Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Kidney Int ; 71(8): 744-54, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17290297

ABSTRACT

Nestin, a marker of multi-lineage stem and progenitor cells, is a member of intermediate filament family, which is expressed in neuroepithelial stem cells, several embryonic cell types, including mesonephric mesenchyme, endothelial cells of developing blood vessels, and in the adult kidney. We used Nestin-green fluorescent protein (GFP) transgenic mice to characterize its expression in normal and post-ischemic kidneys. Nestin-GFP-expressing cells were detected in large clusters within the papilla, along the vasa rectae, and, less prominently, in the glomeruli and juxta-glomerular arterioles. In mice subjected to 30 min bilateral renal ischemia, glomerular, endothelial, and perivascular cells showed increased Nestin expression. In the post-ischemic period, there was an increase in fluorescence intensity with no significant changes in the total number of Nestin-GFP-expressing cells. Time-lapse fluorescence microscopy performed before and after ischemia ruled out the possibility of engraftment by the circulating Nestin-expressing cells, at least within the first 3 h post-ischemia. Incubation of non-perfused kidney sections resulted in a medullary-to-cortical migration of Nestin-GFP-positive cells with the rate of expansion of their front averaging 40 microm/30 min during the first 3 h and was detectable already after 30 min of incubation. Explant matrigel cultures of the kidney and aorta exhibited sprouting angiogenesis with cells co-expressing Nestin and endothelial marker, Tie-2. In conclusion, several lines of circumstantial evidence identify a sub-population of Nestin-expressing cells with the mural cells, which are recruited in the post-ischemic period to migrate from the medulla toward the renal cortex. These migrating Nestin-positive cells may be involved in the process of post-ischemic tissue regeneration.


Subject(s)
Intermediate Filament Proteins/metabolism , Ischemia/metabolism , Kidney/blood supply , Kidney/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Movement , Flow Cytometry , Gene Expression , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Video , Neovascularization, Physiologic , Nestin
2.
Nephron Physiol ; 98(4): p89-96, 2004.
Article in English | MEDLINE | ID: mdl-15627797

ABSTRACT

BACKGROUND/AIM: Endothelin-converting enzyme (ECE) catalyzes the generation of endothelin-1 (ET-1) from its inactive precursor big-ET-1. Previous studies suggested that the ET-1 system is involved in the regulation of sodium excretion by the kidney. In particular, ET-1 via the ET(B) receptor-mediated signaling has been shown to increase renal medullary blood flow and decrease sodium transport in the collecting duct, both acting to promote renal sodium excretion. The present study was designed to evaluate the possibility that alterations in dietary salt intake may regulate the ECE-1. METHODS: Wistar rats were fed for 3 days either with a diet containing low salt (0.01% NaCl), normal salt (0.5% NaCl), or high salt intake, either by high salt diet (4% NaCl) or normal salt diet plus 0.9% saline drinking. The expression of and immunoreactive protein levels of ECE-1 in the renal medulla was studied by RT-PCR, Northern blotting and Western blotting techniques. RESULTS: The expression of ECE-1 mRNA (by RT-PCR and Northern blotting), as well as the immunoreactive levels of ECE-1, were significantly higher in the renal medulla of rats exposed to high salt intake than in rats on normal salt diet. CONCLUSION: The findings suggest that upregulation of ECE-1, leading to increased generation of ET-1 in the renal medulla, may be a compensatory mechanism promoting enhanced sodium excretion by the kidney in response to high salt intake.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Kidney Medulla/metabolism , Metalloendopeptidases/metabolism , Sodium, Dietary/pharmacology , Animals , Aspartic Acid Endopeptidases/genetics , Endothelin-Converting Enzymes , Gene Expression/drug effects , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Medulla/drug effects , Male , Metalloendopeptidases/genetics , Rats , Rats, Wistar , Sodium/blood , Sodium/urine , Up-Regulation
3.
J Histochem Cytochem ; 49(10): 1293-300, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11561014

