Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Acta Physiol (Oxf) ; 213(1): 249-58, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25089004

ABSTRACT

AIM: Tubuloglomerular feedback (TGF) is an important mechanism in control of signal nephron glomerular filtration rate. The oxidative stress in the macula densa, primarily determined by the interactions between nitric oxide (NO) and superoxide (O2-), is essential in maintaining the TGF responsiveness. However, few studies examining the interactions between and amount of NO and O2- generated by the macula densa during normal and hypertensive states. METHODS: In this study, we used isolated perfused juxtaglomerular apparatus to directly measure the amount and also studied the interactions between NO and O2- in macula densa in both physiological and slow pressor Angiotensin II (Ang II)-induced hypertensive mice. RESULTS: We found that slow pressor Ang II at a dose of 600 ng kg(-1) min(-1) for two weeks increased mean arterial pressure by 26.1 ± 5.7 mmHg. TGF response increased from 3.4 ± 0.2 µm in control to 5.2 ± 0.2 µm in hypertensive mice. We first measured O2- generation by the macula densa and found it was undetectable in control mice. However, O2- generation by the macula densa increased to 21.4 ± 2.5 unit min(-1) in Ang II-induced hypertensive mice. We then measured NO generation and found that NO generation by the macula densa was 138.5 ± 9.3 unit min(-1) in control mice. The NO was undetectable in the macula densa in hypertensive mice infused with Ang II. CONCLUSIONS: Under physiological conditions, TGF response is mainly controlled by the NO generated in the macula densa; in Ang II induced hypertension, the TGF response is mainly controlled by the O2- generated by the macula densa.


Subject(s)
Angiotensin II/pharmacology , Hypertension/chemically induced , Kidney Tubules/drug effects , Nitric Oxide/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Hypertension/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Mice, Inbred C57BL , Nephrons/drug effects , Nephrons/metabolism , Oxidative Stress
2.
J Biol Regul Homeost Agents ; 27(2): 569-78, 2013.
Article in English | MEDLINE | ID: mdl-23830406

ABSTRACT

Dehydration and acute reductions of blood pressure increases ADH and Ang II levels. These hormones increase transport along the distal nephron. In the thick ascending limb (TAL) ADH increases transport via cAMP, while Ang II acts via superoxide (O2-). However, the mechanism of interaction of these hormones in this segment remains unclear. The aim of this study was to explore ADH/Ang II interactions on TAL transport. For this, we measured the effects of ADH/Ang II, added sequentially to TAL suspensions from Wistar rats, on oxygen consumption (QO2) -as a transport index-, cAMP and O2-. Basal QO2 was 112+-5 nmol O2/min/mg protein. Addition of ADH (1nM) increased QO2 by 227 percent. In the presence of ADH, Ang II (1nM) elicited a QO2 transient response. During an initial 3.1+-0.7 minutes after adding Ang II, QO2 decreased 58 percent (p less than 0.03 initial vs. ADH) and then rose by 188 percent (p less than 0.03 late vs initial Ang II). We found that Losartan blocked the initial effects of Ang II and the latter blocked ADH and forskolin-stimulated cAMP. The NOS inhibitor L-NAME or the AT2 receptor antagonist PD123319 showed no effect on transported related oxygen consumption. Then, we assessed the late period after adding Ang II. The O2- scavenger tempol blocked the late Ang II effects on QO2, while Ang II increased O2- production during this period. We conclude that 1) Ang II has a transient effect on ADH-stimulated transport; 2) this effect is mediated by AT1 receptors; 3) the initial period is mediated by decreased cAMP and 4) the late period is mediated by O2-.


