroprotective Agents/pharmacology , Ventricular Remodeling/drug effects , Animals , Cardiotonic Agents/therapeutic use , Caspase 3/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Fibrosis/prevention & control , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Heart Failure/drug therapy , Heart Failure/metabolism , Hypertrophy/prevention & control , Indans/therapeutic use , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Ventricular Remodeling/physiology
8.
Circ Heart Fail ; 4(3): 345-54, 2011 May.
Article in English | MEDLINE | ID: mdl-21398416

ABSTRACT

BACKGROUND: The side effects of fluid retention and edema of the thiazolidinedione (TZD) class of peroxisome proliferator-activated receptor-γ agonists limit their use in patients with congestive heart failure (CHF). The present study aims to explore whether chronic treatment with the TZD compound rosiglitazone (RGZ) is associated with worsening of salt and water retention in male Sprague-Dawley rats with aorto-caval fistula, an experimental model of volume-overload CHF. METHODS AND RESULTS: The effects of oral RGZ (30 mg/kg per day for 4 weeks) in CHF rats on plasma volume, cumulative sodium excretion, renal expression of Na(+) channels and transporters, and selected biomarkers of CHF were compared with those in CHF rats and sham-operated control rats treated with vehicle only (n=7 to 10). Additionally, the response to acute saline loading (3.5% of body weight) was evaluated after 2 weeks of treatment by renal clearance methodology. Chronic RGZ treatment caused no further increase in plasma volume compared with vehicle-treated CHF rats. Moreover, no increase in renal expression of Na(+) transport-linked channels/transporters was observed in response to RGZ. Cumulative sodium excretion was enhanced in CHF rats after RGZ and by another TZD compound, pioglitazone. In response to saline loading, RGZ-treated animals displayed a higher natriuretic/diuretic response than did vehicle-treated rats. Chronic RGZ treatment was not associated with any deterioration in selected biomarkers of CHF, whereas indices of cardiac hypertrophy and blood pressure were improved. CONCLUSIONS: Chronic RGZ treatment was not associated with worsening of fluid retention or cardiac status in rats with experimental volume-overload CHF. Rather, RGZ appeared to improve renal handling of salt and water in rats with CHF.


Subject(s)
Heart Failure/drug therapy , Thiazolidinediones/pharmacology , Water-Electrolyte Balance/drug effects , Water-Electrolyte Imbalance/drug therapy , Animals , Disease Models, Animal , Heart Failure/physiopathology , Kidney Function Tests , Male , Rats , Rats, Sprague-Dawley , Rosiglitazone , Sodium Chloride/metabolism , Thiazolidinediones/therapeutic use , Water/metabolism , Water-Electrolyte Imbalance/physiopathology
9.
Br J Pharmacol ; 163(4): 755-69, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21323905

ABSTRACT

BACKGROUND AND PURPOSE: Because myocardial infarction is a major cause of morbidity and mortality worldwide, protecting the heart from the ischaemia and reperfusion (I/R) damage is the focus of intense research. Based on our in vitro findings showing that TVP1022 (the S-enantiomer of rasagiline, an anti-Parkinsonian drug) possesses cardioprotective effects, in the present study we investigated the hypothesis that TVP1022 can attenuate myocardial damage in an I/R model in rats. EXPERIMENTAL APPROACH: The model consisted of 30-min occlusion of the left anterior descending artery followed by 4 or 24 h reperfusion. In addition, we investigated the possible mechanisms of cardioprotection in H9c2 cells and neonatal rat ventricular myocytes (NRVM) exposed to oxidative stress induced by H(2) O(2) . KEY RESULTS: TVP1022 (20 and 40 mg·kg(-1) ) administered 5 min before reperfusion followed by an additional dose 4 h after reperfusion reduced the infarct size and attenuated the decline in ventricular function. TVP1022 also attenuated I/R-induced deterioration in cardiac mitochondrial integrity evaluated by mitochondrial swelling capacity. In vitro, using H9c2 cells and NRVM, TVP1022 attenuated both serum free- and H(2) O(2) -induced damage, preserved mitochondrial membrane potential and Bcl-2 levels, inhibited mitochondrial cytochrome c release and the increase in cleaved caspase 9 and 3 levels, and enhanced the phosphorylation of protein kinase C and glycogen synthase kinase-3ß. CONCLUSIONS AND IMPLICATIONS: TVP1022 provided cardioprotection in a model of myocardial infarction, and therefore should be considered as a novel adjunctive therapy for attenuating myocardial damage resulting from I/R injuries.


