51
|
Tikellis C, Cooper ME, Bialkowski K, Johnston CI, Burns WC, Lew RA, Smith AI, Thomas MC. Developmental expression of ACE2 in the SHR kidney: a role in hypertension? Kidney Int 2006; 70:34-41. [PMID: 16710353 DOI: 10.1038/sj.ki.5000428] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The abnormal development of the intrarenal renin-angiotensin system (RAS) is thought contribute to adult-onset hypertension in the spontaneously hypertensive rat (SHR). Angiotensin-converting enzyme 2 (ACE2) is a novel enzyme with complementary actions to that of ACE. Recent studies have shown that ACE2 expression is reduced in the adult SHR. However, its regulation in pre-hypertensive animals is unknown. In this study, we examine the developmental expression of ACE2 in the rodent kidney and its temporal expression, as it relates to the development of hypertension in the SHR model. Kidneys from SHR and normotensive Wistar Kyoto (WKY) rats (n=8-12/group) at birth, 6 weeks of age, and adulthood (80 days) were examined. Gene expression and activity of ACE2 were determined by real-time reverse transcription-polymerase chain reaction and quenched fluorescence assays, respectively. Renal expression was localized by in situ hybridization and immunohistochemistry. The expression and ACE2 activity are significantly increased in the SHR kidney at birth. With the onset of hypertension, the tubular expression of ACE2 falls in SHR compared to WKY and remains reduced in the adult SHR kidney. Glomerular expression is paradoxically increased in the SHR glomerulus. The overall developmental pattern of ACE2 expression in the SHR kidney is also modified, with declining expression over the course of renal development. The developmental pattern of ACE2 expression in the SHR kidney is altered before the onset of hypertension, consistent with the key role of the RAS in the pathogenesis of adult-onset hypertension. Further research is required to distinguish the contribution of these changes to the development and progression of hypertension in this model.
Collapse
Affiliation(s)
- C Tikellis
- Baker Heart Research Institute, St Kilda Central, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|
52
|
van der Wouden EA, Henning RH, Deelman LE, Roks AJM, Boomsma F, de Zeeuw D. Does angiotensin (1-7) contribute to the anti-proteinuric effect of ACE-inhibitors. J Renin Angiotensin Aldosterone Syst 2006; 6:96-101. [PMID: 16470489 DOI: 10.3317/jraas.2005.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACE-I) reduce proteinuria and protect the kidney in proteinuric renal disease. During ACE-I therapy, circulating levels of angiotensin (1-7) [Ang (1-7)] are increased. As cardiac and renal protective effects of Ang (1-7) have been reported, we questioned whether Ang (1-7) contributes to the anti-proteinuric effects of ACE-I treatment. Therefore, we evaluated whether Ang (1-7) infusion reduces proteinuria in a rat model of adriamycin-induced renal disease. In addition, the effect of a selective Ang (1-7) blocker, [D-Ala7]-Ang (1-7) (A779), was investigated in rats treated with the ACE-I, lisinopril (LIS). Six weeks after induction of proteinuria, therapy was started in four different groups: control, Ang (1-7), LIS, and LIS+A779. After two weeks, the rats were sacrificed. Six weeks after injection of adriamycin, the rats had developed proteinuria of 323+/-40 mg/24 hours. The proteinuria remained stable in the control group and in the Ang (1-7) group, but was reduced in both LIS and LIS+A779-treated groups. Similarly, blood pressure (BP) was unchanged in the control and the Ang (1-7) groups, but reduced in both the LIS and the LIS+A779 groups. Plasma levels of Ang (1-7) were increased in the Ang (1-7) and in both LIS-treated groups. We conclude that systemic Ang (1-7) plays no major role in the anti-proteinuric and BPlowering effects of ACE-I in this rat model of adriamycin-induced nephrosis.
Collapse
Affiliation(s)
- Els A van der Wouden
- Department of Clinical Pharmocology, Division of Nephrology, Groningen University Institute for Drug Exploration (GUIDE), University Medical Center Groningen, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
53
|
Lemos VS, Silva DMR, Walther T, Alenina N, Bader M, Santos RAS. The endothelium-dependent vasodilator effect of the nonpeptide Ang(1-7) mimic AVE 0991 is abolished in the aorta of mas-knockout mice. J Cardiovasc Pharmacol 2006; 46:274-9. [PMID: 16116331 DOI: 10.1097/01.fjc.0000175237.41573.63] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recently, we demonstrated that the endothelium-dependent vasodilator effect of angiotensin(1-7) in the mouse aorta is abolished by genetic deletion of the G protein-coupled receptor encoded by the Mas protooncogene. To circumvent the limitations posed by the possible metabolism of Ang(1-7) in this vessel, in this work we studied the mechanism underlying the vasorelaxant effect of AVE 0991, a nonpeptide mimic of the effects of Ang(1-7), using wild-type and Mas-deficient mice. Ang(1-7) and AVE 0991 induced an equipotent concentration-dependent vasodilator effect in aortic rings from wild-type mice that was dependent on the presence of endothelium. The vasodilator effect of Ang(1-7) and AVE 0991 was completely blocked by 2 specific Ang(1-7) receptor antagonists, A-779 and D-Pro-Ang(1-7), and by inhibition of NO synthase with L-NAME. Moreover, in aortic rings from Mas-deficient mice, the vasodilator effect of both Ang(1-7) and AVE 0991 was abolished. In contrast, the vasodilator effect of acetylcholine and substance P were preserved in Mas-null mice. In addition, the vasoconstriction effect induced by Ang II was slightly increased, and the vasodilation induced by the AT2 agonist CGP 42112A was not altered in Mas-deficient mice. Our results show that Ang(1-7) and AVE 0991 produced an NO-dependent vasodilator effect in the mouse aorta that is mediated by the G protein-coupled receptor Mas.
Collapse
Affiliation(s)
- Virginia S Lemos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil, and Department of Cardiology and Pneumology, University Hospital Benjamin Franklin, Free University, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
54
|
Wilkinson-Berka JL. Angiotensin and diabetic retinopathy. Int J Biochem Cell Biol 2005; 38:752-65. [PMID: 16165393 DOI: 10.1016/j.biocel.2005.08.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 07/25/2005] [Accepted: 08/10/2005] [Indexed: 12/16/2022]
Abstract
Diabetic retinopathy develops in patients with both type 1 and type 2 diabetes and is the major cause of vision loss and blindness in the working population. In diabetes, damage to the retina occurs in the vasculature, neurons and glia resulting in pathological angiogenesis, vascular leakage and a loss in retinal function. The renin-angiotensin system is a causative factor in diabetic microvascular complications inducing a variety of tissue responses including vasoconstriction, inflammation, oxidative stress, cell hypertrophy and proliferation, angiogenesis and fibrosis. All components of the renin-angiotensin system including the angiotensin type 1 and angiotensin type 2 receptors have been identified in the retina of humans and rodents. There is evidence from both clinical and experimental models of diabetic retinopathy and hypoxic-induced retinal angiogenesis that the renin-angiotensin system is up-regulated. In these situations, retinal dysfunction has been linked to angiotensin-mediated induction of growth factors including vascular endothelial growth factor, platelet-derived growth factor and connective tissue growth factor. Evidence to date indicates that blockade of the renin-angiotensin system can confer retinoprotection in experimental models of diabetic retinopathy and ischemic retinopathy. This review examines the role of the renin-angiotensin system in diabetic retinopathy and the potential of its blockade as a treatment strategy for this vision-threatening disease.
Collapse
|
55
|
Der Sarkissian S, Huentelman MJ, Stewart J, Katovich MJ, Raizada MK. ACE2: A novel therapeutic target for cardiovascular diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2005; 91:163-98. [PMID: 16009403 DOI: 10.1016/j.pbiomolbio.2005.05.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypertension afflicts over 65 million Americans and poses an increased risk for cardiovascular morbidity such as stroke, myocardial infarction and end-stage renal disease resulting in significant mortality. Overactivity of the renin-angiotensin system (RAS) has been identified as an important determinant that is implicated in the etiology of these diseases and therefore represents a major target for therapy. In spite of the successes of drugs inhibiting various elements of the RAS, the incidence of hypertension and cardiovascular diseases remain steadily on the rise. This has lead many investigators to seek novel and innovative approaches, taking advantage of new pathways and technologies, for the control and possibly the cure of hypertension and related pathologies. The main objective of this review is to forward the concept that gene therapy and the genetic targeting of the RAS is the future avenue for the successful control and treatment of hypertension and cardiovascular diseases. We will present argument that genetic targeting of angiotensin-converting enzyme 2 (ACE2), a newly discovered member of the RAS, is ideally poised for this purpose. This will be accomplished by discussion of the following: (i) summary of our current understanding of the RAS with a focus on the systemic versus tissue counterparts as they relate to hypertension and other cardiovascular pathologies; (ii) the newly discovered ACE2 enzyme with its physiological and pathophysiological implications; (iii) summary of the current antihypertensive pharmacotherapy and its limitations; (iv) the discovery and design of ACE inhibitors; (v) the emerging concepts for ACE2 drug design; (vi) the current status of genetic targeting of the RAS; (vii) the potential of ACE2 as a therapeutic target for hypertension and cardiovascular disease treatment; and (viii) future perspectives for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Shant Der Sarkissian
- Department of Physiology and Functional Genomics, College of Medicine, and the McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
56
|
Abstract
Angiotensin-(1-7) (Ang-(1-7)) is now considered to be a biologically active member of the renin-angiotensin system. The functions of Ang-(1-7) are often opposite to those attributed to the main effector component of the renin-angiotensin system, Ang II. Chronic administration of angiotensin-converting enzyme inhibitors (ACEI) increases 10- to 25-fold the plasma levels of this peptide, suggesting that part of the beneficial effects of ACEI could be mediated by Ang-(1-7). Ang-(1-7) can be formed from Ang II or directly from Ang I. Other enzymatic pathways for Ang-(1-7) generation have been recently described involving the novel ACE homologue ACE2. This enzyme can form Ang-(1-7) from Ang II or less efficiently by the hydrolysis of Ang I to Ang-(1-9) with subsequent Ang-(1-7) formation. The biological relevance of Ang-(1-7) has been recently reinforced by the identification of its receptor, the G-protein-coupled receptor Mas. Heart and blood vessels are important targets for the formation and actions of Ang-(1-7). In this review we will discuss recent findings concerning the biological role of Ang-(1-7) in the heart and blood vessels, taking into account aspects related to its formation and effects on these tissues. In addition, we will discuss the potential of Ang-(1-7) and its receptor as a target for the development of new cardiovascular drugs.