Subject(s)
Angiotensin II/pharmacology , Cyclic AMP/metabolism , Loop of Henle/metabolism , Oxygen Consumption/drug effects , Oxygen/metabolism , Vasopressins/pharmacology , Animals , Biological Transport , Losartan/pharmacology , Male , Nitric Oxide/physiology , Rats , Rats, Wistar
4.
Am J Physiol Renal Physiol ; 301(2): F263-70, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21543414

ABSTRACT

High salt intake (HS) is a risk factor for cardiovascular and kidney disease. Indeed, HS may promote blood-pressure-independent tissue injury via inflammatory factors. The lipid-lowering 3-hydroxy 3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors exert beneficial lipid-independent effects, reducing the expression and synthesis of inflammatory factors. We hypothesized that HS impairs kidney structure and function in the absence of hypertension, and these changes are reversed by atorvastatin. Four groups of rats were treated for 6 wk in metabolic cages with their diets: normal salt (NS); HS, NS plus atorvastatin and HS plus atorvastatin. We measured basal and final body weight, urinary sodium and protein excretion (U(Prot)V), and systolic blood pressure (SBP). At the end of the experimental period, cholesterolemia, creatinine clearance, renal vascular reactivity, glomerular volume, cortical and glomerular endothelial nitric oxide synthase (eNOS), and transforming growth factor (TGF)-ß1 expression were measured. We found no differences in SBP, body weight, and cholesterolemia. HS rats had increased creatinine clearence, U(Prot)V, and glomerular volume at the end of the study. Acetylcholine-induced vasodilatation decreased by 40.4% in HS rats (P < 0.05). HS decreased cortical and glomerular eNOS and caused mild glomerular sclerosis, interstitial mononuclear cell infiltration, and increased cortical expression of TGF-ß1. All of these salt-induced changes were reversed by atorvastatin. We conclude that long-term HS induces inflammatory and hemodynamic changes in the kidney that are independent of SBP. Atorvastatin corrected all, suggesting that the nitric oxide-oxidative stress balance plays a significant role in the earlier stages of salt induced kidney damage.


Subject(s)
Endothelium, Vascular/drug effects , Heptanoic Acids/pharmacology , Kidney/drug effects , Pyrroles/pharmacology , Sodium Chloride, Dietary/adverse effects , Vasodilation/drug effects , Animals , Atorvastatin , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , Rats , Rats, Wistar
7.
Hemodial Int ; 9(3): 296-302, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16191080

ABSTRACT

Patients on chronic hemodialysis often portray high serum [K+]. Although dietary excesses are evident in many cases, in others, the cause of hyperkalemia cannot be identified. In such cases, hyperkalemia could result from decreased potassium removal during dialysis. This situation could occur if alkalinization of body fluids during dialysis would drive potassium into the cell, thus decreasing the potassium gradient across the dialysis membrane. In 35 chronic hemodialysis patients, we compared two dialysis sessions performed 7 days apart. Bicarbonate or acetate as dialysate buffers were randomly assigned for the first dialysis. The buffer was switched for the second dialysis. Serum [K+], [HCO3-], and pH were measured in samples drawn before dialysis; 60, 120, 180, and 240 min into dialysis; and 60 and 90 min after dialysis. The potassium removed was measured in the dialysate. During the first 2 hr, serum [K+] decreased equally with both types of dialysates but declined more during the last 2 hr with bicarbonate dialysis. After dialysis, the serum [K+] rebounded higher with bicarbonate bringing the serum [K+] up to par with acetate. The lower serum [K+] through the second half of bicarbonate dialysis did not impair potassium removal (295.9 +/- 9.6 mmol with bicarbonate and 299.0 +/- 14.4 mmol with acetate). The measured serum K+ concentrations correlated with serum [HCO3-] and blood pH during bicarbonate dialysis but not during acetate dialysis. Alkalinization induced by bicarbonate administration may cause redistribution of K during bicarbonate dialysis but this does not impair its removal. The more marked lowering of potassium during bicarbonate dialysis occurs late in dialysis, when exchange is negligible because of a low gradient.


Subject(s)
Bicarbonates/blood , Potassium/blood , Renal Dialysis , Adult , Aged , Female , Humans , Hydrogen-Ion Concentration , Male , Middle Aged
8.
Hypertension ; 38(3 Pt 2): 639-44, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566947

ABSTRACT

Hyperkalemia is widely viewed as a common complication of ACE inhibition in azotemic patients. These renal failure patients are the patients who benefit most from ACE inhibition. Because we could not confirm this notion after a retrospective evaluation of 236 azotemic patients, we studied 2 models of renal mass reduction. In the first, we did a 5/6 nephrectomy (Nx) on rats and studied them 2 weeks after surgery (before chronic renal changes had developed). A second group was studied 16 weeks after Nx, once chronic renal failure was established. Rats in both models were treated with quinapril in drinking water. After baseline evaluation, we challenged them either by a high-K(+) diet or by blocking aldosterone receptors. We found that although quinapril blocked the K(+)-induced increase in aldosterone, serum K(+) levels and K(+) balance were maintained before and during high K(+) intake or during simultaneous spironolactone administration. We conclude that in hemodynamically stable rats with reduced renal mass and renal dysfunction, the administration of an ACE inhibitor does not cause severe hyperkalemia.


