1
|
Kratz D, Wilken-Schmitz A, Sens A, Hahnefeld L, Scholich K, Geisslinger G, Gurke R, Thomas D. Post-mortem changes of prostanoid concentrations in tissues of mice: Impact of fast cervical dislocation and dissection delay. Prostaglandins Other Lipid Mediat 2022; 162:106660. [PMID: 35714920 DOI: 10.1016/j.prostaglandins.2022.106660] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/29/2022]
Abstract
Prostanoids are potent lipid mediators involved in a wide variety of physiological functions like blood pressure regulation or inflammation as well as cardiovascular and malign diseases. Elucidation of their modes of action is mainly carried out in pre-clinical animal models by quantifying prostanoids in tissues of interest. Unfortunately, prostanoids are prone to post-mortem artifact formation and de novo synthesis can already be caused by external stimuli during the euthanasia of animals like prolonged hypercapnia or ischemia. Therefore, this study investigates the suitability and impact of fast cervical dislocation for the determination of prostanoids (6-keto-PGF1α, TXB2, PGF2α, PGD2, PGE2) in seven tissues of mice (spinal cord, brain, sciatic nerve, kidney, liver, lung, and spleen) to minimize time-dependent effects and approximate physiological concentrations. Tissues were dissected in a standardized sequence directly or after 10 min to investigate the influence of dissection delays. The enzyme inhibitor indomethacin (10 µM) in combination with low processing temperatures was employed to preserve prostanoid concentrations during sample preparation. Quantification of prostanoids was performed via LC-MS/MS. This study shows, that prostanoids are differentially susceptible to post-mortem artifact formation which is closely connected to their physiological function and metabolic stability in the respective tissues. Prostanoids in the brain, spinal cord, and kidney that are not involved in the regulatory response post-mortem, i.e. blood flow regulation (6-keto-PGF1α, PGE2, PGF2α) showed high reproducibility even after dissection delay and could be assessed after fast cervical dislocation if prerequisites like standardized pre-analytical workflows with immediate dissection and inhibition of residual enzymatic activity are in place. However, in tissues with high metabolic activity (liver, lung) more stable prostanoid metabolites should be used. Moreover, prostanoids in the spleen were strongly affected by dissection delays and presumably the method of euthanasia itself.
Collapse
Affiliation(s)
- D Kratz
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - A Wilken-Schmitz
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - A Sens
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - L Hahnefeld
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - K Scholich
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - G Geisslinger
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - R Gurke
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - D Thomas
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital of Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
2
|
Wu P, Gao ZX, Duan XP, Su XT, Wang MX, Lin DH, Gu R, Wang WH. AT2R (Angiotensin II Type 2 Receptor)-Mediated Regulation of NCC (Na-Cl Cotransporter) and Renal K Excretion Depends on the K Channel, Kir4.1. Hypertension 2018; 71:622-630. [PMID: 29483225 PMCID: PMC5843543 DOI: 10.1161/hypertensionaha.117.10471] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 10/26/2017] [Accepted: 12/21/2017] [Indexed: 02/02/2023]
Abstract
AT2R (AngII [angiotensin II] type 2 receptor) is expressed in the distal nephrons. The aim of the present study is to examine whether AT2R regulates NCC (Na-Cl cotransporter) and Kir4.1 of the distal convoluted tubule. AngII inhibited the basolateral 40 pS K channel (a Kir4.1/5.1 heterotetramer) in the distal convoluted tubule treated with losartan but not with PD123319. AT2R agonist also inhibits the K channel, indicating that AT2R was involved in tonic regulation of Kir4.1. The infusion of PD123319 stimulated the expression of tNCC (total NCC) and pNCC (phosphorylated NCC; Thr53) by a time-dependent way with the peak at 4 days. PD123319 treatment (4 days) stimulated the basolateral 40 pS K channel activity, augmented the basolateral K conductance, and increased the negativity of distal convoluted tubule membrane. The stimulation of Kir4.1 was essential for PD123319-induced increase in NCC because inhibiting AT2R increased the expression of tNCC and pNCC only in wild-type but not in the kidney-specific Kir4.1 knockout mice. Renal clearance study showed that thiazide-induced natriuretic effect was larger in PD123319-treated mice for 4 days than untreated mice. However, this effect was absent in kidney-specific Kir4.1 knockout mice which were also Na wasting under basal conditions. Finally, application of AT2R antagonist decreased the renal ability of K excretion and caused hyperkalemia in wild-type but not in kidney-specific Kir4.1 knockout mice. We conclude that AT2R-dependent regulation of NCC requires Kir4.1 in the distal convoluted tubule and that AT2R plays a role in stimulating K excretion by inhibiting Kir4.1 and NCC.
Collapse
Affiliation(s)
- Peng Wu
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Zhong-Xiuzi Gao
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Xin-Peng Duan
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Xiao-Tong Su
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Ming-Xiao Wang
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Dao-Hong Lin
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Ruimin Gu
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.)
| | - Wen-Hui Wang
- From the Department of Pharmacology, New York Medical College, Valhalla (P.W., Z.-X.G., X.-T.S., M.-X.W., D.-H.L., W.-H.W.); and Department of Physiology, Harbin Medical University, China (X.-P.D., R.G.).
| |
Collapse
|
3
|
Angiotensin II type 2 receptor (AT2R) in renal and cardiovascular disease. Clin Sci (Lond) 2017; 130:1307-26. [PMID: 27358027 DOI: 10.1042/cs20160243] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/06/2016] [Indexed: 12/14/2022]
Abstract
Angiotensin II (Ang II) is well-considered to be the principal effector of the renin-angiotensin system (RAS), which binds with strong affinity to the angiotensin II type 1 (AT1R) and type 2 (AT2R) receptor subtype. However, activation of both receptors is likely to stimulate different signalling mechanisms/pathways and produce distinct biological responses. The haemodynamic and non-haemodynamic effects of Ang II, including its ability to regulate blood pressure, maintain water-electrolyte balance and promote vasoconstriction and cellular growth are well-documented to be mediated primarily by the AT1R. However, its biological and functional effects mediated through the AT2R subtype are still poorly understood. Recent studies have emphasized that activation of the AT2R regulates tissue and organ development and provides in certain context a potential counter-regulatory mechanism against AT1R-mediated actions. Thus, this review will focus on providing insights into the biological role of the AT2R, in particular its actions within the renal and cardiovascular system.
Collapse
|
4
|
Jensen TW, Olsen NV. Angiotensin II during Experimentally Simulated Central Hypovolemia. Front Cardiovasc Med 2016; 3:6. [PMID: 26973842 PMCID: PMC4776081 DOI: 10.3389/fcvm.2016.00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/18/2016] [Indexed: 11/13/2022] Open
Abstract
Central hypovolemia, defined as diminished blood volume in the heart and pulmonary vascular bed, is still an unresolved problem from a therapeutic point of view. The development of pharmaceutical agents targeted at specific angiotensin II receptors, such as the non-peptidergic AT2-receptor agonist compound 21, is yielding many opportunities to uncover more knowledge about angiotensin II receptor profiles and possible therapeutic use. Cardiovascular, anti-inflammatory, and neuroprotective therapeutic use of compound 21 have been suggested. However, there has not yet been a focus on the use of these agents in a hypovolemic setting. We argue that the latest debates on the effect of angiotensin II during hypovolemia might guide for future studies, investigating the effect of such agents during experimentally simulated central hypovolemia. The purpose of this review is to examine the role of angiotensin II during episodes of central hypovolemia. To examine this, we reviewed results from studies with three experimental models of simulated hypovolemia: head up tilt table test, lower body negative pressure, and hemorrhage of animals. A systemic literature search was made with the use of PubMed/MEDLINE for studies that measured variables of the renin–angiotensin system or its effect during simulated hypovolemia. Twelve articles, using one of the three models, were included and showed a possible organ-protective effect and an effect on the sympathetic system of angiotensin II during hypovolemia. The results support the possible organ-protective vasodilatory role for the AT2-receptor during hypovolemia on both the kidney and the splanchnic tissue.
