1
|
Smith D, Layton A. The intrarenal renin-angiotensin system in hypertension: insights from mathematical modelling. J Math Biol 2023; 86:58. [PMID: 36952058 DOI: 10.1007/s00285-023-01891-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2023] [Accepted: 02/21/2023] [Indexed: 03/24/2023]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in the maintenance of volume homeostasis and blood pressure. In addition to the well-studied systemic RAS, local RAS have been documented in various tissues, including the kidney. Given the role of the intrarenal RAS in the pathogenesis of hypertension, a role established via various pharmacologic and genetic studies, substantial efforts have been made to unravel the processes that govern intrarenal RAS activity. In particular, several mechanisms have been proposed to explain the rise in intrarenal angiotensin II (Ang II) that accompanies Ang II infusion, including increased angiotensin type 1 receptor (AT1R)-mediated uptake of Ang II and enhanced intrarenal Ang II production. However, experimentally isolating their contribution to the intrarenal accumulation of Ang II in Ang II-induced hypertension is challenging, given that they are fundamentally connected. Computational modelling is advantageous because the feedback underlying each mechanism can be removed and the effect on intrarenal Ang II can be studied. In this work, the mechanisms governing the intrarenal accumulation of Ang II during Ang II infusion experiments are delineated and the role of the intrarenal RAS in Ang II-induced hypertension is studied. To accomplish this, a compartmental ODE model of the systemic and intrarenal RAS is developed and Ang II infusion experiments are simulated. Simulations indicate that AT1R-mediated uptake of Ang II is the primary mechanism by which Ang II accumulates in the kidney during Ang II infusion. Enhanced local Ang II production is unnecessary. The results demonstrate the role of the intrarenal RAS in the pathogenesis of Ang II-induced hypertension and consequently, clinical hypertension associated with an overactive RAS.
Collapse
Affiliation(s)
- Delaney Smith
- Department of Applied Mathematics, University of Waterloo, 200 University Ave, Waterloo, ON, N2L 3G1, Canada.
| | - Anita Layton
- Department of Applied Mathematics, University of Waterloo, 200 University Ave, Waterloo, ON, N2L 3G1, Canada
- Cheriton School of Computer Science, University of Waterloo, 200 University Ave, Waterloo, ON, N2L 3G1, Canada
- Department of Biology, University of Waterloo, 200 University Ave, Waterloo, ON, N2L 3G1, Canada
- School of Pharmacy, University of Waterloo, 200 University Ave, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
2
|
Interactions between the intrarenal dopaminergic and the renin-angiotensin systems in the control of systemic arterial pressure. Clin Sci (Lond) 2022; 136:1205-1227. [PMID: 35979889 DOI: 10.1042/cs20220338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Systemic arterial hypertension is one of the leading causes of morbidity and mortality in the general population, being a risk factor for many cardiovascular diseases. Although its pathogenesis is complex and still poorly understood, some systems appear to play major roles in its development. This review aims to update the current knowledge on the interaction of the intrarenal renin-angiotensin system (RAS) and dopaminergic system in the development of hypertension, focusing on recent scientific hallmarks in the field. The intrarenal RAS, composed of several peptides and receptors, has a critical role in the regulation of blood pressure (BP) and, consequently, the development of hypertension. The RAS is divided into two main intercommunicating axes: the classical axis, composed of angiotensin-converting enzyme, angiotensin II, and angiotensin type 1 receptor, and the ACE2/angiotensin-(1-7)/Mas axis, which appears to modulate the effects of the classical axis. Dopamine and its receptors are also increasingly showing an important role in the pathogenesis of hypertension, as abnormalities in the intrarenal dopaminergic system impair the regulation of renal sodium transport, regardless of the affected dopamine receptor subtype. There are five dopamine receptors, which are divided into two major subtypes: the D1-like (D1R and D5R) and D2-like (D2R, D3R, and D4R) receptors. Mice deficient in any of the five dopamine receptor subtypes have increased BP. Intrarenal RAS and the dopaminergic system have complex interactions. The balance between both systems is essential to regulate the BP homeostasis, as alterations in the control of both can lead to hypertension.
Collapse
|
3
|
Renin-Angiotensin System Induced Secondary Hypertension: The Alteration of Kidney Function and Structure. Int J Nephrol 2021. [PMID: 31628476 PMCID: PMC8505109 DOI: 10.1155/2021/5599754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Long-term hypertension is known as a major risk factor for cardiovascular and chronic kidney disease (CKD). The Renin-angiotensin system (RAS) plays a key role in hypertension pathogenesis. Angiotensin II (Ang II) enhancement in Ang II-dependent hypertension leads to progressive CKD and kidney fibrosis. In the two-kidney one-clip model (2K1C), more renin is synthesized in the principal cells of the collecting duct than juxtaglomerular cells (JGCs). An increase of renal Ang I and Ang II levels and a decrease of renal cortical and medullary Ang 1–7 occur in both kidneys of the 2K1C hypertensive rat model. In addition, the activity of the angiotensin-converting enzyme (ACE) increases, while ACE2's activity decreases in the medullary region of both kidneys in the 2K1C hypertensive model. Also, the renal prolyl carboxypeptidase (PrCP) expression and its activity reduce in the clipped kidneys. The imbalance in the production of renal ACE, ACE2, and PrCP expression causes the progression of renal injury. Intrarenal angiotensinogen (AGT) expression and urine AGT (uAGT) excretion rates in the unclipped kidney are greater than the clipped kidney in the 2K1C hypertensive rat model. The enhancement of Ang II in the clipped kidney is related to renin secretion, while the elevation of intrarenal Ang II in the unclipped kidney is related to stimulation of AGT mRNA and protein in proximal tubule cells by a direct effect of systemic Ang II level. Ang II-dependent hypertension enhances macrophages and T-cell infiltration into the kidney which increases cytokines, and AGT synthesis in proximal tubules is stimulated via cytokines. Accumulation of inflammatory cells in the kidney aggravates hypertension and renal damage. Moreover, Ang II-dependent hypertension alters renal Ang II type 1 & 2 receptors (AT1R & AT2R) and Mas receptor (MasR) expression, and the renal interstitial fluid bradykinin, nitric oxide, and cGMP response to AT1R, AT2R, or BK B2-receptor antagonists. Based on a variety of sources including PubMed, Google Scholar, Scopus, and Science-Direct, in the current review, we will discuss the role of RAS-induced secondary hypertension on the alteration of renal function.
Collapse
|
4
|
Li XC, Wang CH, Leite APO, Zhuo JL. Intratubular, Intracellular, and Mitochondrial Angiotensin II/AT 1 (AT1a) Receptor/NHE3 Signaling Plays a Critical Role in Angiotensin II-Induced Hypertension and Kidney Injury. Front Physiol 2021; 12:702797. [PMID: 34408663 PMCID: PMC8364949 DOI: 10.3389/fphys.2021.702797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Hypertension is well recognized to be the most important risk factor for cardiovascular diseases, stroke, and end-stage kidney failure. A quarter of the world’s adult populations and 46% of the US adults develop hypertension and currently require antihypertensive treatments. Only 50% of hypertensive patients are responsive to current antihypertensive drugs, whereas remaining patients may continue to develop cardiovascular, stroke, and kidney diseases. The mechanisms underlying the poorly controlled hypertension remain incompletely understood. Recently, we have focused our efforts to uncover additional renal mechanisms, pathways, and therapeutic targets of poorly controlled hypertension and target organ injury using novel animal models or innovative experimental approaches. Specifically, we studied and elucidated the important roles of intratubular, intracellular, and mitochondrial angiotensin II (Ang II) system in the development of Ang II-dependent hypertension. The objectives of this invited article are to review and discuss our recent findings that (a) circulating and intratubular Ang II is taken up by the proximal tubules via the (AT1) AT1a receptor-dependent mechanism, (b) intracellular administration of Ang II in proximal tubule cells or adenovirus-mediated overexpression of an intracellular Ang II fusion protein selectively in the mitochonria of the proximal tubules induces blood pressure responses, and (c) genetic deletion of AT1 (AT1a) receptors or the Na+/H+ exchanger 3 selectively in the proximal tubules decreases basal blood pressure and attenuates Ang II-induced hypertension. These studies provide a new perspective into the important roles of the intratubular, intracellular, and mitochondrial angiotensin II/AT1 (AT1a) receptor signaling in Ang II-dependent hypertensive kidney diseases.
Collapse
Affiliation(s)
- Xiao Chun Li
- Tulane Hypertension and Renal Center of Excellence, Department of Physiology, Tulane University School of Medicine,New Orleans, LA, United States
| | - Chih-Hong Wang
- Tulane Hypertension and Renal Center of Excellence, Department of Physiology, Tulane University School of Medicine,New Orleans, LA, United States
| | - Ana Paula Oliveira Leite
- Tulane Hypertension and Renal Center of Excellence, Department of Physiology, Tulane University School of Medicine,New Orleans, LA, United States
| | - Jia Long Zhuo
- Tulane Hypertension and Renal Center of Excellence, Department of Physiology, Tulane University School of Medicine,New Orleans, LA, United States
| |
Collapse
|
5
|
Leite APO, Li XC, Hassan R, Zheng X, Alexander B, Casarini DE, Zhuo JL. Sex differences in angiotensin II-induced hypertension and kidney injury: role of AT1a receptors in the proximal tubule of the kidney. Clin Sci (Lond) 2021; 135:1825-1843. [PMID: 34282828 PMCID: PMC8969897 DOI: 10.1042/cs20201574] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 01/13/2023]
Abstract
In the present study, we tested the hypothesis that there are significant sex differences in angiotensin II (Ang II)-induced hypertension and kidney injury using male and female wildtype (WT) and proximal tubule-specific AT1a receptor knockout mice (PT-Agtr1a-/-). Twelve groups (n=8-12 per group) of adult male and female WT and PT-Agtr1a-/- mice were infused with a pressor dose of Ang II via osmotic minipump for 2 weeks (1.5 mg/kg/day, i.p.) and simultaneously treated with or without losartan (20 mg/kg/day, p.o.) to determine the respective roles of AT1a receptors in the proximal tubules versus systemic tissues. Basal systolic, diastolic, and mean arterial pressure were approximately 13 ± 3 mmHg lower (P<0.01), while basal 24-h urinary Na+, K+, and Cl- excretion were significantly higher in both male and female PT-Agtr1a-/- mice than WT controls (P<0.01) without significant sex differences between different strains. Both male and female WT and PT-Agtr1a-/- mice developed hypertension (P<0.01), and the magnitudes of the pressor responses to Ang II were similar between male and female WT and PT-Agtr1a-/- mice (n.s.). Likewise, Ang II-induced hypertension was significantly attenuated in both male and female PT-Agtr1a-/- mice (P<0.01). Furthermore, losartan attenuated the hypertensive responses to Ang II to similar extents in both male and female WT and PT-Agtr1a-/- mice. Finally, Ang II-induced kidney injury was attenuated in PT-Agtr1a-/- mice (P<0.01). In conclusion, the present study demonstrates that deletion of AT1a receptors in the proximal tubules of the kidney attenuates Ang II-induced hypertension and kidney injury without revealing significant sex differences.
Collapse
MESH Headings
- Angiotensin II
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Antihypertensive Agents/pharmacology
- Arterial Pressure/drug effects
- Disease Models, Animal
- Female
- Fibrosis
- Hypertension/chemically induced
- Hypertension/metabolism
- Hypertension/physiopathology
- Hypertension/prevention & control
- Kidney Diseases/chemically induced
- Kidney Diseases/metabolism
- Kidney Diseases/physiopathology
- Kidney Diseases/prevention & control
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/physiopathology
- Kidney Tubules, Proximal/ultrastructure
- Losartan/pharmacology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Renin-Angiotensin System/drug effects
- Sex Characteristics
- Sex Factors
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Ana Paula Oliveira Leite
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112-2699, U.S.A
| | - Xiao C. Li
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112-2699, U.S.A
| | - Rumana Hassan
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112-2699, U.S.A
| | - Xiaowen Zheng
- Second Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Barbara Alexander
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, U.S.A
| | - Dulce Elena Casarini
- Division of Nephrology, Department of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jia L. Zhuo
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112-2699, U.S.A
| |
Collapse
|
6
|
Vacková Š, Kikerlová S, Melenovsky V, Kolář F, Imig JD, Kompanowska-Jezierska E, Sadowski J, Červenka L. Altered Renal Vascular Responsiveness to Vasoactive Agents in Rats with Angiotensin II-Dependent Hypertension and Congestive Heart Failure. Kidney Blood Press Res 2019; 44:792-809. [PMID: 31430751 PMCID: PMC10107072 DOI: 10.1159/000501688] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/22/2019] [Indexed: 11/19/2022] Open
Abstract
Objective: We evaluated the hypothesis that the development of renal dysfunction and congestive heart failure (CHF) caused by volume overload in rats with angiotensin II (ANG II)-dependent hypertension is associated with altered renal vascular responsiveness to ANG II and to epoxyeicosatrienoic acids (EETs). Methods: Ren-2 transgenic rats (TGRs) were used as a model of ANG II-dependent hypertension. CHF was induced by volume overload achieved by the creation of the aorto-caval fistula (ACF). Renal blood flow (RBF) responses were determined to renal arterial administration of ANG II, native 11,12-EET, an analog of 14,15-EETs (EET-A), norepinephrine (NE), acetylcholine (Ach) and bradykinin (Bk) in healthy (i.e., sham-operated) TGR and ACF TGR (5 weeks after ACF creation). Results: Selective intrarenal administration of neither vasoactive drug altered mean arterial pressure in any group. Administration of ANG II caused greater decreases in RBF in ACF TGR than in sham-operated TGR, whereas after administration of NE the respective decreases were comparable in the 2 groups. Administration of Ach and Bk elicited significantly higher RBF increases in ACF TGR as compared with sham-operated TGR. In contrast, administration of 11,12-EET and EET-A caused significantly smaller RBF increases in ACF TGR than in sham-operated TGR. Conclusion: The findings show that 5 weeks after creation of ACF, the TGR exhibit exaggerated renal vasoconstrictor responses to ANG II and reduced renal vasodilatory responses to EETs, suggesting that both these alterations might play an important role in the development of renal dysfunction in this model of CHF.