Subject(s)
Hyperkalemia/blood , Peptidyl-Dipeptidase A/drug effects , Renal Insufficiency/drug therapy , Tetrahydroisoquinolines , Adolescent , Adult , Aged , Aged, 80 and over , Aldosterone/blood , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Creatinine/blood , Disease Models, Animal , Female , Glomerular Filtration Rate/drug effects , Humans , Hypertension, Renal/complications , Isoquinolines/pharmacology , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Male , Middle Aged , Nephrectomy , Potassium/blood , Quinapril , Rats , Rats, Sprague-Dawley , Renal Insufficiency/blood , Renal Insufficiency/physiopathology , Retrospective Studies , Uremia/blood , Uremia/drug therapy , Uremia/etiology
9.
Hypertension ; 38(3 Pt 2): 655-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566950

ABSTRACT

Chronically infusing a subpressor dose of angiotensin (Ang) II increases blood pressure via poorly defined mechanisms. We found that this hypertensive response is accompanied by increased oxidant stress and is prevented by blocking endothelin (ET) receptors. Thus, we now tested whether blocking oxidant stress decreases both blood pressure and ET levels. We infused Sprague-Dawley rats (via osmotic pumps) with either vehicle (group 1) or Ang II (5 ng. kg(-1). min(-1); groups 2 to 4) for 15 days. Groups 3 and 4 also received either tempol in the drinking water (1 mmol/L) or vitamin E (5000 IU/kg diet), respectively, for 15 days. We measured systolic blood pressure (SBP) and urinary nitrite excretion every 3 days, and on day 15 we measured systemic and renal venous plasma levels of ET, isoprostanes, and thiobarbituric acid reactive substances (TBARS). SBP in Group 1 did not change throughout the study, whereas Ang II increased SBP (from 132+/-5 to 151+/-7 mm Hg). In addition, Ang II increased the systemic and renal venous levels of isoprostanes, TBARS, and ET and caused a transient decrease in urinary nitrites (that returned to control levels by day 9). Both tempol and vitamin E prevented Ang II-induced hypertension and either prevented or tended to blunt the increase in systemic and renal isoprostanes, TBARS, and ET. Finally, both antioxidants abolished the transient decrease in urinary nitrites. These results together with our previous study suggest that subpressor-dose Ang II increases oxidant stress (and isoprostanes). This in turn increases ET levels, which participate in the hypertensive response to Ang II.


Subject(s)
Angiotensin II/pharmacology , Antioxidants/pharmacology , Blood Pressure/drug effects , Endothelins/drug effects , Animals , Cyclic N-Oxides/pharmacology , Endothelin-1/blood , Endothelin-1/drug effects , Endothelins/blood , Glomerular Filtration Rate/drug effects , Kidney/blood supply , Kidney/physiopathology , Male , Nitrites/urine , Prostaglandins/blood , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Veins/drug effects , Renal Veins/metabolism , Renal Veins/physiopathology , Spin Labels , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin E/pharmacology
10.
Mayo Clin Proc ; 76(7): 758-60, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11444411

ABSTRACT

Enteric hyperoxaluria is a commonly seen adverse event after the jejunoileal bypass procedure. The increased concentration of urinary oxalate predisposes bypass patients to various renal complications such as nephrolithiasis and oxalate nephropathy. If not diagnosed and appropriately treated, these complications can lead to irreversible renal damage. We describe 3 patients in whom severe renal complications developed with irreversible compromise of renal function after a jejunoileal bypass. Patients who undergo a jejunoileal bypass require lifelong follow-up with close monitoring of their renal function. Marked decline in renal function mandates prompt investigation and aggressive intervention, including reversal of the jejunoileal bypass if necessary. Chronic renal failure secondary to oxalate nephropathy is preventable and treatable but may require conversion of a jejunoileal bypass to a more current form of bypass.