Subject(s)
Cardiotonic Agents/pharmacology , Indans/pharmacology , Myocardial Infarction/prevention & control , Reperfusion Injury/prevention & control , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cytochromes c/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
10.
Nephron Physiol ; 115(3): p21-30, 2010.
Article in English | MEDLINE | ID: mdl-20460939

ABSTRACT

BACKGROUND/AIMS: Patients treated with peroxisome proliferator-activated receptor analogs (PPAR) alpha or alpha/gamma may develop a transient and reversible increase in serum creatinine, the mechanism of which remains obscure. This study evaluates whether treatment with either PPAR-alpha or -alpha/gamma analogs, fenofibrate or tesaglitazar, may cause deterioration in renal hemodynamics or exert direct tubular or glomerular nephrotoxic effects in rats. METHODS: Male Sprague-Dawley rats (300-320 g) were treated per os with fenofibrate (300 mg/kg/day), tesaglitazar (1.2 mg/kg/day) or vehicle, for 14 days. Glomerular filtration rate (GFR) and renal blood flow (RBF) were measured by inulin clearance and ultrasonic flowmetry, and cumulative excretion of sodium and creatinine were assessed. Biomarkers of glomerular and tubular injury were measured, including urinary albumin excretion and renal mRNA levels of kidney injury molecule 1 (Kim-1), lipocalin 2 (Lcn2), and osteopontin (Spp1). RESULTS: Fenofibrate and tesaglitazar improved the lipid profile, but caused no detectable decrease in GFR or RBF compared with vehicle-treated rats. Furthermore, the cumulative excretions of sodium and creatinine were not altered by the drugs. Finally, there was no significant difference between drug- and vehicle-treated groups in urinary albumin excretion or in the expression of renal injury biomarkers. CONCLUSIONS: In the rat, no direct nephrotoxic effect or deterioration in renal hemodynamics and function were observed following treatment with fenofibrate or tesaglitazar.


Subject(s)
Alkanesulfonates/pharmacology , Fenofibrate/pharmacology , Kidney Tubules/drug effects , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/pharmacology , Alkanesulfonates/toxicity , Animals , Cell Adhesion Molecules/genetics , Creatinine/urine , Fenofibrate/toxicity , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/toxicity , Inulin/pharmacokinetics , Kidney Tubules/physiology , Lipocalin-2 , Lipocalins/genetics , Male , Osteopontin/genetics , PPAR alpha/metabolism , PPAR gamma/metabolism , Phenylpropionates/toxicity , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Circulation/physiology , Sodium/urine
11.
Nephrol Dial Transplant ; 24(2): 428-36, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18799609

ABSTRACT

BACKGROUND: Ischaemic kidney injury continues to play a dominant role in the pathogenesis of acute renal failure (ARF) in many surgical and medical settings. A major event in the induction of renal injury is related to the generation of oxygen-free radicals. Hyperbaric oxygen therapy (HBO) is indicated for treatment of many ischaemic events but not for ARF. Therefore, the present study examined the effects of HBO on kidney function and renal haemodynamics in rats with ischaemic ARF. METHODS: Renal ischaemia was induced by unilateral renal artery clamping (45 min) in rats. Within 24 h following ischaemia, rats were treated twice with HBO of 100% O(2) at 2.5 absolute atmospheres for 90 min each (+HBO). Untreated rats (-HBO) served as a control. Forty-eight hours later, GFR, RBF and endothelial-dependent vasorelaxation were measured. In addition, the immunoreactive staining of 4-hydroxy-2-noneal (4-HNE), a major product of endogenous lipid peroxidation, and superoxide dismutase (SOD) were assessed. RESULTS: In the -HBO group, GFR was reduced by 94% compared with the untouched normal kidney (ischaemic: 0.06 +/- 0.03 ml/min, normal: 1.02 +/- 0.13 ml). In contrast, in the +HBO group, GFR of the ischaemic kidney (0.36 +/- 0.07 ml/min) was reduced only by 68% compared with the contralateral normal kidney (1.12 +/- 0.12 ml/min). In line with these findings, HBO improved the vasodilatory response to ACh as expressed in enhancement of both total and regional renal blood flow. In addition, HBO reduced the formation of 4-HNE by 33% and 76% and increased SOD by 30% and 70% in the cortex and outer stripe region of the medulla of the ischaemic kidney, respectively. CONCLUSION: HBO attenuates the decline in GFR following renal ischaemia, and improves endothelial-dependent vasorelaxation, suggesting that treatment with HBO may be beneficial in the setting of ischaemic ARF.