Collapse
Affiliation(s)
- A J Ferreira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | | |
Collapse
|
57
|
Bürgelová M, Kramer HJ, Teplan V, Thumová M, Cervenka L. Effects of angiotensin-(1–7) blockade on renal function in rats with enhanced intrarenal Ang II activity. Kidney Int 2005; 67:1453-61. [PMID: 15780097 DOI: 10.1111/j.1523-1755.2005.00222.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Increasing evidence suggests that angiotensin-(1-7) [Ang-(1-7)] acts as an endogenous antagonist of Ang II when the renin-angiotensin system (RAS) is activated. In the present study, we therefore compared the effects of acute intrarenal (i.r.) Ang-(1-7) receptor blockade on renal function under conditions of normal and increased intrarenal Ang II concentration. METHODS Salt-replete Hannover-Sprague Dawley rats (HanSD) served as control animals. As models with enhanced action of Ang II we first used transgenic rats harboring the Ren-2 renin gene (TGR), second, Ang II-infused rats, third, 2-kidney, 1-clip (2K1C) hypertensive rats on normal salt intake, and fourth, salt-depleted TGR and HanSD. RESULTS I.r. Ang-(1-7) receptor blockade elicited significant decreases in glomerular filtration rate (GFR), renal plasma flow (RPF), and sodium excretion in 2K1C rats, and in salt-depleted TGR and HanSD. In contrast, i.r. Ang-(1-7) receptor blockade did not significantly change GFR, RPF, and sodium excretion in salt-replete TGR and HanSD, or in Ang II-infused rats. CONCLUSION These findings suggest that under conditions of normal intrarenal RAS activity and increased intrarenal Ang II action by infusion of Ang II or by insertion of a renin gene in salt-replete conditions, Ang-(1-7) is not an important factor in the regulation of renal function. In contrast, under conditions of endogenous RAS activation due to clipping of the renal artery or to sodium restriction, Ang-(1-7) serves as opponent of the vasoconstrictor actions of Ang II.
Collapse
Affiliation(s)
- Marcela Bürgelová
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
58
|
Giacchetti G, Sechi LA, Rilli S, Carey RM. The renin-angiotensin-aldosterone system, glucose metabolism and diabetes. Trends Endocrinol Metab 2005; 16:120-6. [PMID: 15808810 DOI: 10.1016/j.tem.2005.02.003] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In diabetes mellitus (DM), the circulating renin-angiotensin system (RAS) is suppressed, but the renal tissue RAS is activated. Hyperglycemia increases tissue angiotensin II (Ang II), which induces oxidative stress, endothelial damage and disease pathology including vasoconstriction, thrombosis, inflammation and vascular remodeling. In early DM, the type 1 Ang II (AT(1)) receptor is upregulated but the type 2 Ang II (AT(2)) receptor is downregulated. This imbalance can predispose the individual to tissue damage. Hyperglycemia also increases the production of aldosterone, which has an unknown contribution to tissue damage. The insulin resistance state is associated with upregulation of the AT(1) receptor and an increase in oxygen free radicals in endothelial tissue caused by activation of NAD(P)H oxidase. Treatment with an AT(1) receptor blocker normalizes oxidase activity and improves endothelial function. An understanding of the tissue renin-angiotensin-aldosterone system, which is a crucial factor in the progression of tissue damage in DM, is imperative for protection against tissue damage in this chronic disease.
Collapse
Affiliation(s)
- Gilberta Giacchetti
- The Division of Endocrinology, Department of Internal Medicine, Universita Politecnica della Marche, 60020 Ancona, Italy
| | | | | | | |
Collapse
|
59
|
Fernandes L, Fortes ZB, Casarini DE, Nigro D, Tostes RCA, Santos RAS, de Carvalho MHC. Role of PGI2 and effects of ACE inhibition on the bradykinin potentiation by angiotensin-(1-7) in resistance vessels of SHR. ACTA ACUST UNITED AC 2005; 127:183-9. [PMID: 15680485 DOI: 10.1016/j.regpep.2004.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Accepted: 12/01/2004] [Indexed: 01/09/2023]
Abstract
The present study determined the participation of PGI2 in the angiotensin-(1-7) [Ang-(1-7)]/bradykinin (BK) interaction, in the presence and absence of Angiotensin Converting Enzyme (ACE) inhibition, trying to correlate it with tissue levels of both peptides. The isolated mesenteric arteriolar bed of Spontaneously Hypertensive Rats (SHR) was perfused with Krebs or Krebs plus enalaprilat (10 nM), and drugs were injected alone or in association. BK (10 ng)-induced relaxation was potentiated by Ang-(1-7) (2.2 microg) in the presence or absence of enalaprilat. Ang-(1-7) receptor blockade [A-779 (4.8 microg)] did not interfere with the BK effect in preparations perfused with normal Krebs, but reversed the increased BK relaxation observed after ACE inhibition. PGI2 release by mesenteric vessels was not altered by BK or Ang-(1-7) alone, but was increased when both peptides were injected in association, in the absence or in the presence of enalaprilat. ACE inhibition caused a 2-fold increase in the BK tissue levels, and a significant decrease in the Ang-(1-7) values. We conclude that endogenous Ang-(1-7) has an important contribution to the effect of ACE inhibitors participating in the enhancement of BK response. The mechanism of Ang-(1-7) potentiating effect probably involves an increased production of PGI2. Our results suggest that a different enzymatic pathway (non-related to ACE) is involved in the local Ang-(1-7) metabolism.
Collapse
Affiliation(s)
- Liliam Fernandes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo-SP 05508900, Brazil.
| | | | | | | | | | | | | |
Collapse
|
60
|
Maia LG, Ramos MC, Fernandes L, de Carvalho MHC, Campagnole-Santos MJ, Souza dos Santos RA. Angiotensin-(1-7) antagonist A-779 attenuates the potentiation of bradykinin by captopril in rats. J Cardiovasc Pharmacol 2004; 43:685-91. [PMID: 15071356 DOI: 10.1097/00005344-200405000-00011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We evaluated the possibility that endogenous angiotensin-(1-7) [Ang-(1-7)] could participate in the potentiation of bradykinin (BK) by the angiotensin-converting enzyme inhibitor (ACEI) captopril in conscious Wistar rats. Catheters were introduced into descending aorta (through the left carotid artery) for BK injection, femoral artery for arterial pressure measurement, and both femoral veins for BK injection and vehicle or Ang-(1-7) antagonist, A-779 infusion. Infusion of vehicle or A-779 started 40 to 45 minutes after captopril administration. Sequential BK dose-response curves were made before, 10 minutes after captopril, and within 10 minutes of infusion of vehicle or A-779. To evaluate angiotensin I conversion, dose-response curves for angiotensin I and angiotensin II were made following the same protocol used for BK. Captopril treatment markedly increased the BK hypotensive effect and significantly decreased angiotensin I conversion. Infusion of A-779 did not modify the angiotensin II pressor effect or the effect of captopril on angiotensin I conversion. However, A-779 significantly reduced the potentiating effect of captopril on the hypotensive effect of BK administered intravenously or intra-arterially. These results suggest that endogenous Ang-(1-7) and/ or an Ang-(1-7)-related peptide plays an important role in the BK potentiation by ACEI through a mechanism not dependent upon inhibition of ACE hydrolytic activity.
Collapse
Affiliation(s)
- Luciana Gonçalves Maia
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | |
Collapse
|
61
|
Kabara S, Hirota K, Yoshioka H, Kudo T, Ishihara H, Matsuki A. Differential effects of thiopental on methacholine- and serotonin-induced bronchoconstriction in dogs. Br J Anaesth 2003; 91:379-84. [PMID: 12925478 DOI: 10.1093/bja/aeg190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Thiopental sometimes causes bronchospasm during induction of anaesthesia. In addition, we have reported previously that thiopental produced transient bronchospasm, which was blocked by atropine pretreatment, and worsened histamine-induced bronchoconstriction in dogs. Previous in vitro reports suggest that synthesis of contractile cyclooxygenase products, such as thromboxane A(2), may be involved in the mechanism of bronchospasm. However, the in vivo spastic effects have not been defined comprehensively. METHODS Twenty-seven mongrel dogs were anaesthetized with pentobarbital. Bronchoconstriction was elicited with methacholine (0.5 microg kg(-1)+5.0 microg kg(-1) min(-1); Mch group, n=7) or serotonin (10 microg kg(-1)+1 mg kg(-1) h(-1); 5HT group, n=20), and assessed as percentage changes in bronchial cross-sectional area (BCA, basal=100%) using a bronchoscope. In the 5HT group, dogs were subdivided into four groups of five each: S-5HT, I-5HT, 5HT-S and 5HT-A. In the S-5HT and I-5HT groups, 30 min before serotonin infusion dogs were given saline and indomethacin respectively at 5 mg kg(-1) i.v. In all groups, 30 min after bronchoconstrictor infusion started, dogs were given thiopental at doses between 0 (saline) and 20 mg kg(-1). In the 5HT-S and 5HT-A groups, dogs were given saline or atropine 0.2 mg kg(-1) i.v. 5 min after thiopental 20 mg kg(-1). RESULTS Methacholine and serotonin reduced BCA by about 50 and 40% respectively. Thiopental 20 mg kg(-1) increased and decreased BCA by about 20 and 10% in the Mch and 5HT groups respectively. Indomethacin and atropine did not attenuate the potentiation of serotonin bronchoconstriction produced by thiopental. CONCLUSION The present study indicates that thiopental may attenuate or worsen bronchoconstriction induced by muscarinic or serotonin receptor stimulation, respectively. The synthesis of contractile cyclooxygenase products and cholinergic stimulation may not be involved in the contractile effect of thiopental on serotonin bronchoconstriction.
