1
|
Wilcox CS, Herbert C, Wang C, Ma Y, Sun P, Li T, Verbesey J, Kumar P, Kassaye S, Welch WJ, Choi MJ, Pourafshar N, Wang D. Signals From Inflamed Perivascular Adipose Tissue Contribute to Small-Vessel Dysfunction in Women With Human Immunodeficiency Virus. J Infect Dis 2024; 230:67-77. [PMID: 39052698 PMCID: PMC11272057 DOI: 10.1093/infdis/jiae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND People with the human immunodeficiency virus (PWH) have microvascular disease. Because perivascular adipose tissue (PVAT) regulates microvascular function and adipose tissue is inflamed in PWH, we tested the hypothesis that PWH have inflamed PVAT that impairs the function of their small vessels. METHODS Subcutaneous small arteries were dissected with or without PVAT from a gluteal skin biopsy from 11 women with treated HIV (WWH) aged < 50 years and 10 matched women without HIV, and studied on isometric myographs. Nitric oxide (NO) and reactive oxygen species (ROS) were measured by fluorescence microscopy. Adipokines and markers of inflammation and ROS were assayed in PVAT. RESULTS PVAT surrounding the small arteries in control women significantly (P < .05) enhanced acetylcholine-induced endothelium-dependent relaxation and NO, and reduced contractions to thromboxane and endothelin-1. However, these effects of PVAT were reduced significantly (P < .05) in WWH whose PVAT released less adiponectin but more markers of ROS and inflammation. Moderation of contractions by PVAT were correlated positively with adipose adiponectin. CONCLUSIONS PVAT from WWH has oxidative stress, inflammation, and reduced release of adiponectin, which may contribute to enhanced contractions and therefore could promote small-artery dysfunction.
Collapse
Affiliation(s)
- Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Carly Herbert
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study Combined Cohort Study, Georgetown University, Washington, District of Columbia, USA
| | - Cheng Wang
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Division of Nephrology, The Fifth Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yuchi Ma
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Philena Sun
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Tian Li
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Jennifer Verbesey
- MedStar Georgetown Transplant Institute, Washington, District of Columbia, USA
| | - Princy Kumar
- Division of Infection Disease, Georgetown University, Washington, District of Columbia, USA
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study, Georgetown University, Washington, District of Columbia, USA
| | - Seble Kassaye
- Division of Infection Disease, Georgetown University, Washington, District of Columbia, USA
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study, Georgetown University, Washington, District of Columbia, USA
| | - William J Welch
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Michael J Choi
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Medstar Georgetown University Hospital, Department of Nephrology and Hypertension, Washington, District of Columbia, USA
| | - Negiin Pourafshar
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Medstar Georgetown University Hospital, Department of Nephrology and Hypertension, Washington, District of Columbia, USA
| | - Dan Wang
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
2
|
Stevenson MD, Vendrov AE, Yang X, Chen Y, Navarro HA, Moss N, Runge MS, Arendshorst WJ, Madamanchi NR. Reactivity of renal and mesenteric resistance vessels to angiotensin II is mediated by NOXA1/NOX1 and superoxide signaling. Am J Physiol Renal Physiol 2023; 324:F335-F352. [PMID: 36759130 PMCID: PMC10026993 DOI: 10.1152/ajprenal.00236.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/17/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Activation of NADPH oxidase (NOX) enzymes and the generation of reactive oxygen species and oxidative stress regulate vascular and renal function and contribute to the pathogenesis of hypertension. The present study examined the role of NOXA1/NOX1 function in vascular reactivity of renal and mesenteric resistance arteries/arterioles of wild-type and Noxa1-/- mice. A major finding was that renal blood flow is less sensitive to acute stimulation by angiotensin II (ANG II) in Noxa1-/- mice compared with wild-type mice, with a direct action on resistance arterioles independent of nitric oxide (NO) bioavailability. These functional results were reinforced by immunofluorescence evidence of NOXA1/NOX1 protein presence in renal arteries, afferent arterioles, and glomeruli as well as their upregulation by ANG II. In contrast, the renal vascular response to the thromboxane mimetic U46619 was effectively blunted by NO and was similar in both mouse genotypes and thus independent of NOXA1/NOX1 signaling. However, phenylephrine- and ANG II-induced contraction of isolated mesenteric arteries was less pronounced and buffering of vasoconstriction after acetylcholine and nitroprusside stimulation was reduced in Noxa1-/- mice, suggesting endothelial NO-dependent mechanisms. An involvement of NOXA1/NOX1/O2•- signaling in response to ANG II was demonstrated with the specific NOXA1/NOX1 assembly inhibitor C25 and the nonspecific NOX inhibitor diphenyleneiodonium chloride in cultured vascular smooth muscle cells and isolated mesenteric resistance arteries. Collectively, our data indicate that the NOX1/NOXA1/O2•- pathway contributes to acute vasoconstriction induced by ANG II in renal and mesenteric vascular beds and may contribute to ANG II-induced hypertension.NEW & NOTEWORTHY Renal reactivity to angiotensin II (ANG II) is mediated by superoxide signaling produced by NADPH oxidase (NOX)A1/NOX1. Acute vasoconstriction of renal arteries by ANG was blunted in Noxa1-/- compared with wild-type mice. NOXA1/NOX1/O2•- signaling was also observed in ANG II stimulation of vascular smooth muscle cells and isolated mesenteric resistance arteries, indicating that it contributes to ANG II-induced hypertension. A NOXA1/NOX1 assembly inhibitor (C25) has been characterized that inhibits superoxide production and ameliorates the effects of ANG II.
Collapse
Affiliation(s)
- Mark D Stevenson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Aleksandr E Vendrov
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Xi Yang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Yuenmu Chen
- McAllister Heart Institute, Division of Cardiology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Hernán A Navarro
- Center for Drug Discovery, Organic and Medicinal Chemistry, RTI International, Research Triangle Park, North Carolina, United States
| | - Nicholas Moss
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Marschall S Runge
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - William J Arendshorst
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Nageswara R Madamanchi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
3
|
Moreno C, Llinás MT, Rodriguez F, Moreno JM, Salazar FJ. Nitric oxide, prostaglandins and angiotensin II in the regulation of renal medullary blood flow during volume expansion. J Physiol Biochem 2015; 72:1-8. [PMID: 26611113 DOI: 10.1007/s13105-015-0450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 11/17/2015] [Indexed: 11/29/2022]
Abstract
Regulation of medullary blood flow (MBF) is essential in maintaining renal function and blood pressure. However, it is unknown whether outer MBF (OMBF) and papillary blood flow (PBF) are regulated independently when extracellular volume (ECV) is enhanced. The aim of this study was to determine whether OMBF and PBF are differently regulated and whether there is an interaction between nitric oxide (NO), prostaglandins (PGs) and angiotensin II (Ang II) in regulating OMBF and PBF when ECV is enhanced. To achieve these goals, OMBF and PBF were measured by laser-Doppler in volume-expanded rats treated with a cyclooxygenase inhibitor (meclofenamate, 3 mg/kg) and/or a NO synthesis inhibitor (L-nitro-arginine methyl ester (L-NAME), 3 μg/kg/min) and/or Ang II (10 ng/kg/min). OMBF was unchanged by NO or PGs synthesis inhibition but decreased by 36 % (P < 0.05) when L-NAME and meclofenamate were infused simultaneously. PBF was similarly reduced by L-NAME (12 %), meclofenamate (17 %) or L-NAME + meclofenamate (19 %). Ang II did not modify OMBF, but it led to a similar decrease (P < 0.05) in OMBF when it was administered to rats with reduced NO (32 %), PGs (36 %) or NO and PGs (37 %) synthesis. In contrast, the fall in PBF induced by Ang II (12 %) was enhanced (P < 0.05) by the simultaneous PGs (30 %) or PGs and NO (31 %) synthesis inhibition but not in L-NAME-treated rats (20 %). This study presents novel findings suggesting that blood flows to the outer medulla and renal papilla are differently regulated and showing that there is a complex interaction between NO, PGs and Ang II in regulating OMBF and PBF when ECV is enhanced.
Collapse
Affiliation(s)
- Carol Moreno
- Cardiovascular and Metabolic Diseases, MedImmune, Cambridge, UK
| | - María T Llinás
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - Francisca Rodriguez
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - Juan M Moreno
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain.,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain.,Instituto Murciano de Investigación Biomédica, Murcia, Spain
| | - F Javier Salazar
- Department of Physiology, School of Medicine, University of Murcia, Murcia, 30100, Spain. .,Regional Campus of International Excellence "Mare Nostrum", Murcia, Spain. .,Instituto Murciano de Investigación Biomédica, Murcia, Spain.
| |
Collapse
|
4
|
Abstract
SIGNIFICANCE Renal oxidative stress can be a cause, a consequence, or more often a potentiating factor for hypertension. Increased reactive oxygen species (ROS) in the kidney have been reported in multiple models of hypertension and related to renal vasoconstriction and alterations of renal function. Nicotinamide adenine dinucleotide phosphate oxidase is the central source of ROS in the hypertensive kidney, but a defective antioxidant system also can contribute. RECENT ADVANCES Superoxide has been identified as the principal ROS implicated for vascular and tubular dysfunction, but hydrogen peroxide (H2O2) has been implicated in diminishing preglomerular vascular reactivity, and promoting medullary blood flow and pressure natriuresis in hypertensive animals. CRITICAL ISSUES AND FUTURE DIRECTIONS Increased renal ROS have been implicated in renal vasoconstriction, renin release, activation of renal afferent nerves, augmented contraction, and myogenic responses of afferent arterioles, enhanced tubuloglomerular feedback, dysfunction of glomerular cells, and proteinuria. Inhibition of ROS with antioxidants, superoxide dismutase mimetics, or blockers of the renin-angiotensin-aldosterone system or genetic deletion of one of the components of the signaling cascade often attenuates or delays the onset of hypertension and preserves the renal structure and function. Novel approaches are required to dampen the renal oxidative stress pathways to reduced O2(-•) rather than H2O2 selectivity and/or to enhance the endogenous antioxidant pathways to susceptible subjects to prevent the development and renal-damaging effects of hypertension.
Collapse
Affiliation(s)
- Magali Araujo
- Hypertension, Kidney and Vascular Research Center, Georgetown University , Washington, District of Columbia
| | | |
Collapse
|
5
|
Zhang J, Chandrashekar K, Lu Y, Duan Y, Qu P, Wei J, Juncos LA, Liu R. Enhanced expression and activity of Nox2 and Nox4 in the macula densa in ANG II-induced hypertensive mice. Am J Physiol Renal Physiol 2013; 306:F344-50. [PMID: 24285500 DOI: 10.1152/ajprenal.00515.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
NAD(P)H oxidase (Nox)2 and Nox4 are the isoforms of Nox expressed in the macula densa (MD). MD-derived superoxide (O₂⁻), primarily generated by Nox2, is enhanced by acute ANG II stimulation. However, the effects of chronic elevations in ANG II during ANG II-induced hypertension on MD-derived O₂⁻ are unknown. We infused a slow pressor dose of ANG II (600 ng·min⁻¹·kg⁻¹) for 2 wk in C57BL/6 mice and found that mean arterial pressure was elevated by 22.3 ± 3.4 mmHg (P < 0.01). We measured O₂⁻ generation in isolated and perfused MDs and found that O₂⁻ generation by the MD was increased from 9.4 ± 0.9 U/min in control mice to 34.7 ± 1.8 U/min in ANG II-induced hypertensive mice (P < 0.01). We stimulated MMDD1 cells, a MD-like cell line, with ANG II and found that O₂⁻ generation increased from 921 ± 91 to 3,687 ± 183 U·min⁻¹·10⁵ cells⁻¹, which was inhibited with apocynin, oxypurinol, or NS-398 by 46%, 14%, and 12%, respectively. We isolated MD cells using laser capture microdissection and measured mRNA levels of Nox. Nox2 and Nox4 levels increased by 3.7 ± 0.17- and 2.6 ± 0.15-fold in ANG II-infused mice compared with control mice. In MMDD1 cells treated with Nox2 or Nox4 small interfering (si)RNAs, ANG II-stimulated O₂⁻ generation was blunted by 50% and 41%, respectively. In cells treated with p22(phox) siRNA, ANG II-stimulated O₂⁻ generation was completely blocked. In conclusion, we found that a subpressor dose of ANG II enhances O₂⁻ generation in the MD and that the sources of this O₂⁻ are primarily Nox2 and Nox4.