Collapse
Affiliation(s)
- Šárka Vacková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Soňa Kikerlová
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Vojtěch Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - František Kolář
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Elzbieta Kompanowska-Jezierska
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Centre, Polish Academy of Science, Warsaw, Poland
| | - Janusz Sadowski
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Centre, Polish Academy of Science, Warsaw, Poland
| | - Luděk Červenka
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia,
| |
Collapse
|
7
|
Liu C, Luo R, Wang W, Peng Z, Johnson GVW, Kellems RE, Xia Y. Tissue Transglutaminase-Mediated AT1 Receptor Sensitization Underlies Pro-inflammatory Cytokine LIGHT-Induced Hypertension. Am J Hypertens 2019; 32:476-485. [PMID: 30715101 PMCID: PMC6475879 DOI: 10.1093/ajh/hpz018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/02/2019] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although numerous recent studies have shown a strong link between inflammation and hypertension, the underlying mechanisms by which inflammatory cytokines induce hypertension remain to be fully elucidated. Hypertensive disorders are also associated with elevated pressor sensitivity. Tissue transglutaminase (TG2), a potent cross-linking enzyme, is known to be transcriptionally activated by inflammatory cytokines and stabilize angiotensin II (Ang II) receptor AT1 (AT1R) via ubiquitination-preventing posttranslational modification. Here we sought to investigate the TG2-mediated AT1R stabilization in inflammation-induced hypertension and its functional consequences with a focus on receptor abundance and Ang II responsiveness. METHODS AND RESULTS Using an experimental model of inflammation-induced hypertension established by introducing the pro-inflammatory tumor necrosis factor cytokine LIGHT, we provide pharmacologic and genetic evidence that TG2 is required for LIGHT-induced hypertension (systolic pressure on day 6: LIGHT = 152.3 ± 7.4 vs. LIGHT+ERW1041E [TG2 inhibitor] = 105.8 ± 13.1 or LIGHT+TG2−/− = 114.3 ± 4.3 mm Hg, P < 0.05, n = 4–5) and renal compromise (urine albumin/creatinine: LIGHT = 0.17 ± 0.05 vs. LIGHT+ERW1041E = 0.03 ± 0.01 or LIGHT+TG2−/− = 0.06 ± 0.01 mg/mg; plasma creatinine: LIGHT = 1.11 ± 0.04 vs. LIGHT+ERW1041E = 0.94 ± 0.04 or LIGHT+TG2−/− = 0.88 ± 0.09 mg/dl; urine volume: LIGHT = 0.23 ± 0.1 vs. LIGHT+ERW1041E = 0.84 ± 0.13 or LIGHT+TG2−/− = 1.02 ± 0.09 ml/24 hour on day 14, P < 0.05, n = 4–5). Our mechanistic studies showed that the TG2-mediated AT1R modification and accumulation (relative renal AT1R level: phosphate-buffered saline [PBS] = 1.23 ± 0.22, LIGHT = 3.49 ± 0.37, and LIGHT+ERW1041E = 1.77 ± 0.46, P < 0.05, n = 3; LIGHT+TG2+/+ = 85.28 ± 36.11 vs. LIGHT+TG2−/− = 7.01 ± 5.68, P < 0.05, n = 3) induced by LIGHT is associated with abrogated β-arrestin binding (AT1R/associated β-arrestin ratio: PBS = 2.62 ± 1.07, LIGHT = 38.60 ± 13.91, and LIGHT+ERW1041E = 6.97 ± 2.91, P < 0.05, n = 3; LIGHT+TG2+/+ = 66.43 ± 44.81 vs. LIGHT+TG2−/− = 2.45 ± 1.78, P < 0.01, n = 3) and could be found in renal medulla tubules of kidneys (relative tubular AT1R level: PBS = 5.91 ± 2.93, LIGHT = 92.82 ± 19.54, LIGHT+ERW1041E = 28.49 ± 11.65, and LIGHT+TG2−/− = 0.14 ± 0.10, P < 0.01, n = 5) and the blood vasculature (relative vascular AT1R level: PBS = 0.70 ± 0.30, LIGHT = 13.75 ± 2.49, and LIGHT+ERW1041E = 3.28 ± 0.87, P < 0.01, n = 3), 2 of the tissues highly related to the genesis of hypertension. Our in vitro cellular assays showed that LIGHT stimulation triggered a rapid TG2-dependent increase in the abundance of AT1Rs (relative AT1R level after 2-hour LIGHT treatment: AT1R (WT)+TG2 = 2.21 ± 0.23, AT1R (Q315A)+TG2 = 0.18 ± 0.23, P < 0.05 vs. starting point = 1, n = 2) and downstream calcium signaling (fold increase in NFAT-driven luciferase activity: Saline = 0.02 ± 0.03, Ang II = 0.17 ± 0.08, LIGHT = 0.05 ± 0.04, LIGHT+Ang II = 0.90 ± 0.04 (P < 0.01 vs. Ang II), and LIGHT+Ang II+ERW1041E = 0.15 ± 0.15 (P < 0.01 vs. LIGHT+Ang II), n = 3). CONCLUSIONS Our data indicate an essential and systemic role for TG2 in bridging inflammation to hypertension via its posttranslational modifications stabilizing AT1 receptor and sensitizing Ang II. Our findings also suggest that TG2 inhibitors could be used as a novel group of cardiovascular agents.
Collapse
Affiliation(s)
- Chen Liu
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
| | - Renna Luo
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, PRC
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Zhangzhe Peng
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, McGovern Medical School at Houston, University of Texas, Houston, Texas, USA
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| |
Collapse
|
8
|
Intratubular and intracellular renin-angiotensin system in the kidney: a unifying perspective in blood pressure control. Clin Sci (Lond) 2018; 132:1383-1401. [PMID: 29986878 DOI: 10.1042/cs20180121] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022]
Abstract
The renin-angiotensin system (RAS) is widely recognized as one of the most important vasoactive hormonal systems in the physiological regulation of blood pressure and the development of hypertension. This recognition is derived from, and supported by, extensive molecular, cellular, genetic, and pharmacological studies on the circulating (tissue-to-tissue), paracrine (cell-to-cell), and intracrine (intracellular, mitochondrial, nuclear) RAS during last several decades. Now, it is widely accepted that circulating and local RAS may act independently or interactively, to regulate sympathetic activity, systemic and renal hemodynamics, body salt and fluid balance, and blood pressure homeostasis. However, there remains continuous debate with respect to the specific sources of intratubular and intracellular RAS in the kidney and other tissues, the relative contributions of the circulating RAS to intratubular and intracellular RAS, and the roles of intratubular compared with intracellular RAS to the normal control of blood pressure or the development of angiotensin II (ANG II)-dependent hypertension. Based on a lecture given at the recent XI International Symposium on Vasoactive Peptides held in Horizonte, Brazil, this article reviews recent studies using mouse models with global, kidney- or proximal tubule-specific overexpression (knockin) or deletion (knockout) of components of the RAS or its receptors. Although much knowledge has been gained from cell- and tissue-specific transgenic or knockout models, a unifying and integrative approach is now required to better understand how the circulating and local intratubular/intracellular RAS act independently, or with other vasoactive systems, to regulate blood pressure, cardiovascular and kidney function.
Collapse
|
9
|
Li XC, Zhuo JL. Recent Updates on the Proximal Tubule Renin-Angiotensin System in Angiotensin II-Dependent Hypertension. Curr Hypertens Rep 2017; 18:63. [PMID: 27372447 DOI: 10.1007/s11906-016-0668-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is well recognized that the renin-angiotensin system (RAS) exists not only as circulating, paracrine (cell to cell), but also intracrine (intracellular) system. In the kidney, however, it is difficult to dissect the respective contributions of circulating RAS versus intrarenal RAS to the physiological regulation of proximal tubular Na(+) reabsorption and hypertension. Here, we review recent studies to provide an update in this research field with a focus on the proximal tubular RAS in angiotensin II (ANG II)-induced hypertension. Careful analysis of available evidence supports the hypothesis that both local synthesis or formation and AT1 (AT1a) receptor- and/or megalin-mediated uptake of angiotensinogen (AGT), ANG I and ANG II contribute to high levels of ANG II in the proximal tubules of the kidney. Under physiological conditions, nearly all major components of the RAS including AGT, prorenin, renin, ANG I, and ANG II would be filtered by the glomerulus and taken up by the proximal tubules. In ANG II-dependent hypertension, the expression of AGT, prorenin, and (pro)renin receptors, and angiotensin-converting enzyme (ACE) is upregulated rather than downregulated in the kidney. Furthermore, hypertension damages the glomerular filtration barrier, which augments the filtration of circulating AGT, prorenin, renin, ANG I, and ANG II and their uptake in the proximal tubules. Together, increased local ANG II formation and augmented uptake of circulating ANG II in the proximal tubules, via activation of AT1 (AT1a) receptors and Na(+)/H(+) exchanger 3, may provide a powerful feedforward mechanism for promoting Na(+) retention and the development of ANG II-induced hypertension.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, 2500 North State Street, Jackson, MS, 39216-4505, USA
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, 2500 North State Street, Jackson, MS, 39216-4505, USA.
| |
Collapse
|
10
|
Yao Y, Davis G, Harrison JC, Walker RJ, Sammut IA. Renal functional responses in diabetic nephropathy following chronic bilateral renal denervation. Auton Neurosci 2017; 204:98-104. [DOI: 10.1016/j.autneu.2016.09.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
|
11
|
Zhuo JL, Kobori H, Li XC, Satou R, Katsurada A, Navar LG. Augmentation of angiotensinogen expression in the proximal tubule by intracellular angiotensin II via AT1a/MAPK/NF-кB signaling pathways. Am J Physiol Renal Physiol 2016; 310:F1103-12. [PMID: 26864937 PMCID: PMC4889322 DOI: 10.1152/ajprenal.00350.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 02/02/2016] [Indexed: 11/22/2022] Open
Abstract
Long-term angiotensin II (ANG II) infusion significantly increases ANG II levels in the kidney through two major mechanisms: AT1 receptor-mediated augmentation of angiotensinogen (AGT) expression and uptake of circulating ANG II by the proximal tubules. However, it is not known whether intracellular ANG II stimulates AGT expression in the proximal tubule. In the present study, we overexpressed an intracellular cyan fluorescent ANG II fusion protein (Ad-sglt2-ECFP/ANG II) selectively in the proximal tubule of rats and mice using the sodium and glucose cotransporter 2 (sglt2) promoter. AGT mRNA and protein expression in the renal cortex and 24-h urinary AGT excretion were determined 4 wk following overexpression of ECFP/ANG II in the proximal tubule. Systolic blood pressure was significantly increased with a small antinatriuretic effect in rats and mice with proximal tubule-selective expression of ECFP/ANG II (P < 0.01). AGT mRNA and protein expression in the cortex were increased by >1.5-fold and 61 ± 16% (P < 0.05), whereas urinary AGT excretion was increased from 48.7 ± 5.7 (n = 13) to 102 ± 13.5 (n = 13) ng/24 h (P < 0.05). However, plasma AGT, renin activity, and ANG II levels remained unaltered by ECFP/ANG II. The increased AGT mRNA and protein expressions in the cortex by ECFP/ANG II were blocked in AT1a-knockout (KO) mice. Studies in cultured mouse proximal tubule cells demonstrated involvement of AT1a receptor/MAP kinases/NF-кB signaling pathways. These results indicate that intracellular ANG II stimulates AGT expression in the proximal tubules, leading to increased AGT formation and secretion into the tubular fluid, which contributes to ANG II-dependent hypertension.