Collapse
Affiliation(s)
- Theo Walther Jensen
- Department of Neuroscience and Pharmacology, The Health Faculty, University of Copenhagen , Copenhagen , Denmark
| | - Niels Vidiendal Olsen
- Department of Neuroscience and Pharmacology, The Health Faculty, University of Copenhagen, Copenhagen, Denmark; Department of Neuroanaesthesia, The Neuroscience Centre, University Hospital of Copenhagen (Rigshospitalet), Copenhagen, Denmark
| |
Collapse
|
5
|
Abadir PM, Siragy HM. Angiotensin type 1 receptor mediates renal production and conversion of prostaglandins E2 to F2α in conscious diabetic rats. J Renin Angiotensin Aldosterone Syst 2015. [PMID: 26195268 DOI: 10.1177/1470320315592566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Previous studies demonstrated that stimulation of angiotensin subtype 1 receptor (AT1R) led to increased renal generation of prostaglandins E2 (PGE2) and renal inflammation. In turn, PGE2 increases AT1R activity. The conversion of PGE2 to the less active metabolite prostaglandin F2α (PGF2α) via 9-ketoreductase interrupts this feedback loop. The effects of diabetes on the interface between AT1R, PGE2 and PGF2α are not well established. We hypothesized that in diabetes, an aberrant AT1R activity enhances the biosynthesis of PGE2 and impairs the activity of PGE 9-ketoreductase, leading to accumulation of PGE2. MATERIALS AND METHODS Using microdialysis technique, we monitored renal interstitial fluid levels of angiotensin II (Ang II), PGE2 and PGF2α in control and AT1R blocker, valsartan, treated diabetic rats (N=8 each). We utilized the PGF2α to PGE2 ratio as indirect measure of PGE 9-ketoreductase activity. RESULTS Diabetes increased renal interstitial fluid levels of Ang II, PGE2 and PGF2α. PGF2α/PGE2 ratio increased by the third week, but declined by the sixth week of diabetes. Valsartan reduced PGE2 and PGF2α levels and increased Ang II and the conversion of PGE2 to PGF2α. CONCLUSION Our results suggest that in diabetes, AT1R increases PGE2 generation and reduces conversion of PGE2 to PGF2α with the progression of diabetes.
Collapse
Affiliation(s)
- Peter M Abadir
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, Baltimore, USA
| | - Helmy M Siragy
- University of Virginia School of Medicine, Department of Medicine, Charlottesville, USA
| |
Collapse
|
6
|
Abstract
In the mammalian kidney, prostaglandins (PGs) are important mediators of physiologic processes, including modulation of vascular tone and salt and water. PGs arise from enzymatic metabolism of free arachidonic acid (AA), which is cleaved from membrane phospholipids by phospholipase A2 activity. The cyclooxygenase (COX) enzyme system is a major pathway for metabolism of AA in the kidney. COX are the enzymes responsible for the initial conversion of AA to PGG2 and subsequently to PGH2, which serves as the precursor for subsequent metabolism by PG and thromboxane synthases. In addition to high levels of expression of the "constitutive" rate-limiting enzyme responsible for prostanoid production, COX-1, the "inducible" isoform of cyclooxygenase, COX-2, is also constitutively expressed in the kidney and is highly regulated in response to alterations in intravascular volume. PGs and thromboxane A2 exert their biological functions predominantly through activation of specific 7-transmembrane G-protein-coupled receptors. COX metabolites have been shown to exert important physiologic functions in maintenance of renal blood flow, mediation of renin release and regulation of sodium excretion. In addition to physiologic regulation of prostanoid production in the kidney, increases in prostanoid production are also seen in a variety of inflammatory renal injuries, and COX metabolites may serve as mediators of inflammatory injury in renal disease.
Collapse
Affiliation(s)
- Raymond C Harris
- George M. O'Brien Kidney and Urologic Diseases Center and Division of Nephrology, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee, USA.
| | | |
Collapse
|
7
|
Loria AS, Yamamoto T, Pollock DM, Pollock JS. Early life stress induces renal dysfunction in adult male rats but not female rats. Am J Physiol Regul Integr Comp Physiol 2012; 304:R121-9. [PMID: 23174859 DOI: 10.1152/ajpregu.00364.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Maternal separation (MatSep) is a model of behavioral stress during early life. We reported that MatSep exacerbates ANG II-induced hypertension in adult male rats. The aims of this study were to determine whether exposure to MatSep in female rats sensitizes blood pressure to ANG II infusion similar to male MatSep rats and to elucidate renal mechanisms involved in the response in MatSep rats. Wistar Kyoto (WKY) pups were exposed to MatSep 3 h/day from days 2 to 14, while control rats remained with their mothers. ANG II-induced mean arterial pressure (MAP; telemetry) was enhanced in female MatSep rats compared with control female rats but delayed compared with male MatSep rats. Creatinine clearance (Ccr) was reduced in male MatSep rats compared with control rats at baseline and after ANG II infusion. ANG II infusion significantly increased T cells in the renal cortex and greater histological damage in the interstitial arteries of male MatSep rats compared with control male rats. Plasma testosterone was greater and estradiol was lower in male MatSep rats compared with control rats with ANG II infusion. ANG II infusion failed to increase blood pressure in orchidectomized male MatSep and control rats. Female MatSep and control rats had similar Ccr, histological renal analysis, and sex hormones at baseline and after ANG II infusion. These data indicate that during ANG II-induced hypertension, MatSep sensitizes the renal phenotype in male but not female rats.
Collapse
Affiliation(s)
- Analia S Loria
- Section of Experimental Medicine, Georgia Health Sciences Univ., Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
8
|
Furuya H, Wada M, Shimizu Y, Yamada PM, Hannun YA, Obeid LM, Kawamori T. Effect of sphingosine kinase 1 inhibition on blood pressure. FASEB J 2012; 27:656-64. [PMID: 23109673 DOI: 10.1096/fj.12-219014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Accumulating evidence suggests that sphingosine kinase 1 (SphK1) plays a key role in carcinogenesis by regulating cyclooxygenase-2 (COX-2) expression. Recent clinical studies have revealed that COX-2 inhibitors cause adverse cardiovascular side effects, likely due to inhibition of prostacyclin (PGI(2)). In this work, we investigated the roles of SphK1 inhibition on blood pressure (BP). The results show that lack of SphK1 expression did not exacerbate angiotensin II (Ang II)-induced acute hypertension, whereas celecoxib, a COX-2 inhibitor, augmented and sustained higher BP in mice. Interestingly, SphK1-knockout mice inhibited prostaglandin E(2) (PGE(2)) but not PGI(2) production in response to Ang II, whereas celecoxib blocked both PGE(2) and PGI(2) production. Mechanistically, SphK1 down-regulation by siRNA in human umbilical vein endothelial cells decreased cytokine-induced PGE(2) production primarily through inhibition of microsomal PGE synthase-1 (mPGES-1), not COX-2. SphK1 down-regulation also decreased MKK6 expression, which phosphorylates and activates P38 MAPK, which, in turn, regulates early growth response-1 (Egr-1), a transcription factor of mPGES-1. Together, these data indicate that SphK1 regulates PGE(2) production by mPGES-1 expression via the p38 MAPK pathway, independent of COX-2 signaling, in endothelial cells, suggesting that SphK1 inhibition may be a promising strategy for cancer chemoprevention with lack of the adverse cardiovascular side effects associated with coxibs.