Collapse
Affiliation(s)
- S Kabara
- Department of Anesthesiology, University of Hirosaki School of Medicine, Hirosaki 036-8563, Japan
| | | | | | | | | | | |
Collapse
|
62
|
Abstract
The renin-angiotensin system (RAS) is a coordinated cascade of proteins and peptide hormones, the principal effector of which is angiotensin II (ANG II). Evidence now indicates that the kidney regulates its function via a self-contained RAS in a paracrine fashion. In diabetic nephropathy, the intrarenal generation of ANG II is increased, in spite of suppression of the systemic RAS. This increase can contribute to the progression of diabetic nephropathy via several hemodynamic, tubular and growth-promoting actions. ANG II induces insulin resistance. ANG II type-1 (AT(1)) and type-2 (AT(2)) receptors are downregulated in chronic diabetes, but decreased AT(2) receptor expression might contribute to early diabetic nephropathy by reducing AT(2) receptor-mediated beneficial actions that are counter-regulatory to those of the AT(1) receptor. AT(2) receptor stimulation might account for part of the renal protection seen with AT(1) receptor blockade. A rat model of accelerated diabetic nephropathy is the (mREN-2) 27 renin transgenic rat treated with streptozotocin in which both the intrarenal and extrarenal RAS is activated.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Box 801414, University of Virginia Health System, Charlottesville, VA 22908-1414, USA.
| | | |
Collapse
|
63
|
Santos RAS, e Silva ACS, Maric C, Silva DMR, Machado RP, de Buhr I, Heringer-Walther S, Pinheiro SVB, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss HP, Speth R, Walther T. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci U S A 2003; 100:8258-63. [PMID: 12829792 PMCID: PMC166216 DOI: 10.1073/pnas.1432869100] [Citation(s) in RCA: 1366] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The renin-angiotensin system plays a critical role in blood pressure control and body fluid and electrolyte homeostasis. Besides angiotensin (Ang) II, other Ang peptides, such as Ang III [Ang-(2-8)], Ang IV [Ang-(3-8)], and Ang-(1-7) may also have important biological activities. Ang-(1-7) has become an angiotensin of interest in the past few years, because its cardiovascular and baroreflex actions counteract those of Ang II. Unique angiotensin-binding sites specific for this heptapeptide and studies with a selective Ang-(1-7) antagonist indicated the existence of a distinct Ang-(1-7) receptor. We demonstrate that genetic deletion of the G protein-coupled receptor encoded by the Mas protooncogene abolishes the binding of Ang-(1-7) to mouse kidneys. Accordingly, Mas-deficient mice completely lack the antidiuretic action of Ang-(1-7) after an acute water load. Ang-(1-7) binds to Mas-transfected cells and elicits arachidonic acid release. Furthermore, Mas-deficient aortas lose their Ang-(1-7)-induced relaxation response. Collectively, these findings identify Mas as a functional receptor for Ang-(1-7) and provide a clear molecular basis for the physiological actions of this biologically active peptide.
Collapse
Affiliation(s)
- Robson A. S. Santos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Ana C. Simoes e Silva
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Christine Maric
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Denise M. R. Silva
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Raquel Pillar Machado
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Insa de Buhr
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Silvia Heringer-Walther
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Sergio Veloso B. Pinheiro
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Myriam Teresa Lopes
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Michael Bader
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Elizabeth P. Mendes
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Virgina Soares Lemos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Heinz-Peter Schultheiss
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Robert Speth
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Thomas Walther
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
- To whom correspondence should be addressed at: Benjamin Franklin Medical
Center, Department of Cardiology and Pneumology, Free University of Berlin,
Hindenburgdamm 30, 12200 Berlin, Germany. E-mail:
| |
Collapse
|
64
|
Carey RM, Siragy HM. Newly recognized components of the renin-angiotensin system: potential roles in cardiovascular and renal regulation. Endocr Rev 2003; 24:261-71. [PMID: 12788798 DOI: 10.1210/er.2003-0001] [Citation(s) in RCA: 380] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The renin-angiotensin system (RAS) is a coordinated hormonal cascade in the control of cardiovascular, renal, and adrenal function that governs body fluid and electrolyte balance, as well as arterial pressure. The classical RAS consists of a circulating endocrine system in which the principal effector hormone is angiotensin (ANG) II. ANG is produced by the action of renin on angiotensinogen to form ANG I and its subsequent conversion to the biologically active octapeptide by ANG-converting enzyme. ANG II actions are mediated via the ANG type 1 receptor. Here, we discuss recent advances in our understanding of the components and actions of the RAS, including local tissue RASs, a renin receptor, ANG-converting enzyme-2, ANG (1-7), the function of the ANG type 2 receptor, and ANG receptor heterodimerization. The role of the RAS in the regulation of cardiovascular and renal function is reviewed and discussed in light of these newly recognized components.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia 22908, USA.
| | | |
Collapse
|
65
|
Haulica I, Bild W, Mihaila CN, Ionita T, Boisteanu CP, Neagu B. Biphasic effects of angiotensin (1-7) and its interactions with angiotensin II in rat aorta. J Renin Angiotensin Aldosterone Syst 2003; 4:124-8. [PMID: 12806596 DOI: 10.3317/jraas.2003.013] [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
Using isolated rat aortic rings perfused with Krebs-Henseleit saline, the vascular effects of angiotensin (1-7) (Ang [1-7]) and its interactions with angiotensin II (Ang II) were investigated. Ang (1-7) induced endothelium-dependent relaxation and vasodilating effects in preparations precontracted with phenylephrine. Without preconstriction, Ang (1-7) at high doses (10(-6) 10(-5) M) produced either a significant inhibition of Ang II-induced vasoconstriction or a non-tachyphylactic vasopressor response. While losartan inhibited the vasoconstriction induced by Ang (1-7), A779 blocked only its relaxation. Unlike losartan, blockade of AT(2)-receptors with PD 123319 had no effect. Taking into account the biphasic effects of angiotensin (1-7), we propose that it is one of the active components of the renin-angiotensin system, which is involved as a modulator both in the counter-regulatory actions of Ang II and in the self-regulation of its own vasodilating effects.
Collapse
Affiliation(s)
- Ion Haulica
- Labroatory of Experimental and Applied Physiology, The Romanian Academy Iasi, Iasi, Romania.
| | | | | | | | | | | |
Collapse
|
66
|
Sampaio WO, Nascimento AAS, Santos RAS. Systemic and regional hemodynamic effects of angiotensin-(1-7) in rats. Am J Physiol Heart Circ Physiol 2003; 284:H1985-94. [PMID: 12573992 DOI: 10.1152/ajpheart.01145.2002] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The systemic and regional hemodynamics effects of ANG-(1-7) were examined in urethane-anesthetized rats. The blood flow distribution (kidneys, skin, mesentery, lungs, spleen, brain, muscle, and adrenals), cardiac output, and total peripheral resistance were investigated by using fluorescent microspheres. Blood pressure and heart rate were recorded from the brachial artery. ANG-(1-7) infusion (110 fmol x min(-1) x 10 min(-1) iv) significantly increased blood flow to the kidney (5.10 +/- 1.07 to 8.30 +/- 0.97 ml x min(-1) x g(-1)), mesentery (0.73 +/- 0.16 to 1.17 +/- 0.49 ml x min(-1) x g(-1)), brain (1.32 +/- 0.44 to 2.18 +/- 0.85 ml x min(-1) x g(-1)), and skin (0.07 +/- 0.02 to 0.18 +/- 0.07 ml x min(-1) x g(-1)) and the vascular conductance in these organs. ANG-(1-7) also produced a significant increase in cardiac index (30%) and a decrease in total peripheral resistance (2.90 +/- 0.55 to 2.15 +/- 0.28 mmHg x ml(-1) x min x 100 g). Blood flow to the spleen, muscle, lungs, and adrenals, as well as the blood pressure and heart rate, were not altered by the ANG-(1-7) infusion. The selective ANG-(1-7) antagonist A-779 reduced the blood flow in renal, cerebral, mesenteric, and cutaneous beds and blocked the ANG-(1-7)-induced vasodilatation in the kidney, mesentery, and skin, suggesting a significant role of endogenous ANG-(1-7) in these territories. The effects of ANG-(1-7) on the cerebral blood flow, cardiac index, systolic volume, and total peripheral resistance were partially attenuated by A-779. A high dose of ANG-(1-7) (11 pmol x min(-1) x 10 min(-1)) caused an opposite effect of that produced by the low dose. Our results show for the first time that ANG-(1-7) has a previously unsuspected potent effect in the blood flow distribution and systemic hemodynamics.
Collapse
Affiliation(s)
- Walkyria O Sampaio
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Brazil
| | | | | |
Collapse
|
67
|
Abstract
Increased understanding of the contribution of angiotensin peptides to the physiologic control of arterial pressure and cardiovascular regulation has been made possible with the introduction of agents that either inhibit the activity of angiotensins forming enzymes or block the action of the peptides at their specific receptor subtypes. This review highlights some of the lessons that have been learned from the study of the actions of angiotensin-(1-7) and its inter-relationship with other vasodilator mechanisms that modulate the control systems that determine blood pressure and tissue perfusion. The studies suggest that the renin-angiotensin system acts as a humoral mechanism for blood pressure control through the generation of several distinct forms of angiotensin peptides that may bind to diverse receptor subtypes.
Collapse
Affiliation(s)
- Carlos M Ferrario
- Hypertension and Vascular Disease Center, Wake Forest University Health Science Center, Winston-Salem, NC 27157, USA.
| |
Collapse
|
68
|
Oliveira MA, Carvalho MHC, Nigro D, Passaglia RDCAT, Fortes ZB. Elevated glucose blocks angiotensin-(1-7) and bradykinin interaction: the role of cyclooxygenase products. Peptides 2003; 24:449-54. [PMID: 12732344 DOI: 10.1016/s0196-9781(03)00061-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The interaction between angiotensin-(1-7) [Ang-(1-7)] and bradykinin (BK) was studied in the isolated mesenteric arteriolar bed of control and diabetic rats perfused with either 5.5 or 22 mM of glucose. Prostanoids release after the administration of BK, Ang-(1-7) and Ang-(1-7)+BK was also studied. In control and diabetic preparations perfused with Krebs Henseleit solution with 5.5mM of glucose, Ang-(1-7) potentiates BK-induced vasodilation. On the other hand, the potentiating effect disappeared in control and diabetic preparations perfused with 22 mM of glucose. Prostaglandin F(2alpha) (PGF(2alpha)) release induced by BK and Ang-(1-7)+BK was increased in perfusates of diabetic preparations containing 22 mM of glucose. The release of thromboxane A(2) (TXA(2)) (measured as TXB(2)) or prostaglandin I(2) (PGI(2)) (measured as 6-keto-PGF(1alpha)) did not differ in control and diabetic preparations perfused with 5.5 and 22 mM of glucose. Our data allow us to suggest that hyperglycemia may be involved in the lack of potentiation in control and diabetic preparations; increase in PGF(2alpha) release, but not other cyclooxygenase products, may explain the absence of potentiation in diabetic preparations.