ABSTRACT

The natriuretic peptides are believed to play an important role in the pathophysiology of congestive heart failure (CHF). We utilized a quantitative cytomorphometric method, using double immunocytochemical labeling, to assess the characteristics of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in atrial granules in an experimental model of rats with CHF induced by aortocaval fistula. Rats with CHF were further divided into decompensated (sodium-retaining) and compensated (sodium-excreting) subgroups and compared with a sham-operated control group. A total of 947 granules in myocytes in the right atrium were analyzed, using electron microscopy and a computerized analysis system. Decompensated CHF was associated with alterations in the modal nature of granule content packing, as depicted by moving bin analysis, and in the granule density of both peptides. In control rats, the mean density of gold particles attached to both peptides was 347.0 +/- 103.6 and 306.3 +/- 89.9 gold particles/microm2 for ANP and BNP, respectively. Similar mean density was revealed in the compensated rats (390.6 +/- 81.0 and 351.3 +/- 62.1 gold particles/microm2 for ANP and BNP, respectively). However, in rats with decompensated CHF, a significant decrease in the mean density of gold particles was observed (141.6 +/- 67.3 and 158.0 +/- 71.2 gold particles/microm2 for ANP and BNP, respectively; p<0.05 compared with compensated rats, for both ANP and BNP). The ANP:BNP ratio did not differ between groups. These findings indicate that the development of decompensated CHF in rats with aortocaval fistula is associated with a marked decrease in the density of both peptides in atrial granules, as well as in alterations in the quantal nature of granule formation. The data further suggest that both peptides, ANP and BNP, may be regulated in the atrium by a common secretory mechanism in CHF.


Subject(s)
Atrial Natriuretic Factor/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Natriuretic Peptide, Brain/metabolism , Secretory Vesicles/metabolism , Animals , Heart Atria/metabolism , Heart Atria/ultrastructure , Heart Failure/pathology , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Myocardium/ultrastructure , Rats , Rats, Wistar
4.
News Physiol Sci ; 16: 152-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11479363

ABSTRACT

The kidney is both a source of endothelin (ET) generation and an important target organ of this peptide. The highest concentrations of ET-1 in the body exist in the renal medulla, where it mediates natriuretic and diuretic effects through the ET(B) receptor subtype. It is proposed that aberrations in the renal ET system may lead to sodium and water retention and subsequently to the development of hypertension.


Subject(s)
Endothelins/physiology , Kidney/physiopathology , Animals , Diuresis/physiology , Endothelin-1/physiology , Humans , Hypertension/physiopathology , Kidney Medulla/physiopathology , Natriuresis/physiology , Receptor, Endothelin B , Receptors, Endothelin/physiology
5.
Cardiovasc Res ; 51(3): 567-76, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11476747

ABSTRACT

OBJECTIVE: Surgical closure of a large arteriovenous (A-V) fistula in patients and animals is associated with prompt diuresis and natriuresis. However, the mechanisms underlying these changes remained largely unknown. METHODS: The present study evaluated the hormonal balance between major antinatriuretic systems (plasma renin activity, PRA, and arginine vasopressin, AVP) and natriuretic systems (atrial natriuretic peptide, ANP, and renal nitric oxide, NO) in Wistar rats with an A-V fistula (1.2 mm O.D., side to side) between the abdominal aorta and inferior vena cava. RESULTS: The placement of an A-V fistula caused progressive sodium retention (UNaV decreased from 1500 to 100 microequiv./day), a significant drop in mean arterial blood pressure (MAP) from 127+/-3 to 75+/-2 mmHg (P<0.01), and a significant increase in ANP (from 94+/-12 to 389+/-135 pg/ml, P<0.05), PRA (from 22.1+/-2.0 to 47+/-14 ng angiotensin I [Ang I]/ml/h, P<0.05), AVP (from 14.2+/-3.6 to 37.7+/-9.6 pg/ml, P<0.05), norepinephrine (from 184.2+/-40.5 to 1112.6+/-293.2 pg/ml, P<0.05) and epinephrine (from 667.5+/-175.9 to 2049.8+/-496.9 pg/ml, P<0.05). Furthermore, these changes were associated with a 3-fold increase in the renal medullary immunoreactive levels of endothelial NO synthase (eNOS), an endogenous vasodilator that plays an important role in the regulation of medullary blood flow. After 6 days, rats with A-V fistula and maximal sodium retention underwent surgical closure of the A-V fistula. The A-V fistula closure was associated with dramatic natriuresis (UNaV=2563+/-78 and 1918+/-246 microEq/day on days 3 and 6 following the closure, respectively) and restoration of MAP to normal levels (111+/-6 mmHg); PRA decreased to 29+/-5 ng Ang I/ml/h, AVP to 20.3+/-7.1 pg/ml, and medullary eNOS declined to basal levels, whereas plasma ANP concentrations remained elevated (380+/-90 pg/ml) after 3 days and returned to normal (92+/-12 pg/ml) on day 6. CONCLUSIONS: These results demonstrate that the creation of A-V fistula is associated with activation of both natriuretic and antinatriuretic systems. Closure of A-V fistula is characterized by shifting the balance in favor of the natriuretic substances. Moreover, the observed alterations in medullary eNOS following the creation and closure of A-V fistula suggest that this system, an important determinant of medullary blood flow, may contribute significantly to the regulation of sodium excretion in this model.