Collapse
Affiliation(s)
- Hideki Furuya
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Helmersson-Karlqvist J, Björklund-Bodegård K, Larsson A, Basu S. 24-Hour ambulatory blood pressure associates inversely with prostaglandin F(2α), interleukin-6 and F(2)-isoprostane formation in a Swedish population of older men. Int J Clin Exp Med 2012; 5:145-153. [PMID: 22567175 PMCID: PMC3342711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/06/2012] [Indexed: 05/31/2023]
Abstract
Vasoconstrictive prostaglandins (PGs), such as PGF(2α), F(2)-isoprostanes, and systemic inflammation may be involved in the physiological regulation of blood pressure (BP) and the pathophysiology leading to hypertension. However, studies evaluating these parameters and BP in human populations are sparse. We analysed the cross-sectional associations between 24-hour ambulatory BP and urinary 15-keto-dihydro-PGF(2α) (indicator of PG-mediated vasoconstriction and inflammation), plasma interleukin-6 (IL-6), C-reactive protein (CRP), serum amyloid A (SAA) and urinary F(2)-isoprostanes (indicator of vasoconstriction and oxidative stress) in 619 men in a Swedish older population (Uppsala Longitudinal Study of Adult Men, age 78 years). Both systolic and diastolic 24-hour BP correlated inversely with concentrations of 15-keto-dihydro-PGF(2α) (P<0.01) and F(2)-isoprostanes (P<0.01) independent on other cardiovascular risk factors. Additionally, diastolic 24-hour BP inversely correlated with plasma IL-6 (P<0.05) and 24-hour pulse pressure showed a positive linear correlation with IL-6, CRP and SAA. In conclusion, high BP is associated with decreased formation of vasoconstrictive PGF(2α) and F(2)-isoprostanes in this population of older men. These findings, although unlike our original hypothesis, might have an important physiological function which needs to be further evaluated.
Collapse
|
10
|
Yang R, Smolders I, Dupont AG. Blood pressure and renal hemodynamic effects of angiotensin fragments. Hypertens Res 2011; 34:674-83. [PMID: 21412242 DOI: 10.1038/hr.2011.24] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Angiotensin (Ang) II, the main effector peptide of the renin-Ang system, increases arterial blood pressure through Ang II type 1A (AT(1a)) receptor-dependent arterial vasoconstriction and by decreasing renal salt and water excretion through extrarenal and intrarenal mechanisms. AT(2) receptors are assumed to oppose these responses mediated by AT(1) receptors, thereby attenuating the pressor effects of Ang II. Nevertheless, a possible role of AT(2) receptors in the regulation of renal hemodynamics and sodium homeostasis remains to be unclear. Several other Ang fragments such as Ang III, Ang IV, Ang-(1-7) and Ang A have also been shown to display biological activity. In this review, we focus on the effects of these Ang on blood pressure, renal hemodynamics and sodium water handling, and discuss the receptors involved in these actions.
Collapse
Affiliation(s)
- Rui Yang
- Department of Pharmacology, Brussels, Belgium
| | | | | |
Collapse
|
11
|
Harding P, Yang XP, He Q, Lapointe MC. Lack of microsomal prostaglandin E synthase-1 reduces cardiac function following angiotensin II infusion. Am J Physiol Heart Circ Physiol 2010; 300:H1053-61. [PMID: 21193590 DOI: 10.1152/ajpheart.00772.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our laboratory previously reported that inducible PGE(2) synthase, mPGES-1, contributes to micromolar production of PGE(2) in neonatal ventricular myocytes in vitro, which stimulates their growth. We therefore hypothesized that mPGES-1 contributes to cardiac hypertrophy following angiotensin II (ANG II) infusion. To test this hypothesis, we used 10- to 12-wk-old mPGES-1 knockout mice (mPGES-1 KO) and C57Bl/6 control mice infused for 8 wk with either 1.4 mg · kg(-1) · day(-1) ANG II or vehicle subcutaneously. Blood pressure [systolic blood pressure (SBP)] was measured throughout the study, and cardiac function was assessed by M-mode echocardiography at baseline and at 8 wk of infusion. At the conclusion of the study, immunohistochemistry was used to evaluate collagen fraction, myocyte cross-sectional area (MCSA), and apoptosis. At baseline, there was no difference in SBP between mPGES-1 KO mice and C57BL/6 controls. ANG II infusion increased SBP to similar levels in both strains. In control mice, infusion of ANG II increased MCSA and posterior wall thickness at diastole (PWTd) but had little effect on cardiac function, consistent with compensatory hypertrophy. In contrast, cardiac function was worse in mPGES-1 KO mice after ANG II treatment. Ejection fraction declined from 76.2 ± 2.7 to 63.3 ± 3.4% after ANG II, and left ventricular dimension at systole and diastole increased from 1.29 ± 0.02 to 1.78 ± 0.15 mm and from 2.57 ± 0.03 to 2.90 ± 0.13 mm, respectively. Infusion of ANG II increased both the LV-to-body weight and the mass-to-body weight ratios to a similar extent in both strains. However, PWTd increased by a lesser extent in KO mice, suggesting an impaired hypertrophic response. ANG II infusion increased collagen staining similarly in both strains, but TdT-dUTP nick end labeling staining was greater in mPGES-1 KO mice. Overall, these results are consistent with a beneficial effect for mPGES-1 in the maintenance of cardiac function in ANG II-dependent hypertension.
Collapse
Affiliation(s)
- Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | |
Collapse
|
12
|
Enhanced pressor response to acute Ang II infusion in mice lacking membrane-associated prostaglandin E2 synthase-1. Acta Pharmacol Sin 2010; 31:1284-92. [PMID: 20871624 DOI: 10.1038/aps.2010.99] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
AIM To examine the contribution of vascular membrane-associated prostaglandin E2 synthase-1 (mPGES-1) to acute blood pressure homeostasis. METHODS Angiotensin II (AngII, 75 pmol·kg⁻¹·min⁻¹) was continuously infused via the jugular vein into wild-type and mPGES-1(-/-) mice for 30 min, and blood pressure was measured by carotid arterial catheterization. RT-PCR and immunohistochemistry were performed to detect the expression and localization of mPGES-1 in the mouse arterial vessels. Mesenteric arteries were dissected from mice of both genotypes to study vessel tension and measure vascular PGE2 levels. RESULTS Wild-type and mPGES-1(-/-) mice showed similar blood pressure levels at baseline, and the acute intravenous infusion of AngII caused a greater increase in mean arterial pressure in the mPGES-1(-/-) group, with a similar diuretic and natriuretic response in both groups. mPGES-1 was constitutively expressed in the aortic and mesenteric arteries and vascular smooth muscle cells of wild-type mice. Strong staining was detected in the smooth muscle layer of arterial vessels. Ex vivo treatment of mesenteric arteries with AngII produced more vasodilatory PGE2 in wild-type than in mPGES-1(-/-) mice. In vitro tension assays further revealed that the mesenteric arteries of mPGES-1(-/-) mice exhibited a greater vasopressor response to AngII than those arteries of wild-type mice. CONCLUSION Vascular mPGES-1 acts as an important tonic vasodilator, contributing to acute blood pressure regulation.