Collapse
Affiliation(s)
- Maria A Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av Lineu Prestes 1524, Cidade Universitária, São Paulo, 05508-900 CEP, Brazil
| | | | | | | | | |
Collapse
|
69
|
Clark MA, Tallant EA, Tommasi E, Bosch S, Diz DI. Angiotensin-(1-7) reduces renal angiotensin II receptors through a cyclooxygenase-dependent mechanism. J Cardiovasc Pharmacol 2003; 41:276-83. [PMID: 12548089 DOI: 10.1097/00005344-200302000-00017] [Citation(s) in RCA: 31] [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: 11/26/2022]
Abstract
In the kidney, angiotensin-(1-7) [Ang-(1-7)] exhibits diuretic and natriuretic properties associated with an increase in prostaglandin production. The prohypertensive effects of Ang II are attenuated in rats infused with Ang-(1-7), consistent with recent work showing that Ang-(1-7) downregulates AT1 receptors in Chinese hamster ovary-AT1A or vascular smooth muscle cells. To determine whether exposure to Ang-(1-7) reduces AT1 receptors in the kidney through an increase in prostaglandin production, kidney slices from Sprague-Dawley rats were incubated with 10 n -1 microM Ang-(1-7) in the presence or absence of 5 microM meclofenamate, a cyclooxygenase inhibitor. Following these treatments, the kidney slices were retrieved, frozen, and sectioned for determination of [125I]-Ang II binding using in vitro receptor autoradiography. Greater than 90% of the specific binding was competed for by losartan, indicating that the majority of binding was to the AT1 receptor. Incubation of kidney slices with 1 microM Ang-(1-7) caused a 20% reduction in [125I]-Ang II binding (n = 8) in the cortical tubulointerstitium, which was prevented when Ang-(1-7)-treated slices were incubated in the presence of 5 microM meclofenamate (1 +/- 2% increase, n = 8; p < 0.05). Incubation with 5 microM meclofenamate alone had no effect on [125I]-Ang II binding (-3 +/- 3%). The decrease in [125I]-Ang II binding with Ang-(1-7) was also blocked by the Ang-(1-7) antagonist [d-Ala7]-Ang-(1-7). Treatment with 1 microM [d-Ala7]-Ang-(1-7) alone had no effect on [125I]-Ang II binding (-3 +/- 6% of control). Pretreatment with 1 microM Ang II caused a similar reduction in [125I]-Ang II binding in the cortical tubulointerstitium. Neither Ang-(1-7) nor Ang II had any effect on [125I]-Ang II binding in the glomeruli and the area of the vasa recta of the kidney. These original findings suggest that prior exposure to Ang-(1-7) or Ang II causes a modest decrease in the number of AT1 receptors in the cortical tubulointerstitial area of the kidney. The reduction in Ang II binding by Ang-(1-7) was blocked by meclofenamate and [d-Ala7]-Ang-(1-7), suggesting that cyclooxygenase products released through activation of a novel receptor participate in this effect.
Collapse
Affiliation(s)
- Michelle A Clark
- The Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, U.S.A
| | | | | | | | | |
Collapse
|
70
|
Zhu Z, Zhong J, Zhu S, Liu D, Van Der Giet M, Tepel M. Angiotensin-(1-7) inhibits angiotensin II-induced signal transduction. J Cardiovasc Pharmacol 2002; 40:693-700. [PMID: 12409978 DOI: 10.1097/00005344-200211000-00007] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The inhibitory effects of angiotensin-(1-7) on angiotensin II-induced vasoconstriction, growth of vascular smooth muscle cells, stimulation of protein kinase C, extracellular signal-regulated kinases (ERK), and angiotensin subtype 1 receptor (AT1) and subtype 2 receptor (AT2) mRNA expression were investigated. The hemodynamic effects of angiotensin-(1-7) were measured in Wistar rats. Vasoconstriction was measured using aortic rings. DNA synthesis or protein synthesis was measured in cultured vascular smooth muscle cells using [3H] thymidine or [3H] leucine incorporation, respectively. Angiotensin II stimulated protein kinase C and ERK1/2 were measured by Western blot analysis using phosphospecific protein kinase C and ERK1/2 antibodies. AT1 and AT2 receptor mRNA expression was measured using reverse-transcription polymerase chain reaction. Infusion of angiotensin II significantly increased whereas infusion of angiotensin-(1-7) had no effects on mean arterial blood pressure in Wistar rats. Angiotensin-(1-7) dose-dependently showed partial antagonism on angiotensin II-induced contraction of aortic rings. Angiotensin-(1-7) showed partial antagonism on angiotensin II-induced DNA synthesis and protein synthesis. Angiotensin-(1-7) showed partial antagonism on angiotensin II-induced activation of protein kinase C and ERK1/2. The administration of angiotensin-(1-7) showed partial antagonism on angiotensin II-induced downregulation of AT1 receptor mRNA expression, whereas AT2 receptor mRNA expression was unchanged. Angiotensin-(1-7) showed partial antagonism on angiotensin II-induced intracellular signal transduction and may play a crucial role in the adaptation process of AT1 receptors to sustained stimulation of angiotensin II.
Collapse
Affiliation(s)
- Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing, Peoples Republic of China.
| | | | | | | | | | | |
Collapse
|
71
|
Oliveira MA, Carvalho MHC, Nigro D, Passaglia RDCAT, Fortes ZB. Angiotensin-(1-7) and bradykinin interaction in diabetes mellitus: in vivo study. Peptides 2002; 23:1449-55. [PMID: 12182946 DOI: 10.1016/s0196-9781(02)00080-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interaction between angiotensin-(1-7) (Ang-(1-7)) and bradykinin (BK) was determined in the mesentery of anesthetized Wistar alloxan-diabetic and non-diabetic rats using intravital microscopy. Impaired BK vasodilation observed in arterioles of diabetic rats was restored by acute and chronic insulin treatment as well as by enalapril. Though capable of potentiating BK in non-diabetic rats, Ang-(1-7) did not potentiate BK in diabetic rats. Chronic but not acute insulin treatment restored the potentiation, whereas enalapril did not. Potassium channel blockade (by tetraethylammonium (TEA)) but not nitric oxide (NO) synthase inhibition (by N-omega-nitro-L-arginine-methyl-esther (L-NAME)) abolished the restorative effect of insulin. Our data allow us to suggest that the alteration observed is restored by insulin by a mechanism involving membrane hyperpolarization but not NO release. The beneficial effect of enalapril in diabetes might not involve the potentiation of BK by Ang-(1-7).
Collapse
Affiliation(s)
- Maria A Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 1524, Cidade Universitária, 05508-900 CEP, São Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
72
|
Mahmood A, Jackman HL, Teplitz L, Igić R. Metabolism of angiotensin I in the coronary circulation of normal and diabetic rats. Peptides 2002; 23:1171-5. [PMID: 12126747 DOI: 10.1016/s0196-9781(02)00051-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Formation of metabolites from angiotensin I that passed the coronary vessels in the isolated working rat hearts of normal and streptozotocin-induced diabetes was evaluated. HPLC analysis showed that the levels of angiotensin II and angiotensin 1-7 were unaltered in the diabetic hearts, but the perfusates of the diabetic hearts contained smaller amounts of angiotensin 1-9. It is not clear why the perfusates of diabetic hearts contain less amount of angiotensin 1-9. It is possible that the peptide is metabolized faster or greater internalization takes place in the diabetic heart. The amount of angiotensin II in the perfusates of normal hearts was 5.8 times greater at the perfusion rate of 2 than at 10 ml/min/g wet heart weight. At such conditions, the amount of angiotensin 1-9 and angiotensin 1-7 in the perfusates were increased 2.4 and 1.5 times, respectively. A higher amount of angiotensin II during myocardial hypoperfusion may lead to constriction of the coronary vessels. As a result, myocardial damage may be facilitated.
Collapse
Affiliation(s)
- Asim Mahmood
- Department of Anesthesiology and Pain Management, Cook County Hospital, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
73
|
Tschöpe C, Schultheiss HP, Walther T. Multiple interactions between the renin-angiotensin and the kallikrein-kinin systems: role of ACE inhibition and AT1 receptor blockade. J Cardiovasc Pharmacol 2002; 39:478-87. [PMID: 11904521 DOI: 10.1097/00005344-200204000-00003] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The investigation of therapeutic actions of angiotensin type 1 (AT1) receptor antagonists and ACE inhibitors (ACEI) demonstrated complex interactions between the renin-angiotensin system (RAS) and the kallikrein-kinin system (KKS) in several experimental and clinical studies. They are evidenced by the fact that (1) ACE efficiently catabolizes kinins; (2) angiotensin-derivatives such as ANG-(1-7) exert kininlike effects; and (3) kallikrein probably serves as a prorenin-activating enzyme. (4) Several authors have demonstrated experimentally that the protective effects of ACEI are at least partly mediated by a direct potentiation of kinin receptor response on BK stimulation. (5) Furthermore, studies on AT1 antagonists, which do not directly influence kinin degradation, and studies on angiotensin-receptor transgenic mice have revealed additional interactions between the RAS and the KKS. There is mounting evidence that an autocrine cascade including kinins, nitric oxide, prostaglandins, and cyclic GMP is involved in at least some of the angiotensin type 2 receptor effects. This review discusses multiple possibilities of cross-talks between the RAS and KKS in vascular and cardiac physiology and pathology after ACE inhibition and AT1 receptor blockade.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Department of Cardiology and Pneomology, University Hospital Benjamin Franklin, Free University of Berlin, Hindenburgdamm 30, D-12220 Berlin, Germany.
| | | | | |
Collapse
|
74
|
Bayorh MA, Eatman D, Walton M, Socci RR, Thierry-Palmer M, Emmett N. 1A-779 attenuates angiotensin-(1-7) depressor response in salt-induced hypertensive rats. Peptides 2002; 23:57-64. [PMID: 11814618 DOI: 10.1016/s0196-9781(01)00579-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Chronic infusion of angiotensin-(1-7) [Ang-(1-7)] lowers blood pressure in salt-induced and spontaneously hypertensive (SHR) rats. In the present study, we have examined the acute effect of Ang-(1-7) in salt-induced hypertension using Dahl salt-sensitive rats placed on low (0.3%) or high (8.0% NaCl) salt diets for 2 weeks. Rats fed a high salt diet showed a greater rise in BP than those fed a low salt diet. Ang-(1-7) (24 microg/kg) reduced mean arterial pressure (MAP), enhanced the release of prostacyclin and nitric oxide, and suppressed thromboxane A(2) levels. A-779 (48 microg/kg, i.v), a selective Ang-(1-7) antagonist, partially blocked these effects of Ang-(1-7). The Ang-(1-7)-induced depressor response observed in these animals was related to an increase in vasodilatory prostanoids, a decrease in the constrictor prostanoid thromboxane A(2), and an increase in nitric oxide levels in both plasma and isolated aortic rings.