Subject(s)
Hyperoxaluria/etiology , Jejunoileal Bypass/adverse effects , Kidney Failure, Chronic/etiology , Oxalic Acid , Urinary Calculi/etiology , Aftercare , Causality , Chronic Disease , Humans , Hyperoxaluria/diagnosis , Hyperoxaluria/prevention & control , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/prevention & control , Male , Middle Aged , Recurrence , Urinary Calculi/diagnosis , Urinary Calculi/prevention & control
11.
Hypertension ; 37(2 Pt 2): 505-10, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230326

ABSTRACT

We tested the hypothesis that angiotensin II (Ang II)-induced stimulations of endothelin (ET) and isoprostanes are implicated in the slow pressor responses to Ang II. We infused either vehicle (group 1) or Ang II (groups 2 to 4) intravenously at 5 ng/kg per minute via osmotic pumps for 15 days into Sprague-Dawley rats. Groups 3 and 4 received 30 mg/kg per day of either losartan (Ang II type 1 receptor blocker) or bosentan (ET(A) and ET(B) receptor blocker) in their drinking water. We measured systolic blood pressure (SBP) every 3 days during the infusion. Plasma levels of Ang II, ET, isoprostanes, and urinary nitrites were determined at 15 days. Vehicle infusion did not change SBP (from 138+/-13 to 136+/-2 mm Hg at day 15). Circulating Ang II, ET, and isoprostane levels were 35+/-9, 39+/-3, and 111+/-10 pg/mL, respectively, whereas urinary nitrites were 2.3+/-0.4 microgram/d. Ang II increased SBP (from 133+/-10 to 158+/-8 mm Hg), plasma Ang II (179+/-77 pg/mL), and isoprostanes (156+/-19 pg/mL) without altering ET levels (38+/-5 pg/mL) or urinary nitrites (1.8+/-0.5 microgram/d). Losartan prevented Ang II-induced increases in SBP and isoprostanes (SBP went from 137+/-5 to 120+/-4 mm Hg; isoprostanes were 115+/-15 pg/mL) while increasing urinary nitrite levels (5.2+/-1.1 microgram/d). Losartan did not alter Ang II (141+/-57 pg/mL) or ET (40+/-4 pg/mL) levels. Bosentan also blocked Ang II-induced hypertension (from 135+/-4 to 139+/-3 mm Hg) but did not decrease isoprostanes (146+/-14 pg/mL). Ang II (63+/-11 pg/mL), ET levels (46+/-2 pg/mL), and urinary nitrites (2.8+/-0.4 microgram/d) were not altered. In conclusion, our results suggest that low-dose Ang II increases isoprostanes via its Ang II type 1 receptor and causes an ET-dependent hypertension, without altering circulating ET levels.


Subject(s)
Angiotensin II/pharmacology , Endothelins/metabolism , Prostaglandins/metabolism , Angiotensin II/antagonists & inhibitors , Angiotensin II/blood , Angiotensin Receptor Antagonists , Animals , Blood Pressure/drug effects , Bosentan , Endothelins/antagonists & inhibitors , Endothelins/blood , Glomerular Filtration Rate , Infusions, Intravenous , Losartan/pharmacology , Nitrites/blood , Nitrites/urine , Prostaglandins/blood , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Sulfonamides/pharmacology
12.
Kidney Int ; 58(4): 1632-40, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11012897