Subject(s)
Heart Failure/physiopathology , Natriuresis/physiology , Animals , Arginine Vasopressin/blood , Arteriovenous Shunt, Surgical , Atrial Natriuretic Factor/blood , Blood Pressure/physiology , Heart Failure/metabolism , Hormones/blood , Kidney/enzymology , Male , Nitric Oxide Synthase/metabolism , Rats , Rats, Wistar , Renin/blood , Sodium/urine , Urine
6.
Am J Physiol Renal Physiol ; 280(1): F43-53, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133513

ABSTRACT

The generation of PGs from arachidonic acid is mediated by cyclooxygenase (COX), which consists of a constitutive (COX-1) and an inducible (COX-2) isoform. The present study evaluated the relative expression and immunoreactive levels of COX-1 and COX-2, by means of RT-PCR, Western blot analysis, and immunohistochemistry, in the renal cortex and medulla of rats with congestive heart failure (CHF), induced by the placement of an aortocaval fistula. In addition, we examined the effects of a COX-1 inhibitor (piroxicam), COX-2 inhibitor (nimesulide), and nonselective COX inhibitor (indomethacin) at a dose of 5 mg/kg, on intrarenal blood flow by laser Doppler flowmetry. COX-1 and COX-2 mRNAs were abundantly expressed in the renal medulla of control and CHF rats and only minimally in the cortex. Moreover, both RT-PCR (32-36 cycles) and Western blot techniques revealed upregulation of medullary COX-2, but not of COX-1, in rats with advanced heart failure. In line with these findings, all three tested COX inhibitors provoked significant and sustained decreases (Delta approximately -20%) in medullary blood flow (MBF), which were similar in magnitude and duration in control animals. However, in CHF rats, indomethacin produced a greater reduction in MBF than that obtained with either piroxicam or nimesulide. Taken together, these results indicate that 1) both COX-1 and COX-2 are predominantly expressed in the renal medulla and 2) experimental CHF is associated with selective overexpression of COX-2. The latter may represent a mechanism aimed at defending MBF in the face of a decrease in renal perfusion pressure during the development of CHF.


Subject(s)
Gene Expression Regulation, Enzymologic , Heart Failure/physiopathology , Isoenzymes/genetics , Kidney/enzymology , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Arteriovenous Shunt, Surgical , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Heart Failure/enzymology , Heart Failure/genetics , Indomethacin/pharmacology , Isoenzymes/metabolism , Kidney Cortex/enzymology , Kidney Medulla/blood supply , Kidney Medulla/enzymology , Male , Membrane Proteins , Piroxicam/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Reference Values , Renal Circulation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
7.
J Cardiovasc Pharmacol ; 36(5 Suppl 1): S311-3, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078406

ABSTRACT

The present study examined the effects of A-192621.1, a highly selective endothelin-B- (ETB) receptor antagonist, on the renal hemodynamic and systemic actions of endothelin-1 (ET-1). Intravenous injection of ET-1 (1.0 nmol/kg) into anesthetized rats produced a sustained decrease in renal blood flow (assessed by ultrasonic flowmeter) and a significant increase in renal vascular resistance, as well as an increase in mean arterial pressure. These changes were significantly augmented by pretreatment with A-192621.1 (3.0 mg/kg/h). Analysis of intrarenal blood flow by laser-Doppler flowmeter revealed that ET-1 caused a marked and sustained decrease in cortical blood flow, associated with a transient increase in medullary blood flow. The reduction in cortical blood flow in response to ET-1 was further enhanced by pretreatment with A-192621.1, whereas the ET-1-induced medullary vasodilatation was completely abolished and reversed into a vasoconstrictor response. These findings suggest that the ETB-receptors mediate the systemic and renal vasodilatory actions of ET-1 in the rat, and that their activation may serve as a physiological counterbalance that modulates ET-1-induced vasoconstriction.