Collapse
Affiliation(s)
- Jie Zhang
- Dept. of Physiology and Biophysics, Univ. of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
In vivo, label-free, three-dimensional quantitative imaging of kidney microcirculation using Doppler optical coherence tomography. J Transl Med 2011; 91:1596-604. [PMID: 21808233 PMCID: PMC3312876 DOI: 10.1038/labinvest.2011.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Doppler optical coherence tomography (DOCT) is a functional extension of optical coherence tomography (OCT) and is currently being employed in several clinical arenas to quantify blood flow in vivo. In this study, the objective was to investigate the feasibility of DOCT to image kidney microcirculation, specifically, glomerular blood flow. DOCT is able to capture three-dimensional (3D) data sets consisting of a series of cross-sectional images in real time, which enables label-free and non-destructive quantification of glomerular blood flow. The kidneys of adult, male Munich-Wistar rats were exposed through laparotomy procedure after being anesthetized. Following exposure of the kidney beneath the DOCT microscope, glomerular blood flow was observed. The effects of acute mannitol and angiotensin II infusion were also observed. Glomerular blood flow was quantified for the induced physiological states and compared with baseline measurements. Glomerular volume, cumulative Doppler volume, and Doppler flow range parameters were computed from 3D OCT/DOCT data sets. Glomerular size was determined from OCT, and DOCT readily revealed glomerular blood flow. After infusion of mannitol, a significant increase in blood flow was observed and quantified, and following infusion of angiontensin II, a significant decrease in blood flow was observed and quantified. Also, blood flow histograms were produced to illustrate differences in blood flow rate and blood volume among the induced physiological states. We demonstrated 3D DOCT imaging of rat kidney microcirculation in the glomerulus in vivo. Dynamic changes in blood flow were detected under altered physiological conditions demonstrating the real-time imaging capability of DOCT. This method holds promise to allow non-invasive imaging of kidney blood flow for transplant graft evaluation or monitoring of altered-renal hemodynamics related to disease progression.
Collapse
|
7
|
Polichnowski AJ, Lu L, Cowley AW. Renal injury in angiotensin II+L-NAME-induced hypertensive rats is independent of elevated blood pressure. Am J Physiol Renal Physiol 2011; 300:F1008-16. [PMID: 21270093 DOI: 10.1152/ajprenal.00354.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The balance between angiotensin II (ANG II) and nitric oxide plays an important role in renal function and is thought to contribute to the progression of renal injury in experimental hypertension. In the present study, we investigated the extent of blood pressure (BP)-dependent and BP-independent pathways of renal injury following 2 wk of hypertension produced by intravenous infusion of ANG II (5 ng·kg⁻¹·min⁻¹)+N(ω)-nitro-l-arginine methyl ester (l-NAME; 1.4 μg·kg⁻¹·min⁻¹) in male Sprague-Dawley rats. An aortic balloon occluder was positioned between the renal arteries to maintain (24 h/day) BP to the left kidney (servo-controlled) at baseline levels, whereas the right kidney (uncontrolled) was chronically exposed to elevated BP. Over the 14-day experimental protocol, the average BP to uncontrolled kidneys (152.7 ± 1.8 mmHg) was significantly elevated compared with servo-controlled (113.0 ± 0.2 mmHg) kidneys and kidneys from sham rats (108.3 ± 0.1 mmHg). ANG II+l-NAME infusion led to renal injury that was focal in nature and mainly confined to the outer medulla. Despite the differences in BP between servo-controlled and uncontrolled kidneys, there was a similar ~3.5-fold increase in renal outer medullary tubular injury, ~2-fold increase in outer medullary interstitial fibrosis, ~2-fold increase in outer medullary macrophage infiltration, and a significant increase in renal oxidative stress, all of which are indicative of BP-independent mediated pathways. The results of this study have important implications regarding the pathogenesis of renal injury in various experimental models of hypertension and provide novel insights regarding the variable association observed between hypertension and renal injury in some human populations.
Collapse
Affiliation(s)
- Aaron J Polichnowski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | |
Collapse
|
8
|
Obesity is the major contributor to vascular dysfunction and inflammation in high-fat diet hypertensive rats. Clin Sci (Lond) 2010; 118:291-301. [PMID: 19728860 DOI: 10.1042/cs20090395] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 08/27/2009] [Accepted: 09/03/2009] [Indexed: 12/27/2022]
Abstract
Obesity and hypertension are the two major risk factors that contribute to the progression of end-stage renal disease. To examine whether hypertension further exacerbates oxidative stress and vascular dysfunction and inflammation in obese rats, four groups of male Sprague-Dawley rats were fed either a normal (7% fat) or high-fat (36% fat) diet for 6 weeks and osmotic pumps were implanted to deliver ANG (angiotensin II) or vehicle for an additional 4 weeks.Treatment with the high-fat diet did not alter ANG-induced hypertension compared with the normal diet (174 +/- 6 compared with 170 +/- 5 mmHg respectively). Treatment with the high-fat diet increased body weight gain and plasma leptin levels and induced insulin resistance in normotensive and ANG-induced hypertensive rats. Plasma TBARS (thiobarbituric acid-reacting substances), a measure of oxidative stress, were elevated in high-fat diet-fed rats compared with controls (11.2 +/-1 compared with 8.4 +/- nmol/ml respectively) and was increased further in ANG-induced hypertensive rats fed a high-fat diet (18.8 +/-2.2 nmol/ml). Urinary nitrite excretion was also decreased in rats fed a high-fat diet without or with ANG infusion compared with controls. Afferent arteriolar relaxation to acetylcholine was impaired in rats fed the high-fat diet without or with ANG infusion. Renal cortical TNF-alpha(tumour necrosis factor-alpha), COX-2(cyclo-oxygenase-2) and phospho-IKK (inhibitor of nuclear factor k B kinase) expression increased in high-fat diet-fed rats compared with normal diet-fed rats. The increases in phospho-IKK and COX-2 expression were elevated further in ANG-induced hypertensive rats fed the high-fat diet.These results suggest that ANG-induced hypertension exacerbates oxidative stress and renal inflammation without further impairment in vascular dysfunction in high-fat diet-induced obesity.
Collapse
|
9
|
Sasser JM, Moningka NC, Cunningham MW, Croker B, Baylis C. Asymmetric dimethylarginine in angiotensin II-induced hypertension. Am J Physiol Regul Integr Comp Physiol 2009; 298:R740-6. [PMID: 20018820 DOI: 10.1152/ajpregu.90875.2008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent studies have shown that asymmetric dimethylarginine (ADMA), a nitric oxide synthase inhibitor, is increased in hypertension and chronic kidney disease. However, little is known about the effects of hypertension per se on ADMA metabolism. The purpose of this study was to test the hypothesis that ANG II-induced hypertension, in the absence of renal injury, is associated with increased oxidative stress and plasma and renal cortex ADMA levels in rats. Male Sprague-Dawley rats were treated with ANG II at 200 ng.kg(-1).min(-1) sc (by minipump) for 1 or 3 wk or at 400 ng.kg(-1).min(-1) for 6 wk. Mean arterial pressure was increased after 3 and 6 wk of ANG II; however, renal injury (proteinuria, glomerular sclerosis, and interstitial fibrosis) was only evident after 6 wk of treatment. Plasma thiobarbituric acid reactive substances concentration and renal cortex p22(phox) protein abundance were increased early (1 and 3 wk), but urinary excretion of isoprostane and H(2)O(2) was only increased after 6 wk of ANG II. An increased in plasma ADMA after 6 wk of ANG II was associated with increased lung protein arginine methyltransferase-1 abundance and decreased renal cortex dimethylarginine dimethylaminohydrolase activity. No changes in renal cortex ADMA were observed. ANG II hypertension in the absence of renal injury is not associated with increased ADMA; however, when the severity and duration of the treatment were increased, plasma ADMA increased. These data suggest that elevated blood pressure alone, for up to 3 wk, in the absence of renal injury does not play an important role in the regulation of ADMA. However, the presence of renal injury and sustained hypertension for 6 wk increases ADMA levels and contributes to nitric oxide deficiency and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer M Sasser
- Dept. of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | |
Collapse
|
10
|
Hultström M, Helle F, Iversen BM. AT1 receptor activation regulates the mRNA expression of CAT1, CAT2, arginase-1, and DDAH2 in preglomerular vessels from angiotensin II hypertensive rats. Am J Physiol Renal Physiol 2009; 297:F163-8. [DOI: 10.1152/ajprenal.00087.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Previously, we found increased expression of l-arginine metabolizing enzymes in both kidneys from two-kidney, one-clip (2K1C) hypertensive rats (Helle F, Hultstrom M, Skogstrand T, Palm F, Iversen BM. Am J Physiol Renal Physiol 296: F78–F86, 2009). In the present study, we investigate whether AT1 receptor activation can induce the changes observed in 2K1C. Four groups of rats were infused with 80 ng/min ANG II or saline for 14 days and/or given 60 mg·kg−1·day−1 losartan. Gene expression was studied in isolated preglomerular vessels by RT-PCR. Dose-responses to ANG II were studied in isolated preglomerular vessels with and without acute NOS inhibition [10−4 mol/l NG-nitro-l-arginine methyl ester (l-NAME)]. Expressions of endothelial nitric oxide synthase (eNOS), caveolin-1, and arginase-2 were not changed by ANG II infusion. CAT1 (0.3 8 ± 0.07 to 0.73 ± 0.12, P < 0.05), CAT2 (1.14 ± 0.29 to 2.74 ± 0.48), DDAH2 (1.09 ± 0.27 to 2.3 ± 0.46), and arginase-1 (1.08 ± 0.17 to 1.82 ± 0.22) were increased in ANG II-infused rats. This was prevented by losartan treatment, which reduced the expression of eNOS (0.97 ± 0.26 to 0.37 ± 0.11 in controls; 0.8 ± 0.16 to 0.36 ± 0.1 in ANG II-infused rats) and caveolin-1 (2.49 ± 0.59 to 0.82 ± 0.24 in controls and 2.59 ± 0.61 to 1.1 ± 0.25 in ANG II-infused rats). ANG II (10−10 mol/l) caused vessels from ANG II-infused animals to contract to 53 ± 15% of baseline diameter and 90 ± 5% of baseline diameter in controls ( P < 0.05) and was further enhanced by l-NAME to 4 ± 4% of baseline diameter ( P < 0.05). In vivo losartan treatment reduced the reactivity of isolated vessels to 91 ± 2% of baseline in response to 10−7 mol/l ANG II compared with 82 ± 3% in controls ( P < 0.05) and prevented the increased responsiveness caused by ANG II infusion. In conclusion, CAT1, CAT2, DDAH2, and arginase-1 expression in renal resistance vessels is regulated through the AT1 receptor. This finding may be of direct importance for NOS and the regulation of preglomerular vascular function.
Collapse
|
11
|
Therrien F, Lemieux P, Bélanger S, Agharazii M, Lebel M, Larivière R. Protective effects of angiotensin AT1 receptor blockade in malignant hypertension in the rat. Eur J Pharmacol 2009; 607:126-34. [DOI: 10.1016/j.ejphar.2009.01.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
12
|
Palm F, Onozato ML, Luo Z, Wilcox CS. Dimethylarginine dimethylaminohydrolase (DDAH): expression, regulation, and function in the cardiovascular and renal systems. Am J Physiol Heart Circ Physiol 2007; 293:H3227-45. [PMID: 17933965 DOI: 10.1152/ajpheart.00998.2007] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Asymmetric (N(G),N(G))-dimethylarginine (ADMA) inhibits nitric oxide (NO) synthases (NOS). ADMA is a risk factor for endothelial dysfunction, cardiovascular mortality, and progression of chronic kidney disease. Two isoforms of dimethylarginine dimethylaminohydrolase (DDAH) metabolize ADMA. DDAH-1 is the predominant isoform in the proximal tubules of the kidney and in the liver. These organs extract ADMA from the circulation. DDAH-2 is the predominant isoform in the vasculature, where it is found in endothelial cells adjacent to the cell membrane and in intracellular vesicles and in vascular smooth muscle cells among the myofibrils and the nuclear envelope. In vivo gene silencing of DDAH-1 in the rat and DDAH +/- mice both have increased circulating ADMA, whereas gene silencing of DDAH-2 reduces vascular NO generation and endothelium-derived relaxation factor responses. DDAH-2 also is expressed in the kidney in the macula densa and distal nephron. Angiotensin type 1 receptor activation in kidneys reduces the expression of DDAH-1 but increases the expression of DDAH-2. This rapidly evolving evidence of isoform-specific distribution and regulation of DDAH expression in the kidney and blood vessels provides potential mechanisms for nephron site-specific regulation of NO production. In this review, the recent advances in the regulation and function of DDAH enzymes, their roles in the regulation of NO generation, and their possible contribution to endothelial dysfunction in patients with cardiovascular and kidney diseases are discussed.