Collapse
|
13
|
Abstract
Cyclooxygenase-derived prostanoids exert complex and diverse functions within the kidney. The biological effect of each prostanoid is controlled at multiple levels, including (a) enzymatic reactions catalyzed sequentially by cyclooxygenase and prostanoid synthase for the synthesis of bioactive prostanoid and (b) the interaction with its receptors that mediate its functions. Cyclooxygenase-derived prostanoids act in an autocrine or a paracrine fashion and can serve as physiological buffers, protecting the kidney from excessive functional changes during physiological stress. Through these actions, prostanoids play important roles in maintaining renal function, body fluid homeostasis, and blood pressure. Renal cortical COX2-derived prostanoids, particularly PGI2 and PGE2, play critical roles in maintaining blood pressure and renal function in volume-contracted states. Renal medullary COX2-derived prostanoids appear to have an antihypertensive effect in individuals challenged with a high-salt diet. Loss of EP2 or IP receptor is associated with salt-sensitive hypertension. COX2 also plays a role in maintaining renal medullary interstitial cell viability in the hypertonic environment of the medulla. Cyclooxygenase-derived prostanoids also are involved in certain pathological processes. The cortical COX2-derived PGI2 participates in the pathogenesis of renal vascular hypertension through stimulating renal renin synthesis and release. COX-derived prostanoids also appear to be involved in the pathogenesis of diabetic nephropathy. COXs, prostanoid synthases, and prostanoid receptors should provide fruitful targets for intervention in the pharmacological treatment of renal disease.
Collapse
Affiliation(s)
- Chuan-Ming Hao
- Division of Nephrology, Department of Medicine, Vanderbilt University, and Veterans Affair Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
14
|
Guan Y, Zhang Y, Wu J, Qi Z, Yang G, Dou D, Gao Y, Chen L, Zhang X, Davis LS, Wei M, Fan X, Carmosino M, Hao C, Imig JD, Breyer RM, Breyer MD. Antihypertensive effects of selective prostaglandin E2 receptor subtype 1 targeting. J Clin Invest 2007; 117:2496-505. [PMID: 17710229 PMCID: PMC1940235 DOI: 10.1172/jci29838] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Accepted: 05/29/2007] [Indexed: 11/17/2022] Open
Abstract
Clinical use of prostaglandin synthase-inhibiting NSAIDs is associated with the development of hypertension; however, the cardiovascular effects of antagonists for individual prostaglandin receptors remain uncharacterized. The present studies were aimed at elucidating the role of prostaglandin E2 (PGE2) E-prostanoid receptor subtype 1 (EP1) in regulating blood pressure. Oral administration of the EP1 receptor antagonist SC51322 reduced blood pressure in spontaneously hypertensive rats. To define whether this antihypertensive effect was caused by EP1 receptor inhibition, an EP1-null mouse was generated using a "hit-and-run" strategy that disrupted the gene encoding EP1 but spared expression of protein kinase N (PKN) encoded at the EP1 locus on the antiparallel DNA strand. Selective genetic disruption of the EP1 receptor blunted the acute pressor response to Ang II and reduced chronic Ang II-driven hypertension. SC51322 blunted the constricting effect of Ang II on in vitro-perfused preglomerular renal arterioles and mesenteric arteriolar rings. Similarly, the pressor response to EP1-selective agonists sulprostone and 17-phenyltrinor PGE2 were blunted by SC51322 and in EP1-null mice. These data support the possibility of targeting the EP1 receptor for antihypertensive therapy.
Collapse
Affiliation(s)
- Youfei Guan
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jing Wu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zhonghua Qi
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Guangrui Yang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dou Dou
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yuansheng Gao
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lihong Chen
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiaoyan Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Linda S. Davis
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mingfeng Wei
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xuefeng Fan
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Monica Carmosino
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Chuanming Hao
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John D. Imig
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Richard M. Breyer
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew D. Breyer
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, People’s Republic of China.
Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA.
Department of Pharmacology and
Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW The balance of angiotensin II and nitric oxide determines the sensitivity of the tubuloglomerular feedback mechanism, renal vascular resistance and filtration rate. Angiotensin II induces nitric oxide release, but the role of angiotensin II receptors here is not fully understood. Further, the angiotensin II-nitric oxide interaction can be modulated by reactive oxygen species. This review focuses on the angiotensin II-nitric oxide interaction and their modulation by reactive oxygen species in the control of renal blood flow. RECENT FINDINGS Ideas about the role of angiotensin II type 1 and angiotensin II type 2 receptors are extended by the observation of angiotensin II type 1-mediated nitric oxide release with direct effects on vascular tone, tubuloglomerular feedback and sympathetic neurotransmission. Angiotensin receptors elicit disparate effects on intrarenal circulation. Angiotensin II-nitric oxide interactions are modulated by reactive oxygen species, as shown by angiotensin II type 1-mediated activation of superoxide and depression of antioxidant enzymes leading to reduced nitric oxide concentration - mechanisms that may be also important in angiotensin II-dependent hypertension. SUMMARY Recent studies show that angiotensin II stimulates the nitric oxide system via angiotensin II type 1 and angiotensin II type 2 receptors, whereas receptors exert different effects on renal and medullary flow. The interaction via angiotensin II type 1 is modulated by reactive oxygen species.
Collapse
Affiliation(s)
- Andreas Patzak
- Johannes-Müller-Institute of Physiology, Humboldt-University of Berlin, University Hospital Charité, Berlin, Germany
| | | |
Collapse
|
16
|
Qi Z, Cai H, Morrow JD, Breyer MD. Differentiation of Cyclooxygenase 1- and 2–Derived Prostanoids in Mouse Kidney and Aorta. Hypertension 2006; 48:323-8. [PMID: 16801485 DOI: 10.1161/01.hyp.0000231934.67549.b7] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Accumulating evidence indicates cyclooxygenase (COX) 1 and COX2 differentially regulate cardiovascular and renal function. We have demonstrated previously in mice that COX2 inhibition enhances angiotensin II-induced hypertension, and COX1 inhibition attenuates the pressor effect of angiotensin II. To further elucidate the mechanism underlying the functional difference of COX1 versus COX2 inhibition, the present studies examined the prostaglandin (PG) profiles derived in COX1- or COX2-inhibited mouse kidney and aorta using gas chromatographic/mass spectrometric assays. PGE
2
is the most abundant prostanoid in both renal cortex and medulla in normal C57BL/6J mice, followed by PGI
2
, PGF
2α
and thromboxane A
2
. In contrast PGI
2
was most abundant in aorta followed by thromboxane A
2
, PGE
2
, and PGF
2α
. PGD
2
was undetectable in control kidney or aorta. At baseline, inhibition of COX1 decreased total prostaglandins in renal cortex, medulla, and aorta, whereas COX2 inhibition decreased total prostaglandins only in renal medulla. Angiotensin II infusion significantly increased COX2-dependent/COX1-independent PGE
2
and PGI
2
in renal cortex and medulla. Angiotensin II also significantly increased renal PGF
2α
in cortex, but not in medulla, through both COX1- and COX2-dependent mechanisms. These studies demonstrate that although COX1 primarily contributes to basal prostanoid production in the kidney and aorta, angiotensin II increases renal vasodilator prostanoids predominately via COX2 activity. These effects may contribute to the specific effect of COX2 inhibitors to increase blood pressure.