Subject(s)
Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Hemodynamics/drug effects , Kidney/drug effects , Pyrrolidines/pharmacology , Animals , Kidney/physiology , Rats , Rats, Wistar , Receptor, Endothelin B , Receptors, Endothelin/physiology , Renal Circulation/drug effects , Vascular Resistance
8.
Circulation ; 102(11): 1315-22, 2000 Sep 12.
Article in English | MEDLINE | ID: mdl-10982549

ABSTRACT

BACKGROUND: Chemokines are potent proinflammatory and immune modulators. Increased expression of chemokines, eg, monocyte chemoattractant protein-1 (MCP-1), has recently been described in clinical and experimental heart failure. The present report is aimed at exploring the expression, localization, and binding site regulation of MCP-1, a member of the C-C chemokine family, in a rat model of volume-overload congestive heart failure (CHF). METHODS AND RESULTS: An aortocaval fistula was surgically created between the abdominal aorta and inferior vena cava. Rats with CHF were further subdivided into compensated and decompensated subgroups. Northern blot analysis and real-time quantitative polymerase chain reaction demonstrated upregulation of MCP-1 mRNA expression correlating with the severity of CHF (288+/-22, 502+/-62, and 826+/-138 copies/ng total RNA for sham, compensated, and decompensated animals, respectively; n=5, P:<0.05). MCP-1 protein was localized by immunohistochemistry in cardiomyocytes, vascular endothelium and smooth muscle cells, infiltrating leukocytes, and interstitial fibroblasts, and its intensity increased with severity of CHF. In addition, rats with CHF displayed a significant decrease of (125)I-labeled MCP-1 binding sites to myocardium-derived membranes (384.3+/-57.0, 181.3+/-8.8, and 123.3+/-14.1 fmol/mg protein for sham, compensated, and decompensated animals, respectively). CONCLUSIONS: Volume-overload CHF in rats is associated with alterations in the expression, immunohistochemical localization, and receptor binding of the MCP-1 chemokine in the myocardium. These changes were more pronounced in rats with decompensated CHF. The data suggest that activation of the MCP-1 system may contribute to the progressive cardiac decompensation and development of CHF in rats with aortocaval fistula.


Subject(s)
Chemokine CCL2/metabolism , Heart Failure/metabolism , Animals , Binding, Competitive , Blotting, Northern , Heart Failure/diagnostic imaging , Immunohistochemistry , In Vitro Techniques , Male , Myocardium/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Wistar , Ultrasonography , Up-Regulation
9.
Hypertension ; 35(3): 732-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720587

ABSTRACT

Endothelin-1 (ET-1) at high concentrations has marked antidiuretic and antinatriuretic activities, whereas its precursor, big endothelin-1 (big ET-1), has surprisingly potent diuretic and natriuretic actions. The mechanisms underlying the excretory effects of big ET-1 have not been fully elucidated. To explore these mechanisms, we examined the effects of a highly selective ET(B) antagonist (A-192621.1), a calcium channel blocker (verapamil), a nitric oxide synthase inhibitor (N-nitro-L-arginine methyl ester [L-NAME]), and a cyclooxygenase inhibitor (indomethacin) on the systemic and renal actions of big ET-1 in anesthetized rats. An intravenous bolus injection of incremental doses of big ET-1 (0.3, 1. 0, and 3.0 nmol/kg) produced a significant hypertensive effect that was dose dependent and prolonged (from 113+/-7 mm Hg to a maximum of 148+/-6 mm Hg). The administration of big ET-1 induced marked diuretic and natriuretic responses (urinary flow rate increased from 8.5+/-1 to 110+/-14 microL/min, and fractional excretion of sodium increased from 0.38+/-0.13% to 7.51+/-1.24%). Glomerular filtration rate and renal plasma flow significantly decreased only at the highest dose of big ET-1. Pretreatment with A-192621.1 (3 mg/kg plus 3 mg. kg(-1). h(-1)) significantly abolished the diuretic (17+/-5 microL/min to a maximum of 19+/-3 microL/min) and natriuretic (0. 29+/-0.1% to a maximum of 1.93+/-0.37%) responses induced by big ET-1. Moreover, A-192621.1 potentiated the decline in glomerular filtration rate and renal plasma flow and the increase in mean arterial blood pressure produced by the low doses of big ET-1. Similar to A-192621.1, pretreatment with a nitric oxide synthase inhibitor (L-NAME, 10 mg/kg plus 5 mg. kg(-1). h(-1)) significantly and comparably reduced the diuretic and natriuretic actions of big ET-1 and augmented the hypoperfusion/hypofiltration and systemic vasoconstriction induced by high doses of the peptide. Pretreatment with verapamil (2 mg. kg(-1). h(-1)) slightly inhibited the diuretic/natriuretic effects of the high-dose big ET-1 and completely prevented the increase in mean arterial blood pressure provoked by the peptide. Unlike verapamil and L-NAME, only indomethacin administration was associated with significant natriuretic/diuretic responses and did not influence the pressor effect and renal actions of big ET-1. Taken together, these results suggest that big ET-1-induced diuretic and natriuretic responses are mediated mainly by stimulation of nitric oxide production coupled to ET(B) receptor subtype activation.