Collapse
Affiliation(s)
- Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - H Kobori
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - R Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - A Katsurada
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
12
|
VOKURKOVÁ M, RAUCHOVÁ H, ŘEZÁČOVÁ L, VANĚČKOVÁ I, ZICHA J. NADPH Oxidase Activity and Reactive Oxygen Species Production in Brain and Kidney of Adult Male Hypertensive Ren-2 Transgenic Rats. Physiol Res 2015; 64:849-56. [DOI: 10.33549/physiolres.933254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM) play an important role in brain control of blood pressure (BP). One of the important mechanisms involved in the pathogenesis of hypertension is the elevation of reactive oxygen species (ROS) production by nicotine adenine dinucleotide phosphate (NADPH) oxidase. The aim of our present study was to investigate NADPH oxidase-mediated superoxide (O2-) production and to search for the signs of lipid peroxidation in hypothalamus and medulla oblongata as well as in renal medulla and cortex of hypertensive male rats transgenic for the murine Ren-2 renin gene (Ren-2 TGR) and their age-matched normotensive controls ‒ Hannover Sprague Dawley rats (HanSD). We found no difference in the activity of NADPH oxidase measured as a lucigenin-mediated O2- production in the hypothalamus and medulla oblongata. However, we observed significantly elevated NADPH oxidase in both renal cortex and medulla of Ren-2 TGR compared with HanSD. Losartan (LOS) treatment (10 mg/kg body weight/day) for 2 months (Ren-2 TGR+LOS) did not change NADPH oxidase-dependent O2- production in the kidney. We detected significantly elevated indirect markers of lipid peroxidation measured as thiobarbituric acid-reactive substances (TBARS) in Ren-2 TGR, while they were significantly decreased in Ren-2 TGR+LOS. In conclusion, the present study shows increased NADPH oxidase activities in renal cortex and medulla with significantly increased TBARS in renal cortex. No significant changes of NADPH oxidase and markers of lipid peroxidation were detected in the studied brain regions.
Collapse
Affiliation(s)
| | - H. RAUCHOVÁ
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | | | | | | |
Collapse
|
13
|
Filyushina EE, Shmerling MD, Buzueva II, Lazarev VA, Markel AL, Yakobson GS. Structural characteristics of renomedullary interstitial cells of hypertensive ISIAH rats. Bull Exp Biol Med 2015; 155:408-12. [PMID: 24137614 DOI: 10.1007/s10517-013-2164-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The structural characteristics of the renal medulla and its interstitial cells were studied in hypertensive ISIAH rats (in comparison with normotensive WAG rats) in order to clear out the role of the renomedullary interstitial cells in the mechanisms of AP regulation. Morphometric electron microscopic analysis and immunohistochemical studies in ISIAH rats detected the initial signs of the renomedullary sclerosis. The renomedullary interstitial cells of ISIAH rats were characterized by higher numerical density and were larger in size, with a higher volumic share of their secretory granules. These structural features of the studied cells were regarded as signs of their more intense functional activity aimed at hypertension suppression.
Collapse
Affiliation(s)
- E E Filyushina
- Institute of Physiology, Siberian Division of the Russian Academy of Medical Sciences; Institute of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia.
| | | | | | | | | | | |
Collapse
|
14
|
Vavrinec P, Henning RH, Goris M, Vavrincova-Yaghi D, Buikema H, van Dokkum RPE. Vascular smooth muscle function of renal glomerular and interlobar arteries predicts renal damage in rats. Am J Physiol Renal Physiol 2012; 303:F1187-95. [PMID: 22791345 DOI: 10.1152/ajprenal.00653.2011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, it was shown that individuals with good baseline (a priori) endothelial function in isolated (in vitro) renal arteries developed less renal damage after 5/6 nephrectomy (5/6Nx; Gschwend S, Buikema H, Navis G, Henning RH, de Zeeuw D, van Dokkum RP. J Am Soc Nephrol 13: 2909-2915, 2002). In this study, we investigated whether preexisting glomerular vascular integrity predicts subsequent renal damage after 5/6Nx, using in vivo intravital microscopy and in vitro myogenic constriction of small renal arteries. Moreover, we aimed to elucidate the role of renal ANG II type 1 receptor (AT1R) expression in this model. Anesthetized rats underwent intravital microscopy to visualize constriction to ANG II of glomerular afferent and efferent arterioles, with continuous measurement of blood pressure, heart rate, and renal blood flow. Thereafter, 5/6Nx was performed, interlobar arteries were isolated from the extirpated kidney, and myogenic constriction was assessed in a perfused vessel setup. Blood pressure and proteinuria were assessed weekly for 12 wk, and focal glomerulosclerosis (FGS) was determined at the end of study. Relative expression AT1R in the kidney cortex obtained at 5/6Nx was determined by PCR. Infusion of ANG II induced significant constriction of both afferent and efferent glomerular arterioles, which strongly positively correlated with proteinuria and FGS at 12 wk after 5/6Nx. Furthermore, in vitro measured myogenic constriction of small renal arteries negatively correlated with proteinuria 12 wk after 5/6Nx. Moreover, in vivo vascular reactivity negatively correlated with in vitro reactivity. Additionally, relative expression of AT1R positively correlated with responses of glomerular arterioles and with markers of renal damage. Both in vivo afferent and efferent responses to ANG II and in vitro myogenic constriction of small renal arteries in the healthy rat predict the severity of renal damage induced by 5/6Nx. This vascular responsiveness is highly dependent on AT1R expression. Intraorgan vascular integrity may provide a useful tool to guide the prevention and treatment of renal end-organ damage.
Collapse
Affiliation(s)
- Peter Vavrinec
- Department of Clinical Pharmacology, Groningen University Institute for Drug Exploration, University Medical Center Groningen, A. Deusinglaan 1, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
15
|
Eräranta A, Riutta A, Fan M, Koskela J, Tikkanen I, Lakkisto P, Niemelä O, Parkkinen J, Mustonen J, Pörsti I. Dietary phosphate binding and loading alter kidney angiotensin-converting enzyme mRNA and protein content in 5/6 nephrectomized rats. Am J Nephrol 2012; 35:401-8. [PMID: 22517117 DOI: 10.1159/000337942] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 02/25/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vitamin D receptor activation with paricalcitol can modulate the transcription of renin-angiotensin system components in the surgical 5/6 nephrectomy rat model (5/6 NX) of chronic renal insufficiency. We tested the hypothesis whether dietary modification of phosphate influences kidney renin-angiotensin system gene expression at the mRNA level in 5/6 NX rats. METHODS Fifteen weeks after surgery, rats were given control diet (0.3% calcium, 0.5% phosphate), phosphate-lowering diet (3% calcium as carbonate) or high-phosphate diet (1.5%) for 12 weeks. Sham-operated rats were on control diet. RESULTS Blood pressure, plasma phosphate, parathyroid hormone, glomerulosclerosis, tubulointerstitial damage, and FGF-23 were increased in remnant kidney rats, whereas creatinine clearance was decreased. Phosphate, parathyroid hormone, glomerulosclerosis, tubulointerstitial damage, and FGF-23 were further elevated by the high-phosphate diet, but were reduced by the phosphate-lowering diet. Plasma calcium was increased with the phosphate-lowering diet and decreased with the high-phosphate diet. Remnant kidney rats on control diet showed upregulated kidney angiotensin-converting enzyme (ACE) and angiotensin (Ang) IV receptor (AT(4)) transcription, while ACE2, Ang II type 2 receptor and renin receptor transcription were downregulated in comparison with sham rats. Phosphate-lowering diet reduced whereas high-phosphate diet increased kidney ACE, and these effects were observed at both mRNA and protein levels. Dietary phosphate loading also resulted in lower AT(1a) gene transcription. CONCLUSION Dietary phosphate loading was associated with elevated kidney ACE expression, increased tissue damage and lower AT(1a) transcription in 5/6 NX rats. Phosphate binding with 3% calcium carbonate had opposite effects on ACE and kidney damage.
Collapse
Affiliation(s)
- Arttu Eräranta
- School of Medicine, University of Tampere, Tampere, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Palanisamy N, Venkataraman Anuradha C. Soy protein prevents renal damage in a fructose-induced model of metabolic syndrome via inhibition of NF-kB in male rats. Pediatr Nephrol 2011; 26:1809-21. [PMID: 21533627 DOI: 10.1007/s00467-011-1882-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/25/2011] [Accepted: 03/17/2011] [Indexed: 12/15/2022]
Abstract
The study determines the effect of soy protein on inflammatory status and expression of nuclear factor-kappa B (NF-κB P(65)) and receptor for advanced glycation end products (RAGE) in a metabolic syndrome (MS) model. MS was induced in adult male rats by feeding them a high fructose diet (60 g/100 g diet). The rats were randomised into six groups by feeding one of the following semi-synthetic diets for 60 days: corn starch (60%) and casein (20%; CCD), fructose (60%) and casein (20%; FCD), fructose (60%) and soy protein (20%; FSD) or corn starch (60%) and soy protein (20%; CSD). The expression of NF-κB P(65), transforming growth factor-β1 (TGF-β1) and RAGE, histochemical localization of α-smooth muscle actin (α-SMA), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) assays, collagen deposition and ultrastructural analysis were performed. FCD rats displayed inflammatory changes and increased expression of growth factors and nuclear factors. FSD rats showed reduction in inflammation, fibrogenesis, collagen deposition, NF-κB activation and mitigated the ultrastructural changes. Soy protein prevents inflammation and early nephropathic changes in the MS model secondary to the attenuation of NF-κB activation.
Collapse
Affiliation(s)
- Nallasamy Palanisamy
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar, 608 002 Tamil Nadu, India.
| | | |
Collapse
|
17
|
Palanisamy N, Kannappan S, Anuradha CV. Genistein modulates NF-κB-associated renal inflammation, fibrosis and podocyte abnormalities in fructose-fed rats. Eur J Pharmacol 2011; 667:355-64. [PMID: 21704028 DOI: 10.1016/j.ejphar.2011.06.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/26/2011] [Accepted: 06/07/2011] [Indexed: 12/21/2022]
Abstract
The study determines the effect of genistein on inflammatory status and expression of nuclear factor-kappa B (NF-κB p65), transforming growth factor-β1 (TGF-β1) and receptor for advanced glycation end products (RAGE) in kidney of fructose-fed rats. Adult male Wistar rats were fed a diet containing either starch or fructose as the source of carbohydrate. Fifteen days later, after confirming the development of insulin resistance in fructose-fed rats, the rats in each dietary group were divided into two and treated with either genistein (1 mg/kg/day) in 30% dimethylsulfoxide (DMSO) or 30% DMSO alone for the next 45 days. The expression of NF-κB P(65), TGF-β1 and RAGE, histochemical localization of α-smooth muscle actin (α-SMA), levels of tumour necrosis factor-α (TNF-α) and interleukin-6(IL-6) and ultrastructural analysis were performed at the end of the experimental period. Fructose-fed rats displayed inflammatory changes in kidney. Increased expression of TGF-β1 and RAGE in cytosol and NF-κB p65 in nuclear fraction were observed. α-SMA expression was higher in fructose-fed rat kidney. Proliferation of connective tissue was evident from increased collagen deposition in perivascular and intraglomerular regions. Administration of genistein to fructose-fed rats reduced inflammation, fibrogenesis and NF-κB activation. Genistein also mitigated the structural changes such as basement membrane thickening, reduction in podocyte number and loss of glomerular filtration barrier integrity. These findings suggest that genistein prevents inflammation, fibrosis and early nephropathic changes in fructose-fed insulin resistant rats secondary to the attenuation of NF-κB activation.