Collapse
Affiliation(s)
- Zhonghua Qi
- Division of Nephrology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
17
|
Tirapelli CR, Fukada SY, de Godoy MAF, de Oliveira AM. Analysis of the mechanisms underlying the vasorelaxant action of angiotensin II in the isolated rat carotid. Life Sci 2006; 78:2676-82. [PMID: 16386763 DOI: 10.1016/j.lfs.2005.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2005] [Accepted: 10/17/2005] [Indexed: 11/18/2022]
Abstract
It has been suggested that low concentrations of angiotensin II cause vasoconstriction whereas high concentrations evoke vasodilation. Thus, this work aimed to functionally characterize the mechanisms underlying the relaxation induced by angiotensin II at high concentrations in isolated rat carotid rings. Experiments using standard muscle bath procedures showed that angiotensin II (0.01-3 microM) concentration dependently induces relaxation of phenylephrine-pre-contracted rings. No differences between intact or denuded endothelium were found. The angiotensin II-induced relaxation was strongly inhibited by saralasin, the non-selective antagonist of angiotensin II receptors but not by the selective antagonists of AT1 and AT2 receptors, losartan and PD123319, respectively. However, A-779, a selective angiotensin-(1-7) receptor antagonist, reduced the relaxation induced by angiotensin II. Administration of exogenous angiotensin-(1-7) on pre-contracted tissues produced concentration-dependent relaxation, which was also inhibited by A-779. HOE-140, the selective antagonist of the bradykinin in B2 receptor did not produce any significant effect on angiotensin II-induced relaxation. Pre-incubation of denuded-rings with N G-nitro-L-arginine methyl ester (L-NAME) or 1H-[1,2,4] Oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) reduced angiotensin II-induced relaxation. On the other hand, neither indomethacin nor tetraethylammonium (TEA) produced any significant effect. The major new finding of this work is that high concentrations of angiotensin II induce relaxation of the rat carotid via activation of the NO-cGMP pathway through a mechanism that seems to be partially dependent on activation of angiotensin-(1-7) receptors.
Collapse
Affiliation(s)
- Carlos R Tirapelli
- Department of Pharmacology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (USP), SP, Brazil
| | | | | | | |
Collapse
|
18
|
Batenburg WW, Tom B, Schuijt MP, Danser AHJ. Angiotensin II type 2 receptor-mediated vasodilation. Focus on bradykinin, NO and endothelium-derived hyperpolarizing factor(s). Vascul Pharmacol 2005; 42:109-18. [PMID: 15792928 DOI: 10.1016/j.vph.2005.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Angiotensin (Ang) II type 1 (AT(1)) receptors account for the majority of the cardiovascular effects Ang II, including vasoconstriction and growth stimulation. Recent evidence, mainly obtained in animals, suggests that Ang II type 2 (AT(2)) receptors counteract some or all of these effects. This review summarizes the current knowledge on the vasodilator effects induced by AT(2) receptors in humans and animals, focussing not only on the mediators of this effect, but also on the modulatory role of age, gender, and endothelial function. It is concluded that AT(2) receptor-mediated vasodilation most likely depends on the bradykinin-bradykinin type 2 (B(2)) receptor-NO-cGMP pathway, although evidence for a direct link between AT(2) and B(2) receptors is currently lacking. If indeed B(2) receptors are involved, this would imply that, in addition to NO, also the wide range of non-NO 'endothelium-derived hyperpolarizing factors' (EDHFs) that is released following B(2) receptor activation (e.g., K(+), cytochrome P450 products from arachidonic acid, H(2)O(2) and S-nitrososothiols), could contribute to AT(2) receptor-induced vasodilation.
Collapse
Affiliation(s)
- Wendy W Batenburg
- Department of Pharmacology, room EE1418b, Erasmus MC, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
19
|
Gonzalez M, Lobos L, Castillo F, Galleguillos L, Lopez NC, Michea L. High-Salt Diet Inhibits Expression of Angiotensin Type 2 Receptor in Resistance Arteries. Hypertension 2005; 45:853-9. [PMID: 15809360 DOI: 10.1161/01.hyp.0000161990.98383.ad] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies suggested that type 2 angiotensin receptor (AT2R) could contribute to regulation of blood pressure and/or vascular remodeling. A key question relates to the effects of potential modulators of vascular AT2R expression. In the present work, we evaluated if high salt intake (70 mmol/L NaCl in drinking water) could modulate rat mesenteric artery AT2R function and expression. Angiotensin II dose-response curves were studied in rat perfused pressurized small-diameter arteries in the presence of losartan (AT1R antagonist). Arteries were precontracted with phenylephrine, yielding ≈30% decrease in resting diameter. AT2R activation by angiotensin-induced dose-dependent relaxation of precontracted arteries (60.1±9.1% of phenylephrine-induced contraction,
P
<0.05). In contrast, AT2R-dependent relaxation was not observed in arteries obtained from rats on high-salt diet. Semi-quantitative reverse-transcription polymerase chain reaction experiments demonstrated reduced amount of AT2R mRNA in arteries of rats on high-salt diet (65.5±7.5% of control levels,
P
<0.05). Western blot studies demonstrated decreased AT2R in mesenteric artery protein fractions of high-salt diet rats (60.0±18.0 of control levels,
P
<0.05). In a second set of experiments, adrenalectomy (4 days) blunted AT2R-mediated vasorelaxation and decreased AT2R mRNA (72.0±11.0% of control levels,
P
<0.05). AT2R abundance in protein fractions of mesenteric arteries of ADX rats was also diminished (64.0±13% of control levels,
P
<0.05). Both, AT2R mRNA and protein downregulation were prevented by mineralocorticoid replacement therapy. Finally, physiological concentrations of aldosterone caused a dose-dependent increase in AT2R mRNA of small diameter mesenteric artery explants. The results are consistent with aldosterone-mediated upregulation AT2R.