Collapse
Affiliation(s)
- Mohamed A Bayorh
- Department of Pharmacology & Toxicology, Morehouse School of Medicine, 720 Westview Drive, S.W., Atlanta, GA 30310-1495, USA.
| | | | | | | | | | | |
Collapse
|
75
|
Machado RD, Santos RA, Andrade SP. Mechanisms of angiotensin-(1-7)-induced inhibition of angiogenesis. Am J Physiol Regul Integr Comp Physiol 2001; 280:R994-R1000. [PMID: 11247819 DOI: 10.1152/ajpregu.2001.280.4.r994] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiotensin-(1-7) [ANG-(1-7)], an endogenous bioactive peptide constituent of the renin-angiotensin system, acts as an inhibitory growth factor in vitro and in vivo. In this study, we evaluated whether the antiangiogenic effect of ANG-(1-7) in the mouse sponge model of angiogenesis might be receptor mediated and involved in the release of nitric oxide (NO). The hemoglobin content (microg/mg wet tissue) of 7-day-old sponge implants was used as an index of the vascularization and showed that daily injections of ANG-(1-7) (20 ng) inhibited significantly the angiogenesis in the implants relative to the saline-treated group. The specific receptor antagonist D-Ala(7)-ANG-(1-7); A-779 prevented ANG-(1-7)-induced inhibition of angiogenesis. The antiangiogenic effect was also abolished by pretreatment with NO synthase inhibitors aminoguanidine (1 mg/ml) or N(G)-nitro-L-arginine methyl ester (0.3 mg/ml). Selective AT1 and AT2 angiotensin-receptor antagonists and an angiotensin-converting enzyme inhibitor, in combination with ANG-(1-7) or alone, did not alter angiogenesis in the implants. These results establish that the regulation of the vascular tissue growth by ANG-(1-7) is associated with NO release by activation of an angiotensin receptor distinct from AT1 and AT2.
Collapse
Affiliation(s)
- R D Machado
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627-Campus Pampulha, Belo Horizonte, Brazil 31270-901
| | | | | |
Collapse
|
76
|
Clark MA, Tallant EA, Diz DI. Downregulation of the AT1A receptor by pharmacologic concentrations of Angiotensin-(1-7). J Cardiovasc Pharmacol 2001; 37:437-48. [PMID: 11300657 DOI: 10.1097/00005344-200104000-00011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Angiotensin (Ang)-(1-7), the amino terminal heptapeptide fragment of Ang II, is an endogenous Ang peptide with vasodilatory and antiproliferative actions. Because Ang II causes vasoconstriction and promotes growth through activation of Ang type 1 (AT1) receptors, we investigated whether the actions of Ang-(1-7) are due to its regulation of these receptors. Studies were performed in CHO cells stably transfected with the AT1A receptor. Ang-(1-7) competed poorly with [125I]-Ang II for the AT1A binding site and was ineffective at shifting the IC50 for Ang II competition with [125I]-Ang II for binding to the AT1A receptor. However, if CHO-AT1A cells were pretreated with Ang-(1-7) and then treated with acidic glycine to remove surface-bound ligand, the heptapeptide caused a concentration-dependent reduction in Ang II binding, with a maximal inhibition to 67.8 +/- 4.6% of total (p < 0.05) at 1 microM Ang-(1-7) compared with a reduction to 24% of total by 10 nM Ang II. Ang-(1-7) pretreatment caused a small but significant decrease in the affinity of [125I]-Ang II for the AT1A receptor and a significant reduction in the total number of binding sites. The Ang-(1-7)-induced reduction in binding was rapid (occurring as early as 5 min after exposure to the peptide), was maintained for 30 min during continued exposure of the cells to Ang-(1-7), and rapidly recovered after removal of the heptapeptide. The AT1 receptor antagonist L-158,809 reduced the Ang-(1-7)-induced downregulation of the AT1A receptor, suggesting that interactions with AT1A receptors mediate the regulatory events. Pretreatment with 1 microM or 10 microM Ang-(1-7) significantly reduced inositol phosphate production in response to 10 nM Ang II. The decrease in binding and responsiveness of the AT1A receptor after exposure to micromolar concentrations of Ang-(1-7) suggests that the heptapeptide downregulates the AT1A receptor to reduce responses to Ang II. Because downregulation of the receptor only occurred at micromolar concentrations of the heptapeptide, our findings suggest that Ang-(1-7) is not a potent antagonist at the AT1A receptor. However, when the balance between Ang II and Ang-(1-7) is shifted in favor of Ang-(1-7), such as during inhibition of Ang-converting enzyme, some contribution of this mechanism may come into play.
Collapse
Affiliation(s)
- M A Clark
- Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1032, USA.
| | | | | |
Collapse
|
77
|
Komers R, Komersova K. Therapeutic potential of ACE inhibitors for the treatment of hypertension in Type 2 diabetes. Expert Opin Investig Drugs 2000; 9:2601-17. [PMID: 11060823 DOI: 10.1517/13543784.9.11.2601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Type 2 diabetes mellitus is associated with hypertension. If untreated, hypertension has a major impact on the clinical course of Type 2 diabetes and its vascular complications. In this review, we discuss rationale for the use of ACE inhibitors (ACEI) in hypertensive Type 2 diabetic patients and compare those theoretical assumptions with results of recent major clinical trials. Furthermore, possible directions for future clinical and experimental research are outlined. The RAS and its effector angiotensin II are important players in a number of cardiovascular and renal disorders. Recent evidence suggests that RAS and factors functionally linked to RAS are activated in Type 2 diabetes. Therefore, there is a theoretical basis for the use of ACEI in the treatment of hypertension in diabetic patients. Some recent studies reported superior outcome in patients treated with ACEI-based antihypertensive regimens compared with non-ACEI based treatments in reducing the risk of macrovascular disease (CAPPP, FACET, ABCD) or both micro- and macrovascular complications in Type 2 diabetes (HOPE). However, at least two of the large prospective studies discussed in this review (UKPDS 38, HOT), supported by results from previously published SHEP study, have recently suggested that the degree of reduction of blood pressure, rather than the choice of a particular class of antihypertensive agent, is associated with decreased risk of cardiovascular events. Studies focusing on renal end-points suggest that ACEI have a superior antiproteinuric effect than the other agents. However, whether ACEI are more nephroprotective, as assessed by the rate of decline in renal function, still remains to be elucidated. Despite promising results from recent trials, large numbers of patients progress despite ACEI treatment. Incomplete inhibition of the RAS may underlie this phenomenon. Treatment strategies that could enhance the degree of RAS inhibition represent one possible direction for clinical research in the near future. However, it is unlikely that the course of such a complex syndrome as Type 2 diabetes could be dramatically changed by just one class of antihypertensive agents. This goal is more likely to be achieved by multifactorial intervention, reflecting the complexity of metabolic syndrome. ACEI should be viewed as an important, but not the only, part of this complex approach.
Collapse
Affiliation(s)
- R Komers
- Division of Nephrology and Hypertension, Oregon Health Sciences University, PP262, 3314 SW US Veterans Hospital Road, Portland, Oregon, 97201-2940, USA.
| | | |
Collapse
|
78
|
Komers R, Anderson S. Optimal strategies for preventing progression of renal disease: should angiotensin converting enzyme inhibitors and angiotensin receptor blockers be used together? Curr Hypertens Rep 2000; 2:465-72. [PMID: 10995522 DOI: 10.1007/s11906-000-0029-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Interruption of the renin-angiotensin system (RAS) with angiotensin converting enzyme (ACE) inhibitors or angiotensin AT(1) receptor blockers has been shown to delay progression in a variety of renal diseases, suggesting that the RAS, and its major effector molecule, angiotensin II, are important players in renal pathophysiology. Both antagonists combine inhibition of deleterious effects of angiotensin II with activation of potentially beneficial pathways mediated by nitric oxide and prostaglandins. Some concerns have been raised about the completeness of the RAS blockade achieved by these agents. ACE-independent pathways can generate angiotensin II, whereas increases in angiotensin II levels may compete with the AT(1) receptor blocker at the receptor site. It has been suggested that an ACE inhibitor/AT(1) receptor blocker combination offers a better therapeutic effect than treatment with either agent alone. In this review, we focus on mechanisms of actions of ACE inhibitors and AT(1) receptor blockers, implicate them in the rationale for the use of an ACE inhibitor/AT(1) receptor blocker combination, and discuss evidence evaluating the renal effects of the combination as compared to the effects of a single agent. There is a surprising lack of information about the nephroprotective potential of the combination, allowing no consistent conclusions about the superiority of the combination over the single agent. Several experimental and clinical reports suggest that in some conditions, the combination may be beneficial. Rather than providing unequivocal evidence for the use of combination treatment in the renal disease, these studies should be considered as stimuli for more detailed exploration of this issue.
Collapse
Affiliation(s)
- R Komers
- Division of Nephrology and Hypertension, Oregon Health Sciences University, PP262, 3314 SW US Veterans Hospital Road, Portland, OR 97201-2940, USA
| | | |
Collapse
|
79
|
Feterik K, Smith L, Katusic ZS. Angiotensin-(1-7) causes endothelium-dependent relaxation in canine middle cerebral artery. Brain Res 2000; 873:75-82. [PMID: 10915812 DOI: 10.1016/s0006-8993(00)02482-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The heptapeptide, angiotensin-(1-7), is an active member of the renin-angiotensin system. The present study was designed to characterize the role of endothelium in relaxations of large cerebral arteries to angiotensin-(1-7). Rings of canine middle cerebral arteries were suspended in organ chambers for isometric force recording. The levels of cyclic guanosine 3',5'-monophosphate (cGMP) were assessed by radioimmunoassay. During contraction to uridine 5'-triphosphate (UTP, 3x10(-6) to 10(-5) mol/l), angiotensin-(1-7) (10(-9) to 3x10(-5) mol/l) caused concentration-dependent relaxations in arteries with endothelium, but not in endothelium-denuded vessels. Angiotensin-(1-7) significantly increased formation of cGMP. Nitric oxide synthase inhibitor, N-omega-nitro-L-arginine methyl ester (L-NAME, 3x10(-4) mol/l), and selective soluble guanylate cyclase inhibitor, 1 H-[1,2, 4]oxadiazolo[4,3-a]quinozalin-1-one (ODQ, 3x10(-6) mol/l), abolished angiotensin-(1-7)-induced relaxations. Angiotensin receptor antagonists, losartan (10(-5) mol/l), PD 123319 (10(-5) mol/l), [Sar(1),Thr(8)]-angiotensin II (10(-5) mol/l) [Sar(1),Val(5), Ala(8)]-angiotensin II (10(-5) mol/l) or [7-D-Ala]-angiotensin 1-7 (10(-6) mol/l) did not affect these relaxations. However, angiotensin-converting enzyme inhibitor, captopril (10(-5) mol/l) augmented relaxations to angiotensin-(1-7). Finally, bradykinin B(2) receptor antagonist, [D-Arg(0),Hyp(3),Thi(5),D-Tic(7), Oic(8)]-bradykinin (HOE 140, 5x10(-8) mol/l) significantly reduced the effect of angiotensin-(1-7), while bradykinin B(1) receptor antagonist, des-Arg(9), [Leu(8)]-bradykinin (6x10(-9) mol/l) did not influence the vascular response to the heptapeptide. These findings indicate that (1) angiotensin-(1-7) produces relaxation of canine middle cerebral arteries by the release of nitric oxide from endothelial cells, (2) angiotensin receptors do not mediate endothelium-dependent relaxations to the heptapeptide, and (3) this effect appears to be dependent on activation of local production of kinins. Our studies support the concept that angiotensin-(1-7), as a natural vasodilator hormone, may counterbalance the hemodynamic actions of angiotensin II.