Collapse
Affiliation(s)
- Nallasamy Palanisamy
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar-608 002, Tamil Nadu, India
| | | | | |
Collapse
|
18
|
Gonzalez-Villalobos RA, Billet S, Kim C, Satou R, Fuchs S, Bernstein KE, Navar LG. Intrarenal angiotensin-converting enzyme induces hypertension in response to angiotensin I infusion. J Am Soc Nephrol 2011; 22:449-59. [PMID: 21115616 PMCID: PMC3060439 DOI: 10.1681/asn.2010060624] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 09/25/2010] [Indexed: 01/13/2023] Open
Abstract
The contribution of the intrarenal renin-angiotensin system to the development of hypertension is incompletely understood. Here, we used targeted homologous recombination to generate mice that express angiotensin-converting enzyme (ACE) in the kidney tubules but not in other tissues. Mice homozygous for this genetic modification (ACE 9/9 mice) had low BP levels, impaired ability to concentrate urine, and variable medullary thinning. In accord with the ACE distribution, these mice also had reduced circulating angiotensin II and high plasma renin concentration but maintained normal kidney angiotensin II levels. In response to chronic angiotensin I infusions, ACE 9/9 mice displayed increased kidney angiotensin II, enhanced rate of urinary angiotensin II excretion, and development of hypertension. These findings suggest that intrarenal ACE-derived angiotensin II formation, even in the absence of systemic ACE, increases kidney angiotensin II levels and promotes the development of hypertension.
Collapse
Affiliation(s)
- Romer A Gonzalez-Villalobos
- Departments of Physiology and Hypertension, Renal Center, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Navar LG, Kobori H, Prieto MC, Gonzalez-Villalobos RA. Intratubular renin-angiotensin system in hypertension. Hypertension 2011; 57:355-62. [PMID: 21282552 PMCID: PMC3073668 DOI: 10.1161/hypertensionaha.110.163519] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- L Gabriel Navar
- Department of Physiology, SL39, Tulane University Health Science Center, 1430 Tulane Ave, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
20
|
Vaajanen A, Lakkisto P, Virtanen I, Kankuri E, Oksala O, Vapaatalo H, Tikkanen I. Angiotensin receptors in the eyes of arterial hypertensive rats. Acta Ophthalmol 2010; 88:431-8. [PMID: 19758401 DOI: 10.1111/j.1755-3768.2009.01563.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of the present study was to determine whether the eye tissues of arterial hypertensive rats evince expression of angiotensin receptors (AT(1) and AT(2)) as well as the novel Mas receptor, whose endogenous ligand is vasorelaxing Angiotensin (1-7) [Ang (1-7)]. METHODS Enucleated eyes from spontaneously hypertensive rats (SHR) and double transgenic rats harbouring human renin and angiotensinogen genes (dTGR) and their normotensive controls were used. Half of the rats were pretreated orally with an Angiotensin II (Ang II) type 1 receptor blocker (ARB). The eyes were snap-frozen in isopentane at -40 degrees and stored at -70 degrees for subsequent reverse transcriptase polymerase chain reaction (RT-PCR) analysis or in vitro autoradiography. RESULTS The mRNA expression of AT(1a) and AT(2) as well as the novel Mas receptor was detected in all rat groups, being markedly higher in the retina than in the ciliary body. dTGR had significantly more receptors than SHR, but no direct relation to blood pressure level was seen. According to the autoradiography, treatment with ARB blocked a part of AT(1) receptors but had no clear effect on AT(2) receptors. CONCLUSION The novel Mas receptor was found by RT-PCR in eye tissue for the first time. Its specific ligand, Ang (1-7), may be involved in the regulation of intraocular pressure--as recently demonstrated by us--and in the pathogenesis of retinal diseases as a counter-regulatory component for the vascular and proliferative actions of Ang II. The results suggest that the density of AT(1) receptors in the eye is independent of the blood pressure level of the animal.
Collapse
MESH Headings
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensinogen/genetics
- Animals
- Animals, Genetically Modified
- Autoradiography
- Blood Pressure
- Ciliary Body/metabolism
- Gene Expression Regulation/physiology
- Hypertension/genetics
- Intraocular Pressure
- Male
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 2/genetics
- Receptors, G-Protein-Coupled/genetics
- Renin/genetics
- Retina/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Anu Vaajanen
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
21
|
Castrop H, Höcherl K, Kurtz A, Schweda F, Todorov V, Wagner C. Physiology of Kidney Renin. Physiol Rev 2010; 90:607-73. [PMID: 20393195 DOI: 10.1152/physrev.00011.2009] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The protease renin is the key enzyme of the renin-angiotensin-aldosterone cascade, which is relevant under both physiological and pathophysiological settings. The kidney is the only organ capable of releasing enzymatically active renin. Although the characteristic juxtaglomerular position is the best known site of renin generation, renin-producing cells in the kidney can vary in number and localization. (Pro)renin gene transcription in these cells is controlled by a number of transcription factors, among which CREB is the best characterized. Pro-renin is stored in vesicles, activated to renin, and then released upon demand. The release of renin is under the control of the cAMP (stimulatory) and Ca2+(inhibitory) signaling pathways. Meanwhile, a great number of intrarenally generated or systemically acting factors have been identified that control the renin secretion directly at the level of renin-producing cells, by activating either of the signaling pathways mentioned above. The broad spectrum of biological actions of (pro)renin is mediated by receptors for (pro)renin, angiotensin II and angiotensin-( 1 – 7 ).
Collapse
Affiliation(s)
- Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Klaus Höcherl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Vladimir Todorov
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
22
|
Hayden MR, Habibi J, Whaley-Connell A, Sowers D, Johnson M, Tilmon R, Jain D, Ferrario C, Sowers JR. Nebivolol attenuates maladaptive proximal tubule remodeling in transgenic rats. Am J Nephrol 2010; 31:262-72. [PMID: 20110666 DOI: 10.1159/000278757] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 12/14/2009] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS The impact of nebivolol therapy on the renal proximal tubular cell (PTC) structure and function was investigated in a transgenic (TG) rodent model of hypertension and the cardiometabolic syndrome. The TG Ren2 rat develops nephropathy with proteinuria, increased renal angiotensin II levels and oxidative stress, and PTC remodeling. Nebivolol, a beta(1)-antagonist, has recently been shown to reduce albuminuria, in part, through reductions in renal oxidative stress. Accordingly, we hypothesized that nebivolol therapy would attenuate PTC damage and tubulointerstitial fibrosis. METHODS Young Ren2 (R2-N) and SD (SD-N) rats were treated with nebivolol (10 mg/kg/day) or vehicle (R2-C; SD-C) for 3 weeks. PTC structure and function were tested using transmission electron microscopy and functional measurements. RESULTS Nebivolol treatment decreased urinary N-acetyl-beta-D-glucosaminidase, tubulointerstitial ultrastructural remodeling and fibrosis, NADPH oxidase activity, 3-nitrotyrosine levels, and increased megalin and lysosomal-associated membrane protein-2 immunostaining in PTCs. Ultrastructural abnormalities that were improved with therapy included altered canalicular structure, reduced endosomes/lysosomes and PTC vacuoles, basement membrane thickening, and mitochondrial remodeling/fragmentation. CONCLUSION These observations support the notion that nebivolol may improve PTC reabsorption of albumin and other glomerular filtered small molecular weight proteins in association with the attenuation of oxidative stress, tubulointerstitial injury and fibrosis in this rat model of metabolic kidney disease.
Collapse
MESH Headings
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Benzopyrans/pharmacology
- Blood Pressure/drug effects
- Disease Models, Animal
- Ethanolamines/pharmacology
- Fibrosis
- Hypertension, Renal/drug therapy
- Hypertension, Renal/metabolism
- Hypertension, Renal/pathology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Lysosomes/metabolism
- Lysosomes/ultrastructure
- Male
- Microscopy, Electron
- Nebivolol
- Oxidative Stress/drug effects
- Proteinuria/drug therapy
- Proteinuria/pathology
- Rats
- Rats, Sprague-Dawley
- Rats, Transgenic
- Renin/genetics
- Vacuoles/metabolism
- Vacuoles/ultrastructure
Collapse
Affiliation(s)
- Melvin R Hayden
- Department of Internal Medicine, University of Missouri-Columbia School of Medicine, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gonzalez-Villalobos RA, Satou R, Ohashi N, Semprun-Prieto LC, Katsurada A, Kim C, Upchurch GM, Prieto MC, Kobori H, Navar LG. Intrarenal mouse renin-angiotensin system during ANG II-induced hypertension and ACE inhibition. Am J Physiol Renal Physiol 2010; 298:F150-F157. [PMID: 19846570 PMCID: PMC2806115 DOI: 10.1152/ajprenal.00477.2009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 10/14/2009] [Indexed: 01/07/2023] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibition (ACEi) ameliorates the development of hypertension and the intrarenal ANG II augmentation in ANG II-infused mice. To determine if these effects are associated with changes in the mouse intrarenal renin-angiotensin system, the expression of angiotensinogen (AGT), renin, ACE, angiotensin type 1 receptor (AT(1)R) mRNA (by quanitative RT-PCR) and protein [by Western blot (WB) and/or immunohistochemistry (IHC)] were analyzed. C57BL/6J male mice (9-12 wk old) were distributed as controls (n = 10), ANG II infused (ANG II = 8, 400 ng x kg(-1) x min(-1) for 12 days), ACEi only (ACEi = 10, lisinopril, 100 mg/l), and ANG II infused + ACEi (ANG II + ACEi = 11). When compared with controls (1.00), AGT protein (by WB) was increased by ANG II (1.29 +/- 0.13, P < 0.05), and this was not prevented by ACEi (ACEi + ANG II, 1.31 +/- 0.14, P < 0.05). ACE protein (by WB) was increased by ANG II (1.21 +/- 0.08, P < 0.05), and it was reduced by ACEi alone (0.88 +/- 0.07, P < 0.05) or in combination with ANG II (0.80 +/- 0.07, P < 0.05). AT(1)R protein (by WB) was increased by ANG II (1.27 +/- 0.06, P < 0.05) and ACEi (1.17 +/- 0.06, P < 0.05) but not ANG II + ACEi [1.15 +/- 0.06, not significant (NS)]. Tubular renin protein (semiquantified by IHC) was increased by ANG II (1.49 +/- 0.23, P < 0.05) and ACEi (1.57 +/- 0.15, P < 0.05), but not ANG II + ACEi (1.10 +/- 0.15, NS). No significant changes were observed in AGT, ACE, or AT(1)R mRNA. In summary, reduced responses of intrarenal tubular renin, ACE, and the AT(1)R protein to the stimulatory effects of chronic ANG II infusions, in the presence of ACEi, are associated with the effects of this treatment to ameliorate augmentations in blood pressure and intrarenal ANG II content during ANG II-induced hypertension.
Collapse
Affiliation(s)
- Romer A Gonzalez-Villalobos
- Dept. of Physiology, Tulane Univ. Health Sciences Center, 1430 Tulane Ave., SL39, New Orleans, LA 70112, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Beierwaltes WH. The role of calcium in the regulation of renin secretion. Am J Physiol Renal Physiol 2010; 298:F1-F11. [PMID: 19640903 PMCID: PMC2806121 DOI: 10.1152/ajprenal.00143.2009] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/23/2009] [Indexed: 12/20/2022] Open
Abstract
Renin is the enzyme which is the rate-limiting step in the formation of the hormone angiotensin II. Therefore, the regulation of renin secretion is critical in understanding the control of the renin-angiotensin-aldosterone system and its many biological and pathological actions. Renin is synthesized, stored in, and released from the juxtaglomerular (JG) cells of the kidney. While renin secretion is positively regulated by the "second messenger" cAMP, unlike most secretory cells, renin secretion from the JG cell is inversely related to the extracellular and intracellular calcium concentrations. This novel relationship is referred to as the "calcium paradox." This review will address observations made over the past 30 years regarding calcium and the regulation of renin secretion, and focus on recent observations which address this scientific conundrum. These include 1) receptor-mediated pathways for changing intracellular calcium; 2) the discovery of a calcium-inhibitable isoform of adenylyl cyclase associated with renin in the JG cells; 3) calcium-sensing receptors in the JG cells; 4) calcium-calmodulin-mediated signals; 5) the role of phosphodiesterases; and 6) connexins, gap junctions, calcium waves, and the cortical extracellular calcium environment. While cAMP is the dominant second messenger for renin secretion, calcium appears to modulate the integrated activities of the enzymes, which balance cAMP synthesis and degradation. Thus this review concludes that calcium modifies the amplitude of cAMP-mediated renin-signaling pathways. While calcium does not directly control renin secretion, increased calcium inhibits and decreased calcium amplifies cAMP-stimulated renin secretion.