Collapse
Affiliation(s)
- Fredrik Palm
- Division of Nephrology and Hypertension, Georgetown University, 3800 Reservoir Road N.W., Washington, DC 20007, USA
| | | | | | | |
Collapse
|
13
|
Abstract
Extracellular matrix (ECM) remodeling with successive tissue fibrosis is a key feature of chronic cardiovascular diseases, including atherosclerosis and restenosis. The atherogenic changes underlying these pathologies result from chronification of an acute repair response towards injurious and inflammatory stimuli. Thereby functional tissue is replaced by excessive ECM deposition. In the kidney, impaired remodeling is a major cause of perivascular, interstitial, and glomerular fibrosis but also a common complication of chronic hypertension. Experimental evidence points to the matrix metalloproteases (MMPs) and their intrinsic inhibitors, the tissue inhibitors of MMPs as key mediators of atherogenic and fibrotic pathologies. Mechanistically, a deregulation in ECM turnover tightly correlates with an increased production and release of proinflammatory and profibrotic factors including interleukin-1beta, transforming growth factor beta, angiotensin II, and reactive oxygen species. Unlike these factors the pleiotropic messenger molecule nitric oxide (NO) by acting as the major physiological vasodilator has emerged as one of the most atheroprotective factors. However, under inflammatory conditions NO does acquire proatherogenic and profibrotic properties thereby exacerbating tissue fibrosis. In this review, the mechanisms underlying both opposing properties of NO on perivascular ECM remodeling will exemplarily be discussed for renal fibrosis with a particular focus on the MMPs and intrinsic protease inhibitors.
Collapse
Affiliation(s)
- W Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7 Frankfurt am Main, Germany.
| | | |
Collapse
|
14
|
Brands MW, Bell TD, Fleming C, Labazi H, Sturgis LC. LACK OF BLOOD PRESSURE SALT-SENSITIVITY SUPPORTS A PREGLOMERULAR SITE OF ACTION OF NITRIC OXIDE IN TYPE I DIABETIC RATS. Clin Exp Pharmacol Physiol 2007; 34:475-9. [PMID: 17439418 DOI: 10.1111/j.1440-1681.2007.04597.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
1. The relationship between sodium intake and blood pressure is affected differently by changes in angiotensin (Ang) II and preglomerular resistance, and this study measured that relationship to evaluate the link between nitric oxide and blood pressure early in diabetes. 2. Rats were chronically instrumented, placed on high-sodium (HS = 12 mEq/d) or low-sodium (LS = 0.07 mEq/d) intake diets and assigned to either vehicle- (V) or Nomega-nitro-L-arginine methyl ester- (L-NAME; L) treated groups. Mean arterial pressure (MAP) was measured 18 h/day for a 6-day control and 14-day streptozotocin diabetic period in each animal. 3. The MAP of the control period averaged 95 +/- 1 and 94 +/- 1 mmHg in the LSV and HSV rats and 116 +/- 2 and 124 +/- 1 mmHg in the LSL and HSL rats, respectively (LSL vs HSL was significant at P < 0.05). Diabetes increased MAP only in the LSL and HSL rats to 141 +/- 2 mmHg and 152 +/- 2, respectively, similar to our previous reports, and those respective 25 and 28 mmHg increases were a parallel shift in the pressure natriuresis relationship. However, the apparent difference between the LSL and HSL groups when compared was a parallel of the control MAP difference. Plasma renin activity (PRA) in the control period averaged 1.5 +/- 0.5 and 8.1 +/- 1.8 ng AI/mL per h in the HSV and LSV rats, and 0.8 +/- 0.2 and 2.8 +/- 0.5 ng AI/mL per h in the HSL and LSL rats, respectively, and increased similarly by 4.6-fold in the HSL and 4.8-fold in the LSL rats during diabetes. Glomerular filtration rate (GFR) increased in the vehicle but not the L-NAME-treated groups, consistent with our previous reports. 4. Thus, the hypertension caused by the onset of diabetes in L-NAME-treated rats was not salt-sensitive. The normal modulation of PRA by salt intake and the failure of GFR to increase are consistent with our hypothesis that nitric oxide may protect against hypertension early in diabetes by preventing preglomerular vasoconstriction by AngII.
Collapse
Affiliation(s)
- Michael W Brands
- Department of Physiology and the Vascular Biology Center, Medical College of Georgia, Augusta, Georgia 30912-3000, USA.
| | | | | | | | | |
Collapse
|
15
|
Toda N, Ayajiki K, Okamura T. Interaction of Endothelial Nitric Oxide and Angiotensin in the Circulation. Pharmacol Rev 2007; 59:54-87. [PMID: 17329548 DOI: 10.1124/pr.59.1.2] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Discovery of the unexpected intercellular messenger and transmitter nitric oxide (NO) was the highlight of highly competitive investigations to identify the nature of endothelium-derived relaxing factor. This labile, gaseous molecule plays obligatory roles as one of the most promising physiological regulators in cardiovascular function. Its biological effects include vasodilatation, increased regional blood perfusion, lowering of systemic blood pressure, and antithrombosis and anti-atherosclerosis effects, which counteract the vascular actions of endogenous angiotensin (ANG) II. Interactions of these vasodilator and vasoconstrictor substances in the circulation have been a topic that has drawn the special interest of both cardiovascular researchers and clinicians. Therapeutic agents that inhibit the synthesis and action of ANG II are widely accepted to be essential in treating circulatory and metabolic dysfunctions, including hypertension and diabetes mellitus, and increased availability of NO is one of the most important pharmacological mechanisms underlying their beneficial actions. ANG II provokes vascular actions through various receptor subtypes (AT1, AT2, and AT4), which are differently involved in NO synthesis and actions. ANG II and its derivatives, ANG III, ANG IV, and ANG-(1-7), alter vascular contractility with different mechanisms of action in relation to NO. This review article summarizes information concerning advances in research on interactions between NO and ANG in reference to ANG receptor subtypes, radical oxygen species, particularly superoxide anions, ANG-converting enzyme inhibitors, and ANG receptor blockers in patients with cardiovascular disease, healthy individuals, and experimental animals. Interactions of ANG and endothelium-derived relaxing factor other than NO, such as prostaglandin I2 and endothelium-derived hyperpolarizing factor, are also described.
Collapse
Affiliation(s)
- Noboru Toda
- Department of Pharmacology, Shiga University of Medical Science, Seta, Otsu, Japan.
| | | | | |
Collapse
|
16
|
Cheng HF, Zhang MZ, Harris RC. Nitric oxide stimulates cyclooxygenase-2 in cultured cTAL cells through a p38-dependent pathway. Am J Physiol Renal Physiol 2006; 290:F1391-7. [PMID: 16380459 DOI: 10.1152/ajprenal.00315.2005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
To examine the interaction of nitric oxide (NO) and cyclooxygenase (COX-2) and the signaling pathway involved, primary cultured rabbit cortical thick ascending limb (cTAL) were used. In these cells, immunoreactive COX-2 and vasodilatory prostaglandins were increased by a NO donor, S-nitros- N-acetylpenicillamine (SNAP; 2.5 ± 0.3-fold control, n = 6, P < 0.01). SNAP increased expression of phosphorylated p38 (pp38; 2.4 ± 0.3-fold control; n = 5; P < 0.01), which was inhibited by the p38 inhibitor SB-203580 (1.3 ± 0.1-fold control, n = 5, P < 0.01). SB-203580 inhibited SNAP-induced COX-2 expression [1.4 ± 0.2-fold control, n = 6, not significant (NS) vs. control] and levels of PGE2significantly. In cTAL cells transfected with a luciferase reporter driven by the wild-type mouse COX-2 promoter, SNAP stimulated luciferase activity, which was reversed by SB-203580 (control vs. SNAP vs. SNAP + SB-203580: 1.4 ± 0.2-, 8.3 ± 1.4-, and 0.4 ± 0.1-fold control, respectively, n = 4, P < 0.01). Electrophoretic mobility shift assay indicated that SNAP stimulated nuclear factor (NF)-κB binding activity in cTAL that was also inhibited by the p38 inhibitor. SNAP was not able to stimulate a mutant COX-2 promoter construct that is not activated by NF-κB (0.9 ± 0.1, 1.2 ± 0.1, and 1.0 ± 0.2 respectively, n = 4, NS). Low chloride increased COX-2 expression (2.7 ± 0.4-fold control, n = 6, P < 0.01) and pp38 expression (2.8 ± 0.3-fold; n = 5, P < 0.01), which were reversed by the specific NO synthase (NOS) inhibitor 7-nitroindazole. Administration of a low-salt diet increased immunoreactive COX-2 and neuronal NOS (nNOS) in the macula densa and surrounding cTAL of kidneys of wild-type mice but did not significantly elevate COX-2 expression in nNOS−/−mice. In summary, these studies indicate that, in cTAL, NO can increase COX-2 expression in cTAL and macula densa through p38-dependent signaling pathways via activation of NF-κB.
Collapse
Affiliation(s)
- Hui-Fang Cheng
- George M. O'Brien Kidney and Urologic Diseases Center and Division of Nephrology, Vanderbilt Univ. School of Medicine, and Nashville Veterans Affairs Hospital, Nashville, TN 37232, USA
| | | | | |
Collapse
|
17
|
Wilcox CS. Oxidative stress and nitric oxide deficiency in the kidney: a critical link to hypertension? Am J Physiol Regul Integr Comp Physiol 2005; 289:R913-35. [PMID: 16183628 DOI: 10.1152/ajpregu.00250.2005] [Citation(s) in RCA: 357] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is growing evidence that oxidative stress contributes to hypertension. Oxidative stress can precede the development of hypertension. In almost all models of hypertension, there is oxidative stress that, if corrected, lowers BP, whereas creation of oxidative stress in normal animals can cause hypertension. There is overexpression of the p22(phox) and Nox-1 components of NADPH oxidase and reduced expression of extracellular superoxide dismutase (EC-SOD) in the kidneys of ANG II-infused rodents, whereas there is overexpression of p47(phox) and gp91(phox) and reduced expression of intracellular SOD with salt loading. Several mechanisms have been identified that can make oxidative stress self-sustaining. Reactive oxygen species (ROS) can enhance afferent arteriolar tone and reactivity both indirectly via potentiation of tubuloglomerular feedback and directly by microvascular mechanisms that diminish endothelium-derived relaxation factor/nitric oxide responses, generate a cyclooxygenase-2-dependent endothelial-derived contracting factor that activates thromboxane-prostanoid receptors, and enhance vascular smooth muscle cells reactivity. ROS can diminish the efficiency with which the kidney uses O(2) for Na(+) transport and thereby diminish the P(O(2)) within the kidney cortex. This may place a break on further ROS generation yet could further enhance vasculopathy and hypertension. There is a tight relationship between oxidative stress in the kidney and the development and maintenance of hypertension.
Collapse
Affiliation(s)
- Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University Medical Center, 3800 Reservoir Rd., NW, Washington, DC 20007, USA.
| |
Collapse
|
18
|
Welch WJ, Blau J, Xie H, Chabrashvili T, Wilcox CS. Angiotensin-induced defects in renal oxygenation: role of oxidative stress. Am J Physiol Heart Circ Physiol 2005; 288:H22-8. [PMID: 15598867 DOI: 10.1152/ajpheart.00626.2004] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that superoxide anion (O2−·) generated in the kidney by prolonged angiotensin II (ANG II) reduces renal cortical Po2and the use of O2for tubular sodium transport (TNa:QO2). Groups ( n = 8–11) of rats received angiotensin II (ANG II, 200 ng·kg−1·min−1sc) or vehicle for 2 wk with concurrent infusions of a permeant nitroxide SOD mimetic 4-hydroxy-2,2,6,6-tetramethylpiperidine 1-oxyl (Tempol, 200 nmol·kg−1·min−1) or vehicle. Rats were studied under anesthesia with measurements of renal oxygen usage and Po2in the cortex and tubules with a glass electrode. Compared with vehicle, ANG II increased mean arterial pressure (107 ± 4 vs. 146 ± 6 mmHg; P < 0.001), renal vascular resistance (42 ± 3 vs. 65 ± 7 mmHg·ml−1·min−1·100 g−1; P < 0.001), renal cortical NADPH oxidase activity (2.3 ± 0.2 vs. 3.6 ± 0.4 nmol O2−··min−1·mg−1protein; P < 0.05), mRNA and protein expression for p22phox(2.1- and 1.8-fold respectively; P < 0.05) and reduced the mRNA for extracellular (EC)-SOD (−1.8 fold; P < 0.05). ANG II reduced the Po2in the proximal tubule (39 ± 1 vs. 34 ± 2 mmHg; P < 0.05) and throughout the cortex and reduced the TNa:QO2(17 ± 1 vs. 9 ± 2 μmol/μmol; P < 0.001). Tempol blunted or prevented all these effects of ANG II. The effects of prolonged ANG II to cause hypertension, renal vasoconstriction, renal cortical hypoxia, and reduced efficiency of O2usage for Na+transport, activation of NADPH oxidase, increased expression of p22phox, and reduced expression of EC-SOD can be ascribed to O2−· generation because they are prevented by an SOD mimetic.