Collapse
Affiliation(s)
- K Feterik
- Departments of Anesthesiology, Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
80
|
Abstract
The renin-angiotensin system is a major physiological regulator of arterial pressure and hydro-electrolyte balance. Evidence has now been accumulated that in addition to angiotensin (Ang) II other Ang peptides [Ang III, Ang IV and Ang-(1-7)], formed in the limited proteolysis processing of angiotensinogen, are importantly involved in mediating several actions of the RAS. In this article we will review our knowledge of the biological actions of Ang-(1-7) with focus on the puzzling aspects of the mediation of its effects and the interaction Ang-(1-7)-kinins. In addition, we will attempt to summarize the evidence that Ang-(1-7) takes an important part of the mechanisms aimed to counteract the vasoconstrictor and proliferative effects of Ang II.
Collapse
Affiliation(s)
- R A Santos
- Departamento de Fisiologia e Biofísica, Av. Antonio Carlos, 6627 - Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG Brazil.
| | | | | |
Collapse
|
81
|
Almeida AP, Frábregas BC, Madureira MM, Santos RJ, Campagnole-Santos MJ, Santos RA. Angiotensin-(1-7) potentiates the coronary vasodilatatory effect of bradykinin in the isolated rat heart. Braz J Med Biol Res 2000; 33:709-13. [PMID: 10829099 DOI: 10.1590/s0100-879x2000000600012] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It has been shown that angiotensin-(1-7) (Ang-(1-7)) infusion potentiates the bradykinin (BK)-induced hypotensive response in conscious rats. The present study was conducted to identify Ang-(1-7)-BK interactions in the isolated rat heart perfused according to the Langendorff technique. Hearts were excised and perfused through the aortic stump under a constant flow with Krebs-Ringer solution and the changes in perfusion pressure and heart contractile force were recorded. Bolus injections of BK (2.5, 5, 10 and 20 ng) produced a dose-dependent hypotensive effect. Ang-(1-7) added to the perfusion solution (2 ng/ml) did not change the perfusion pressure or the contractile force but doubled the hypotensive effect of the lower doses of BK. The BK-potentiating Ang-(1-7) activity was blocked by pretreatment with indomethacin (5 mg/kg, ip) or L-NAME (30 mg/kg, ip). The Ang-(1-7) antagonist A-779 (50 ng/ml in Krebs-Ringer) completely blocked the effect of Ang-(1-7) on BK-induced vasodilation. These data suggest that the potentiation of the BK-induced vasodilation by Ang-(1-7) can be attributed to the release of nitric oxide and vasodilator prostaglandins through an Ang-(1-7) receptor-mediated mechanism.
Collapse
Affiliation(s)
- A P Almeida
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | | | | | | | | | | |
Collapse
|
82
|
Israel A, Cierco M, Sosa B. Angiotensin AT(2) receptors mediate vasodepressor response to footshock in rats. Role of kinins, nitric oxide and prostaglandins. Eur J Pharmacol 2000; 394:103-8. [PMID: 10771041 DOI: 10.1016/s0014-2999(00)00133-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Footshocks increased mean arterial pressure and heart rate. Systemic administration of losartan, a specific angiotensin AT(1) receptor antagonist, not only inhibited the pressor response to footshocks, but also resulted in vasodepression. Administration of 1-[[4-(dimethylamino)3-methylphenyl]methyl]-5 (diphenylacetyl)-4,5,6, 7-tetrahydro-1H imidazol (4,5-c] pyridine-6-carboxilic acid, ditrifluoro acetatemonohydrate (PD 123319), a specific angiotensin AT(2) receptor antagonist, did not alter the hemodynamic response to footshocks. Simultaneous blockade of angiotensin AT(1) and AT(2) receptors by combined administration of losartan and PD 123319, eliminated the vasodepressor response to footshocks unmasked in losartan-pretreated rats. Saralasin, a non-specific angiotensin receptor antagonist, abolished the cardiovascular response to footshocks similarly like the losartan+PD 123319 treatment. Our data suggest that the vasodepressor response to footshocks in the presence of an angiotensin AT(1) receptor antagonist is triggered by activation of angiotensin AT(2) receptors. We studied the role of kinins, nitric oxide and prostaglandins in the vasodepressor response observed after footshocks. The decrease in mean arterial pressure observed after footshocks in losartan-treated rats was blunted by icatibant (HOE 140), N(G)-nitro-L-arginine-methyl ester (L-NAME) or indomethacin, indicating that kinins, nitric oxide and prostaglandins appear to be involved in the pressure response to footshocks during angiotensin AT(1) receptor blockade.
Collapse
Affiliation(s)
- A Israel
- School of Pharmacy, Universidad Central de Venezuela and Schools of Medicine, Apartado Postal 50176, Sabana Grande 1050, Caracas, Venezuela.
| | | | | |
Collapse
|
83
|
Abstract
Using the murine sponge model of angiogenesis, associated to functional and morphological parameters we have demonstrated opposing actions of angiotensin II (Ang II) and angiotensin-(1-7;Ang-1-7) in modulating fibrovascular tissue growth. Angiogenesis in the implants was assessed at day 7 postimplantation by extracting the hemoglobin content, by determining the outflow rate of sodium fluorescein applied intraimplant and by histological analysis. Furthermore, the proliferative activity of control and angiotensin-treated implants was established using the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2(4 -sulfonyl)2H-tetrazolium)assay. The hemoglobin content in the control implants was 2.4 +/- 0.14 (microg/mg wet weight) versus 3.6 +/- 0.27(Ang II;100 ng) and 0.86 +/- 0.07 Ang-(1-7); 20 ng. Blood flow in the implants as determined by t1/2 values (time taken for the fluorescence to reach 50% of the peak in the systemic circulation) showed that Ang II stimulated angiogenesis, whereas Ang-(1-7) inhibited it. The proliferative activity of the sponge-induced fibrovascular tissue was enhanced by Ang II and diminished by Ang-(1-7). These results show the pro-versus anti-angiogenic effects of these angiotensin molecules, providing evidence for their opposing effects on vascular tissue growth and wound healing in vivo.
Collapse
Affiliation(s)
- R D Machado
- Department of Physiology and Biophysics Institute of Biological Sciences-Federal University of Minas Gerais, Belo Horizonte/MG-Brazil
| | | | | |
Collapse
|
84
|
Dendorfer A, Wolfrum S, Dominiak P. Pharmacology and cardiovascular implications of the kinin-kallikrein system. JAPANESE JOURNAL OF PHARMACOLOGY 1999; 79:403-26. [PMID: 10361880 DOI: 10.1254/jjp.79.403] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Kinins are peptide hormones that can exert a significant influence on the regulation of blood pressure and vascular tone due to their vasodilatatory, natriuretic and growth modulating activity. Their cardiovascular involvement in physiological and pathophysiological situations has been studied intensively since inhibitors for angiotensin I-converting enzyme and selective receptor antagonists have become available for pharmacologically potentiating or inhibiting kinin-mediated reactions. Molecular biological analysis and the establishment of genetically modified animal models have also allowed newer information to be acquired on this subject. In this review, the components and cardiovascularly relevant mechanisms of the kinin-kallikrein system shall be described. Organ-specific effects concerning the kidneys, the vascular system, the heart and nervous tissue shall also be illustrated. On this issue, the physiological functions and pathophysiological implications of the kinin-kallikrein system should be clearly distinguished from the many, mostly endothelium-mediated protective effects which occur during ACE inhibition due to the potentiation of kinin effects. Finally, a view shall also be cast upon newly discovered targets of action, which could be exploited for therapeutically altering the kinin-kallikrein system.
Collapse
Affiliation(s)
- A Dendorfer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical University Lübeck, Germany
| | | | | |
Collapse
|
85
|
Oliveira MA, Fortes ZB, Santos RA, Kosla MC, De Carvalho MH. Synergistic effect of angiotensin-(1-7) on bradykinin arteriolar dilation in vivo. Peptides 1999; 20:1195-201. [PMID: 10573291 DOI: 10.1016/s0196-9781(99)00123-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interaction between angiotensin [Ang-(1-7)] and bradykinin (BK) was determined in the mesentery of anesthetized Wistar rats using intravital microscopy. Topical application of BK and Ang-(1-7) induced vasodilation that was abolished by the BK B2 receptor antagonist HOE-140 and the Ang-(1-7) antagonist A-779, respectively. BK (1 pmol)-induced vasodilation, but not SNP and ACh responses, was potentiated by Ang-(1-7) 10 pmol and 100 pmols. The effect of 100 pmol of Ang-(1-7) on BK-induced vasodilation was abolished by A-779, indomethacin, and L-nitroarginine methyl esther, whereas losartan was without effect. Enalaprilat treatment enhanced the BK- and Ang-(1-7)-induced vasodilation and the potentiating effect of Ang-(1-7) on BK vasodilation. The potentiation of BK-induced vasodilation by Ang-(1-7) is a receptor-mediated phenomenon dependent on cyclooxygenase-related products and NO release.