Collapse
Affiliation(s)
- William H Beierwaltes
- Dept. of Medicine, Hypertension and Vascular Research Div, Henry Ford Hospital, 7121 E&R Bldg, 2799 W. Grand Blvd, Detroit, MI 48202, USA.
| |
Collapse
|
25
|
Angiotensin II receptor blocker is a renoprotective remedy for metabolic syndrome. Hypertens Res 2009; 32:735-7. [PMID: 19662021 DOI: 10.1038/hr.2009.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Jones ES, Vinh A, McCarthy CA, Gaspari TA, Widdop RE. AT2 receptors: functional relevance in cardiovascular disease. Pharmacol Ther 2008; 120:292-316. [PMID: 18804122 PMCID: PMC7112668 DOI: 10.1016/j.pharmthera.2008.08.009] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 08/07/2008] [Indexed: 12/24/2022]
Abstract
The renin angiotensin system (RAS) is intricately involved in normal cardiovascular homeostasis. Excessive stimulation by the octapeptide angiotensin II contributes to a range of cardiovascular pathologies and diseases via angiotensin type 1 receptor (AT1R) activation. On the other hand, tElsevier Inc.he angiotensin type 2 receptor (AT2R) is thought to counter-regulate AT1R function. In this review, we describe the enhanced expression and function of AT2R in various cardiovascular disease settings. In addition, we illustrate that the RAS consists of a family of angiotensin peptides that exert cardiovascular effects that are often distinct from those of Ang II. During cardiovascular disease, there is likely to be an increased functional importance of AT2R, stimulated by Ang II, or even shorter angiotensin peptide fragments, to limit AT1R-mediated overactivity and cardiovascular pathologies.
Collapse
Key Words
- angiotensin ii
- at2 receptor
- at1 receptor
- cardiovascular disease
- ace, angiotensin converting enzyme
- ace2, angiotensin converting enzyme 2
- ang ii, angiotensin ii
- ang iii, angiotensin iii
- ang iv, angiotensin iv
- ang (1–7), angiotensin (1–7)
- atbp50, at2r-binding protein of 50 kda
- atip-1, at2 receptor interacting protein-1
- at1r, angiotensin ii type 1 receptor
- at2r, angiotensin ii type 2 receptor
- at4r, angiotensin ii type 4 receptor
- bk, bradykinin
- bp, blood pressure
- cgmp, cyclic guanine 3′,5′-monophosphate
- ecm, extracellular matrix
- enos, endothelial nitric oxide synthase
- erk-1/2, extracellular-regulated kinases-1,2
- irap, insulin-regulated aminopeptidase
- l-name, ng-nitro-l arginine methyl ester
- lvh, left ventricular hypertrophy
- mapk, mitogen-activated protein kinase
- mcp-1, monocyte chemoattractant protein-1
- mi, myocardial infarction
- mmp, matrix metalloproteinase
- mrna, messenger ribonucleic acid
- nf-κβ, nuclear transcription factor-κβ
- no, nitric oxide
- o2−, superoxide
- pc12w, rat pheochromocytoma cell line
- ras, renin angiotensin system
- ros, reactive oxygen species
- shr, spontaneously hypertensive rat
- timp-1, tissue inhibitor of metalloproteinase-1
- tnfα, tumour-necrosis factor α
- vsmc, vascular smooth muscle cell
- wky, wistar-kyoto rat
Collapse
Affiliation(s)
- Emma S Jones
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | |
Collapse
|
27
|
Oxonic acid-induced hyperuricemia elevates plasma aldosterone in experimental renal insufficiency. J Hypertens 2008; 26:1661-8. [DOI: 10.1097/hjh.0b013e328303205d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Li XC, Zhuo JL. Intracellular ANG II directly induces in vitro transcription of TGF-beta1, MCP-1, and NHE-3 mRNAs in isolated rat renal cortical nuclei via activation of nuclear AT1a receptors. Am J Physiol Cell Physiol 2008; 294:C1034-45. [PMID: 18256274 DOI: 10.1152/ajpcell.00432.2007] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The present study tested the hypothesis that intracellular ANG II directly induces transcriptional effects by stimulating AT(1a) receptors in the nucleus of rat renal cortical cells. Intact nuclei were freshly isolated from the rat renal cortex, and transcriptional responses to ANG II were studied using in vitro RNA transcription assays and semiquantitative RT-PCR. High-power phase-contrast micrographs showed that isolated nuclei were encircled by an intact nuclear envelope and stained strongly by the DNA marker 4',6-diamidino-2-phenylindole, but not by the membrane or endosomal markers. Fluorescein isothiocyanate-labeled ANG II and [(125)I]Val(5)-ANG II binding confirmed the presence of ANG II receptors in the nuclei with a predominance of AT(1) receptors. RT-PCR showed that AT(1a) mRNA expression was threefold greater than AT(1b) receptor mRNAs in these nuclei. In freshly isolated nuclei, ANG II increased in vitro [alpha-(32)P]CTP incorporation in a concentration-dependent manner, and the effect was confirmed by autoradiography and RNA electrophoresis. ANG II markedly increased in vitro transcription of mRNAs for transforming growth factor-beta1 by 143% (P < 0.01), macrophage chemoattractant protein-1 by 89% (P < 0.01), and the sodium and hydrogen exchanger-3 by 110% (P < 0.01). These transcriptional effects of ANG II on the nuclei were completely blocked by the AT(1) receptor antagonist losartan (P < 0.01). By contrast, ANG II had no effects on transcription of angiotensinogen and glyceraldehyde-3-phosphate dehydrogenase mRNAs. Because these transcriptional effects of ANG II in isolated nuclei were induced by ANG II in the absence of cell surface receptor-mediated signaling and completely blocked by losartan, we concluded that ANG II may directly stimulate nuclear AT(1a) receptors to induce transcriptional responses that are associated with tubular epithelial sodium transport, cellular growth and hypertrophy, and proinflammatory cytokines.
Collapse
Affiliation(s)
- Xiao C Li
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | | |
Collapse
|
29
|
Li XC, Zhuo JL. In vivo regulation of AT1a receptor-mediated intracellular uptake of [125I]Val5-ANG II in the kidneys and adrenals of AT1a receptor-deficient mice. Am J Physiol Renal Physiol 2007; 294:F293-302. [PMID: 18045833 DOI: 10.1152/ajprenal.00398.2007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Using type 1a angiotensin receptor (AT1a) receptor-deficient (Agtr1a-/-) mice and in vivo autoradiography, we tested the hypothesis that intracellular uptake of ANG II in the kidney and adrenal glands is primarily mediated by AT1a receptors and that the response is regulated by prevailing endogenous ANG II. After pretreatment of wild-type (Agtr1a+/+) and Agtr1a-/- mice (n = 6-9 each group) with or without captopril (25 mg.kg(-1).day(-1)) or losartan (10 mg.kg(-1).day(-1)) for 2 wk, [125I]Val5-ANG II was infused for 60 min. Intracellular uptake of [125I]Val5-ANG II was determined by quantitative in vivo autoradiography after washout of circulating [125I]Val5-ANG II. Basal intracellular ANG II levels were 65% lower in the kidney (P < 0.001), but plasma ANG II levels were threefold higher, in Agtr1a-/- than wild-type mice (P < 0.01). Although plasma [125I]Val5-ANG II levels were similar, urinary excretion of [125I]Val5-ANG II was fourfold higher in Agtr1a-/- mice (P < 0.001). By contrast, intracellular [125I]Val5-ANG II levels were approximately 80% lower in the kidney and adrenal glands of Agtr1a-/- mice (P < 0.01). Captopril decreased endogenous plasma and renal ANG II levels (P < 0.01) but increased intracellular uptake of [125I]Val5-ANG II in the kidney and adrenal glands of wild-type and Agtr1a-/- mice (P < 0.01). Losartan largely blocked renal and adrenal uptake of [125I]Val5-ANG II in wild-type and Agtr1a-/- mice. Thus 80% of intracellular ANG II uptake in the kidney and adrenal glands is mediated by AT1a receptors, whereas AT1b receptor- and other non-receptor-mediated mechanisms account for 20% of the response. Our results suggest that AT1a receptor-mediated uptake of extracellular ANG II may play a physiological role in the kidney and adrenal glands.
Collapse
|
30
|
Bäcklund T, Lakkisto P, Palojoki E, Grönholm T, Saraste A, Finckenberg P, Mervaala E, Tikkanen I, Laine M. Activation of protective and damaging components of the cardiac renin-angiotensin system after myocardial infarction in experimental diabetes. J Renin Angiotensin Aldosterone Syst 2007; 8:66-73. [PMID: 17703432 DOI: 10.3317/jraas.2007.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
INTRODUCTION Diabetes is associated with prolonged apoptotic cell death of cardiac myocytes and adverse remodelling after myocardial infarction (MI). Because the renin-angiotensin system (RAS) has a major role in the remodelling, we studied whether diabetes is associated with altered regulation of RAS after MI in rats. METHODS Male Wistar rats were randomised to receive either streptozotocin (diabetic group) or citrate buffer (control group) intravenously. MI was produced four weeks later by ligating the left descending coronary artery. The rats were sacrificed 1, 4 and 12 weeks after the operation. Angiotensin-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE 2), angiotensin type 1 and 2 receptors (AT(1)-receptor, AT(2)-receptor), and connective tissue growth factor (CTGF) mRNA expression were determined. RESULTS The expression of both protective and damaging components of RAS increased after MI. However, myocardial ACE 2 and AT(2)-receptor messenger ribonucleic acid (mRNA) expression levels were significantly lower in diabetic compared to non-diabetic rats 1 week after MI. In contrast, AT(1)-receptor, ACE and CTGF mRNA levels were up-regulated in diabetic as compared with non-diabetic rats 12 weeks after MI. CONCLUSION The activation of the protective components of RAS (ACE 2 and AT(2)-receptor) was blunted early after MI in diabetic rats, whereas the levels of ACE, AT(1)-receptor and CTGF mRNA leading to adverse effects on myocardium, were elevated in diabetic as compared with non-diabetic rats. This unbalanced activation of the RAS may influence the pathophysiology of myocardial injury in diabetes after MI.
Collapse
Affiliation(s)
- Tom Bäcklund
- Minerva Foundation Institute for Medical Research, Biomedicum, Helsinki, Haartmaninkatu 8, FIN-00029 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kobori H, Nangaku M, Navar LG, Nishiyama A. The intrarenal renin-angiotensin system: from physiology to the pathobiology of hypertension and kidney disease. Pharmacol Rev 2007; 59:251-87. [PMID: 17878513 DOI: 10.1124/pr.59.3.3] [Citation(s) in RCA: 886] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In recent years, the focus of interest on the role of the renin-angiotensin system (RAS) in the pathophysiology of hypertension and organ injury has changed to a major emphasis on the role of the local RAS in specific tissues. In the kidney, all of the RAS components are present and intrarenal angiotensin II (Ang II) is formed by independent multiple mechanisms. Proximal tubular angiotensinogen, collecting duct renin, and tubular angiotensin II type 1 (AT1) receptors are positively augmented by intrarenal Ang II. In addition to the classic RAS pathways, prorenin receptors and chymase are also involved in local Ang II formation in the kidney. Moreover, circulating Ang II is actively internalized into proximal tubular cells by AT1 receptor-dependent mechanisms. Consequently, Ang II is compartmentalized in the renal interstitial fluid and the proximal tubular compartments with much higher concentrations than those existing in the circulation. Recent evidence has also revealed that inappropriate activation of the intrarenal RAS is an important contributor to the pathogenesis of hypertension and renal injury. Thus, it is necessary to understand the mechanisms responsible for independent regulation of the intrarenal RAS. In this review, we will briefly summarize our current understanding of independent regulation of the intrarenal RAS and discuss how inappropriate activation of this system contributes to the development and maintenance of hypertension and renal injury. We will also discuss the impact of antihypertensive agents in preventing the progressive increases in the intrarenal RAS during the development of hypertension and renal injury.
Collapse
Affiliation(s)
- Hiroyuki Kobori
- Department of Medicine, Director of the Molecular Core in Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112-2699, USA.
| | | | | | | |
Collapse
|
32
|
Vanecková I, Kopkan L, Husková Z, Vanourková Z, Schejbalová S, Cervenka L, Kramer HJ. Long-Term Prevention of Hypertension and End-Organ Damage in Ren-2 Transgenic Rats Is Achieved Only with Persistent but Not Transient AT 1 Receptor Blockade. Kidney Blood Press Res 2007; 30:38-44. [PMID: 17237618 DOI: 10.1159/000098869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Accepted: 11/28/2006] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was first to evaluate the effects of persistent or transient blockade of the angiotensin II (ANG II) receptor AT(1) on the development of hypertension and end-organ damage in hypertensive Ren-2 transgenic rats (TGR), and second to assess the potential role of AT(2) receptors in the control of blood pressure (BP) in this monogenetic model of hypertension. Male heterozygous TGR and Hannover Sprague-Dawley (HanSD) rats fed a normal salt diet were treated from day 32 of age either persistently until the end of the experiment (day 100 of age) or transiently until day 56 of age with the selective AT(1) receptor antagonist candesartan or with the combination of candesartan and the AT(2) receptor antagonist PD 123319. Persistent treatment with candesartan completely prevented the rise in BP, proteinuria and the increase in left ventricular weight/body weight ratio, whereas transient treatment with candesartan was effective only as long as the drug was administered. In the presence of candesartan, PD 123319 was without effect. Our results show that in male heterozygous TGR persistent candesartan treatment completely prevented hypertension and end-organ damage as long as the drug was administered, whereas transient AT(1 )receptor blockade had no long-term effects.