Collapse
MESH Headings
- Adrenalectomy
- Aldosterone/blood
- Aldosterone/pharmacology
- Angiotensin II Type 2 Receptor Blockers
- Animals
- Blood Pressure/drug effects
- Desoxycorticosterone/pharmacology
- Dose-Response Relationship, Drug
- Electrolytes/blood
- In Vitro Techniques
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Sodium Chloride, Dietary/administration & dosage
- Sodium Chloride, Dietary/pharmacology
- Vascular Resistance
- Vasodilation/drug effects
- Vasodilation/physiology
Collapse
Affiliation(s)
- Magdalena Gonzalez
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, University Los Andes, Chile
| | | | | | | | | | | |
Collapse
|
20
|
Rincón-Sánchez AR, Covarrubias A, Rivas-Estilla AM, Pedraza-Chaverrí J, Cruz C, Islas-Carbajal MC, Panduro A, Estanes A, Armendáriz-Borunda J. PGE2 alleviates kidney and liver damage, decreases plasma renin activity and acute phase response in cirrhotic rats with acute liver damage. ACTA ACUST UNITED AC 2005; 56:291-303. [PMID: 15816358 DOI: 10.1016/j.etp.2004.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In this study, we evaluated the effect of prostaglandin E2 (PGE2) on renal and hepatic function using an experimental cirrhosis model plus acute liver damage (ALD). Male Wistar rats treated with carbon tetrachloride (CCl4) for 8 weeks were used for the cirrhosis model. Cirrhotic rats were further exposed to an additional acute dose of CCl4 to induce ALD and then treated with PGE2 intramuscularly twice a day for 7 days (200 microg/Kg/day). PGE2 administration started 3 h after the additional dosing of CCl4 and PGE2 effect on hepatorenal function was examined on days 1, 2, 3, and 7. PGE2-treatment ameliorated the decrease in urinary sodium excretion, and normalized serum activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT) and plasma renin observed in cirrhotic rats with ALD. In addition, PGE2-treatment decreased mean arterial pressure, glomerular hypercellularity and thickening of the kidney capillary wall, and liver steatosis and cellular necrosis. Also, PGE2 increased the number of regenerative nodules. Finally, PGE2-treatment inhibited the increase in Alpha 1-acid glycoprotein (pAGP), fibrinogen, and Apo A-1 mRNA expression by 83%, 59%, and 77%, respectively. These results suggest that PGE2 administration may decrease the expression of acute phase proteins. In conclusion, PGE2-treatment improved hepatic and renal function and may be useful to down-regulate the acute phase response in cirrhotic rats presenting ALD induced by CCl4.
Collapse
Affiliation(s)
- Ana Rosa Rincón-Sánchez
- Institute of Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Jalisco 44281, México.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Silvestre JS, Lévy BI. Hormones and the neovascularization process: role of angiotensin II. EXS 2004:77-93. [PMID: 15617472 DOI: 10.1007/3-7643-7311-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
|
22
|
Abstract
The renin angiotensin system plays an important role in the control of body fluid and electrolyte homeostasis and blood pressure regulation. Angiotensin II is the most effector hormone of this system and functions mainly through stimulation of its subtype receptors, namely, the AT1 and AT2 receptors. Most of the known physiological and pathologic effects of angiotensin II are mediated through stimulation of the AT1 receptor. The knowledge about the involvement of the AT2 receptor in physiological and pathologic processes is still evolving. In the kidney, both the AT1 and AT2 receptors contribute to the regulation of renal hemodynamic and tubular functions. Also, these receptors regulate renal cellular growth and matrix formation. However, AT2 receptor possesses functions that counteract the effects of the AT1 receptor. The balance between the AT1 and AT2 receptors can determine the renal status in health and disease.
Collapse
Affiliation(s)
- Helmy M Siragy
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, 22908, USA.
| |
Collapse
|
23
|
Widdop RE, Jones ES, Hannan RE, Gaspari TA. Angiotensin AT2 receptors: cardiovascular hope or hype? Br J Pharmacol 2003; 140:809-24. [PMID: 14530223 PMCID: PMC1574085 DOI: 10.1038/sj.bjp.0705448] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2003] [Revised: 06/30/2003] [Accepted: 07/10/2003] [Indexed: 02/02/2023] Open
Abstract
British Journal of Pharmacology (2003) 140, 809–824. doi:10.1038/sj.bjp.0705448
Collapse
Affiliation(s)
- Robert E Widdop
- Department of Pharmacology, Monash University, Melbourne, Victoria 3800, Australia.
| | | | | | | |
Collapse
|
24
|
Carey RM, Siragy HM. Newly recognized components of the renin-angiotensin system: potential roles in cardiovascular and renal regulation. Endocr Rev 2003; 24:261-71. [PMID: 12788798 DOI: 10.1210/er.2003-0001] [Citation(s) in RCA: 378] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The renin-angiotensin system (RAS) is a coordinated hormonal cascade in the control of cardiovascular, renal, and adrenal function that governs body fluid and electrolyte balance, as well as arterial pressure. The classical RAS consists of a circulating endocrine system in which the principal effector hormone is angiotensin (ANG) II. ANG is produced by the action of renin on angiotensinogen to form ANG I and its subsequent conversion to the biologically active octapeptide by ANG-converting enzyme. ANG II actions are mediated via the ANG type 1 receptor. Here, we discuss recent advances in our understanding of the components and actions of the RAS, including local tissue RASs, a renin receptor, ANG-converting enzyme-2, ANG (1-7), the function of the ANG type 2 receptor, and ANG receptor heterodimerization. The role of the RAS in the regulation of cardiovascular and renal function is reviewed and discussed in light of these newly recognized components.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia 22908, USA.
| | | |
Collapse
|
25
|
Sandberg K, Ji H. Sex and the renin angiotensin system: implications for gender differences in the progression of kidney disease. ADVANCES IN RENAL REPLACEMENT THERAPY 2003; 10:15-23. [PMID: 12616459 DOI: 10.1053/jarr.2003.50006] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Two recognized risk factors implicated in the pathogenesis of progressive renal disease are overactivation of the renin angiotensin system and male gender. The peptide hormone, angiotensin II, produced by the renin angiotensin system cascade, plays a crucial role in maintaining blood pressure and electrolyte homeostasis. Medications that block the action of angiotensin II by either inhibiting its synthesis or by blocking its ability to bind its receptor are in wide clinical use because of their ability to significantly retard the progression of kidney disease. Analysis of data from national end-stage renal disease registries, clinical trials, and experimental animal models suggest that the progression of chronic kidney disease from several etiologies is more rapid in men than in women. In this review, we examine the data supporting the hypothesis that modulation of the activity of the renin angiotensin system by sex steroids markedly contributes to the gender differences observed in the pathophysiology of progressive kidney disease.
Collapse
Affiliation(s)
- Kathryn Sandberg
- Division of Nephrology and Hypertension, Department of Medicine, Center for Hypertension and Renal Disease Research, Georgetown University, Washington, DC, USA.
| | | |
Collapse
|
26
|
Ichihara S, Senbonmatsu T, Price E, Ichiki T, Gaffney FA, Inagami T. Targeted deletion of angiotensin II type 2 receptor caused cardiac rupture after acute myocardial infarction. Circulation 2002; 106:2244-9. [PMID: 12390955 DOI: 10.1161/01.cir.0000033826.52681.37] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Accumulating evidence has suggested that the cardiac renin-angiotensin system is activated during the remodeling process after myocardial infarction (MI). Although 2 types of angiotensin II receptors (AT(1) and AT(2)) are upregulated in the infarcted tissue, the contribution of AT(2) to the subsequent fibrogenetic phase of wound healing is less certain. This study was conducted to evaluate the role of AT(2) in wound healing after MI using an in vivo intervention study in mice with MI. METHODS AND RESULTS We examined myocardial hypertrophy, cardiac fibrosis, and morphological evidence of fibrillar collagen accumulation at the infarcted and noninfarcted regions in male mice lacking the AT(2) receptor (Agtr2-/Y) and age-matched wild-type (WT) animals. Of the Agtr2-/Y mice, 63.6% died of cardiac rupture, whereas 23.5% of the WT mice died of the same cause within 1 week. The extent of fibrosis and that of collagen gene expression in Agtr2-/Y mice were significantly reduced compared with WT mice at 1 week after coronary ligation. Furthermore, MI resulted in a marked increase in the prostaglandin E(2) (PGE(2)) level at 4 days after surgery in Agtr2-/Y mice. In WT mice, the PGE(2) level was also elevated after MI but to a significantly lesser extent than in Agtr2-/Y mice. CONCLUSIONS A chronic loss of AT(2) by gene targeting prevented the collagen deposition and caused cardiac rupture. The markedly elevated PGE(2) may be a mechanism that inhibits collagen synthesis in the infarcted region of Agtr2-/Y mice.