Collapse
Affiliation(s)
- William J Welch
- Division of Nephrology and Hypertension and Cardiovascular Kidney Institute, Georgetown University, Washington, DC, USA
| | | | | | | | | |
Collapse
|
19
|
Abstract
Although initially adaptive, the changes that accompany hypertension, namely, cell growth, endothelial dysfunction, and extracellular matrix deposition, eventually can become maladaptive and lead to end-organ disease such as heart failure, coronary artery disease, and renal failure. A functional imbalance between angiotensin II (Ang II) and nitric oxide (NO) plays an important pathogenetic role in hypertensive end-organ injury. NO, an endogenous vasodilator, inhibitor of vascular smooth muscle and mesangial cell growth, and natriuretic agent, is synthesized in the endothelium by a constitutive NO synthase. NO antagonizes the effects of Ang II on vascular tone, cell growth, and renal sodium excretion, and also down-regulates the synthesis of angiotensin-converting enzyme (ACE) and Ang II type 1 receptors. On the other hand, Ang II decreases NO bioavailability by promoting oxidative stress. A better understanding of the pathophysiologic mechanisms involved in hypertensive end-organ damage may aid in identifying markers of cardiovascular susceptibility to injury and in developing therapeutic interventions. We propose that those antihypertensive agents that lower blood pressure and concomitantly restore the homeostatic balance of vasoactive agents such as Ang II and NO within the vessel wall would be more effective in preventing or arresting end-organ disease.
Collapse
Affiliation(s)
- Ming-Sheng Zhou
- Nephrology-Hypertension Division, Vascular Biology Institute, University of Miami School of Medicine, Veterans Affairs Medical Center, FL 33125, USA
| | | | | |
Collapse
|
20
|
Kawada N, Dennehy K, Solis G, Modlinger P, Hamel R, Kawada JT, Aslam S, Moriyama T, Imai E, Welch WJ, Wilcox CS. TP receptors regulate renal hemodynamics during angiotensin II slow pressor response. Am J Physiol Renal Physiol 2004; 287:F753-9. [PMID: 15213069 DOI: 10.1152/ajprenal.00423.2003] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the hypothesis that thromboxane A2(TxA2)-prostaglandin H2receptors (TP-Rs) mediate the hemodynamic responses and increase in reactive oxygen species (ROS) to ANG II (400 ng·kg−1·min−1sc for 14 days) using TP-R knockout (TP −/−) and wild-type (+/+) mice. TP −/− had normal basal mean arterial blood pressure (MAP) and glomerular filtration rate but reduced renal blood flow and increased filtration fraction (FF) and renal vascular resistance (RVR) and markers of ROS (thiobarbituric acid-reactive substances and 8-isoprostane PGF2α) and nitric oxide (NOx). Infusion of ANG II into TP +/+ increased ROS and thromboxane B2(TxB2) and increased RVR and FF. ANG II infusion into TP −/− mice reduced ANG I and increased aldosterone but caused a blunted increase in MAP (TP −/−: +6 ± 2 vs. TP +/+: +15 ± 3 mmHg) and failed to increase FF, ROS, or TxB2but increased NOx and paradoxically decreased RVR (−2.1 ± 1.7 vs. +2.6 ± 0.8 mmHg·ml−1·min−1·g−1). Blockade of AT1receptor of TP −/− mice infused with ANG II reduced MAP (−8 mmHg) and aldosterone but did not change the RVR or ROS. In conclusion, during an ANG II slow pressor response, AT1receptors activate TP-Rs that generate ROS and prostaglandins but inhibit NO. TP-Rs mediate all of the increase in RVR and FF, part of the increase in MAP, but are not implicated in the suppression of ANG I or increase in aldosterone. TP −/− mice have a basal increase in RVR and FF associated with ROS.
Collapse
Affiliation(s)
- Noritaka Kawada
- Cardiovascular Kidney Institute and Division of Nephrology and Hypertension, Georgetown University, Washington, DC 20007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Chabrashvili T, Kitiyakara C, Blau J, Karber A, Aslam S, Welch WJ, Wilcox CS. Effects of ANG II type 1 and 2 receptors on oxidative stress, renal NADPH oxidase, and SOD expression. Am J Physiol Regul Integr Comp Physiol 2003; 285:R117-24. [PMID: 12609817 DOI: 10.1152/ajpregu.00476.2002] [Citation(s) in RCA: 254] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oxidative stress accompanies angiotensin (ANG) II infusion, but the role of ANG type 1 vs. type 2 receptors (AT1-R and AT2-R, respectively) is unknown. We infused ANG II subcutaneously in rats for 1 wk. Excretion of 8-isoprostaglandin F2alpha (8-Iso) and malonyldialdehyde (MDA) were related to renal cortical mRNA abundance for subunits of NADPH oxidase and superoxide dismutases (SODs) using real-time PCR. Subsets of ANG II-infused rats were given the AT1-R antagonist candesartan cilexetil (Cand) or the AT2-R antagonist PD-123,319 (PD). Compared to vehicle (Veh), ANG II increased 8-Iso excretion by 41% (Veh, 5.4 +/- 0.8 vs. ANG II, 7.6 +/- 0.5 pg/24 h; P < 0.05). This was prevented by Cand (5.6 +/- 0.5 pg/24 h; P < 0.05) and increased by PD (15.8 +/- 2.0 pg/24 h; P < 0.005). There were similar changes in MDA excretion. Compared to Veh, ANG II significantly (P < 0.005) increased the renal cortical mRNA expression of p22phox (twofold), Nox-1 (2.6-fold), and Mn-SOD (1.5-fold) and decreased expression of Nox-4 (2.1-fold) and extracellular (EC)-SOD (2.1-fold). Cand prevented all of these changes except for the increase in Mn-SOD. PD accentuated changes in p22phox and Nox-1 and increased p67phox. We conclude that ANG II infusion stimulates oxidative stress via AT1-R, which increases the renal cortical mRNA expression of p22phox and Nox-1 and reduces abundance of Nox-4 and EC-SOD. This is offset by strong protective effects of AT2-R, which are accompanied by decreased expression of p22phox, Nox-1, and p67phox.
Collapse
Affiliation(s)
- Tina Chabrashvili
- Division of Nephrology and Hypertension, Georgetown Univ. Hospital, 3800 Reservoir Rd, N.W., PHC F6003, Washington, DC 20007, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Cowley AW, Mori T, Mattson D, Zou AP. Role of renal NO production in the regulation of medullary blood flow. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1355-69. [PMID: 12736168 DOI: 10.1152/ajpregu.00701.2002] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The unique role of nitric oxide (NO) in the regulation of renal medullary function is supported by the evidence summarized in this review. The impact of reduced production of NO within the renal medulla on the delivery of blood to the medulla and on the long-term regulation of sodium excretion and blood pressure is described. It is evident that medullary NO production serves as an important counterregulatory factor to buffer vasoconstrictor hormone-induced reduction of medullary blood flow and tissue oxygen levels. When NO synthase (NOS) activity is reduced within the renal medulla, either pharmacologically or genetically [Dahl salt-sensitive (S) rats], a super sensitivity to vasoconstrictors develops with ensuing hypertension. Reduced NO production may also result from reduced cellular uptake of l-arginine in the medullary tissue, resulting in hypertension. It is concluded that NO production in the renal medulla plays a very important role in sodium and water homeostasis and the long-term control of arterial pressure.
Collapse
Affiliation(s)
- Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | |
Collapse
|
23
|
Wilcox CS, Welch WJ. Thromboxane synthase and TP receptor mRNA in rat kidney and brain: effects of salt intake and ANG II. Am J Physiol Renal Physiol 2003; 284:F525-31. [PMID: 12517736 DOI: 10.1152/ajprenal.00256.2002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A TP receptor (TP-R) mimetic causes salt-sensitive hypertension and renal afferent arteriolar vasoconstriction. TP-Rs mediate effects of ANG II on renal vascular resistance and drinking. Therefore, we investigated the hypothesis that thromboxane A(2) synthase (TxA(2)-S) and/or TP-R expression is regulated by salt and/or ANG II. Rats (n = 6) received high-salt (HS) or low-salt (LS) diets. Additional HS-diet rats received ANG II while other HS- and LS-diet rats received the AT(1) receptor (AT(1)-R) antagonist losartan. Excretion of thromboxane B(2) by conscious rats was increased with the HS diet compared with the LS diet (126 +/- 10 vs. 48 +/- 5 pmol/24 h, respectively; P < 0.01). The mRNA abundance for TP-Rs (relative to beta-actin) in the kidney cortex was enhanced 30% by the HS diet (P < 0.001) and was reduced 50% by the addition of ANG II (P < 0.001). However, during losartan administration, the effects of salt were reversed; mRNA more than doubled during the LS diet (P < 0.001). Similarly, the mRNA abundance for TP-Rs in the brain stem was reduced by 50% with the addition of ANG II (P < 0.001) and during losartan administration was almost doubled by the LS diet (P < 0.001). The mRNA abundance for TxA(2)-S in the kidney cortex also was increased many times with the HS diet (P < 0.001). In contrast, the mRNA for TxA(2)-S in the brain was unaffected by salt. ANG II did not affect TxA(2)-S at either site. During losartan administration, TxA(2)-S increased modestly in the brain stem with the LS diet. mRNA abundance for TP-Rs in the kidney cortex and brain stem is suppressed by ANG II acting on AT(1)-Rs. In the absence of AT(1)-Rs, expression of TP-Rs at both sites is enhanced by LS intake. In contrast, ANG II does not affect the mRNA abundance for TxA(2)-S. Expression of TxA(2)-S is enhanced by HS intake in the kidney cortex but by LS intake in the brain stem only during losartan administration. Thus TP-Rs are strongly dependent on ANG II acting on AT(1)-Rs, whereas TxA(2)-S is regulated differentially in the kidney cortex and brain stem by salt intake.
Collapse
Affiliation(s)
- Christopher S Wilcox
- Division of Nephrology and Hypertension and Center for Hypertension and Renal Disease Research, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | |
Collapse
|
24
|
Matsuda H, Hayashi K, Arakawa K, Kubota E, Honda M, Tokuyama H, Suzuki H, Yamamoto T, Kajiya F, Saruta T. Distinct modulation of superficial and juxtamedullary arterioles by prostaglandin in vivo. Hypertens Res 2002; 25:901-10. [PMID: 12484515 DOI: 10.1291/hypres.25.901] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Renal afferent (AFF) and efferent arteriolar (EFF) responsiveness to angiotensin II (ANG II) in superficial and juxtamedullary nephrons in vivo remains undetermined, nor has it been clarified what role intrarenal autocrines/paracrines play in modulating the renal microvascular response. The present study characterized the responsiveness to ANG II (1-30 ng/kg/min) of AFF and EFF of canine superficial and juxtamedullary nephrons under pentobarbital anesthesia, using intravital CCD-videomicroscopy that allowed direct in vivo visualization of the renal microcirculation. Furthermore, the effect of prostaglandins (PG) and nitric oxide (NO) on ANG II-induced tone was examined. In superficial nephrons, ANG II induced a similar dose-dependent constriction of both AFF (46 +/- 5% constriction) and EFF (53 +/- 3%). In juxtamedullary arterioles, ANG II induced a dose-dependent constriction of EFF, whereas AFF responses were diminished (17 +/- 4% vs. 37 +/- 4% at 10 ng/kg/min). The PG inhibition by indomethacin enhanced the ANG II-induced constriction of juxtamedullary AFF, whereas no augmentation was observed in other arterioles. In contrast, NO inhibition by nitro-L-arginine methylester (L-NAME) enhanced the ANG II-induced constriction, with greater augmentation in juxtamedullary AFF and EFF. Finally, renal interstitial PG and nitrite/nitrate contents were greater in the medulla than the superficial cortex under basal and ANG II-stimulated conditions. Taken together, the results of the intravital CCD-videomicroscopy reveal that the renal microvascular action of ANG II had both zonal (juxtamedullary vs. superficial nephrons) and segmental (AFF vs. EFF) heterogeneity under the present experimental conditions. This heterogeneity was associated with a difference in the intrarenal production of prostaglandin E2 (PGE2) and NO; PGE2 contributed to segmental and zonal differences whereas NO was responsible for the zonal heterogeneity in arteriolar responsiveness.