Subject(s)
Endothelins/pharmacology , Natriuresis/drug effects , Natriuresis/physiology , Protein Precursors/pharmacology , Receptors, Endothelin/physiology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cardiovascular Agents/pharmacology , Cytosol/metabolism , Endothelin Receptor Antagonists , Endothelin-1 , Enzyme Inhibitors/pharmacology , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Indomethacin/pharmacology , Kidney/chemistry , Kidney/drug effects , Kidney/physiology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Prostaglandins/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Receptor, Endothelin B , Urination/drug effects , Urination/physiology , Verapamil/pharmacology
10.
J Lab Clin Med ; 134(3): 232-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10482307

ABSTRACT

Long-term furosemide therapy is associated with increased urinary loss of thiamine. To examine the mechanism of furosemide-induced urinary thiamine loss, we measured urinary excretion of thiamine in rats in response to increasing doses of furosemide, acetazolamide, chlorothiazide, amiloride, mannitol, and extracellular fluid (ECF) volume loading by saline infusion. All animals were in normal thiamine balance as reflected by a thiamine pyrophosphate effect (TPPE) of 2.25% +/- 0.60% (mean +/- SEM), and all had normal renal function. Urinary flow increased in response to diuretic administration in a dose-dependent manner, reaching (mean) peak urinary flow rates of 283 to 402 microL/min. Fractional excretion of sodium (FE(Na)) exhibited the same pattern, reaching peak values of 12.3% to 23.2%. Urinary thiamine excretion increased in proportion to the incremental doses of diuretic agents, reaching (mean) maximal values of 7.44 to 9.34 pmol/min, with no significant difference (P = .11) between the various diuretics tested nor in response to saline loading. None of the diuretics tested differed in the effect on thiamine excretion, which was clearly flow dependent and only partially related to fractional sodium excretion. Urinary flow rate, being the single significant predictor, explained 78% (R2 = 0.78) of the variability in thiamine excretion rates. These findings indicate that urinary thiamine loss is caused by a nonspecific, flow-dependent mechanism common to all of the diuretics tested.


Subject(s)
Diuretics/toxicity , Furosemide/toxicity , Thiamine/urine , Animals , Diuresis/drug effects , Diuretics/administration & dosage , Furosemide/administration & dosage , Heart Failure/complications , Heart Failure/drug therapy , Humans , Kidney/drug effects , Kidney/metabolism , Male , Mannitol/administration & dosage , Natriuresis/drug effects , Rats , Rats, Sprague-Dawley , Thiamine Deficiency/chemically induced , Thiamine Deficiency/etiology , Thiamine Deficiency/metabolism
11.
Gen Pharmacol ; 33(3): 277-81, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10480661

ABSTRACT

The effects of 2-mercaptoacetyl-L-leucyl-L-phenylalanine (MA-LF) on the activity of neutral endopeptidase and on renal hemodynamics and excretory function were investigated in experiments in vitro and in vivo. In vitro studies showed that the compound effectively inhibited purified bovine kidney neutral endopeptidase (Ki = 0.012 microM), while having slight influence on the activity of angiotensin I converting enzyme (Ki = 0.14 microM). In experiments on normal anesthetized rats (thiobutabarbital sodium salt, 100 mg/kg), IV administration of MA-LF (20 and 60 mg/kg) produced a dose-dependent increase in absolute rate and fractional excretion of sodium (+324% and +299%, respectively) and urinary flow rate (+261%), but did not change renal and systemic hemodynamics. Renal excretory effects of the new compound were comparable to those of the selective neutral endopeptidase inhibitor SQ 28,603. These results demonstrate that MA-LF is a potent neutral endopeptidase inhibitor with prominent natriuretic and diuretic properties.


Subject(s)
Alanine/analogs & derivatives , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Kidney/drug effects , Neprilysin/antagonists & inhibitors , Alanine/pharmacology , Animals , Blood Pressure/drug effects , Cattle , Kidney/blood supply , Kidney/physiology , Male , Neprilysin/metabolism , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Renal Circulation/drug effects , Sodium/urine , Urodynamics/drug effects , Vascular Resistance/drug effects
12.
Nephron ; 80(4): 458-67, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832646