Collapse
Affiliation(s)
- Sahoko Ichihara
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tenn. 37232-0146, USA
| | | | | | | | | | | |
Collapse
|
27
|
Xu J, Carretero OA, Liu YH, Shesely EG, Yang F, Kapke A, Yang XP. Role of AT2 receptors in the cardioprotective effect of AT1 antagonists in mice. Hypertension 2002; 40:244-50. [PMID: 12215461 DOI: 10.1161/01.hyp.0000029095.23198.ad] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Angiotensin II (Ang II) acts mainly on two receptor subtypes: AT1 and AT2. Most of the known biological actions of Ang II are mediated by AT1 receptors; however, the role of AT2 receptors remains unclear. We tested the hypothesis that the cardioprotective effects of AT1 receptor antagonists (AT1-ant) after myocardial infarction (MI) are partially mediated by activation of AT2 receptors; thus in AT2 receptor gene knockout mice (AT2-/Y), the effect of AT1-ant will be diminished or absent. MI was induced by ligating the left anterior descending coronary artery. Four weeks later, AT2-/Y and their wild-type littermates (AT2+/Y) were started on vehicle, AT1-ant (valsartan, 50 mg/kg per day), or ACE inhibitor (enalapril, 20 mg/kg per day) for 20 weeks. Basal blood pressure and cardiac function as well as remodeling after MI did not differ between AT2+/Y and AT2-/Y. AT1-ant increased ejection fraction and cardiac output and decreased left ventricular diastolic dimension, myocyte cross-sectional area, and interstitial collagen deposition in AT2+/Y, and these effects were significantly diminished in AT2-/Y. ACE inhibitors improved cardiac function and remodeling similarly in both strains. We concluded that (1) activation of AT2 during AT1 blockade plays an important role in the therapeutic effect of AT1-ant and (2) the AT2 receptor may not play an important role in regulation of cardiac function, either under basal conditions after MI remodeling or in the therapeutic effect of ACE inhibitors.
Collapse
Affiliation(s)
- Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Mich 48202-2689, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Gross V, Plehm R, Tank J, Jordan J, Diedrich A, Obst M, Luft FC. Heart rate variability and baroreflex function in AT2 receptor-disrupted mice. Hypertension 2002; 40:207-13. [PMID: 12154115 DOI: 10.1161/01.hyp.0000027279.69240.75] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We adapted telemetry and sequence analysis employed in humans to mice and measured heart rate variability and the spontaneous baroreflex sensitivity in angiotensin II type 2 (AT2) receptor-deleted (AT2 -/-) and wild-type (AT2 +/+) mice with either deoxycorticosterone acetate (DOCA)-salt hypertension or N(omega)-nitro-L-arginine methylester hydrochloride (L-NAME) hypertension. Mean arterial pressure leveled during the day at 101+/-1 mm Hg and during the night at 109+/-1 mm Hg in AT2 receptor-deleted mice, compared with 98+/-2 mm Hg (day) and 104+/-2 mm Hg (night) in wild-type mice. Mean arterial pressure increased in AT2 receptor-deleted mice with L-NAME to 114+/-1 mm Hg (day) and 121+/-1 mm Hg (night), compared with 105+/-2 mm Hg (day) and 111+/-2 mm Hg (night), respectively. DOCA-salt also increased day and night blood pressures in AT2 receptor-deleted mice to a greater degree than in wild-type mice. Heart rate variability in the time and frequency domain was not different between AT2 receptor-deleted mice and AT2 receptor-deleted mice at baseline. Systolic blood pressure variability in the low frequency band was lower in AT2 receptor-deleted mice (0.6+/-0.1 ms2 versus 3.9+/-1.3 ms2) than in wild-type mice. Baroreceptor-heart rate reflex sensitivity was significantly increased in AT2 receptor-deleted mice compared with wild-type mice (3.4+/-0.6 versus 2.1+/-0.5 ms/mm Hg). These differences remained after DOCA-salt and L-NAME treatments. We conclude that activation of the AT2 receptor impairs arterial baroreceptor reflex function, probably by a central action. These data support the existence of an inhibitory central effect of the AT2 receptor on baroreflex function.
Collapse
Affiliation(s)
- Volkmar Gross
- Max-Delbrück-Center for Molecular Medicine, Helios-Klinikum-Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Qi Z, Hao CM, Langenbach RI, Breyer RM, Redha R, Morrow JD, Breyer MD. Opposite effects of cyclooxygenase-1 and -2 activity on the pressor response to angiotensin II. J Clin Invest 2002. [DOI: 10.1172/jci0214752] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
30
|
Qi Z, Hao CM, Langenbach RI, Breyer RM, Redha R, Morrow JD, Breyer MD. Opposite effects of cyclooxygenase-1 and -2 activity on the pressor response to angiotensin II. J Clin Invest 2002; 110:61-9. [PMID: 12093889 PMCID: PMC151026 DOI: 10.1172/jci14752] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Therapeutic use of cyclooxygenase-inhibiting (COX-inhibiting) nonsteroidal antiinflammatory drugs (NSAIDs) is often complicated by renal side effects including hypertension and edema. The present studies were undertaken to elucidate the roles of COX1 and COX2 in regulating blood pressure and renal function. COX2 inhibitors or gene knockout dramatically augment the pressor effect of angiotensin II (Ang II). Unexpectedly, after a brief increase, the pressor effect of Ang II was abolished by COX1 deficiency (either inhibitor or knockout). Ang II infusion also reduced medullary blood flow in COX2-deficient but not in control or COX1-deficient animals, suggesting synthesis of COX2-dependent vasodilators in the renal medulla. Consistent with this, Ang II failed to stimulate renal medullary prostaglandin E(2) and prostaglandin I(2) production in COX2-deficient animals. Ang II infusion normally promotes natriuresis and diuresis, but COX2 deficiency blocked this effect. Thus, COX1 and COX2 exert opposite effects on systemic blood pressure and renal function. COX2 inhibitors reduce renal medullary blood flow, decrease urine flow, and enhance the pressor effect of Ang II. In contrast, the pressor effect of Ang II is blunted by COX1 inhibition. These results suggest that, rather than having similar cardiovascular effects, the activities of COX1 and COX2 are functionally antagonistic.