Collapse
MESH Headings
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Animals, Genetically Modified
- Benzimidazoles/pharmacology
- Biphenyl Compounds
- Blood Pressure
- Hypertension, Renal/drug therapy
- Kidney/physiopathology
- Male
- Proteinuria/urine
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/blood
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Angiotensin, Type 2/physiology
- Renin/genetics
- Tetrazoles/pharmacology
Collapse
Affiliation(s)
- Ivana Vanecková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
33
|
Hayden MR, Chowdhury NA, Cooper SA, Whaley-Connell A, Habibi J, Witte L, Wiedmeyer C, Manrique CM, Lastra G, Ferrario C, Stump C, Sowers JR. Proximal tubule microvilli remodeling and albuminuria in the Ren2 transgenic rat. Am J Physiol Renal Physiol 2006; 292:F861-7. [PMID: 17032939 DOI: 10.1152/ajprenal.00252.2006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TG(mRen2)27 (Ren2) transgenic rats overexpress the mouse renin gene, with subsequent elevated tissue ANG II, hypertension, and nephropathy. The proximal tubule cell (PTC) is responsible for the reabsorption of 5-8 g of glomerular filtered albumin each day. Excess filtered albumin may contribute to PTC damage and tubulointerstitial disease. This investigation examined the role of ANG II-induced oxidative stress in PTC structural remodeling: whether such changes could be modified with in vivo treatment with ANG type 1 receptor (AT(1)R) blockade (valsartan) or SOD/catalase mimetic (tempol). Male Ren2 (6-7 wk old) and age-matched Sprague-Dawley rats were treated with valsartan (30 mg/kg), tempol (1 mmol/l), or placebo for 3 wk. Systolic blood pressure, albuminuria, N-acetyl-beta-D-glucosaminidase, and kidney tissue malondialdehyde (MDA) were measured, and x60,000 transmission electron microscopy images were used to assess PTC microvilli structure. There were significant differences in systolic blood pressure, albuminuria, lipid peroxidation (MDA and nitrotyrosine staining), and PTC structure in Ren2 vs. Sprague-Dawley rats (each P < 0.05). Increased mean diameter of PTC microvilli in the placebo-treated Ren2 rats (P < 0.05) correlated strongly with albuminuria (r(2) = 0.83) and moderately with MDA (r(2) = 0.49), and there was an increase in the ratio of abnormal forms of microvilli in placebo-treated Ren2 rats compared with Sprague-Dawley control rats (P < 0.05). AT(1)R blockade, but not tempol treatment, abrogated albuminuria and N-acetyl-beta-d-glucosaminidase; both therapies corrected abnormalities in oxidative stress and PTC microvilli remodeling. These data indicate that PTC structural damage in the Ren2 rat is related to the oxidative stress response to ANG II and/or albuminuria.
Collapse
Affiliation(s)
- Melvin R Hayden
- Department of Internal Medicine, University of Missouri, Columbia, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Whaley-Connell AT, Chowdhury NA, Hayden MR, Stump CS, Habibi J, Wiedmeyer CE, Gallagher PE, Tallant EA, Cooper SA, Link CD, Ferrario C, Sowers JR. Oxidative stress and glomerular filtration barrier injury: role of the renin-angiotensin system in the Ren2 transgenic rat. Am J Physiol Renal Physiol 2006; 291:F1308-14. [PMID: 16788142 DOI: 10.1152/ajprenal.00167.2006] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TG(mRen2)27 (Ren2) transgenic rats overexpress the mouse renin gene, manifest hypertension, and exhibit increased tissue ANG II levels and oxidative stress. Evidence indicates that elevated tissue ANG II contributes to oxidative stress, increases in glomerular macromolecular permeability, and consequent albuminuria. Furthermore, angiotensin type 1 receptor (AT1R) blockers reduce albuminuria and slow progression of renal disease. However, it is not known whether improvements in glomerular filtration barrier integrity and albuminuria during treatment are related to reductions in oxidative stress and/or kidney renin-angiotensin system (RAS) activity. To investigate the renal protective effects of AT1R blockade, we treated young (6-7 wk old) male Ren2 rats with valsartan (Ren2-V; 30 mg/kg) for 3 wk and measured urine albumin, kidney malondialdehyde (MDA), RAS component mRNAs, and NADPH oxidase subunits (gp91(phox) and Rac1) compared with age-matched untreated Ren2 and Sprague-Dawley (S-D) rats. Basement membrane thickness, slit pore diameter and number, and foot process base width were measured by transmission electron microscopy (TEM). Results indicate that AT1R blockade lowered systolic blood pressure (30%), albuminuria (91%), and kidney MDA (80%) in Ren2-V compared with untreated Ren2 rats. Increased slit pore number and diameter and reductions in basement membrane thickness and podocyte foot process base width were strongly associated with albuminuria and significantly improved following AT1R blockade. AT1R blockade was also associated with increased angiotensin-converting enzyme-2 and neprilysin expression, demonstrating a beneficial shift in balance of renal RAS. Thus reductions in blood pressure, albuminuria, and tissue oxidative stress with AT1R blockade were associated with improved indexes of glomerular filtration barrier integrity and renal RAS in Ren2 rats.
Collapse
Affiliation(s)
- Adam T Whaley-Connell
- Department of Internal Medicine, University of Missouri School of Medicine, 1 Hospital Dr., MA410 Medical Science Bldg., Columbia, MO 65212, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Li XC, Carretero OA, Zhuo JL. Cross-talk between angiotensin II and glucagon receptor signaling mediates phosphorylation of mitogen-activated protein kinases ERK 1/2 in rat glomerular mesangial cells. Biochem Pharmacol 2006; 71:1711-9. [PMID: 16643859 PMCID: PMC2276839 DOI: 10.1016/j.bcp.2006.03.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Revised: 03/16/2006] [Accepted: 03/16/2006] [Indexed: 12/26/2022]
Abstract
We have recently shown that the pancreatic hormone glucagon-induced phosphorylation of mitogen-activated protein (MAP) kinase ERK 1/2 as well as growth and proliferation of rat glomerular mesangial cells (MCs) via activation of cAMP-dependent protein kinase A (PKA)- and phospholipase C (PLC)/Ca2+-mediated signaling pathways. Since circulating glucagon and tissue angiotensin II (Ang II) levels are inappropriately elevated in type 2 diabetes, we tested the hypothesis that glucagon induces phosphorylation of ERK 1/2 in MCs by interacting with Ang II receptor signaling. Stimulation of MCs by glucagon (10 nM) induced a marked increase in intracellular [Ca2+]i that was abolished by [Des-His1, Glu9]-glucagon (1 microM), a selective glucagon receptor antagonist. Both glucagon and Ang II-induced ERK 1/2 phosphorylation (glucagon: 214+/-14%; Ang II: 174+/-16%; p<0.001 versus control), and these responses were inhibited by the AT1 receptor blocker losartan (glucagon + losartan: 77+/-14%; Ang II + losartan: 84+/-18%; p<0.01 versus glucagon or Ang II) and the AT2 receptor blocker PD 123319 (glucagon + PD: 78+/-7%; Ang II + PD: 87+/-7%; p<0.01 versus glucagon or Ang II). Inhibition of cAMP-dependent PKA with H89 (1 microM) or PLC with U73122 (1 microM) also markedly attenuated the phosphorylation of ERK 1/2 induced by glucagon (glucagon + U73122: 109+/-15%; glucagon + H89: 113+/-16%; p<0.01 versus glucagon) or Ang II (Ang II + U73122: 111+/-13%; Ang II + H89: 86+/-10%; p<0.01 versus Ang II). Wortmannin (1 microM), a selective PI 3-kinase inhibitor, also blocked glucagon- or Ang II-induced ERK 1/2 phosphorylation. These results suggest that AT1 receptor-activated cAMP-dependent PKA, PLC and PI 3-kinase signaling is involved in glucagon-induced MAP kinase ERK 1/2 phosphorylation in MCs. The inhibitory effect of PD 123319 on glucagon-induced ERK 1/2 phosphorylation further suggests that AT2 receptors also play a similar role in this response.
Collapse
Affiliation(s)
- Xiao C. Li
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Oscar A. Carretero
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Jia L. Zhuo
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- * Corresponding author at: Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA. Tel.: +1 313 916 4958; fax: +1 313 916 1479. E-mail address: (J.L. Zhuo)
| |
Collapse
|
36
|
Li XC, Carretero OA, Navar LG, Zhuo JL. AT1 receptor-mediated accumulation of extracellular angiotensin II in proximal tubule cells: role of cytoskeleton microtubules and tyrosine phosphatases. Am J Physiol Renal Physiol 2006; 291:F375-83. [PMID: 16478976 PMCID: PMC2276851 DOI: 10.1152/ajprenal.00405.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Long-term angiotensin II (ANG II) administration is associated with increased ANG II accumulation in the kidney, but intrarenal compartment(s) involved in this response remains to be determined. We tested the hypothesis that 1) extracellular ANG II is taken up by proximal tubule cells (PTCs) through AT(1) receptor-mediated endocytosis, 2) this process is regulated by cytoskeleton microtubule- and tyrosine phosphatase-dependent mechanisms, and 3) AT(1) receptor-mediated endocytosis of ANG II has a functional relevance by modulating intracellular cAMP signaling. In cultured PTCs, [(125)I]Tyr-labeled ANG II and fluorescein labeled-ANG II were internalized in a time-dependent manner and colocalized with the endosome marker Alexa Fluor 594-transferrin. Endocytosis of extracellular ANG II was inhibited by the AT(1) receptor blocker losartan (16.5 +/- 4.6%, P < 0.01 vs. ANG II, 78.3 +/- 6.2%) and by the tyrosine phosphatase inhibitor phenylarsine oxide (PAO; 30.0 +/- 3.5%, P < 0.05 vs. ANG II). Intracellular ANG II levels were increased by approximately 58% (basal, 229.8 +/- 11.4 vs. ANG II, 361.3 +/- 11.8 pg ANG II/mg protein, P < 0.01), and the responses were blocked by losartan (P < 0.01), the cytoskeleton microtubule inhibitor colchicine (P < 0.05), and PAO (P < 0.01), whereas depletion of clathrin-coated pits with hyperosmotic sucrose had no effect (356.1 +/- 25.5 pg ANG II/mg protein, not significant). ANG II accumulation was associated with significant inhibition of both basal (control, 15.5 +/- 2.8 vs. ANG II, 9.1 +/- 2.4 pmol/mg protein, P < 0.05) and forskolin-stimulated cAMP signaling (forskolin, 68.7 +/- 8.6 vs. forskolin + ANG II, 42.8 +/- 13.8 pmol/mg protein, P < 0.01). These effects were blocked by losartan and PAO. We conclude that extracellular ANG II is internalized in PTCs through AT(1) receptor-mediated endocytosis and that internalized ANG II may play a functional role in proximal tubule cells by inhibiting intracellular cAMP signaling.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Arsenicals/pharmacology
- Cells, Cultured
- Colchicine/pharmacology
- Colforsin/pharmacology
- Cyclic AMP/physiology
- Cytoskeleton/drug effects
- Cytoskeleton/metabolism
- Endocytosis/physiology
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation/physiology
- Imidazoles/pharmacology
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/metabolism
- Losartan/pharmacology
- Microtubules/drug effects
- Microtubules/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatases/antagonists & inhibitors
- Protein Tyrosine Phosphatases/physiology
- Pyridines/pharmacology
- Rabbits
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/analysis
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/physiology
- Signal Transduction/physiology
- Sucrose/pharmacology
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit 48202, USA
| | | | | | | |
Collapse
|
37
|
Pendergrass KD, Averill DB, Ferrario CM, Diz DI, Chappell MC. Differential expression of nuclear AT1 receptors and angiotensin II within the kidney of the male congenic mRen2. Lewis rat. Am J Physiol Renal Physiol 2006; 290:F1497-506. [PMID: 16403834 DOI: 10.1152/ajprenal.00317.2005] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We established a new congenic model of hypertension, the mRen(2). Lewis rat and assessed the intracellular expression of angiotensin peptides and receptors in the kidney. The congenic strain was established from the backcross of the (mRen2)27 transgenic rat that expresses the mouse renin 2 gene onto the Lewis strain. The 20-wk-old male congenic rats were markedly hypertensive compared with the Lewis controls (systolic blood pressure: 195 +/- 2 vs. 107 +/- 2 mmHg, P < 0.01). Although plasma ANG II levels were not different between strains, circulating levels of ANG-(1-7) were 270% higher and ANG I concentrations were 40% lower in the mRen2. Lewis rats. In contrast, both cortical (CORT) and medullary (MED) ANG II concentrations were 60% higher in the mRen2. Lewis rats, whereas tissue ANG I was 66 and 84% lower in CORT and MED. For both strains, MED ANG II, ANG I, and ANG-(1-7) were significantly higher than CORT levels. Intracellular ANG II binding distinguished nuclear (NUC) and plasma membrane (PM) receptor using the ANG II radioligand 125I-sarthran. Isolated CORT nuclei exhibited a high density (Bmax >200 fmol/mg protein) and affinity for the sarthran ligand (KD<0.5 nM); the majority of these sites (>95%) were the AT1 receptor subtype. CORT ANG II receptor Bmax and KD values in nuclei were 75 and 50% lower, respectively, for the mRen2. Lewis vs. the Lewis rats. In the MED, the PM receptor density (Lewis: 50 +/- 4 vs. mRen2. Lewis: 21 +/- 5 fmol/mg protein) and affinity (Lewis: 0.31 +/- 0.1 vs. 0.69 +/- 0.1 nM) were lower in the mRen2. Lewis rats. In summary, the hypertensive mRen2. Lewis rats exhibit higher ANG II in both CORT and MED regions of the kidney. Evaluation of intracellular ANG II receptors revealed lower CORT NUC and MED PM AT1 sites in the mRen2. Lewis. The downregulation of AT1 sites in the mRen2. Lewis rats may reflect a compensatory response to dampen the elevated levels of intrarenal ANG II.