ABSTRACT

BACKGROUND: In hepatic cirrhosis, renal sodium and water retention can occur prior to decreases in renal blood flow (RBF). This may be explained in part by redistribution of the intrarenal microcirculation toward the juxtamedullary nephrons. To appreciate this three-dimensional spatial redistribution better, we examined the intrarenal microcirculatory changes using microcomputed tomography (micro-CT) in rats subjected to chronic bile duct ligation (CBDL). METHODS: Six kidneys from control rats and eight kidneys from rats that had undergone CBDL for 21 days were perfusion fixed in situ at physiological pressure, perfused with silicon-based Microfil containing lead chromate, embedded in plastic, and scanned by micro-CT. The microvasculature in the reconstructed three-dimensional renal images was studied using computerized image-analysis techniques. To determine the physiological condition of the rats, parallel experiments were conducted on six control and six CBDL rats to measure mean arterial pressure (MAP), RBF, glomerular filtration rate (GFR), urine flow (UF) rate, and sodium excretion by conventional methods. RESULTS: The percentage of vasculature in the renal cortex from CBDL rats was significantly decreased (10.8 +/- 0.4% vs. 16.8 +/- 2.7% control values). However, the vascular volume fractions of the medullary tissues were not significantly altered. There were no significant differences in the number of glomeruli between groups (36,430 +/- 1908 CBDLs, 36,609 +/- 3167 controls). The CBDL rats had a similar GFR than the controls but a reduced MAP, RBF, UF, and sodium excretion. CONCLUSIONS: The results indicate that after CBDL, there is a selective decrease in cortical vascular filling, which may contribute to the salt and water retention that accompanies cirrhosis.


Subject(s)
Cholestasis/diagnostic imaging , Hepatorenal Syndrome/diagnostic imaging , Tomography, X-Ray Computed/methods , Animals , Blood Pressure , Cholestasis/complications , Chronic Disease , Glomerular Filtration Rate , Hepatorenal Syndrome/etiology , Kidney Cortex/blood supply , Kidney Cortex/physiology , Ligation , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , Microcirculation , Rats , Rats, Sprague-Dawley , Renal Circulation/physiology , Sodium/metabolism , Water/metabolism
13.
Am J Kidney Dis ; 35(5): 941-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10793031

ABSTRACT

We report a woman with a history of allergies, polyuria, polydipsia, proteinuria, renal loss of electrolytes, renal tubular acidosis, nephrocalcinosis, and palpable purpura. A proximal defect was excluded by a normal bicarbonate reabsorption curve, and a distal tubular defect was shown because urine pH did not decrease to less than 6.4 despite ammonium chloride-induced systemic acidosis. Moreover, furosemide failed to improve urinary acidification. Urine-to-blood PCO(2) gradient was less than 14 mm Hg, although the urine bicarbonate level reached values as high as 89 mEq/L. Combining bicarbonate and neutral phosphate infusions increased the urine-to-blood PCO(2) gradient to only 20 mm Hg. These subnormal PCO(2) gradient values point to proton-pump dysfunction in the collecting tubule. Histological evidence of tubulointerstitial disease accompanied the tubular defects. The striking histological feature was the presence of immunoglobulin E (IgE) deposits in glomeruli, tubuli, and vessels. Concurrent with these findings, she had high serum IgE titers and CD23 levels. IgE antibodies from her serum were reactive against human renal tubuli, with binding to two regions that matched two different proteins present in cortex and medulla. One of these proteins corresponded to carbonic anhydrase II (31 kd); the second, to an unidentified protein that seems attached to cell membranes. We suggest that these IgE antibodies could have had a pathogenic role in this patient's glomerular, tubular, and small-vessel disease.


Subject(s)
Acidosis, Renal Tubular/etiology , Blood Vessels/metabolism , Immunoglobulin E/metabolism , Kidney/metabolism , Vasculitis/etiology , Acidosis, Renal Tubular/immunology , Adult , Female , Humans , Immunoglobulin E/blood , Vasculitis/immunology
14.
Am J Physiol Regul Integr Comp Physiol ; 278(1): R28-33, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644618