Collapse
Affiliation(s)
- M A Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | | | | | | |
Collapse
|
86
|
Paula RD, Lima CV, Britto RR, Campagnole-Santos MJ, Khosla MC, Santos RA. Potentiation of the hypotensive effect of bradykinin by angiotensin-(1-7)-related peptides. Peptides 1999; 20:493-500. [PMID: 10458520 DOI: 10.1016/s0196-9781(99)00031-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this study, we evaluated the bradykinin potentiating activity and ACE inhibitory activity of several Ang-(1-7)-related peptides: Ang-(2-7), Ang-(3-7), Ang-(4-7), Ang-(1-6), Ang-(1-5) and the selective antagonist of Ang-(1-7): D-[Ala7]Ang-(1-7) (A-779). In vivo experiments were performed in freely moving Wistar rats. ACE activity was evaluated by a fluorometric assay in rat plasma using Hip-His-Leu as a substrate. Intravenous injections of Ang-(1-7) (2.2 nmol) transformed the effect of a single dose of bradykinin (1 nmol) into the effect produced by a double dose. A similar bradykinin potentiating activity was demonstrated for Ang-(2-7) and Ang-(3-7). On the other hand, Ang-(1-5), Ang-(1-6), Ang-(4-7) and A-779 did not change the hypotensive effect of bradykinin in doses ranging from 8 up to 25 nmols. The hypotensive effect of bradykinin was increased by intravenous infusion (0.3 ng/min) of Ang-(1-7) > Ang-(2-7) > Ang-(3-7). Conversely, Ang-(1-5), Ang-(1-6), Ang-(4-7) or A-779 did not change the hypotensive effect of bradykinin. ACE inhibition with Ang-(1-7) related peptides occurred in the order: Ang-(2-7) > or = Ang-(3-7) > Ang-(1-7) [>>] Ang-(1-5) > Ang-(4-7) > or = Ang-(1-6) > or = A-779. A-779 in concentrations up to 10(-5) M did not change the ACE inhibitory activity of Ang-(1-7). These results suggest that Ang-(1-7), Ang-(2-7) and Ang-(3-7) can modulate bradykinin actions in vivo. More important, our data pointed out that alternative mechanisms besides interaction with ACE are required to explain the bradykinin potentiating activity of Ang-(1-7).
Collapse
Affiliation(s)
- R D Paula
- Departamento de Fisiologia e Biofísica Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | |
Collapse
|
87
|
Bomtempo CA, Santos GF, Santos RA, Campagnole-Santos MJ. Interaction of bradykinin and angiotensin-(1-7) in the central modulation of the baroreflex control of the heart rate. J Hypertens 1998; 16:1797-804. [PMID: 9869014 DOI: 10.1097/00004872-199816120-00013] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Previous studies have shown that angiotensin-(1-7) potentiates the vascular actions of bradykinin. In the present study, we evaluated the interaction of bradykinin and angiotensin-(1-7) in the central modulation of baroreflex control of the heart rate. MATERIALS AND METHODS Blood pressure and reflex bradycardia, elicited by intravenous injection of phenylephrine, were evaluated in conscious male Wistar rats before and at the end of 1 h of an intracerebroventricular infusion of angiotensin-(1-7) at 0.5 or 1.0 microg/h combined with bradykinin at 2.5 microg/h; or angiotensin-(1-7) at 2.0 microg/h combined with bradykinin at 4.0 microg/h; or angiotensin-(1-7) alone at 2.0 or 4.0 microg/h; or bradykinin alone at 4.0 or 8.0 microg/h; or saline at 8 microl/h. In addition, baroreflex bradycardia was evaluated before and at the end of 1 and 2 h of intracerebroventricular infusion of angiotensin-(1-7) at 4 microg/h for 2 h; or saline at 8 microl/h in the first hour followed by HOE 140 at 90 ng/h in the second hour; or angiotensin-(1-7) at 4 microg/h in the first hour followed by angiotensin-(1-7) at 4 microg combined with HOE 140 at 90 ng/h in the second hour; or HOE 140 at 90 ng/h in the first hour followed by HOE 140 at 90th ng/h combined with angiotensin-(1-7) at 4 microg/h in the second hour; or saline at 8 microl/h for 2 h. RESULTS The intracerebroventricular infusion of angiotensin-(1-7) or bradykinin alone required a dose of 4.0 and 8.0 microg/h, respectively, to facilitate baroreflex control of the heart. However, a simultaneous infusion of these peptides at subeffective rates was able to produce a significant increase in baroreflex sensitivity. In addition, the facilitation of the baroreflex control of the heart rate induced by angiotensin-(1-7) at 4.0 microg/h was inhibited by HOE 140. CONCLUSIONS These results suggest that centrally, bradykinin and angiotensin-(1-7) can interact in order to modulate baroreflex control of the heart rate. In addition, our data indicate that the central modulatory effect of angiotensin-(1-7) on the baroreflex is mediated, at least in part, by the release of kinins.
Collapse
Affiliation(s)
- C A Bomtempo
- Departamento de Fisiologia e Biofisica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | | | | | | |
Collapse
|
88
|
Ferrario CM, Iyer SN. Angiotensin-(1-7): a bioactive fragment of the renin-angiotensin system. REGULATORY PEPTIDES 1998; 78:13-8. [PMID: 9879742 DOI: 10.1016/s0167-0115(98)00134-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Accumulating evidence suggests that angiotensin-(1-7) [Ang-(1-7)] is an important component of the renin-angiotensin system. As the most pleiotropic metabolite of angiotensin I (Ang I) it manifest actions which are most often the opposite of those described for angiotensin II (Ang II). Ang-(1-7) is produced from Ang I bypassing the prerequisite formation of Ang II. The generation of Ang-(1-7) is under the control of at least three enzymes, which include neprilysin, thimet oligopeptidase, and prolyl oligopeptidase depending on the tissue compartment. Both neprilysin and thimet oligopeptidase are also involved in the metabolism of bradykinin and the atrial natriuretic peptide. Moreover, recent studies suggest that in addition to Ang I and bradykinin, Ang-(1-7) is an endogenous substrate for angiotensin converting enzyme. This suggests that there is a complex relationship between the enzymatic pathways forming angiotensin II and other various vasodepressor peptides from either the renin-angiotensin system or other peptide systems. The antihypertensive actions of angiotensin-(1-7) are mediated by an angiotensin receptor that is distinct from the pharmacologically characterized AT1 or AT2 receptor subtypes. Ang-(1-7) mediates it antihypertensive effects by stimulating synthesis and release of vasodilator prostaglandins, and nitric oxide and potentiating the hypotensive effects of bradykinin.
Collapse
Affiliation(s)
- C M Ferrario
- The Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1032, USA.
| | | |
Collapse
|
89
|
Abstract
Accumulating evidence suggests that angiotensin-(1-7)(Ang-(1-7)) is an important component of the renin-angiotensin system and that the actions of the peptide may either contribute to or oppose those of Ang II. Ang-(1-7) can be converted directly from Ang I bypassing prerequisite formation of Ang II. Formation of Ang-(1-7) is under the control of at least three endopeptidases depending on the tissue compartment and include neprilysin, thimet oligopeptidase and prolyl oligopeptidase. Both neprilysin and thimet oligopeptidase are also involved in the metabolism of bradykinin and the atrial natriuretic peptide. Moreover, recent studies suggest that in addition to Ang I and bradykinin, Ang-(1-7) is an endogenous substrate for angiotensin converting enzyme. These enzymatic pathways may contribute to a complex relationship between the hypertensive actions of Ang II and various vasodepressor peptides from either the renin-angiotensin system or other peptide systems. Ang-(1-7) is devoid of the vasoconstrictor, central pressor, or thirst-stimulating actions associated with Ang II. In fact, new findings reveal depressor, vasodilator, and antihypertensive actions that may be more apparent in hypertensive animals or humans. Thus, Ang-(1-7) may oppose the actions of Ang II directly or as a result of increasing prostaglandins or nitric oxide. In this review, we examine the mechanisms by which Ang-(1-7) may contribute to cardiovascular regulation.
Collapse
Affiliation(s)
- M C Chappell
- Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1032, USA.
| | | | | | | |
Collapse
|
90
|
Gillies LK, Werstiuk ES, Lee RM. Cross-over study comparing effects of treatment with an angiotensin converting enzyme inhibitor and an angiotensin II type 1 receptor antagonist on cardiovascular changes in hypertension. J Hypertens 1998; 16:477-86. [PMID: 9797193 DOI: 10.1097/00004872-199816040-00009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To compare the blood-pressure-lowering effects of an angiotensin converting enzyme inhibitor, perindopril, with those of an angiotensin II type 1 receptor antagonist, L-158,809, for adult spontaneously hypertensive rats. DESIGN A cross-over design was used, to treat adult spontaneously hypertensive rats with one drug for 10 weeks, and then with the other for 5 weeks. METHODS Adult, male spontaneously hypertensive rats (aged 15 weeks) were treated daily by gavage for 10 weeks with perindopril (P group) or L-158,809 (L group), then treatment was crossed over so that rats in the P group were treated with L-158,809 (P/L group) and rats in the L group were treated with perindopril (L/P group) for 5 weeks. Blood pressure was measured weekly. Plasma angiotensin converting enzyme activity, renal angiotensin receptor density, and arterial structure and functioning were measured after the single and crossover treatment periods. RESULTS Treatment lowered the blood pressure from 206 +/- 2 mmHg in rats in the control group, to 126 +/- 2 in rats in the P group and 150 +/- 2 in rats in the L group. After the cross-over period, blood pressure decreased further from 150 +/- 2 to 129 +/- 3 mmHg in rats in the L/P group, whereas blood pressure of spontaneously hypertensive rats in the P/L group increased from 126 +/- 2 to 148 +/- 2 mmHg. Perindopril treatment almost abolished plasma angiotensin converting enzyme activity, whereas L-158,809 treatment had no effect. Renal angiotensin II receptor density was decreased versus baseline in rats in the P and L groups. The affinity of binding was decreased versus baseline in rats in the L group. A positive correlation to blood pressure was found for mesenteric artery wall thickness and wall: lumen ratio. Concentration for half-maximal effect for the response of mesenteric arteries from rats in the P group to norepinephrine was lower than that of the control group rats. Angiotensin II potentiated the norepinephrine-stimulated contraction of arteries from rats in the control and P groups, but not that of arteries from rats in the groups treated with L-158,809. CONCLUSION Perindopril was more effective than was L-158,809 at lowering the blood pressure of adult spontaneously hypertensive rats, and at altering the structure and functioning of the arteries.