Collapse
Affiliation(s)
- Hiroto Matsuda
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Romano L, Coviello A, Jerez S, Peral de Bruno M. Role of nitric oxide on the vasorelaxant effect of atrial natriuretic peptide on rabbit aorta basal tone. Can J Physiol Pharmacol 2002; 80:1022-9. [PMID: 12450070 DOI: 10.1139/y02-130] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of nitric oxide (NO) on the vasorelaxant effect of atrial natriuretic peptide (ANP) on the basal tone of rabbit aortic rings conditioned to angiotensin II (Ang II) was studied. ANP aortic relaxation and nitrite release were measured in the presence and absence of endothelium and a NO-synthase inhibitor. Ang II at 10(-8) M triggered a contractile response, conditioning the vessel to a vasorelaxant effect of ANP (10(-8) M). This effect was significantly enhanced by endothelium removal, NG-nitro-L-arginine methyl ester (L-NAME, 10(-4) M), and methylene blue (10(-5) M). ANP decrease of basal tone in Ang-II-sensitized aortic rings was improved when a higher concentration of Ang II was used (l0(-6) M). Basal and Ang-II-stimulated nitrite release were measured in stretched (S) and nonstretched (NS) aortic rings. Nitrite release was significantly increased in S rings (p < 0.001). L-NAME (10(-4) M) partially inhibited nitrite release in both basal and Ang-II-stimulated S aortic rings. In NS aortic rings, the NO inhibitor did not inhibit basal nitrite release but blunted the Ang-II-stimulated nitrite level. A significant negative correlation between nitrite release and the ANP vasorelaxant effect on basal tone was dependent on the Ang-II-sensitizing dose. The present results demonstrate that ANP relaxant effects on aortic basal tone are related to NO levels, which are regulated by S- and Ang-II-concentration-dependent NO generation and quenching.
Collapse
Affiliation(s)
- Liliana Romano
- Department of Physiology, Instituto Superior de Investigaciones Biológicas and Facultad de Medicina, Universidad Nacional de Tucumán, CC-69 Suc. 2, (4000) Tucumán, Argentina
| | | | | | | |
Collapse
|
26
|
Lansang MC, Hollenberg NK. Renal perfusion and the renal hemodynamic response to blocking the renin system in diabetes: are the forces leading to vasodilation and vasoconstriction linked? Diabetes 2002; 51:2025-8. [PMID: 12086929 DOI: 10.2337/diabetes.51.7.2025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In three groups of subjects, those with type 2 diabetes and nephropathy, those with type 1 diabetes without nephropathy, and healthy volunteers subjected to short-term hyperglycemia, we observed a counterintuitive relationship. In all three groups, baseline renal plasma flow (RPF) was positively correlated with the RPF response to blocking the renin-angiotensin system (RAS). This seems paradoxical in that an opposite result would have been expected if angiotensin-dependent renal vasoconstriction was responsible for the renal vasodilator response to RAS blockade. This suggests a link between the renal vasodilator response, mediated by nitric oxide (NO), and the activation of the intrarenal RAS. The complex interrelationships between hyperglycemia, insulin, NO, and the RAS may result in phenotypes that indicate varying risk of diabetic nephropathy and underlying genetic polymorphisms.
Collapse
Affiliation(s)
- M Cecilia Lansang
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | |
Collapse
|
27
|
Mourad JJ, Ducailar G, Rudnicki A, Lajemi M, Mimran A, Safar ME. Age-related increase of pulse pressure and gene polymorphisms in essential hypertension: a preliminary study. J Renin Angiotensin Aldosterone Syst 2002; 3:109-15. [PMID: 12228851 DOI: 10.3317/jraas.2002.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Genes may modulate the changes of blood pressure (BP) with age; this possibility has never been studied for the age-related increase of pulse pressure (PP), although in older populations, PP is considered the stronger mechanical factor predicting cardiovascular mortality. In humans, the presence of the mutant allele C of the angiotensin II (Ang II) AT(1)-receptor or of the mutant allele T of the eNOS G(298) T gene polymorphisms is associated with enhanced contractile properties of conduit arteries in response to vasoconstrictive agents. In this study, we evaluated, in subjects with untreated essential hypertension, whether the presence of these mutant alleles or their combination might influence the age-related increase of PP. Three main findings emerged from the study and were particularly observed in women: 1) the presence of the C and/or of the T mutant alleles or their combination were associated with a steeper slope of the age versus PP curve, compared with subjects without the mutant allele; 2) the slope was more significantly enhanced when the two mutant alleles were associated in the same genotype; and 3) no comparable age- and gender-related changes in systolic, diastolic or mean BP were found according to this genetic classification. In subjects with essential hypertension, genes may modulate the age-mediated increase of PP. This finding gives new insights in the interactions between genes, mechanical factors and cardiovascular risk.
Collapse
Affiliation(s)
- Jean-Jacques Mourad
- Department of Internal Medicine and INSERM U337, Broussais Hospital, Paris, France
| | | | | | | | | | | |
Collapse
|
28
|
Sener A, Smith FG. Glomerular and tubular responses to N(G)-nitro-L-arginine methyl ester are age dependent in conscious lambs. Am J Physiol Regul Integr Comp Physiol 2002; 282:R1512-20. [PMID: 11959695 DOI: 10.1152/ajpregu.00628.2001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present experiments were carried out to investigate the role of endogenously produced NO in modulating renal function during postnatal maturation under physiological conditions. In conscious, chronically instrumented lambs aged approximately 1 (n = 8) and approximately 6 wk (n = 8) of postnatal life, various parameters of glomerular and tubular function were measured for 1 h before and 1 h after intravenous injection of 20 mg/kg of N(G)-nitro-L-arginine methyl ester (L-NAME; experiment 1) or its inactive isomer D-NAME (experiment 2). After administration of L-NAME to 1-wk-old lambs, glomerular filtration rate (GFR) and filtration factor (FF) decreased by approximately 50% at 20 min, remaining decreased at 60 min. In 6-wk-old lambs, GFR and FF remained constant after L-NAME. Proximal fractional Na(+) reabsorption decreased after L-NAME administration to lambs aged 6 wk, resulting in a prompt natriuresis; this was sustained for 60 min. There were no effects of L-NAME on proximal fractional Na(+) reabsorption in 1-wk-old lambs. In 6-wk-old lambs, urinary flow rate increased by approximately 500%, free water clearance increased by approximately 50%, and urinary osmolality decreased by approximately 60% after L-NAME administration; no effects on these variables were measured in 1-wk-old lambs. The diuresis after L-NAME administration to 6-wk-old lambs was unaccompanied by any changes in plasma levels of arginine vasopressin. There were no effects of D-NAME on any of the measured variables. We conclude that endogenously produced nitric oxide modulates glomerular and tubular function in an age-dependent manner.
Collapse
Affiliation(s)
- Alp Sener
- Departments of Physiology and Biophysics/Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | | |
Collapse
|
29
|
Rahman M, Kimura S, Yoneyama H, Kosaka H, Fukui T, Nishiyama A, Abe Y. Effects of angiotensin II on the renal interstitial concentrations of NO2/NO3 and cyclic GMP in anesthetized rats. JAPANESE JOURNAL OF PHARMACOLOGY 2002; 88:436-41. [PMID: 12046987 DOI: 10.1254/jjp.88.436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study was conducted to determine whether exogenous angiotensin II (Ang II) may increase the renal interstitial fluid concentrations of NO2/NO3 (NOx) and cyclic guanosine monophosphate (cGMP) concomitantly and which Ang II receptor subtypes may induce these changes in anesthetized rats, using a microdialysis method. Ang II (50 ng/kg per min, i.v.) significantly increased mean blood pressure (MBP), extraction rates of renal interstitial NOx from 23.9+/-1.0 to 31.2+/-1.9 pmol/min, and cGMP from 4.1+/-0.3 to 6.4+/-0.5 fmol/min, and decreased renal blood flow (RBF). The AT1-receptor antagonist CV11974 alone significantly increased RBF, but did not alter MBP, renal interstitial concentrations of NOx and cGMP. A superimposition of Ang II on CV11974 did not affect MBP and RBF, but significantly increased renal interstitial concentrations of NOx and cGMP. The AT2-receptor antagonist PD123319 alone did not change any of the parameters. However, superimposition of Ang II on PD123319 increased MBP and decreased RBF without any effects on renal interstitial concentrations of NOx and cGMP. These results suggest that Ang II stimulates NO production via the AT2-receptor in the kidney.
Collapse
Affiliation(s)
- Matlubur Rahman
- Department of Pharmacology, Kagawa Medical University, Japan.
| | | | | | | | | | | | | |
Collapse
|
30
|
Moreno C, López A, Llinás MT, Rodríguez F, López-Farré A, Nava E, Salazar FJ. Changes in NOS activity and protein expression during acute and prolonged ANG II administration. Am J Physiol Regul Integr Comp Physiol 2002; 282:R31-7. [PMID: 11742820 DOI: 10.1152/ajpregu.2002.282.1.r31] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to assess the effects of acute or prolonged increases of ANG II on nitric oxide synthase (NOS) activities and protein expression in mesenteric resistance vessels, left ventricle, renal cortex, and renal medulla. The response of NOS activities to ANG II is compared with that induced by phenylephrine. ANG II or phenylephrine were infused over either 3 h or 3 days to conscious rats. NOS activity was examined by measuring the rate of conversion of L-[14C]arginine to L-[14C]citrulline. Protein levels of endothelial (e) and neuronal (n) NOS were determined by Western blot analysis. Arterial pressure (AP) increased (P < 0.05) during acute and prolonged ANG II infusion. Ca2+-dependent NOS activity values (pmol of citrulline x min(-1) x g wet wt(-1)) for control rats were 21 +/- 9 in mesenteric arteries, 13 +/- 7 in left ventricle, 14 +/- 8 in renal cortex, and 411 +/- 70 in renal medulla. Acute ANG II infusion increased (P < 0.05) Ca2+-dependent NOS activity in renal cortex and renal medulla (81 +/- 18 and 611 +/- 48, respectively), but no differences were found in mesenteric arteries and left ventricle with respect to control rats. In contrast to the renal changes in NOS activity, acute ANG II infusion did not modify eNOS or nNOS expression in any of the tissues examined. Prolonged ANG II infusion increased (P < 0.05) Ca2+-dependent NOS activity in mesenteric arteries (70 +/- 17), renal cortex (104 +/- 31), and left ventricle (49 +/- 8) and did not elicit changes in renal medulla. After a prolonged ANG II infusion, eNOS and nNOS levels increased in all tissues examined with the exception of eNOS in the mesenteric arteries and nNOS in the left ventricle, which were not altered. Acute and prolonged phenylephrine infusion elevated AP to a similar extent as ANG II but only elicited significant increments of Ca2+-dependent NOS activity in renal cortex. These data indicate that acute and prolonged elevations in ANG II upregulate Ca2+-dependent NOS activity and protein expression in different tissues related to the control of blood pressure. However, these ANG II effects are heterogeneous with respect to the tissue implicated, the time course of the stimulation, and the NOS isoform involved. Phenylephrine only induces a significant elevation of Ca2+-dependent NOS activity in renal cortex.
Collapse
Affiliation(s)
- Carol Moreno
- Department of Physiology, University of Murcia School of Medicine, E-30100 Murcia, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Schuijt MP, de Vries R, Saxena PR, Jan Danser AH. Prostanoids, but not nitric oxide, counterregulate angiotensin II mediated vasoconstriction in vivo. Eur J Pharmacol 2001; 428:331-6. [PMID: 11689191 DOI: 10.1016/s0014-2999(01)01349-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To evaluate the modulating effects of nitric oxide and prostanoids during angiotensin II-mediated vasoconstriction, male Wistar rats (n=25) were infused with increasing doses of angiotensin II following pretreatment with the cyclooxygenase inhibitor indomethacin, the nitric oxide-synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME) plus sodium nitroprusside to restore mean arterial blood pressure, or saline. Hemodynamics were studied with the radioactive microsphere method. Indomethacin did not alter systemic or regional hemodynamics. L-NAME+sodium nitroprusside reduced cardiac output, as well as systemic and renal vascular conductance. Angiotensin II increased mean arterial blood pressure and heart rate, and decreased systemic vascular conductance as well as vascular conductance in gastrointestinal tract, kidney, skeletal muscle, skin, mesentery+pancreas, spleen and adrenal. Indomethacin enhanced the angiotensin II-mediated effects in all vascular beds, whereas L-NAME+sodium nitroprusside enhanced its effect in mesentery+pancreas only. In conclusion, vasodilator prostanoids, but not nitric oxide, counterregulate angiotensin II-mediated vasoconstriction in vivo.