ABSTRACT

Cross-linked hemoglobin (XL-Hb) infused into dogs increases mean arterial pressure (MAP) but decreases blood flow to the renal (RBF), mesenteric (MBF), and iliac (IBF) circulations. These actions differ markedly from dextran infusion (which increases RBF, MBF, and IBF without altering MAP) and may be due to scavenging of nitric oxide by XL-Hb. However, because the hormonal milieu regulating regional circulation is altered during hemorrhage (when XL-Hb may be used), we studied whether systemic hemodynamics, RBF, MBF, IBF, and renal excretory function in hemorrhaged dogs was altered when resuscitated with XL-Hb compared with dextran (n = 6 each). Hemorrhage decreased MAP by 25% due to a 75% decline in cardiac output. RBF, MBF, and IBF all fell by 33, 64, and 72%, respectively (P<0.05 each). There was also a fall in glomerular filtration rate (GFR), urinary flow, and sodium excretion (P<0.05 each). After resuscitation, MAP, cardiac output, RBF, MBF, IBF, and GFR all recovered to basal values with either XL-Hb or dextran. Urinary flow and sodium excretion increased to above basal levels with dextran (both by 3.5-fold; P<0.05) or XL-Hb (by 7.5- and 10-fold, respectively; P<0.05). We conclude that resuscitation with XL-Hb after hemorrhage not only increases MAP, but also restores RBF, MBF, IBF, GFR, and urinary sodium and volume excretion analogously to dextran. The results contrast with those in normal dogs and suggest that nitric oxide inhibition does not impair hemodynamic and renal function recovery during hemorrhage.


Subject(s)
Aspirin/analogs & derivatives , Hemodynamics/drug effects , Hemoglobins/therapeutic use , Hemorrhage/physiopathology , Hemorrhage/therapy , Kidney/drug effects , Kidney/physiopathology , Resuscitation , Animals , Aspirin/therapeutic use , Cardiovascular System/drug effects , Dextrans/therapeutic use , Diuresis/drug effects , Dogs , Ilium/blood supply , Natriuresis/drug effects , Plasma Substitutes/therapeutic use , Regional Blood Flow/drug effects , Renal Circulation/drug effects , Splanchnic Circulation/drug effects
15.
Hypertension ; 34(4 Pt 2): 983-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523395

ABSTRACT

Chronic intravenous infusion of subpressor doses of angiotensin II causes blood pressure to increase progressively over the course of several days. The mechanisms underlying this response, however, are poorly understood. Because high-dose angiotensin II increases oxidative stress, and some compounds that result from the increased oxidative stress (eg, isoprostanes) produce vasoconstriction and antinatriuresis, we tested the hypothesis that a subpressor dose of angiotensin II also increases oxidative stress, as measured by 8-epi-prostaglandin F(2alpha) (isoprostanes), which may contribute to the slow pressor response to angiotensin II. To test this hypothesis, we infused angiotensin II (10 ng/kg per minute for 28 days via an osmotic pump) into 6 conscious normotensive female pigs (30 to 35 kg). We recorded mean arterial pressure continuously with a telemetry system and measured plasma isoprostanes before starting the angiotensin II infusion (baseline) and again after 28 days with an enzyme immunoassay. Angiotensin II infusion significantly increased mean arterial pressure from 121+/-4 to 153+/-7 mm Hg (P<0. 05) without altering total plasma isoprostane levels (180.0+/-24.3 versus 147.0+/-29.2 pg/mL; P=NS). However, the plasma concentrations of free isoprostanes increased significantly, from 38.3+/-5.8 to 54.7+/-10.4 pg/mL (P<0.05). These results suggest that subpressor doses of angiotensin II increase oxidative stress, as implied by the increased concentration of free isoprostanes, which accompany the elevation in mean arterial pressure elevation. Thus, isoprostane-induced vasoconstriction and antinatriuresis may contribute to the hypertension induced by the slow pressor responses of angiotensin II.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Dinoprost/analogs & derivatives , Vasoconstrictor Agents/pharmacology , Animals , Blood Pressure/physiology , Dinoprost/blood , F2-Isoprostanes , Female , Oxidative Stress , Swine
16.
Am J Kidney Dis ; 33(1): 43-51, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9915266