ABSTRACT

The contributions of nitric oxide (NO) and renal blood flow (RBF) were examined in ischemia-reperfusion injury in the rat kidney. The function of both kidneys was assessed by glomerular filtration rate (GFR), and fractional excretion of sodium (FENa), calculated before, during unilateral renal artery clamping (45 min), and following reperfusion (90 min). RBF was measured in the same model by ultrasonic flowmetry. Intrarenal NO levels were modulated by administration of S-nitroso-N-acetylpenicillamine (SNAP), L-arginine, acetylcholine, and the NO synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME). SNAP increased GFR from 0.20 +/- 0.04 ml/min in control ischemic kidney to 0.38 +/- 0.06 ml/min and reduced FENa from 19.3 +/- 3.4 to 9.5 +/- 1.8%. Similar results were observed when L-arginine was administered. Acetylcholine had no effect on GFR or FENa. RBF was fully restored within 60 min following reperfusion, with no change in the rate of recovery by L-arginine. L-NAME aggravated the ischemia-reperfusion injury, preventing full restoration of RBF, further reducing GFR and worsening FENa. In conclusion, ischemia-reperfusion injury ends in low intrarenal levels of NO. We propose that this low NO level results from damage to the endothelial receptor signal transduction process and is not due to impaired NO synthase activity or to changes in RBF.


Subject(s)
Ischemia/physiopathology , Kidney/blood supply , Nitric Oxide/physiology , Renal Circulation/physiology , Reperfusion Injury/physiopathology , Acetylcholine/pharmacology , Animals , Arginine/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Enzyme Inhibitors/pharmacology , Female , Glomerular Filtration Rate/drug effects , Ischemia/metabolism , Kidney/drug effects , Kidney/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Reperfusion Injury/metabolism , Sodium/urine , Vasodilator Agents/pharmacology
13.
Hypertension ; 32(4): 746-52, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9774374

ABSTRACT

Activation of the renin-angiotensin system may contribute to the derangement in renal and cardiac function in congestive heart failure. The present study evaluated the effects of eprosartan, a selective angiotensin II receptor antagonist, on renal hemodynamic and excretory parameters and on the development of cardiac hypertrophy in rats with aortocaval fistula, an experimental model of congestive heart failure. Infusion of eprosartan (1.0 mg/kg) in rats with aortocaval fistula produced a significant increase (+34%) in total renal blood flow and a sustained decrease (-33%) in the calculated renal vascular resistance. These effects on renal hemodynamics were more pronounced than those observed in sham-operated control rats and occurred despite a significant fall (-12%) in mean arterial blood pressure. Moreover, eprosartan caused a preferential increase in renal cortical blood perfusion and significantly increased glomerular filtration in rats with congestive heart failure. Chronic administration of eprosartan (5.0 mg/kg per day for 7 days through osmotic minipumps inserted intraperitoneally on the day of operation) resulted in a significant enhancement of urinary sodium excretion compared with nontreated rats with heart failure. Moreover, administration of eprosartan to salt-retaining rats with congestive heart failure resulted in a progressive increase and ultimate recovery in urinary sodium excretion. Finally, early treatment with eprosartan blocked the development of cardiac hypertrophy in rats with aortocaval fistula to a larger extent than the angiotensin-converting enzyme inhibitor enalapril. These findings emphasize the importance of angiotensin II in mediating the impairment in renal function and induction of cardiac hypertrophy in heart failure and further suggest that angiotensin II receptor blockade may be a useful treatment of these consequences in severe cardiac failure.


Subject(s)
Acrylates/therapeutic use , Angiotensin Receptor Antagonists , Antihypertensive Agents/therapeutic use , Cardiomegaly/prevention & control , Heart Failure/drug therapy , Hemodynamics/drug effects , Imidazoles/therapeutic use , Kidney/drug effects , Thiophenes , Acrylates/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antihypertensive Agents/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Enalapril/therapeutic use , Heart Failure/physiopathology , Imidazoles/pharmacology , Male , Rats , Rats, Wistar , Renal Circulation/drug effects , Sodium/urine
14.
J Cardiovasc Pharmacol ; 31 Suppl 1: S31-4, 1998.
Article in English | MEDLINE | ID: mdl-9595392

ABSTRACT

The expression and immunoreactivity of endothelin-converting enzyme (ECE) were examined in the renal tissue of rats with experimental congestive heart failure (CHF). Reverse transcriptase polymerase chain reaction (RT-PCR) revealed that ECE mRNA was more abundant (about twofold) in the renal medulla than in the cortex. Induction of heart failure caused a significant enhancement in the expression of this key enzyme in the renal cortex of rats with compensated CHF (delta + 28%) and in animals with decompensated heart failure (delta + 58%). An identical trend was also observed in the renal medulla, although these increases were moderate compared to those in the cortex. Similar findings were observed with Western blot techniques applying two monoclonal antibodies to rat ECE (AEC32-236 and AEC27-121). Taken together, these data suggest that upregulation of ECE is an important component in the activated renal ET system in CHF.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Heart Failure/enzymology , Kidney/enzymology , Metalloendopeptidases/metabolism , Animals , Blotting, Western , Endothelin-Converting Enzymes , Kidney Cortex/enzymology , Kidney Medulla/enzymology , Male , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Wistar
15.
Am J Physiol ; 274(4): F766-74, 1998 04.
Article in English | MEDLINE | ID: mdl-9575902