Collapse
Affiliation(s)
- M P Schuijt
- Department of Pharmacology, Erasmus University Rotterdam, Room EE1418b, Dr. Molewaterplein 50, 3015 GE, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
32
|
Kohagura K, Arima S, Endo Y, Chiba Y, Ito O, Abe M, Omata K, Ito S. Involvement of cytochrome P450 metabolites in the vascular action of angiotensin II on the afferent arterioles. Hypertens Res 2001; 24:551-7. [PMID: 11675950 DOI: 10.1291/hypres.24.551] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies have demonstrated that cytochrome P450-dependent metabolites of arachidonic acid (CYP450-AA) play important roles in the control of renal vascular resistance (RVR). In the present study, we examined the possible involvement of CYP450-AA in the vasoconstrictor action of angiotensin II (Ang II) on the afferent arterioles (Af-Arts), a vascular segment crucial to the control of RVR. Rabbit Af-Arts were microperfused at 60 mmHg in vitro, and the vasoconstrictor action of Ang II (10(-11)-10(-8) M, added to both the bath and lumen) was examined with or without blocking the activity of CYP450 epoxygenase or hydroxylase. Ang II decreased the luminal diameter of Af-Arts in a dose-dependent manner (34+/-2% of control diameter at 10(-8) M, n=9, p<0.0001). Pretreatment with miconazole, an inhibitor of CYP450 epoxygenase, at 10(-6) M decreased the basal diameter by 14+/-1% (n=6, p<0.01) and augmented the vasoconstrictor action of Ang II (7+/-3% of control diameter at 10(-8) M, p<0.001 vs. without miconazole). This augmentation was abolished by blocking the Ang II type 2 (AT2) receptor with PD 123319 at 10(-7) M. In contrast, pretreatment with 17-octadecynoic acid (17-ODYA, 10(-6) M), which inhibits both epoxygenase and hydroxylase activity, had no effect on the basal diameter but attenuated the vasoconstrictor action of Ang 11(46+/-2% of control diameter at 10(-8) M, p<0.01 vs. without 17-ODYA). Our results demonstrate that in the Af-Art, endogenous CYP450-AA are involved not only in the control of basal tone but also in the action of Ang II. Further, it appears that the CYP450 epoxygenase pathway attenuates Ang II action via AT2 receptors.
Collapse
Affiliation(s)
- K Kohagura
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Lohmeier TE, Lohmeier JR, Reckelhoff JF, Hildebrandt DA. Sustained influence of the renal nerves to attenuate sodium retention in angiotensin hypertension. Am J Physiol Regul Integr Comp Physiol 2001; 281:R434-43. [PMID: 11448845 DOI: 10.1152/ajpregu.2001.281.2.r434] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies indicate that baroreflex suppression of renal sympathetic nerve activity is sustained for up to 5 days of ANG II infusion; however, steady-state conditions are not associated with ANG II hypertension of this short duration. Thus the major goal of this study was to determine whether neurally induced increments in renal excretory function during chronic intravenous infusion of ANG II are sustained under more chronic conditions when hypertension is stable and sodium balance is achieved. Experiments were conducted in five conscious dogs subjected to unilateral renal denervation and surgical division of the urinary bladder into hemibladders to allow separate 24-h urine collection from denervated (Den) and innervated (Inn) kidneys. ANG II was infused after control measurements for 10 days at a rate of 5 ng. kg(-1). min(-1). Twenty-four-hour control values for mean arterial pressure (MAP) and the ratio for urinary sodium excretion from Den and Inn kidneys (Den/Inn) were 92 +/- 4 mmHg and 0.99 +/- 0.05, respectively. On days 8-10 of ANG II infusion, MAP was stable (+30 +/- 3 mmHg) and sodium balance was achieved. Whereas equal amounts of sodium were excreted from the kidneys during the control period, throughout ANG II infusion there was a greater rate of sodium excretion from Inn vs. Den kidneys (day 10 Den/Inn sodium = 0.56 +/- 0.05), indicating chronic suppression of renal sympathetic nerve activity. The greater rate of sodium excretion in Inn vs. Den kidneys during renal sympathoinhibition also revealed a latent impairment in sodium excretion from Den kidneys. Although the Den/Inn for sodium and the major metabolites of nitric oxide (NO) decreased in parallel during ANG II hypertension, the Den/Inn for cGMP, a second messenger of NO, remained at control levels throughout this study. This disparity fails to support the notion that a deficiency in NO production and action in Den kidneys accounts for the impaired sodium excretion. Most importantly, these results support the contention that baroreflex suppression of renal sympathetic nerve activity is sustained during chronic ANG II hypertension, a response that may play an important role in attenuating the rise in arterial pressure.
Collapse
Affiliation(s)
- T E Lohmeier
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State St., Jackson, MS 39216, USA.
| | | | | | | |
Collapse
|
34
|
Nishiyama A, Fujisawa Y, Fukui T, Rahman M, Kondo N, Ogawa Y, Fanzhu L, Guoxing Z, Kimura S, Abe Y. Role of nitric oxide in regional blood flow in angiotensin II-induced hypertensive rats. Hypertens Res 2001; 24:421-7. [PMID: 11510755 DOI: 10.1291/hypres.24.421] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The present study was designed to evaluate the contribution of nitric oxide (NO) to regional hemodynamics during the early phase of angiotensin II (Ang II)-induced hypertension. The responses of regional blood flow to chronic NO synthase inhibition with N(G)-nitro-L-arginine methyl ester (L-NAME) were assessed using radioactive microspheres in conscious Ang II-infused hypertensive rats. Ang II-infused rats (270 ng/kg/min, subcutaneously for 12 days: n=11) showed higher mean arterial pressure (MAP: 153+/-4 mmHg) and total peripheral resistance (TPR: 1.61+/-0.06 mmHg/min/ml), and lower cardiac output (CO: 102+/-3 ml/min) than vehicle-infused normotensive rats (115+/-2 mmHg, 0.96+/-0.05 mmHg/min/ml and 130+/-7 ml/min, n=11, respectively). The blood flow rates in the brain, spleen, large intestine and skin were significantly reduced in Ang III-infused rats compared with vehicle-infused rats, while those in the lung, heart, liver, kidney, adrenal gland, small intestine, and skeletal muscle were similar. Treating Ang II-infused rats with L-NAME (75 mg/l in drinking water for 10 days, n=11) resulted in higher MAP (166+/-6 mmHg) and TPR (1.89+/-0.18 mmHg/min/ml) and lower CO (87+/-7 m/min) than untreated Ang II-infused rats. L-NAME-treated Ang II-infused rats showed widespread increases in regional vascular resistance and reduced blood flow rates in the kidney (3.81+/-0.27 ml/min/g) and skeletal muscle (0.20+/-0.03 ml/min/g) compared with untreated Ang II-infused rats (6.88+/-0.27 and 0.33+/-0.04 ml/min/g, respectively). However, there were no significant differences in the flow rates of other organs investigated between these animals. An NO donor, (+/-)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (FK409: 30 microg/kg/min, i.v.), significantly decreased MAP (110+/-6 mmHg) and TPR (1.23+/-0.18 mmHg/min/ml) without significant changes in CO (89+/-9 ml/min) in L-NAME-treated Ang II-infused rats. Furthermore, FK409 partially reversed blood flow rates in the kidney (4.72+/-0.40 ml/min/g) and skeletal muscle (0.25+/-0.02 ml/min/g)in these animals. These results suggest that NO counteracts, at least in part, the vasoconstrictor effects of elevated Ang II levels in renal and skeletal muscle vascular beds, and is an important modulator in the regulation of blood flow to these organs during the development of Ang II-induced hypertension.
Collapse
Affiliation(s)
- A Nishiyama
- Department of Pharmacology, Kagawa Medical University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Chronic azotemic renovascular disease is common in patients with atherosclerosis. Its prevalence appears to be increasing in the aging population. How often it is the primary cause of end-stage renal disease (ESRD) is not yet certain. Some studies suggest that 10%-40% of elderly hypertensive patients with newly documented ESRD and no demonstrable primary renal disease have significant renal artery stenosis (RAS). Atherosclerotic vascular occlusive disease of the renal arteries does progress, but current rates of progression and occlusion are lower than those reported a decade ago. Methods of identifying patients whose renal function is at true risk from vascular occlusive disease and determining who will benefit from intervention remain elusive. The presence of RAS in an azotemic patient can be assessed with noninvasive and risk-free radiologic techniques, including Duplex doppler velicometry and magnetic resonance angiography. Functional tests that predict the change in renal function after revascularization are not yet available. However, a renal length of greater than 7.5 cm in the absence of renal cysts and a short history of renal functional deterioration indicate a good prognosis. Patients with recent deterioration in renal function, those with bilateral renal artery stenosis or stenosis to a single functioning kidney, those with flash pulmonary edema, advanced chronic renal failure, or ESRD (who have much to gain), those with reversible azotemia during angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor antagonist (ARB) therapy, and those whose conditions cannot be managed medically should be considered for revascularization. Results from recent controlled clinical trials of the response to percutaneous transluminal renal artery angioplasty (PTRA) and stenting indicate that improvement in blood pressure control or renal function is not a predictable outcome of renal revascularization. In azotemic groups, 25%-30% of patients achieve important recovery of renal function. Thus, significant progress has been made recently in determining whether RAS is a frequent, treatable cause of renal failure. The decision to recommend revascularization remains a difficult balance between the risks and expense of the procedure and the undoubted benefits that accrue if renal function is successfully stabilized.
Collapse
Affiliation(s)
- S C Textor
- Divisions of Hypertension and Nephrology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA.
| | | |
Collapse
|
36
|
Stulak JM, Lerman A, Caccitolo JA, Wilson SH, Romero JC, Schaff HV, Rodriguez Porcel M, Lerman LO. Impaired renal vascular endothelial function in vitro in experimental hypercholesterolemia. Atherosclerosis 2001; 154:195-201. [PMID: 11137100 DOI: 10.1016/s0021-9150(00)00462-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hypercholesterolemia (HC) induces alterations in systemic vascular reactivity, which can manifest as an attenuated endothelium-dependent relaxation, partly consequent to an impairment in nitric oxide (NO) activity. To determine whether experimental HC has a similar effect on renal vascular function, renal artery segments obtained from pigs fed a HC (n=5) or normal (n=5) diet were studied in vitro. Endothelium-dependent relaxation was examined using increasing concentrations of acetylcholine (Ach), calcium ionophore A23187, and Ach following pre-incubation with N(G)-monomethyl-L-arginine or L-arginine (L-ARG). The NO-donor diethylamine (DEA) was used to examine smooth muscle relaxation response and cyclic GMP generation in endothelium-denuded vessels. The expression of endothelial NO synthase (eNOS) in the renal arteries was examined using Western blotting. Endothelium-dependent relaxation to Ach was significantly attenuated in the HC group compared to normal (53.3+/-9.1 vs. 98.8+/-3.7%, P<0.005), but normalized after pre-incubation with L-ARG (82.3+/-13.8%, P=0.21). Receptor-independent endothelium-dependent relaxation to A23187 was also significantly blunted in HC (75.2+/-10.5 vs. 115.5+/-4.2%, P<0. 017). Smooth muscle relaxation and cyclic GMP generation in response to DEA were greater in denuded HC vessels, while relaxation of intact vessels to nitroprusside was unaltered. In the HC vessels eNOS was almost undetectable. In conclusion, experimental HC attenuates in vitro endothelium-dependent relaxation of the porcine renal artery, possibly due to low bioavailability of NO. These vascular alterations in HC could play a role in the pathogenesis of renal disease or hypertension, supporting a role for HC as a risk factor for renovascular disease.
Collapse
Affiliation(s)
- J M Stulak
- Department of Physiology and Biophysics, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Tojo A, Kimoto M, Wilcox CS. Renal expression of constitutive NOS and DDAH: separate effects of salt intake and angiotensin. Kidney Int 2000; 58:2075-83. [PMID: 11044228 DOI: 10.1111/j.1523-1755.2000.00380.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Nitric oxide (NO) is generated from NO synthase (NOS) isoforms. These enzymes can be inhibited by asymmetric dimethylarginine, which is inactivated by N(G)-N(G)-dimethylarginine dimethylaminohydrolase (DDAH). The neuroneal (nNOS) type I and endothelial (eNOS) type III constitutive NOS isoforms are expressed predominantly in the macula densa and microvascular endothelium of the renal cortex, respectively. DDAH is expressed at sites of NOS expression. Since NO may coordinate the renal responses to angiotensin II (Ang II) and changes in salt intake, we tested the hypothesis that salt intake regulates the expression of nNOS, eNOS and DDAH by Ang II acting on type 1 (AT(1)) receptors. METHODS Groups (N = 6) of rats were adapted to low-salt (LS) or high-salt (HS) intakes for 10 days. Other groups of LS and HS rats received the AT(1) receptor antagonist losartan for six days (to test the effects of salt independent of AT(1) receptors). A further group of HS rats received an infusion of Ang II for six days (to test the effect of Ang II independent of salt intake). RESULTS Compared with HS rats, there was a significant (P < 0.05) increase in LS rats of nNOS protein in kidney and immunohistochemical expression in the macula densa, and of eNOS protein expression and immunohistochemical expression in the microvascular endothelium, and of DDAH protein expression. Losartan prevented these effects of salt on the expression of eNOS or DDAH, both of which were also increased by Ang II infusions in HS rats. In contrast, losartan did not prevent the effects of salt on nNOS expression, which was unresponsive to Ang II infusion. The generation of NO(2)(-) released by slices of renal cortex, in the presence of saturating concentrations of L-arginine, was increased by LS, compared to HS, independent of losartan and by Ang II during HS. CONCLUSION The expressions of eNOS in cortical microvascular endothelium and DDAH in kidney are enhanced by Ang II acting on AT(1) receptors. The expression of nNOS in the macula densa is enhanced by salt restriction independent of Ang II or AT(1) receptors.