Subject(s)
Glomerular Filtration Rate , Hyperbaric Oxygenation , Kidney/blood supply , Kidney/injuries , Reperfusion Injury/therapy , Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Animals , Antioxidants/metabolism , Endothelium, Vascular/physiopathology , Ischemia/metabolism , Ischemia/physiopathology , Kidney/physiopathology , Lipid Peroxidation , Male , Oxidants/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation , Reperfusion Injury/physiopathology , Superoxide Dismutase/metabolism , Vasodilation
12.
Nephrol Dial Transplant ; 21(5): 1205-11, 2006 May.
Article in English | MEDLINE | ID: mdl-16396970

ABSTRACT

BACKGROUND: Urotensin II (U-II) and its receptor GPR-14 are expressed in the kidney and the cardiovascular system of various mammalian species. Recent studies suggested that the U-II/GPR-14 system is upregulated in patients with congestive heart failure (CHF). However, the involvement of the peptide in the alterations of renal function in CHF remains unknown. METHODS: The effects of incremental doses (1.0-100.0 nmol/kg) of human U-II (hU-II) on renal haemodynamic and clearance parameters were assessed in rats with an aorto-caval fistula, an experimental model of CHF, and sham controls. Additionally, the effects of pre-treatment with the nitric oxide (NO) synthase blocker, nitro-L-arginine methyl ester (L-NAME), and the cyclooxygenase inhibitor, indomethacin, on the renal haemodynamic response to hU-II were studied in CHF rats. RESULTS: hU-II caused a decrease in mean arterial pressure in control and CHF rats. In controls, hU-II did not alter renal blood flow (RBF), and caused a minimal decrease (-12.5%) in renal vascular resistance (RVR). However, in CHF rats, the peptide induced a marked increase in RBF (+28%) and a decrease in RVR (-21.5%). These effects were attenuated by L-NAME, but not by indomethacin. Furthermore, hU-II caused a significant increase (+29%) in glomerular filtration rate (GFR) in CHF rats, whereas GFR tended to decrease in controls. Sodium excretion was not altered in control or in CHF rats in response to hU-II. CONCLUSIONS: hU-II exerts an NO-dependent renal vasodilatation that is more pronounced in rats with CHF. The data further suggest that the U-II/GPR-14 system may be involved in the regulation of renal haemodynamics in CHF.


Subject(s)
Heart Failure/physiopathology , Hemodynamics/drug effects , Urotensins/pharmacology , Analysis of Variance , Animals , Disease Models, Animal , Glomerular Filtration Rate/drug effects , Hemodynamics/physiology , Humans , Indomethacin/pharmacology , Kidney Function Tests , Male , NG-Nitroarginine Methyl Ester/pharmacology , Probability , Rats , Rats, Wistar , Reference Values , Renal Circulation/drug effects , Risk Factors , Sensitivity and Specificity , Up-Regulation , Urodynamics , Urotensins/metabolism
13.
Am J Physiol Heart Circ Physiol ; 290(5): H2007-14, 2006 May.
Article in English | MEDLINE | ID: mdl-16361369