ABSTRACT

Congestive heart failure(CHF) is associated with a marked decrease in cortical blood flow and preservation of medullary blood flow. In the present study we tested the hypothesis that changes in the endothelin (ET) and nitric oxide (NO) systems in the kidney may contribute to the altered intrarenal hemodynamics in rats with aortocaval fistula, an experimental model of CHF. Cortical and medullary blood flow were measured simultaneously by laser-Doppler flowmetry in controls and rats with compensated and decompensated CHF. As previously reported [K. Gurbanov, I. Rubinstein, A. Hoffman, Z. Abassi, O. S. Better, and J. Winaver. Am. J Physiol. 271 (Renal Fluid Electrolyte Physiol. 40): F1166-F1172, 1996], administration of ET-1 in control rats produced a sustained cortical vasoconstriction and a transient medullary vasodilatory response. In rats with decompensated CHF, cortical vasoconstriction was severely blunted, whereas ET-1-induced medullary vasodilation was significantly prolonged. This prolonged response was mimicked by IRL-1620, a specific ETB agonist, and partially abolished by NO synthase (NOS) blockade. In line with these findings, expression of ET-1, ETA and ETB receptors, and endothelial NOS (eNOS), assessed by RT-PCR, and eNOS immunoreactivity, assessed by Western blotting, was significantly higher in the medulla than in the cortex. Moreover, expression of ET-1 mRNA in the cortex and eNOS mRNA in the cortex and the medulla increased in proportion to the severity of heart failure. These findings indicate that CHF is associated with altered regulation of intrarenal blood flow, which reflects alterations in expression and activity of the ET and NO systems. It is further suggested that exaggerated NO activity in the medulla contributes to preservation of medullary blood flow in the face of cortical vasoconstriction in CHF.


Subject(s)
Endothelins/physiology , Heart Failure/physiopathology , Nitric Oxide/physiology , Renal Circulation/physiology , Animals , Endothelin-1/pharmacology , Endothelins/genetics , Endothelins/pharmacology , Endothelium, Vascular/enzymology , Kidney Cortex/blood supply , Kidney Medulla/blood supply , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Peptide Fragments/pharmacology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Endothelin/genetics , Regional Blood Flow/drug effects , Transcription, Genetic
16.
J Clin Invest ; 101(6): 1325-33, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9502774

ABSTRACT

Muscle crush injury is often complicated by hemodynamic shock, electrolyte disorders, and myoglobinuric renal failure. In this study, we examined the involvement of the nitric oxide (NO) system in the development of muscle damage in an experimental model of crush injury induced by exertion of standardized mechanical pressure on tibialis muscle of rat. The intact limb served as a control. Four days after injury, the crushed muscle was characterized by extreme capillary vasodilatation as demonstrated by histological morphometric analysis. These changes were accompanied by muscle hyperperfusion as evaluated by measurements of femoral blood flow (ultrasonic flowmetry) and capillary blood flow (laser-doppler flowmetry). Treatment with Nomega-nitro-L-arginine methyl ester, a NO synthase (NOS) inhibitor, largely decreased the hyperperfusion. Furthermore, the expression of the different NOS isoforms, assessed by reverse transcription-PCR and immunoreactive levels, determined by Western blot, revealed a remarkable induction of the inducible NOS in the crushed limb. Similarly, endothelial NOS mRNA increased gradually after the induction of muscle damage. In contrast, the major muscular NOS, i.e., neuronal isoform remained unchanged. In line with the alterations in the mRNA levels, Western blot analysis revealed parallel changes in the immunoreactive levels of the various NOS. These findings indicate that muscle crush is associated with activation of the NO system mainly due to enhancement of iNOS. This may contribute to NO-dependent extreme vasodilatation in the injured muscle and aggravate the hypovolemic shock after crush injury.