ABSTRACT

In congestive heart failure (CHF), the neurohormonal mechanisms that cause renal vasoconstriction, particularly those depending on the renin-angiotensin system, could interfere with renal vasodilating mechanisms. To elucidate this issue, we studied the kidney response to an amino acid infusion (known to cause renal vasodilation in healthy individuals) in eight patients with CHF. We found that the amino acid infusion (0.7 mL/kg/h of a 10% solution) elicited no renal hemodynamic response, in marked contrast to healthy subjects. We next hypothesized that the renin-angiotensin system (known to be activated in heart failure) has a role in the lack of response to the amino acid infusion. To test this hypothesis, we repeated the study after two 5-mg doses of enalapril, an inhibitor of the angiotensin-converting enzyme, administered 12 hours apart. After enalapril treatment, the amino acid infusion caused a 45% increase in mean renal blood flow (RBF) from 383 +/- 55 to 557 +/- 51 mL/min at the fifth hour (P < 0.05). This normalization of the renal response to the amino acid infusion occurred without changes in cardiac output or in systemic vascular resistance. Hence, the renal fraction of the cardiac output increased during the amino acid infusion. The recovery of the renal vascular response was not accompanied by an increase in glomerular filtration rate (GFR; filtration fraction decreased), suggesting a predominant efferent arteriole dilatation. Our study shows that, in heart failure, the kidney loses its ability to increase RBF in response to an amino acid load. This lack of renal vascular response can be restored by inhibiting the renin-angiotensin system and is unrelated to changes in systemic hemodynamics.


Subject(s)
Amino Acids/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Enalapril/administration & dosage , Heart Failure/physiopathology , Renal Artery/drug effects , Vasodilation/drug effects , Aged , Analysis of Variance , Female , Heart Failure/metabolism , Hemodynamics/drug effects , Hemodynamics/physiology , Humans , Infusions, Intravenous , Male , Middle Aged , Renal Artery/physiopathology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Time Factors , Vasodilation/physiology
18.
Hypertension ; 32(2): 287-92, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9719056

ABSTRACT

Bradykinin plays an important role in the regulation of renal hemodynamics. However, there have been few studies of the effect of bradykinin on isolated afferent arterioles, vascular segments that are important for the regulation of renal blood flow and glomerular filtration rate. Our purpose was to study (1) the effects of bradykinin on isolated perfused rabbit afferent arterioles and (2) the mechanisms of actions. Afferent arterioles dissected from rabbits were perfused in vitro at 60 mm Hg. In afferent arterioles preconstricted with phenylephrine, 10(-12) to 10(-10) mol/L bradykinin increased luminal diameter from 9.0+/-1.0 to 14.3+/-1.2 microm (P<0.003). In contrast, 10(-9) and 10(-8) mol/L bradykinin decreased luminal diameter to 10.8+/-1.4 and 9.7+/-1.2 microm, respectively (P<0.001). Bradykinin added to the bath had no effect on preconstricted afferent arterioles. The addition of [des-Arg9]-bradykinin (10(-9) and 10(-8) mol/L), a B1 receptor agonist, to the lumen decreased diameter from 9.7+/-1.2 to 6.7+/-1.2 microm at 10(-8) mol/L (P<0.002). Icatibant (Hoe 140), a B2 receptor antagonist, blocked both the vasodilation and vasoconstriction induced by bradykinin as well as the vasoconstriction induced by [des-Arg9]-bradykinin. L-NAME had no effect on bradykinin-induced dilation or constriction. Indomethacin blocked both the dilation induced by 10(-12) to 10(-10) mol/L bradykinin and the constriction induced by 10(-9) to 10(-8) mol/L bradykinin. In fact, in the presence of indomethacin, 10(-9) and 10(-8) mol/L bradykinin increased luminal diameter from 6.2+/-0.7 to 10.7+/-0.6 microm at 10(-8) mol/L (P<0.001), which was attenuated by L-NAME. Finally, in the presence of SQ29548, a prostaglandin H2/thromboxane A2 receptor antagonist, bradykinin caused dilation at all concentrations tested. In conclusion, bradykinin has a biphasic effect on afferent arterioles. Both dilation and constriction may be mediated by bradykinin B2 receptors. The mechanisms of vasodilation and vasoconstriction are due to cyclooxygenase products, not nitric oxide.