Collapse
Affiliation(s)
- Karl D Pendergrass
- Hypertension and Vascular Disease Ctr., Wake Forest Univ. Health Sciences, Medical Center Blvd., Winston-Salem, NC 27157-1095, USA
| | | | | | | | | |
Collapse
|
38
|
Li XC, Campbell DJ, Ohishi M, Yuan S, Zhuo JL. AT1 receptor-activated signaling mediates angiotensin IV-induced renal cortical vasoconstriction in rats. Am J Physiol Renal Physiol 2005; 290:F1024-33. [PMID: 16380463 PMCID: PMC2276856 DOI: 10.1152/ajprenal.00221.2005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Angiotensin IV (ANG IV), an active ANG II fragment, has been shown to induce systemic and renal cortical effects by binding to ANG IV (AT(4)) receptors and activating unique signaling transductions unrelated to classical type 1 (AT(1)) or type 2 (AT(2)) receptors. We tested whether ANG IV exerts systemic and renal cortical effects on blood pressure, renal microvascular smooth muscle cells (VSMCs), and glomerular mesangial cells (MC) and, if so, whether AT(1) receptor-activated signaling is involved. In anesthetized rats, systemic infusion of ANG II, ANG III, or ANG IV (0.01, 0.1, and 1.0 nmol.kg(-1).min(-1) iv) caused dose-dependent increases in mean arterial pressure (MAP) and decreases in renal cortical blood flow (CBF; P < 0.01). ANG II also induced dose-dependent reductions in renal medullary blood flow (P < 0.01), whereas ANG IV did not. ANG IV-induced pressor and renal cortical vasoconstriction were completely abolished by AT(1) receptor blockade with losartan (5 mg/kg iv; P < 0.05). When ANG IV (1 nmol.kg(-1).min(-1)) was infused directly in the renal artery, CBF was reduced by >30%, and the response was also blocked by losartan (P < 0.01). In the renal cortex, unlabeled ANG IV displaced (125)I-labeled [Sar(1),Ile(8)]ANG II binding, whereas unlabeled ANG II (10 microM) inhibited (125)I-labeled Nle(1)-ANG IV (AT(4)) binding in a concentration-dependent manner (P < 0.01). In freshly isolated renal VSMCs, ANG IV (100 nM) increased intracellular Ca(2+) concentration, and the effect was blocked by losartan and U-73122, a selective inhibitor of phospholipase C/inositol trisphosphate/Ca(2+) signaling (1 microM). In cultured rat MCs, ANG IV (10 nM) induced mitogen-activated protein kinase extracellular/signal-regulated kinase 1/2 phosphorylation via AT(1) receptor- and phospholipase C-activated signaling. These results suggest that, at nanomolar concentrations, ANG IV can increase MAP and induce renal cortical effects by interacting with AT(1) receptor-activated signaling.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA
| | | | | | | | | |
Collapse
|
39
|
Zhuo JL, Li XC, Garvin JL, Navar LG, Carretero OA. Intracellular ANG II induces cytosolic Ca2+ mobilization by stimulating intracellular AT1 receptors in proximal tubule cells. Am J Physiol Renal Physiol 2005; 290:F1382-90. [PMID: 16380461 PMCID: PMC2276850 DOI: 10.1152/ajprenal.00269.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intracellular ANG II induces biological effects in nonrenal cells, but it is not known whether it plays a physiological role in renal proximal tubule cells (PTCs). PTCs express angiotensinogen, renin, and angiotensin-converting enzyme mRNAs, suggesting the presence of high levels of intracellular ANG II. We determined if microinjection of ANG II directly in single PTCs increases intracellular calcium concentration ([Ca2+]i) and, if so, elucidated the cellular mechanisms involved. Changes in [Ca2+]i responses were studied by fluorescence imaging using the Ca2+ indicator fluo 3. ANG II (1 nM) was microinjected directly in the cells, whereas cell-surface angiotensin type 1 (AT1) receptors were blocked by losartan (10 microM). When ANG II (1 nM) was added to the perfusate, there was a marked increase in [Ca2+]i that was blocked by extracellular losartan. With losartan in the perfusate, intracellular microinjection of ANG II elicited a robust increase in cytoplasmic [Ca2+]i that peaked at 30 s (basal: 2.2 +/- 0.3 vs. ANG II: 14.9 +/- 0.4 relative fluorescence units; P < 0.01). Chelation of extracellular Ca2+ with EGTA (2 mM) did not alter microinjected ANG II-induced [Ca2+]i responses (Ca2+ free + ANG II: 12.3 +/- 2.6 relative fluorescence units, not significant vs. ANG II); however, pretreatment with thapsigargin to deplete intracellular Ca2+ stores or with U-73122 to inhibit phospholipase C (1 microM each) markedly attenuated microinjected ANG II-induced [Ca2+]i responses. Combined microinjection of ANG II and losartan abolished [Ca2+]i responses, whereas a combination of ANG II and PD-123319 had no effect. These data demonstrate for the first time that direct microinjection of ANG II in single PTCs increases [Ca2+]i by stimulating intracellular AT1 receptors and releases Ca2+ from intracellular stores, suggesting that intracellular ANG II may play a physiological role in PTC function.
Collapse
MESH Headings
- Angiotensin II/analysis
- Angiotensin II/pharmacology
- Animals
- Blotting, Western
- Calcium/metabolism
- Calcium/pharmacology
- Cells, Cultured
- Cytosol/metabolism
- Enzyme Activation/drug effects
- Kidney Tubules, Proximal/chemistry
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/ultrastructure
- Microinjections
- Rabbits
- Receptor, Angiotensin, Type 1/analysis
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/physiology
- Thapsigargin/pharmacology
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Jia L Zhuo
- Division of Hypertension and Vascular Research, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA.
| | | | | | | | | |
Collapse
|
40
|
Pörsti I, Fan M, Kööbi P, Jolma P, Kalliovalkama J, Vehmas TI, Helin H, Holthöfer H, Mervaala E, Nyman T, Tikkanen I. High calcium diet down-regulates kidney angiotensin-converting enzyme in experimental renal failure. Kidney Int 2005; 66:2155-66. [PMID: 15569305 DOI: 10.1111/j.1523-1755.2004.66006.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Calcium salts are used as phosphate binders in renal failure, while high calcium diet also improves vasorelaxation and enhances natriuresis. The influences of calcium intake on renal renin-angiotensin system (RAS) are largely unknown. METHODS Four weeks after NTX, rats were put on 3.0% or 0.3% calcium diet for 8 weeks (12-week study). In additional experiments, 15 weeks after NTX, rats were put on similar diets for 12 weeks (27-week study). Appropriate blood, urine, and kidney samples were taken. Renal angiotensin-converting enzyme (ACE) and angiotensin II receptors (AT1, AT2) were examined using autoradiography, ACE also using Western blotting, and connective tissue growth factor (CTGF) using immunohistochemistry. RESULTS In the 12-week study, albuminuria increased 5-fold in NTX rats, but only 2-fold in calcium NTX rats on 3.0% calcium. In the 27-week study, high calcium intake decreased blood pressure, retarded progression of renal failure, reduced glomerulosclerosis, interstitial damage, and aortic calcifications, and improved survival from 50% to 92% in NTX rats. In both experiments plasma parathyroid hormone and phosphate were elevated after NTX, and suppressed by high calcium diet, while kidney ACE was down-regulated by 40% or more after increased calcium intake. In the 27-week study renal CTGF was decreased and cortical AT1 receptor density reduced after high calcium diet. CONCLUSION High calcium diet down-regulated kidney ACE, reduced albuminuria and blood pressure, and favorably influenced kidney morphology in experimental renal failure. These findings suggest a link between calcium metabolism and kidney ACE expression, which may play a role in the progression of renal damage.
Collapse
Affiliation(s)
- Ilkka Pörsti
- Department of Internal Medicine, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ichihara A, Hayashi M, Hirota N, Okada H, Koura Y, Tada Y, Kaneshiro Y, Tsuganezawa H, Saruta T. Angiotensin II type 2 receptor inhibits prorenin processing in juxtaglomerular cells. Hypertens Res 2004; 26:915-21. [PMID: 14714584 DOI: 10.1291/hypres.26.915] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Long-term treatment with an angiotensin II type 1 receptor blocker (ARB) has been shown to decrease the plasma renin activity (PRA) of hypertensive patients, whereas PRA remains elevated during angiotensin-converting enzyme inhibitor (ACEI) treatment. In the present study, we used rat juxtaglomerular (JG) cells to elucidate the mechanism(s) involved in the differential regulation of PRA between ARB and ACEI treatment. Addition of 100 nmol/l angiotensinogen (Aogen) to JG cells (n=6 primary cultures) significantly increased the medium angiotensin (Ang) II levels from 14 +/- 2 to 440 +/- 9 pg/ml and suppressed the renin secretion rate (RSR) from 39.6 +/- 5.4% to 6.3 +/- 1.8% without affecting active renin content (ARC) or total renin content (TRC). In the Aogen-treated cells, the ACEI, delapril hydrochloride (CV3317, 10 micromol/l), significantly decreased the medium Ang II levels to 58 +/- 14 pg/ml and increased RSR to 39.8 +/- 4.1% without affecting ARC or TRC. The ARB, an active metabolite of candesartan cilexetil (CV11974, 10 micromol/l), however, significantly increased the medium Ang II levels and RSR to 486 +/- 15 pg/ml and 40.9 +/- 9.8%, respectively, and decreased ARC from 63.2 +/- 6.8 to 21.6 +/- 3.6 ng of Ang l x h(-1) x million cells(-1) without affecting TRC. The decreases in ARC of the Aogen+CV11974-treated cells (n=6 primary cultures) were inhibited by an Ang II type 2 receptor blocker, PD123319 (10 micromol/l). JG cells (n=6 primary cultures) were also treated with an Ang II type 2 receptor agonist, CGP42212A (0.1 micromol/l). CGP42212A significantly increased RSR from 38.2 +/- 1.6% to 49.7 +/- 4.7% and decreased ARC from 60.8 +/- 3.0 to 25.3 +/- 2.8 ng of Ang l x h(-1) million cells(-1) without affecting TRC. Addition of CV11974 did not alter the RSR, ARC, or TRC of the CGP42212A-treated cells; however, PD123319 abolished the effects of CGP42212A. These results indicate that, distinct from ACEIs, ARBs inhibit prorenin processing of JG cells through Ang II type 2 receptors. Long-term treatment with an ARB may decrease PRA in part by diminishing the storage of active renin in JG cells.
Collapse
Affiliation(s)
- Atsuhiro Ichihara
- Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Elevations in intrarenal angiotensin II (Ang II) cause reductions in renal function and sodium excretion that contribute to progressive hypertension and lead to renal and vascular injury. Augmentation of intrarenal Ang II occurs by several processes, leading to levels much greater than can be explained from the circulating levels. In Ang II-dependent hypertension, Ang II is internalized via an AT1 receptor mechanism, but there is also sustained intrarenal production of Ang II. Ang II exerts a positive feedback action on intrarenal angiotensinogen (AGT) mRNA and protein. The increased intrarenal AGT production is associated with increased intrarenal and intracellular Ang II contents and urinary AGT excretion rates. The increased urinary AGT indicates spillover of AGT into distal nephron segments supporting enhanced distal Ang II formation and sodium reabsorption. The augmentation of intrarenal Ang II provides the basis for sustained actions on renal function, sodium excretion, and maintenance of hypertension.