Subject(s)
Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Blood Flow Velocity , Blotting, Western , Capillaries/physiopathology , Dilatation, Pathologic/physiopathology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Femoral Artery/physiopathology , Hindlimb/injuries , Hindlimb/metabolism , Isoenzymes/genetics , Isoenzymes/immunology , Isoenzymes/metabolism , Muscle, Skeletal/blood supply , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
17.
Pflugers Arch ; 435(3): 432-4, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9426302

ABSTRACT

Nitric oxide (NO) has been implicated as a modulator of the vascular effects of angiotensin II (ANG II) in the kidney. We used a NO-sensitive microelectrode to study the effect of ANG II on NO release, and to determine the effect of selective inhibition of the ANG II subtype I receptor (AT1) with losartan (LOS) and candesartan (CAN). NO release from isolated and perfused renal resistance arteries was measured with a porphyrin-electroplated, carbon fiber. The vessels were microdissected from isolated perfused rat kidneys and perfused at constant flow and pressure in vitro. The NO-electrode was placed inside the glass collection cannula to measure vessel effluent NO concentration. ANG II stimulated NO release in a dose-dependent fashion: 0.1 nM, 10 nM and 1000 nM ANG II increased NO-oxidation current by 85+/-18 pA (n = 11), 148+/-22 pA (n = 11), and 193+/-29 pA (n = 11), respectively. These currents correspond to changes in effluent NO concentration of 3.4+/-0.5 nM, 6.1+/-1.1 nM, and 8.2+/-1.3 nM, respectively. Neither LOS (1 muM) nor CAN (1 nM) significantly affected basal NO production, but both AT1-receptor blockers markedly blunted NO release in response to ANG II (10 nM): 77+/-6% inhibition with LOS (n = 8) and 63+/-9% with CAN (n = 8). These results are the first to demonstrate that ANG II stimulates NO release in isolated renal resistance arteries, and that ANG II-induced NO release is blunted by simultaneous AT1-receptor blockade. Our findings suggest that endothelium-dependent modulation of ANG II-induced vasoconstriction in renal resistance arteries is mediated, at least in part, by AT1-receptor-dependent NO release.


Subject(s)
Angiotensin II/pharmacology , Nitric Oxide/metabolism , Renal Artery/drug effects , Renal Artery/metabolism , Tetrazoles , Angiotensin Receptor Antagonists , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Electric Conductivity , Endothelium, Vascular/physiology , Losartan/pharmacology , Male , Microelectrodes , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology , Vascular Resistance
18.
Circulation ; 96(10): 3655-64, 1997 Nov 18.
Article in English | MEDLINE | ID: mdl-9396468

ABSTRACT

BACKGROUND: Congestive heart failure (CHF) is associated with a decrease in renal perfusion. Because endothelium-derived NO is important in the regulation of renal blood flow (RBF), we tested the hypothesis that an impairment in the NO system may contribute to the decrease in RBF in rats with experimental CHF. METHODS AND RESULTS: Studies were performed in rats with experimental high-output CHF induced by aortocaval (AV) fistula and sham-operated controls. In controls, incremental doses of acetylcholine (ACh, 1 to 100 microg x kg(-1) x min(-1)) increased RBF and caused a dose-related decrease in renal vascular resistance (RVR). However, the increase in RBF and decrease in RVR were markedly attenuated in rats with CHF. Likewise, the effects of ACh on urinary sodium and cGMP excretion were also diminished in CHF rats, as was the renal vasodilatory effect of the NO donor S-nitroso-N-acetylpenicillamine (SNAP). These attenuated responses to endothelium-dependent and -independent renal vasodilators in CHF rats occurred despite a normal baseline and stimulated NO2+NO3 excretion and normal expression of renal endothelial NO synthase (eNOS), as determined by eNOS mRNA levels and immunoreactive protein. Infusion of the NO precursor L-arginine did not affect baseline RBF or the response to ACh in rats with CHF. However, administration of the nonpeptide angiotensin II receptor antagonist A81988 before ACh completely restored the renal vasodilatory response to ACh in CHF rats. CONCLUSIONS: This study demonstrates that despite a significant attenuation in the NO-related renal vasodilatory responses, the integrity of the renal NO system is preserved in rats with chronic AV fistula. This impairment in NO-mediated renal vasodilation in experimental CHF appears to be related to increased activity of the renin-angiotensin system and may contribute further to the decrease in renal perfusion seen in CHF.


Subject(s)
Angiotensin II/physiology , Arteriovenous Fistula/physiopathology , Heart Failure/physiopathology , Nitric Oxide/physiology , Renal Circulation/physiology , Vasodilation/physiology , Venae Cavae/physiopathology , Acetylcholine/pharmacology , Angiotensin Receptor Antagonists , Animals , Aortic Diseases/physiopathology , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Kidney/drug effects , Male , Nitric Oxide Synthase/metabolism , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats , Rats, Wistar , Renal Circulation/drug effects , S-Nitroso-N-Acetylpenicillamine , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...