Subject(s)
Arterioles/drug effects , Bradykinin/pharmacology , Renal Circulation/drug effects , Renal Circulation/physiology , Vasodilation/drug effects , Animals , Arterioles/physiology , Bridged Bicyclo Compounds, Heterocyclic , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated , Hydrazines/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Rabbits
19.
Hypertension ; 30(3 Pt 2): 672-6, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9323003

ABSTRACT

While arterial hypertension and renal dysfunction are well recognized complications of renal irradiation, the mechanisms that trigger the development of these complications are unknown. Recently, it was reported that the endothelium is a major target in radiation injury. Because dysfunction of the endothelial cells may lead or contribute to the development of hypertension and renal dysfunction in radiation nephropathy, we tested the hypothesis that endothelium-dependent vasodilation is impaired in radiated kidneys prior to the onset of hypertension. To test this hypothesis, we used Long-Evans rats that had undergone left nephrectomy (3 weeks earlier) and irradiation (3000 r's) to the right kidney 8 days earlier (mean blood pressures in the irradiated rats were not different than in the controls). We then measured the changes in renal blood flow (RBF) induced by endothelium-dependent (acetylcholine and bradykinin) and -independent (nitroprusside, norepinephrine, and angiotensin II) vasoactive agents. We found that the increases in RBF induced by the endothelium-dependent but not independent vasodilators were markedly impaired in the irradiated kidneys. Blocking nitric oxide synthesis with nitro L-arginine methyl ester in sham rats mimicked the blunted responsiveness of the irradiated rats, whereas indomethacin (an inhibitor of prostaglandin synthesis) had no effect on either sham or irradiated rats. Finally, the RBF responses to the endothelium-independent vasoconstrictors, norepinephrine and angiotensin II, were not altered in the irradiated kidneys. These results suggest that renal irradiation causes endothelial dysfunction (prior to the onset of hypertension) but spares the vascular smooth muscle cells.


Subject(s)
Endothelium, Vascular/radiation effects , Kidney/radiation effects , Acetylcholine/pharmacology , Angiotensin II/pharmacology , Animals , Blood Pressure/radiation effects , Endothelium, Vascular/physiology , Indomethacin/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Renal Circulation/radiation effects , Vasodilation/drug effects
20.
Kidney Int ; 49(2): 374-81, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8821820

ABSTRACT

We have recently presented evidence that endogenous nitric oxide (NO) and prostaglandins (PGs) modulate angiotensin II (Ang II) action in microperfused afferent arterioles (Af-Arts). Because flow may be a physiological stimulus of endothelial release of NO and PGs, we tested the hypothesis that flow through the lumen of the Af-Art stimulates the endothelium to produce NO and PGs, which in turn modulate the action of Ang II. We microdissected the terminal segment of an interlobular artery together with two Af-Arts, their glomeruli and efferent arterioles (Ef-Art). The two Af-Arts were perfused simultaneously from the interlobular artery, while one Ef-Art was occluded. Since the arteriolar perfusate contained 5% albumin, oncotic pressure built up in the glomerulus with the occluded Ef-Art and opposed the force of filtration, resulting in little or no flow through the corresponding Af-Art. Thus this preparation allowed us to observe Ang II action in free-flow and non-flow Af-Arts simultaneously. Ang II-induced constriction was weaker in free-flow than non-flow Af-Arts, with the luminal diameter decreasing by 8 +/- 2% and 23 +/- 3% at 10(-9) M, respectively (P < 0.013 free-flow vs. non-flow; N = 9). Disrupting the endothelium augmented Ang II action in free-flow (33 +/- 5.1%; P < 0.01 vs. intact endothelium) but not non-flow Af-Arts (31 +/- 5.3%), thus abolishing the differences between them (N = 8). Pretreatment with an inhibitor of either NO synthase (N-nitro-L-arginine methyl ester) or cyclooxygenase (indomethacin) augmented Ang II action more in free-flow than non-flow Af-Arts, likewise abolishing the differences between them. These results suggest that intraluminal flow modulates the vasoconstrictor action of Ang II in Af-Arts via endothelium-derived NO and PGs. Thus flow may be important in the fine control of glomerular hemodynamics.


Subject(s)
Angiotensin II/pharmacology , Kidney Glomerulus/blood supply , Perfusion/methods , Animals , Arterioles/drug effects , Arterioles/physiology , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Endothelium/metabolism , Factor VIII/antagonists & inhibitors , In Vitro Techniques , Indomethacin/pharmacology , Kidney Glomerulus/cytology , Kidney Glomerulus/enzymology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Norepinephrine/pharmacology , Rabbits , Regional Blood Flow
SELECTION OF CITATIONS
SEARCH DETAIL
...