Collapse
Affiliation(s)
- L Gabriel Navar
- Department of Physiology SL39, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
43
|
Lemos VS, Côrtes SF, Silva DMR, Campagnole-Santos MJ, Santos RAS. Angiotensin-(1-7) is involved in the endothelium-dependent modulation of phenylephrine-induced contraction in the aorta of mRen-2 transgenic rats. Br J Pharmacol 2002; 135:1743-8. [PMID: 11934815 PMCID: PMC1573295 DOI: 10.1038/sj.bjp.0704630] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The contribution of the local vascular production of angiotensin-(1-7) [Ang-(1-7)] to the control of alpha-adrenergic-induced contractions in the aorta of Sprague-Dawley (SD) and TGR(mRen-2)27 [mRen-2] rats was studied. 2. In mRen-2 rats, contractile responses to phenylephrine were diminished as compared to control SD rats in endothelium containing but not in endothelium-denuded vessels. L-NAME increased contractile responses to phenylephrine in mRen-2 rats and, after nitric oxide synthase blockade, responses to phenylephrine became comparable in both strains. 3. Inhibition of angiotensin-converting enzyme (ACE) by captopril potentiated contractile responses in mRen-2 rats and diminished contractile responses in SD rats, both effects being dependent on the presence of a functional endothelium. The effect of captopril in mRen-2 rats was abolished in vessels pre-incubated with Ang-(1-7). 4. Blockade of Ang-(1-7) and bradykinin (BK) receptors by A-779 and HOE 140 respectively, increased phenylephrine-induced contraction in mRen-2, but not in SD rats. This effect was seen only in endothelium-containing vessels. 5. Angiotensin II AT(1) and AT(2) receptor blockade by CV 11974 and PD 123319 did not affect the contractile responses to phenylephrine in aortas of transgenic animals but diminished the response in SD rats. This effect was only seen in the presence of a functional endothelium. 6. It is concluded that the decreased contractile responses to phenylephrine in aortas of mRen-2 rats was dependent on an intact endothelium, the local release and action of Ang-(1-7) and bradykinin.
Collapse
MESH Headings
- Analysis of Variance
- Angiotensin I/pharmacology
- Angiotensin I/physiology
- Angiotensin Receptor Antagonists
- Animals
- Animals, Genetically Modified
- Antihypertensive Agents/pharmacology
- Aorta/drug effects
- Aorta/physiology
- Bradykinin Receptor Antagonists
- Captopril/pharmacology
- Drug Interactions
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- In Vitro Techniques
- Male
- Mice
- Peptide Fragments/pharmacology
- Peptide Fragments/physiology
- Phenylephrine/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Adrenergic, alpha/physiology
- Receptors, Angiotensin/physiology
- Receptors, Bradykinin/physiology
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
Collapse
Affiliation(s)
- Virgínia S Lemos
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | | | | | | |
Collapse
|
44
|
Harrison-Bernard LM, Zhuo J, Kobori H, Ohishi M, Navar LG. Intrarenal AT(1) receptor and ACE binding in ANG II-induced hypertensive rats. Am J Physiol Renal Physiol 2002; 282:F19-25. [PMID: 11739108 PMCID: PMC2575663 DOI: 10.1152/ajprenal.0335.2000] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The intrarenal expression of angiotensin II (ANG II) type 1 (AT(1)) receptors and angiotensin-converting enzyme (ACE) was determined in ANG II-induced hypertensive rats (80 ng/min; 2 wk). Systolic blood pressure averaged 184 +/- 3 and 125 +/- 1 mmHg in ANG II-infused compared with Sham rats on day 12. Total kidney AT(1) receptor protein levels were not altered significantly. AT(1) receptor binding mapped by quantitative in vitro autoradiography was significantly decreased in glomeruli (172 +/- 25 vs. 275 +/- 34 disintegrations. min(-1). mm(-2)) and the inner stripe of the outer medulla (121 +/- 17 vs. 178 +/- 19 disintegrations. min(-1). mm(-2)), but not proximal convoluted tubules (48 +/- 9 vs. 58 +/- 6 disintegrations. min(-1). mm(-2)) of ANG II-infused compared with Sham rats. Proximal tubule ACE binding was significantly augmented (132 +/- 4 vs. 97 +/- 3 disintegrations. min(-1). mm(-2)) in ANG II-infused rats. In summary, during ANG II-induced hypertension, glomeruli and inner stripe of the outer medulla have reduced AT(1) receptor binding. Proximal convoluted tubules exhibit maintained AT(1) receptor density and increased ACE binding, which together with the elevated ANG II levels suggest that ANG II exerts a sustained influence on tubular reabsorption and consequently contributes to the development and maintenance of ANG II-dependent hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Autoradiography
- Blood Pressure
- Blotting, Western
- Hypertension, Renal/chemically induced
- Hypertension, Renal/metabolism
- Kidney Glomerulus/chemistry
- Kidney Glomerulus/metabolism
- Kidney Medulla/chemistry
- Kidney Medulla/metabolism
- Kidney Tubules, Proximal/chemistry
- Kidney Tubules, Proximal/metabolism
- Male
- Osmotic Pressure
- Peptidyl-Dipeptidase A/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Angiotensin/analysis
- Receptors, Angiotensin/metabolism
- Renin/blood
- Vasoconstrictor Agents
Collapse
Affiliation(s)
- Lisa M Harrison-Bernard
- Department of Physiology, Tulane University Health Sciences Center, New Orleans, Louisiana 70112, USA.
| | | | | | | | | |
Collapse
|
45
|
Uhlenius N, Vuolteenaho O, Tikkanen I. Renin-angiotensin blockade improves renal cGMP production via non-AT(2)-receptor mediated mechanisms in hypertension-induced by chronic NOS inhibition in rat. J Renin Angiotensin Aldosterone Syst 2001; 2:233-9. [PMID: 11881129 DOI: 10.3317/jraas.2001.037] [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: 11/01/2022] Open
Abstract
BACKGROUND To investigate the changes in the angiotensin II (Ang II) receptors and nitric oxide (NO)-cGMP pathway in the rat kidney after nitric oxide synthase (NOS)blockade. METHODS Captopril, an angiotensin-converting enzyme (ACE)inhibitor, 20 mg/100 ml; and/or L-158,809 (an Ang II AT1-receptor antagonist, 5 mg/100 ml) and L-NAME (NOS inhibitor, 50 mg/100 ml) were administered orally for 12 weeks. Blood pressure (BP),urinary albumin, urinary cGMP excretion, plasma ANP, and plasma renin activity were measured. In vitro autoradiography was used to locate the Ang II receptors in the kidney. RESULTS Captopril and L- 158,809 treatments normalised BP and prevented the appearance of albuminuria in rats receiving L-NAME. Urinary cGMP excretion was significantly increased in L-158,809-treated rats compared with the non-treated group, suggesting that the dysfunctional NO system may be activated by the treatment. AT1-receptor binding in the kidney was inhibited to about 40% of the control value after administration of L- 158,809. The AT2-receptor binding was inhibited to less than 15% of the control value. NOS inhibition had no effect on receptor binding. CONCLUSION Blockade of NOS causes hypertension and renal damage. Treatment with an ACE inhibitor and/or Ang II receptor antagonist prevented these changes equally effectively. The stimulatory effect of AT1-receptor antagonism on cGMP production was not mediated by AT2-receptor-dependent mechanisms, since renalAT2-receptor binding density was suppressed following treatment with L-158,809. AT1-receptor blockade per se favours activation of humoral pathways that stimulate cGMP production potentially contributing to renal and vascular protection in hypertension and chronic renal disease.
Collapse
Affiliation(s)
- N Uhlenius
- Minerva Foundation Institute for Medical Research, Biomedicum Helsink, Helsinki, Finland.
| | | | | |
Collapse
|
46
|
Ohishi M, Ueda M, Tanabe S, Han YS, Mendelsohn FA, Zhuo JL. Increased angiotensin-converting enzyme and type 1 angiotensin receptors in cortical vasculature and tubulointerstitium of chronically rejected human kidney allografts. Nephrology (Carlton) 2001. [DOI: 10.1046/j.1440-1797.2001.00045.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Lemmer B, Rueff T, Reiter S, Huser L, Hauptfleisch S, Witte K. Influence of circadian time and age on glomerular angiotensin II receptors in normotensive Sprague-Dawley and transgenic hypertensive TGR(mREN2)27 rats. Chronobiol Int 2001; 18:447-59. [PMID: 11475415 DOI: 10.1081/cbi-100103968] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In male heterozygous transgenic hypertensive rats, TGR(mREN2)27 (TGR), exhibiting an inverse blood pressure profile and in normotensive Sprague-Dawley (SPRD) controls, the density and affinity of angiotensin II receptors were determined at six circadian times in glomeruli of animals 11 weeks old kept under light-dark 12h:12 (LD 12:12) conditions. Angiotensin II receptors were also studied in rats 18-20 weeks old of both strains at 2h after light onset. As a measure of renal excretory functions, diuresis, creatinine, and protein excretion were monitored using metabolic cages. The expression of angiotensin II receptor mRNA was determined in renal arteries 2h-4h after light onset. The following results were obtained: (1) Renal excretory functions showed significant daily variation, with higher excretion rates in the dark span in both TGR and SPRD rats. (2) No circadian phase dependency was found in the glomerular angiotensin II receptors in both rat strains. However, receptor density was significantly lower in TGR than in SPRD rats. In both strains, receptor number increased with aging. (3) In renal arteries, the angiotensin II receptor mRNA of the main receptor subtype AT1A was neither strain nor age dependent, AT1B- and AT2-receptor mRNAs were significantly lower in TGR than SPRD rats. In conclusion, the results demonstrate that the overactive renin-angiotensin system in TGR rats led to a down-regulation of glomerular angiotensin II receptors that was not accompanied by a down-regulation of the mRNA of the dominant AT1A- receptor subtype. Circadian short-term variations in blood pressure in both TGR and SPRD rats are not reflected by daily variation in angiotensin II receptor density of renal glomeruli or by variation in receptor expression in renal vascular tissue.
Collapse
Affiliation(s)
- B Lemmer
- Institute of Pharmacology and Toxicology, Faculty of Clinical Medicine Mannheim, University of Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
48
|
Tanabe S, Nakatani T, Han Y, Kishimoto T, Ohishi M, Ogami M, Ikura Y, Ueda M. Enhanced expression of angiotensin II type 1 receptor in the neointima of transplant renal arteriosclerosis in human renal allografts. Transplant Proc 2001; 33:1172-1174. [PMID: 11267243 DOI: 10.1016/s0041-1345(00)02448-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- S Tanabe
- Department of Urology, Osaka City University Medical School, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhuo JL. Renomedullary interstitial cells: a target for endocrine and paracrine actions of vasoactive peptides in the renal medulla. Clin Exp Pharmacol Physiol 2000; 27:465-73. [PMID: 10874500 DOI: 10.1046/j.1440-1681.2000.03277.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
1. The renal medulla plays an important role in regulating body sodium and fluid balance and blood pressure homeostasis through its unique structural relationships and interactions between renomedullary interstitial cells (RMIC), renal tubules and medullary vasculature. 2. Several endocrine and/or paracrine factors, including angiotensin (Ang)II, endothelin (ET), bradykinin (BK), atrial natriuretic peptide (ANP) and vasopressin (AVP), are implicated in the regulation of renal medullary function and blood pressure by acting on RMIC, tubules and medullary blood vessels. 3. Renomedullary interstitial cells express multiple vasoactive peptide receptors (AT1, ETA, ETB, BK B2, NPRA and NPRB and V1a) in culture and in tissue. 4. In cultured RMIC, AngII, ET, BK, ANP and AVP act on their respective receptors to induce various cellular responses, including contraction, prostaglandin synthesis, cell proliferation and/or extracellular matrix synthesis. 5. Infusion of vasoactive peptides or their antagonists systemically or directly into the medullary interstitium modulates medullary blood flow, sodium excretion and urine osmolarity. 6. Overall, expression of multiple vasoactive peptide receptors in RMIC, which respond to various vasoactive peptides and paracrine factors in vitro and in vivo, supports the hypothesis that RMIC may be an important paracrine target of various vasoactive peptides in the regulation of renal medullary function and long-term blood pressure homeostasis.
Collapse
Affiliation(s)
- J L Zhuo
- Howard Florey Institute of Experimental Physiology and Medicine, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|