ABSTRACT

Rho-dependent kinases serve as downstream effectors of several vasoconstrictor systems, the activities of which are upregulated in congestive heart failure (CHF). We evaluated renal and cardiac effects of Y-27632, a highly selective Rho kinase inhibitor, in an experimental model of volume-overload CHF. Effects of acute administration of Y-27632 (0.3 mg/kg) on renal hemodynamic and clearance parameters and effects of chronic treatment (10.0 mg.kg(-1).day(-1) for 7 days via osmotic minipumps) on cardiac hypertrophy and cumulative Na+ excretion were studied in male Wistar rats with aortocaval fistula and control rats. The Y-27632-induced decrease in renal vascular resistance (from 40.4 +/- 4.6 to 26.0 +/- 3.1 resistance units, P < 0.01) in CHF rats was associated with a significant increase in total renal blood flow (+34%) and cortical and medullary blood flow (approx +37 and +27%, respectively). These values were significantly higher than those in control rats and occurred despite a decrease in mean arterial pressure (-15 mmHg). Despite the marked renal vasodilatory effect, Y-27632 did not alter glomerular filtration rate and renal Na+ excretion. Chronic administration of Y-27632 did not alter daily or cumulative renal Na+ excretion in CHF rats but was associated with a significant decrease in heart-to-body weight ratio, an index of cardiac hypertrophy: 0.32 +/- 0.007, 0.46 +/- 0.017, and 0.37 +/- 0.006% in control, CHF, and CHF + Y-27632 rats, respectively. The findings suggest that Rho kinase-dependent pathways are involved in the mechanisms of renal vasoconstriction and cardiac hypertrophy in rats with volume-overload heart failure. Selective blockade of these signaling pathways may be considered an additional tool to improve renal perfusion and attenuate cardiac hypertrophy in heart failure.


Subject(s)
Cardiomegaly/physiopathology , Disease Models, Animal , Heart Failure/physiopathology , Kidney/blood supply , Kidney/physiopathology , Protein Serine-Threonine Kinases/metabolism , Renal Circulation , Vasoconstriction , Animals , Cardiomegaly/complications , Heart Failure/complications , Intracellular Signaling Peptides and Proteins , Male , Rats , Rats, Wistar , Signal Transduction , rho-Associated Kinases
14.
Clin Sci (Lond) ; 103 Suppl 48: 245S-248S, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12193096

ABSTRACT

The present study examined the effects of two highly selective endothelin-1 (ET-1) receptor antagonists, ABT-627 (ET(A) blocker) and A-192621 (ET(B) blocker), on the systemic and renal haemodynamic effects of ET-1 in normal rats and in rats with experimental congestive heart failure (CHF) produced by aortocaval fistula. Intravenous injection of ET-1 (1.0 nmol x kg(-1) of body weight) to anaesthetized normal rats produced sustained decreases in renal blood flow (RBF) (assessed by ultrasonic flowmetry) and glomerular filtration rate (GFR), and significant increases in renal vascular resistance (RVR) and mean arterial pressure (MAP). Pretreatment with ABT-627 (1 mg x h(-1) x kg(-1) of body weight) abolished the pressor response to ET-1 without affecting the depressor phase, and significantly impaired the renal vasoconstriction. The systemic and renal vasoconstrictive effects of ET-1 in normal rats were significantly augmented by pretreatment with 3.0 mg x h(-1) x kg(-1) of A-192621. Baseline RBF and GFR in rats with CHF were reduced significantly compared with control rats, whereas RVR was elevated. The hypertensive effect of ET-1 was attenuated in rats with CHF. In the presence of ET(A) blockade, the pressor response to ET-1 was completely abolished in CHF rats. Furthermore, pretreatment with ABT-627 enhanced the recovery from ET-1- dependent vasoconstriction and remarkably reversed the ET-1-induced hypofiltration. Blockade of ET(B) receptors in rats with CHF further exposed the exaggerated ET-1-induced renal vasoconstriction. Our data demonstrate that experimental CHF is associated with altered responsiveness to ET(A)- and ET(B)-mediated systemic and renal effects of ET-1. Furthermore, in CHF, as in control rats, the ET(B)-mediated vasodilatory response may serve as an important compensatory counterbalance to the adverse ET(A)-mediated effects.


Subject(s)
Endothelin Receptor Antagonists , Heart Failure/physiopathology , Kidney/physiopathology , Pyrrolidines/pharmacology , Animals , Atrasentan , Blood Pressure/drug effects , Endothelin-1 , Glomerular Filtration Rate/drug effects , Kidney/drug effects , Male , Rats , Rats, Wistar , Receptor, Endothelin A , Receptor, Endothelin B , Renal Circulation/drug effects , Vascular Resistance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...