Collapse
MESH Headings
- Amidohydrolases
- Angiotensin II/physiology
- Animals
- Diet, Sodium-Restricted
- Endothelium, Vascular/enzymology
- Hydrolases/metabolism
- Kidney/enzymology
- Kidney Cortex/blood supply
- Kidney Tubules, Distal/cytology
- Kidney Tubules, Distal/enzymology
- Male
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type I
- Nitric Oxide Synthase Type III
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Angiotensin/physiology
- Tissue Distribution
Collapse
Affiliation(s)
- A Tojo
- Division of Nephrology and Endocrinology, Department of Internal Medicine, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
38
|
Abstract
We have shown that NO production, assessed by measuring changes in plasma nitrate concentration, is down-regulated when blood pressure falls. This study intended to determine first, whether NO-derived plasma nitrate varies in response to increases in blood pressure induced by different mechanical and pharmacologic stimuli, including angiotensin II and catecholamines; and second, specifically to study the interaction between angiotensin II and NO production. An intravenous infusion (4-10 min) of norepinephrine (7.5 microg/kg/min), phenylephrine (30 microg/kg/min), or angiotensin II (0.3 and 3 microg/kg/min) caused hypertension accompanied by an increase in plasma nitrate, as assessed by high-performance capillary electrophoresis. Mechanical hypertension elicited by aortic occlusion also was accompanied by an increase in plasma nitrate. Angiotensin II (0.03, 0.3, and 3 microg/kg/min, 10 min) dose-dependently increased blood pressure. The intermediate and high dose, but not the low dose, of angiotensin II increased plasma nitrate concentration. N(G)-nitro-L-arginine methyl ester (L-NAME) lowered the basal concentration of plasma nitrate, abolished the increase in plasma nitrate elicited by angiotensin II and norepinephrine, and potentiated the pressor effect of the low dose of angiotensin II, although this dose did not increase NO production. L-NAME also potentiated the pressor effects of the intermediate dose of angiotensin II. This study demonstrates that an augmented systemic production of NO, measured as an increase in plasma nitrate, takes place after acute hypertension. The results of this study suggest that an increase in NO generation occurs when angiotensin II hypertension exceeds a certain limit, below which the basal production of NO is sufficient to compensate the vasoconstriction.
Collapse
Affiliation(s)
- E Nava
- Department of Physiology, University of Murcia School of Medicine, Spain.
| | | | | | | | | |
Collapse
|
39
|
Romero JC, Reckelhoff JF. Oxidative stress may explain how hypertension is maintained by normal levels of angiotensin II. Braz J Med Biol Res 2000; 33:653-60. [PMID: 10829093 DOI: 10.1590/s0100-879x2000000600006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well known that essential hypertension evolves in most patients with "near normal" levels of plasma renin activity. However, these levels appear to be responsible for the high levels of arterial pressure because they are normalized by the administration of angiotensin II converting inhibitors or angiotensin receptor antagonist. In experimental animals, hypertension can be induced by the continuous intravenous infusion of small doses of angiotensin II that are not sufficient to evoke an immediate pressor response. However, this condition resembles the characteristics of essential hypertension because the high levels of blood pressure exist with normal plasma levels of angiotensin II. It is suggested that small amounts of angiotensin whose plasma levels are inappropriate for the existing size of extracellular volume stimulate oxidative stress which binds nitric oxide forming peroxynitrite. The latter compound oxidizes arachidonic acid producing isoprostaglandin F2alpha (an isoprostane) which is characterized by a strong antinatriuretic vasoconstrictor renal effect. In this chain of reactions the vasoconstrictor effects derived from oxygen quenching of nitric oxide and increased isoprostane synthesis could explain how hypertension is maintained with normal plasma levels of renin.
Collapse
Affiliation(s)
- J C Romero
- Department of Physiology and Biophysics, Mayo School of Medicine and Division of Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
40
|
Navar LG, Ichihara A, Chin SY, Imig JD. Nitric oxide-angiotensin II interactions in angiotensin II-dependent hypertension. ACTA PHYSIOLOGICA SCANDINAVICA 2000; 168:139-47. [PMID: 10691792 DOI: 10.1046/j.1365-201x.2000.00630.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Many studies indicate that renal haemodynamic function in angiotensin II- (ANG II) dependent hypertension is not reduced as much as would be predicted from the elevated ANG II levels suggesting that counteracting renoprotective mechanisms are activated. One important renoprotective effect is mediated by increased levels of nitric oxide. Recent studies using the ANG II-infused hypertensive rat model have shown that inhibition of nitric oxide synthesis (NOS) causes greater decreases in renal blood flow and glomerular filtration rate in ANG II-infused hypertensive rats than in control rats. This augmented nitric oxide-dependent influence is localized primarily in the cortex and to the preglomerular vasculature. The differential effects on the renal cortex and medulla are also reflected by the differences in NOS activities and protein expression. Ca2+-dependent NOS activity was significantly greater in the cortex but not the medulla of the ANG II-infused hypertensive rats compared with control rats. This was associated with marked activation of endothelial NOS protein levels and smaller increases in neuronal NOS protein levels in the cortex but not in the medulla. In contrast, the Ca2+-independent NOS activity and the inducible NOS protein levels in the cortex were significantly lower in the ANG II-infused hypertensive rats. These data support the hypothesis that cortical Ca2+-dependent NOS, primarily endothelial NOS, is stimulated during the early phases of ANG II-induced hypertension and exerts a renoprotective effect on cortical haemodynamics.
Collapse
Affiliation(s)
- L G Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
41
|
Fernández-Alfonso M, González García C. Interrelaciones sistema reninaangiotensina/sistema NO-GMPc. HIPERTENSION Y RIESGO VASCULAR 2000. [DOI: 10.1016/s1889-1837(00)71038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
42
|
Fernández-Alfonso MS, González C. Nitric oxide and the renin-angiotensin system. Is there a physiological interplay between the systems? J Hypertens 1999; 17:1355-61. [PMID: 10526894 DOI: 10.1097/00004872-199917100-00001] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Opposed actions for nitric oxide (NO) and angiotensin II (Ang II) in vascular contraction and vascular smooth muscle cell proliferation and apoptosis are well documented. In addition, various experimental approaches have shown that NO negatively modulates the renin-angiotensin system by inhibiting angiotensin-converting enzyme (ACE) activity and down-regulating AT1 receptors. On the other hand, Ang II and Ang-(1-7) positively stimulate NO synthesis and release. In this review, we analyse the data suggesting a mutual regulation between the renin-angiotensin and the nitric oxide-generating systems, and we propose a homeostatic interplay between both factors aimed at regulating cardiovascular function.
Collapse
Affiliation(s)
- M S Fernández-Alfonso
- Departamento de Farmacologia, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | | |
Collapse
|
43
|
Symons JD, Stebbins CL, Musch TI. Interactions between angiotensin II and nitric oxide during exercise in normal and heart failure rats. J Appl Physiol (1985) 1999; 87:574-81. [PMID: 10444615 DOI: 10.1152/jappl.1999.87.2.574] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We hypothesized that nitric oxide (NO) opposes ANG II-induced increases in arterial pressure and reductions in renal, splanchnic, and skeletal muscle vascular conductance during dynamic exercise in normal and heart failure rats. Regional blood flow and vascular conductance were measured during treadmill running before (unblocked exercise) and after 1) ANG II AT(1)-receptor blockade (losartan, 20 mg/kg ia), 2) NO synthase (NOS) inhibition [N(G)-nitro-L-arginine methyl ester (L-NAME); 10 mg/kg ia], or 3) ANG II AT(1)-receptor blockade + NOS inhibition (combined blockade). Renal conductance during unblocked exercise (4.79 +/- 0.31 ml x 100 g(-1) x min(-1) x mmHg(-1)) was increased after ANG II AT(1)-receptor blockade (6.53 +/- 0.51 ml x 100 g(-1) x min(-1) x mmHg(-1)) and decreased by NOS inhibition (2.12 +/- 0.20 ml x 100 g(-1) x min(-1) x mmHg(-1)) and combined inhibition (3.96 +/- 0.57 ml x 100 g(-1) x min(-1) x mmHg(-1); all P < 0.05 vs. unblocked). In heart failure rats, renal conductance during unblocked exercise (5.50 +/- 0.66 ml x 100 g(-1) x min(-1) x mmHg(-1)) was increased by ANG II AT(1)-receptor blockade (8.48 +/- 0.83 ml x 100 g(-1) x min(-1) x mmHg(-1)) and decreased by NOS inhibition (2.68 +/- 0.22 ml x 100 g(-1) x min(-1) x mmHg(-1); both P < 0.05 vs. unblocked), but it was unaltered during combined inhibition (4.65 +/- 0.51 ml x 100 g(-1) x min(-1) x mmHg(-1)). Because our findings during combined blockade could be predicted from the independent actions of NO and ANG II, no interaction was apparent between these two substances in control or heart failure animals. In skeletal muscle, L-NAME-induced reductions in conductance, compared with unblocked exercise (P < 0.05), were abolished during combined inhibition in heart failure but not in control rats. These observations suggest that ANG II causes vasoconstriction in skeletal muscle that is masked by NO-evoked dilation in animals with heart failure. Because reductions in vascular conductance between unblocked exercise and combined inhibition were less than would be predicted from the independent actions of NO and ANG II, an interaction exists between these two substances in heart failure rats. L-NAME-induced increases in arterial pressure during treadmill running were attenuated (P < 0.05) similarly in both groups by combined inhibition. These findings indicate that NO opposes ANG II-induced increases in arterial pressure and in renal and skeletal muscle resistance during dynamic exercise.
Collapse
Affiliation(s)
- J D Symons
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California 95616, USA.
| | | | | |
Collapse
|
44
|
Symons JD, Musch TI, Hageman KS, Stebbins CL. Regional blood flow responses to acute ANG II infusion: effects of nitric oxide synthase inhibition. J Cardiovasc Pharmacol 1999; 34:116-23. [PMID: 10413077 DOI: 10.1097/00005344-199907000-00019] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We hypothesized that nitric oxide (NO) opposes regional vasoconstriction caused by acute angiotensin II (ANG II) infusion in conscious rats. Mean arterial pressure (MAP), blood flow, and vascular conductance (regional blood flow/ MAP; ml/min/100 g/mm Hg) were measured and/or calculated before and at 2 min of ANG II infusion (0.05 or 1 microg/kg/min, i.a.) in the absence and presence of NO synthase (NOS) inhibition [N(G)-nitro-L-arginine methyl ester (L-NAME), 0.25 or 1 mg/kg, i.a.]. ANG II reduced stomach and hindlimb conductance only after NOS inhibition. For example, whereas 0.05 microg/kg/min ANG II did not attenuate conductance in the stomach (i.e., 1.04+/-0.08 to 0.93+/-0.12 ml/min/100 g/mm Hg), this variable was reduced (i.e., 0.57+/-0.14 to 0.34-/+0.05 ml/min/100 g/mm Hg; p < 0.05) when ANG II was infused after 0.25 mg/kg L-NAME. In addition, whereas hindlimb conductance was similar before and after administering 1 microg/kg/min ANG II (i.e., 0.13+/-0.01 and 0.09+/-0.02, respectively), this variable was reduced (i.e., 0.07+/-0.01 and 0.02+/-0.00, respectively; p < 0.05) when ANG II was infused after 1 mg/kg L-NAME. These findings indicate that NO opposes ANG II-induced vasoconstriction in the stomach and hindlimb. In contrast, whereas both doses of ANG II decreased (p < 0.05) vascular conductance in the kidneys and small and large intestine regardless of whether NOS inhibition was present, absolute vascular conductance was lower (p < 0.05) after L-NAME. For example, 1 microg/kg ANG II reduced renal conductance from 3.34+/-0.31 to 1.22+/-0.14 (p < 0.05). After 1 mg/kg L-NAME, renal conductance decreased from 1.39+/-0.18 to 0.72+/-0.16 (p < 0.05) during ANG II administration. Therefore the constrictor effects of NOS inhibition and ANG II are additive in these circulations. Taken together, our results indicate that the ability of NO to oppose ANG II-induced constriction is not homogeneous among regional circulations.