Collapse
Affiliation(s)
- L K Gillies
- Department of Anaesthesia, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
91
|
Hollenberg NK, Guidi E. ACE Inhibitors and Renal Vascular Responses in the Spontaneously Hypertensive Rat. J Cardiovasc Pharmacol Ther 1998; 3:161-170. [PMID: 10684494 DOI: 10.1177/107424849800300209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Substantial evidence has accumulated for the intrarenal generation of functionally important quantities of angiotensin II (Ang II). To assess the possibility that Ang II generation occurs beyond a barrier to diffusion from the vascular compartment, six angiotensin-converting enzyme (ACE) inhibitors varying widely in their lipid solubility were employed in the spontaneously hypertensive rat (SHR) and their normotensive controls (WKY). The biological end points were renal blood flow and its response to Ang II. RESULTS: Two ACE inhibitors, ramipril and captopril, induced a larger increase in renal blood flow and enhanced the renal vascular response to Ang II substantially more than did enalapril and lisinopril. The two prodrugs, enalapril and ramipril, which are substantially more lipophilic than the respective active drugs, enalaprilat and ramiprilat, showed equivalent responses. The partial agonist saralasin virtually abolished the renal vasodilator response to ramipril. The pattern of response was similar in WKY, but the responses were substantially smaller. CONCLUSIONS: The results support the concept that a functionally important compartment for intrarenal Ang II formation exists in the healthy rat and that this process is amplified in the SHR.
Collapse
Affiliation(s)
- NK Hollenberg
- Departments of Medicine and Radiology, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
92
|
Lima CV, Paula RD, Resende FL, Khosla MC, Santos RA. Potentiation of the hypotensive effect of bradykinin by short-term infusion of angiotensin-(1-7) in normotensive and hypertensive rats. Hypertension 1997; 30:542-8. [PMID: 9322979 DOI: 10.1161/01.hyp.30.3.542] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In this study we evaluated the effect of angiotensin-(1-7) on the hypotensive action of bradykinin (BK) in normotensive rats, renal hypertensive rats (RHR), and spontaneously hypertensive rats (SHR). In addition, we evaluated the effect of angiotensin-converting enzyme (ACE) inhibition with enalaprilat treatment (10 mg/kg I.V.) on the BK-potentiating activity of Ang-(1-7). Renal hypertension was produced by aorta coarctation between the origin of renal arteries. Ang-(1-7) (0.3 pmol/min) or saline (0.9% NaCl, 5 microL/min) was infused intravenously in conscious male Wistar rats, adult SHR, or RHR. Intravenous bolus injections of BK (0.1 to 1.6 nmol in RHR and SHR; 0.625 to 5 nmol in Wistar rats) were made before and within 30 and 60 minutes of Ang-(1-7) infusion. Ang-(1-7) infusion did not change mean arterial pressure (MAP) of Wistar rats (MAP=97+/-3 mm Hg), RHR (MAP=173+/-3 mm Hg), or SHR (MAP=177+/-5 mm Hg). In Wistar rats, Ang-(1-7) increased the BK hypotensive effect by 24+/-6% within 60 minutes of infusion. No significant changes were observed at 30 minutes of infusion. In additional groups of rats, Ang-(1-7) (5 pmol/min, n=5) was infused alone or combined with its selective antagonist D-Ala7-Ang-(1-7) (A-779) (5 pmol/min, n=6). The bradykinin-potentiating activity of Ang-(1-7) was completely abolished by A-779. In SHR and RHR, Ang-(1-7) significantly increased the hypotensive effect of BK by 59+/-8% and 57+/-9.8%, respectively, within 60 minutes of infusion. No significant changes were observed with saline infusion. In Wistar rats, enalaprilat treatment increased the BK-potentiating activity of Ang-(1-7) transforming the effect of 0.3 pmol/min into that observed with a rate 16-fold higher (5 pmol/min). On the other hand, in SHR enalaprilat did not change the Ang-(1-7) effect, while it abolished the BK potentiation in RHR. Our data show that the BK-potentiating activity of Ang-(1-7) is preserved and even augmented in hypertensive rats. The finding that the BK-potentiating activity of Ang-(1-7) could be demonstrated at a very low infusion rate suggests that this angiotensin can act as an endogenous modulator of the vascular actions of kinins. ACE inhibition can influence differently the BK-potentiating activity of Ang-(1-7) in normotensive and hypertensive rats.
Collapse
Affiliation(s)
- C V Lima
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federale de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | |
Collapse
|
93
|
Hecker M, Blaukat A, Bara AT, Müller-Esterl W, Busse R. ACE inhibitor potentiation of bradykinin-induced venoconstriction. Br J Pharmacol 1997; 121:1475-81. [PMID: 9257930 PMCID: PMC1564840 DOI: 10.1038/sj.bjp.0701281] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. Angiotensin-converting enzyme (ACE) inhibitors exert their cardiovascular effects not only by preventing the formation of angiotensin II (AII), but also by promoting the accumulation of bradykinin in or at the vessel wall. In addition, certain ACE inhibitors have been shown to augment the vasodilator response to bradykinin, presumably by an interaction at the level of the B2 receptor. We have investigated whether this is a specific effect of the ACE inhibitor class of compounds in isolated endothelium-denuded segments of the rabbit jugular vein where bradykinin elicits a constrictor response which is exclusively mediated by activation of the B2 receptor. 2. Moexiprilat and ramiprilat (< or = 3 nM) enhanced the constrictor response to bradykinin three to four fold. Captopril and enalaprilat were less active by approximately one and quinaprilat by two orders of magnitude. Moexiprilat and ramiprilat, on the other hand, had no effect on the constrictor response to AII or the dilator response to acetylcholine. 3. The bradykinin-potentiating effect of the ACE inhibitors was not mimicked by inhibitors of amino-, carboxy-, metallo- or serine peptidases or the synthetic ACE substrate, hippuryl-L-histidyl-L-leucine, at a concentration which almost abolished the residual ACE activity in the vessel wall. In contrast, angiotensin-(1-7) (10 microM), an angiotensin I metabolite, significantly enhanced the constrictor response to bradykinin. 4. Ramiprilat did not alter the binding of [3H]-bradykinin to a membrane fraction prepared from endothelium-denuded rabbit jugular veins or to cultured fibroblasts, and there was no ACE inhibitor-sensitive, bradykinin-induced cleavage of the B2 receptor in cultured endothelial cells. 5. These findings demonstrate that ACE inhibitors selectively potentiate the B2 receptor-mediated vascular effects of bradykinin. Their relative efficacy appears to be independent of their ACE-inhibiting properties and might be related to differences in molecule structure. Moreover, the potentiation of the biological activity of bradykinin by this class of compounds does not seem to be mediated by a shift in affinity of the B2 receptor or a prevention of its desensitization, but may involve an increase in the intrinsic activity of unoccupied B2 receptor molecules.
Collapse
Affiliation(s)
- M Hecker
- Institute of Physiological Chemistry and Pathological Biochemistry, University of Mainz, Germany
| | | | | | | | | |
Collapse
|
94
|
Farhy RD, Peterson E, Scicli AG. Kinins and the events influenced by an angiotensin-converting enzyme inhibitor during neointima formation in the rat carotid artery. J Hypertens 1997; 15:421-9. [PMID: 9211177 DOI: 10.1097/00004872-199715040-00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE In balloon-injured rat carotid arteries, angiotensin-converting enzyme inhibitors (ACEI) decrease neointima formation, and a kinin receptor antagonist partially reverses this inhibitory effect. We studied which of the events leading to neointima formation are involved in the effects of ACEI and kinins. METHODS We administered 5 mg/kg per day ramipril, either alone or combined with the kinin receptor antagonist icatibant (Hoe 140), on the days each wave occurred and studied the effects on neointima formation 14 days after balloon injury. Ramipril alone or combined with icatibant had no effect on neointima formation when administered from 2 days before to 3 or 5 days after balloon injury. In contrast, ramipril inhibited neointima formation when administered from day 7 to day 14. Treatment with icatibant had a small effect, which was sufficient to abolish the effects of ramipril (control 0.11 +/- 0.01 mm2, ramipril 0.08 +/- 0.01 mm2; P < 0.05; ramipril plus icatibant 0.09 +/- 0.01 mm2; NS, ramipril plus icatibant versus control). Thus ramipril was not effective when treatment was stopped after 3 or 5 days, but was mildly effective when treatment was administered during the second week. The effect on migration was studied by counting the number of neointimal cells in rats treated from 2 days before to 4 days after injury. Ramipril decreased the number of cells by 93% compared with controls (control 65.0 +/- 13.5 cells/slice, ramipril 4.7 +/- 2.0 cells/slice; P < 0.001), and this effect was blunted significantly by icatibant (19.5 +/- 5.7 cells/slice; P < 0.009, versus ramipril; P < 0.007, versus controls). The influence of treatment on the rate of proliferation (the 5'-bromo-2'-deoxyuridine index) was studied in the media 3 days, and in the neointima 7 and 10 days after balloon injury. Although proliferation peaked in the neointima after 7 days, there were no differences among the groups at any time. Thus neither ramipril nor icatibant affected the rate of proliferation at the times sampled. Ramipril increased cell density (cells/mm2) in the neointima, and this effect was abolished by cotreatment with icatibant (P < 0.05). CONCLUSION The ACEI needs to be present throughout the experimental period to be most effective. ACEI act on neointima formation in part by inhibiting migration; thus, because ramipril was mildly effective when administered from 7 to 10 days after injury, it is likely that vascular smooth muscle cell migration also occurs continuously. Kinins help mediate roughly 30% of the effect of ACEI on migration. In addition, ACEI, through kinins, affect a process that increases the density of the cells in the neointima, perhaps by decreasing extracellular matrix deposition.
Collapse
Affiliation(s)
- R D Farhy
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | |
Collapse
|
95
|
Lima DX, Fontes MA, Oliveira RC, Campagnole-Santos MJ, Khosla MC, Santos RA. Pressor action of angiotensin I at the ventrolateral medulla: effect of selective angiotensin blockade. IMMUNOPHARMACOLOGY 1996; 33:305-7. [PMID: 8856170 DOI: 10.1016/0162-3109(96)00049-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In this study we explored the possibility that angiotensin-(1-7) (Ang-(1-7)) is involved in the control of blood pressure at the rostral ventrolateral medulla (RVLM) by determining the effect of angiotensin antagonists (DuP 753 and A-779) and the effect of the angiotensin converting enzyme inhibitor, ramiprilat on the pressor action produced by angiotensin I (Ang I). The pressor effect produced by bilateral microinjection of Ang I into the RVLM of anesthetized rats was not significantly altered by DuP 753 or by the ACE inhibitor ramiprilat. Conversely, the Ang-(1-7) antagonist, A-779, reduced significantly the pressor effect produced by Ang I. These data suggest that in our experimental condition Ang I was preferentially converted to Ang-(1-7) at RVLM, or that Ang I and/or one of its fragments acts through a receptor blocked by A-779.
Collapse
Affiliation(s)
- D X Lima
- Departamento de Fisiologia and Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | |
Collapse
|