Collapse
Affiliation(s)
- J D Symons
- Department of Internal Medicine, University of California, Davis 95616, USA.
| | | | | | | |
Collapse
|
45
|
Millatt LJ, Abdel-Rahman EM, Siragy HM. Angiotensin II and nitric oxide: a question of balance. REGULATORY PEPTIDES 1999; 81:1-10. [PMID: 10395403 DOI: 10.1016/s0167-0115(99)00027-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The vasoconstrictor peptide angiotensin II (Ang II) and the endogenous vasodilator nitric oxide (NO) have many antagonistic effects, as well as influencing each other's production and functioning. In the short-term, Ang II stimulates NO release, thus modulating the vasoconstrictor actions of the peptide. In the long-term, Ang II influences the expression of all three NO synthase (NOS) isoforms, while NO downregulates the Ang II Type I (AT1) receptor, contributing to the protective role of NO in the vasculature. Within the cardiovascular system, Ang II and NO also have antagonistic effects on vascular remodeling and apoptosis. In the kidney, the distribution of the NOS isoforms coincides with the sites of the components of the renin-angiotensin system. NO influences renin secretion from the kidney, and NO-Ang II interactions are important in the control of glomerular and tubular function. In the adrenal gland, NO has been shown to affect Ang II-induced aldosterone synthesis, while in the brain NO appears to influence Ang II-induced drinking behavior, although conflicting data have been reported. In this review, we focus on the diverse ways in which Ang II and NO interact, and on the importance of maintaining a balance between these two important mediators.
Collapse
Affiliation(s)
- L J Millatt
- Department of Anesthesiology, University of Virginia, Health Sciences Center, Charlottesville 22908, USA
| | | | | |
Collapse
|
46
|
Abstract
Nitric oxide (NO) plays critical roles in the control of renal and glomerular hemodynamics, tubuloglomerular feedback response, release of renin and sympathetic transmitters, tubular ion transport, and renal water and sodium excretion. This paper explores the importance of NO in the control of renal water and sodium excretion and in the long-term control of arterial blood pressure. Synthesis of NO, characteristics of NO tissue redox forms, NO synthase activity, and NO synthase isoforms in the kidney are reviewed. To define the role of NO as a natriuretic and antihypertensive factor, the most supportive evidence is summarized, and some contradictory results are also noted. Given the evidence that high salt intake results in high NO concentrations and great NO synthase expression and activity selectively in the renal medulla of the kidney, as well as evidence of a deficiency of the NO synthase activity in Dahl salt-sensitive rats confined in the renal medulla, this report emphasizes the mechanisms by which the renal medullary l-arginine/NO system controls sodium excretion and arterial blood pressure. Other mechanisms for the action of NO on sodium homeostasis such as the action on glomerular filtration rate and the direct effect on tubules are also discussed. We conclude that there is strong evidence that under physiologic conditions, NO plays an important role in the regulation of renal blood flow to the renal medulla and in the tubular regulation of sodium excretion. It is thereby involved in the long-term control of arterial blood pressure, and inhibition or deficiency of NO synthase may result in a sustained hypertension.
Collapse
Affiliation(s)
- A P Zou
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | |
Collapse
|
47
|
Roczniak A, Fryer JN, Levine DZ, Burns KD. Downregulation of neuronal nitric oxide synthase in the rat remnant kidney. J Am Soc Nephrol 1999; 10:704-13. [PMID: 10203353 DOI: 10.1681/asn.v104704] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Chronic renal failure is associated with disturbances in nitric oxide (NO) production. This study was conducted to determine the effect of 5/6 nephrectomy (5/6 Nx) on expression of intrarenal neuronal nitric oxide synthase (nNOS) in the rat. In normal rat kidney, nNOS protein was detected in the macula densa and in the cytoplasm and nuclei of cells of the inner medullary collecting duct by both immunofluorescence and electron microscopy. Western blot analysis revealed that 2 wk after 5/6 Nx, there were significant decreases in nNOS protein expression in renal cortex (sham: 95.42+/-15.60 versus 5/6 Nx: 47.55+/-12.78 arbitrary units, P<0.05, n = 4) and inner medulla (sham: 147.70+/-26.96 versus 5/6 Nx: 36.95+/-17.24 arbitrary units, P<0.005, n = 8). Losartan treatment was used to determine the role of angiotensin II (AngII) AT1 receptors in the inhibition of nNOS expression in 5/6 Nx. Losartan had no effect on the decreased expression of nNOS in the inner medulla, but partially increased nNOS protein expression in the cortex of 5/6 Nx rats. In contrast, in sham rats losartan significantly inhibited nNOS protein expression in the cortex (0.66+/-0.04-fold of sham values, P<0.05, n = 6) and inner medulla (0.74+/-0.12-fold of sham values, P<0.05, n = 6). nNOS mRNA was significantly decreased in cortex and inner medulla from 5/6 Nx rats, and the effects of losartan on nNOS mRNA paralleled those observed on nNOS protein expression. These data indicate that 5/6 Nx downregulates intrarenal nNOS mRNA and protein expression. In normal rats, AngII AT1 receptors exert a tonic stimulatory effect on expression of intrarenal nNOS. These findings suggest that the reduction in intrarenal nNOS expression in 5/6 Nx may play a role in contributing to hypertension and altered tubular transport responses in chronic renal failure.
Collapse
Affiliation(s)
- A Roczniak
- Department of Medicine, University of Ottawa and Ottawa General Hospital, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Farquhar WB, Morgan AL, Zambraski EJ, Kenney WL. Effects of acetaminophen and ibuprofen on renal function in the stressed kidney. J Appl Physiol (1985) 1999; 86:598-604. [PMID: 9931196 DOI: 10.1152/jappl.1999.86.2.598] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise, salt restriction, and/or dehydration causes transient reductions in renal function that may be buffered by vasodilatory prostaglandins (PGs). Over-the-counter (OTC) analgesics have the potential to alter renal hemodynamics by inhibiting renal PGs. Therefore, we tested the renal effects of the maximal recommended dose of acetaminophen (Acet, 4 g/day) and ibuprofen (Ibu, 1.2 g/day) vs. a placebo (Pl) in humans subjected to progressive renal stresses. After baseline measurements, 12 fit young (25 +/- 1 yr) men and women underwent 3 days of a low (10 meq/day)-sodium diet while taking one of the drugs or Pl (crossover design). Day 4 involved dehydration (-1.6% body wt) followed by 45 min of treadmill exercise (65% maximal O2 uptake) in the heat (36 degreesC). These combined stressors caused dramatic decreases in effective renal plasma flow, glomerular filtration rate (GFR), and sodium excretion. Baseline GFR (range: 118-123 ml/min) decreased to 78 +/- 4, 73 +/- 5, and 82 +/- 5 ml/min postexercise in the Acet, Ibu, and Pl trials, respectively, with a significantly greater decrease in GFR in the Ibu trial (P < 0. 05 vs. Pl). OTC Ibu has small but statistically significant effects on GFR during exercise in a sodium- and volume-depleted state; OTC Acet was associated with no such effects.
Collapse
Affiliation(s)
- W B Farquhar
- Noll Physiological Research Center, Pennylvania State University, University Park, Pennsylvania 16802-6900, USA
| | | | | | | |
Collapse
|
49
|
Kurtz A, Wagner C. Role of nitric oxide in the control of renin secretion. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:F849-62. [PMID: 9843901 DOI: 10.1152/ajprenal.1998.275.6.f849] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Because of the significant constitutive expression of NO synthases in the juxtaglomerular apparatus, nitric oxide (NO) is considered as a likely modulator of renin secretion. In most instances, NO appears as a tonic enhancer of renin secretion, acting via inhibition of cAMP degradation through the action of cGMP. Depending on as yet unknown factors, the stimulatory effect of NO on renin secretion may also switch to an inhibitory one that is compatible with the inhibition of renin secretion by cGMP-dependent protein kinase activity. Whether NO plays a direct regulatory role or a more permissive role in the control of renin secretion remains to be answered.
Collapse
Affiliation(s)
- A Kurtz
- Institut für Physiologie, Universität Regensburg, Regensburg D-93040, Germany
| | | |
Collapse
|
50
|
Clayton JS, Clark KL, Johns EJ, Drew GM. Effects of prostaglandins and nitric oxide on the renal effects of angiotensin II in the anaesthetized rat. Br J Pharmacol 1998; 124:1467-74. [PMID: 9723960 PMCID: PMC1565550 DOI: 10.1038/sj.bjp.0702003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The potential influences of nitric oxide (NO) and prostaglandins on the renal effects of angiotensin II (Ang II) have been investigated in the captopril-treated anaesthetized rat by examining the effect of indomethacin or the NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME), on the renal responses obtained during infusion of Ang II directly into the renal circulation. 2. Intrarenal artery (i.r.a.) infusion of Ang II (1-30 ng kg(-1) min(-1)) elicited a dose-dependent decrease in renal vascular conductance (RVC; -38+/-3% at 30 ng kg(-1) min(-1); P < 0.01) and increase in filtration fraction (FF; +49+/-8%; P < 0.05) in the absence of any change in carotid mean arterial blood pressure (MBP). Urine output (Uv), absolute (UNaV) and fractional sodium excretion (FENa), and glomerular filtration rate (GFR) were unchanged during infusion of Ang II 1-30 ng kg(-1) min(-1) (+6+/-17%, +11+/-17%, +22+/-23%, and -5+/-9%, respectively, at 30 ng kg(-1) min(-1)). At higher doses, Ang II (100 and 300 ng kg(-1) min(-1)) induced further decreases in RVC, but with associated increases in MBP, Uv and UNaV. 3. Pretreatment with indomethacin (10 mg kg(-1) i.v.) had no significant effect on basal renal function, or on the Ang II-induced reduction in RVC (-25+/-7% vs -38+/-3% at Ang II 30 ng kg(-1) min(-1)). In the presence of indomethacin, Ang II tended to cause a dose-dependent decrease in GFR (-38+/-10% at 30 ng kg(-1) min(-1)); however, this effect was not statistically significant (P=0.078) when evaluated over the dose range of 1-30 ng kg(-1) min(-1), and was not accompanied by any significant changes in Uv, UNaV or FENa (-21+/-12%, -18+/-16% and +36+/-38%, respectively). 4. Pretreatment with L-NAME (10 microg kg(-1) min(-1) i.v.) tended to reduce basal RVC (control -11.8+/-1.4, +L-NAME -7.9+/-1.8 ml min(-1) mmHg(-1) x 10(-2)), and significantly increased basal FF (control +15.9+/-0.8, +L-NAME +31.0+/-3.7%). In the presence of L-NAME, renal vasoconstrictor responses to Ang II were not significantly modified (-38+/-3% vs -35+/-13% at 30 ng kg(-1) min(-1)), but Ang II now induced dose-dependent decreases in GFR, Uv and UNaV (-51+/-11%, -41+/-14% and -31+/-17%, respectively, at an infusion rate of Ang II, 30 ng kg(-1) min(-1)). When evaluated over the range of 1-30 ng kg(-1) min(-1), the effect of Ang II on GFR and Uv were statistically significant (P < 0.05), but on UNaV did not quite achieve statistical significance (P=0.066). However, there was no associated change in FENa observed, suggesting a non-tubular site of interaction between Ang II and NO. 5. In contrast to its effects after pretreatment with L-NAME alone, Ang II (1-30 ng kg(-1) min(-1)) failed to reduce renal vascular conductance in rats pretreated with the combination of L-NAME and the selective angiotensin AT1 receptor antagonist, GR117289 (1 mg kg(-1) i.v.). This suggests that the renal vascular effects of Ang II are mediated through AT1 receptors. Over the same dose range, Ang II also failed to significantly reduce GFR or Uv. 6. In conclusion, the renal haemodynamic effects of Ang II in the rat kidney appear to be modulated by cyclooxygenase-derived prostaglandins and NO. The precise site(s) of such an interaction cannot be determined from the present data, but the data suggest complex interactions at the level of the glomerulus.
Collapse
Affiliation(s)
- J S Clayton
- Systems Biology, Glaxo Wellcome Medicines Research Centre, Stevenage, Herts
| | | | | | | |
Collapse
|