Collapse
Affiliation(s)
- Zhonghua Qi
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Guan Y, Stillman BA, Zhang Y, Schneider A, Saito O, Davis LS, Redha R, Breyer RM, Breyer MD. Cloning and expression of the rabbit prostaglandin EP2 receptor. BMC Pharmacol 2002; 2:14. [PMID: 12097143 PMCID: PMC117438 DOI: 10.1186/1471-2210-2-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2002] [Accepted: 06/27/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prostaglandin E2 (PGE2) has multiple physiologic roles mediated by G protein coupled receptors designated E-prostanoid, or "EP" receptors. Evidence supports an important role for the EP2 receptor in regulating fertility, vascular tone and renal function. RESULTS The full-length rabbit EP2 receptor cDNA was cloned. The encoded polypeptide contains 361 amino acid residues with seven hydrophobic domains. COS-1 cells expressing the cloned rabbit EP2 exhibited specific [3H]PGE2 binding with a Kd of 19.1 +/- 1.7 nM. [3H]PGE2 was displaced by unlabeled ligands in the following order: PGE2>>PGD2=PGF2alpha=iloprost. Binding of [3H]PGE2 was also displaced by EP receptor subtype selective agonists with a rank order of affinity consistent with the EP2 receptor (butaprost>AH13205>misoprostol>sulprostone). Butaprost free acid produced a concentration-dependent increase in cAMP accumulation in rabbit EP2 transfected COS-1 cells with a half-maximal effective concentration of 480 nM. RNase protection assay revealed high expression in the ileum, spleen, and liver with lower expression in the kidney, lung, heart, uterus, adrenal gland and skeletal muscle. In situ hybridization localized EP2 mRNA to the uterine endometrium, but showed no distinct localization in the kidney. EP2 mRNA expression along the nephron was determined by RT-PCR and its expression was present in glomeruli, MCD, tDL and CCD. In cultured cells EP2 receptor was not detected in collecting ducts but was detected in renal interstitial cells and vascular smooth muscle cells. EP2 mRNA was also detected in arteries, veins, and preglomerular vessels of the kidney. CONCLUSION EP2 expression pattern is consistent with the known functional roles for cAMP coupled PGE2 effects in reproductive and vascular tissues and renal interstitial cells. It remains uncertain whether it is also expressed in renal tubules.
Collapse
Affiliation(s)
- Youfei Guan
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Brett A Stillman
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Yahua Zhang
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - André Schneider
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Osamu Saito
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Linda S Davis
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Reyadh Redha
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Richard M Breyer
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| | - Matthew D Breyer
- Division of Nephrolgy, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
- Department of Molecular Physiology and Biophysics, Veterans Administration Medical Center, and Vanderbilt University School of Medicine, Nashville, Tennessee, USA37232-2372, USA
| |
Collapse
|
32
|
|
33
|
Katada J, Majima M. AT(2) receptor-dependent vasodilation is mediated by activation of vascular kinin generation under flow conditions. Br J Pharmacol 2002; 136:484-91. [PMID: 12055126 PMCID: PMC1573373 DOI: 10.1038/sj.bjp.0704731] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2002] [Revised: 03/12/2002] [Accepted: 03/22/2002] [Indexed: 12/13/2022] Open
Abstract
Physiological roles of angiotensin II type 2 receptor (AT(2)) are not well defined. This study was designed to investigate the mechanisms of AT(2)-dependent vascular relaxation by studying vasodilation in pressurized and perfused rat mesenteric arterial segments. Perfusion of angiotensin II in the presence of AT(1) antagonist elicited vascular relaxation, which was completely dependent on AT(2) receptors on endothelium. FR173657 (>1 microM), a bradykinin (BK) B(2)-specific antagonist, significantly suppressed AT(2)-dependent vasodilation (maximum inhibition: 68.5% at 10 microM). Kininogen-deficient Brown Norway Katholiek rats showed a significant reduction in AT(2)-mediated vasodilatory response compared with normal wild-type Brown Norway rats. Indomethacin (>1 microM), aprotinin (10 microM) and soybean trypsin inhibitor (10 microM) also reduced AT(2)-dependent vasodilation. Our results demonstrated that stimulation of AT(2) receptors caused a significant vasodilation through local production of BK in resistant arteries of rat mesentery in a flow-dependent manner. Such vasodilation counterbalances AT(1)-dependent vasoconstriction to regulate the vascular tone.
Collapse
Affiliation(s)
- Jun Katada
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Sagamihara, Kanagawa 228-8555, Japan.
| | | |
Collapse
|
34
|
Berry C, Touyz R, Dominiczak AF, Webb RC, Johns DG. Angiotensin receptors: signaling, vascular pathophysiology, and interactions with ceramide. Am J Physiol Heart Circ Physiol 2001; 281:H2337-65. [PMID: 11709400 DOI: 10.1152/ajpheart.2001.281.6.h2337] [Citation(s) in RCA: 183] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiotensin II (ANG II) is a pleiotropic vasoactive peptide that binds to two distinct receptors: the ANG II type 1 (AT(1)) and type 2 (AT(2)) receptors. Activation of the renin-angiotensin system (RAS) results in vascular hypertrophy, vasoconstriction, salt and water retention, and hypertension. These effects are mediated predominantly by AT(1) receptors. Paradoxically, other ANG II-mediated effects, including cell death, vasodilation, and natriuresis, are mediated by AT(2) receptor activation. Our understanding of ANG II signaling mechanisms remains incomplete. AT(1) receptor activation triggers a variety of intracellular systems, including tyrosine kinase-induced protein phosphorylation, production of arachidonic acid metabolites, alteration of reactive oxidant species activities, and fluxes in intracellular Ca(2+) concentrations. AT(2) receptor activation leads to stimulation of bradykinin, nitric oxide production, and prostaglandin metabolism, which are, in large part, opposite to the effects of the AT(1) receptor. The signaling pathways of ANG II receptor activation are a focus of intense investigative effort. We critically appraise the literature on the signaling mechanisms whereby AT(1) and AT(2) receptors elicit their respective actions. We also consider the recently reported interaction between ANG II and ceramide, a lipid second messenger that mediates cytokine receptor activation. Finally, we discuss the potential physiological cross talk that may be operative between the angiotensin receptor subtypes in relation to health and cardiovascular disease. This may be clinically relevant, inasmuch as inhibitors of the RAS are increasingly used in treatment of hypertension and coronary heart disease, where activation of the RAS is recognized.
Collapse
Affiliation(s)
- C Berry
- Department of Medicine and Therapeutics, Western Infirmary, University of Glasgow, G11 6NT Glasgow, United Kingdom.
| | | | | | | | | |
Collapse
|
35
|
Schuijt MP, de Vries R, Saxena PR, Jan Danser AH. Prostanoids, but not nitric oxide, counterregulate angiotensin II mediated vasoconstriction in vivo. Eur J Pharmacol 2001; 428:331-6. [PMID: 11689191 DOI: 10.1016/s0014-2999(01)01349-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To evaluate the modulating effects of nitric oxide and prostanoids during angiotensin II-mediated vasoconstriction, male Wistar rats (n=25) were infused with increasing doses of angiotensin II following pretreatment with the cyclooxygenase inhibitor indomethacin, the nitric oxide-synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME) plus sodium nitroprusside to restore mean arterial blood pressure, or saline. Hemodynamics were studied with the radioactive microsphere method. Indomethacin did not alter systemic or regional hemodynamics. L-NAME+sodium nitroprusside reduced cardiac output, as well as systemic and renal vascular conductance. Angiotensin II increased mean arterial blood pressure and heart rate, and decreased systemic vascular conductance as well as vascular conductance in gastrointestinal tract, kidney, skeletal muscle, skin, mesentery+pancreas, spleen and adrenal. Indomethacin enhanced the angiotensin II-mediated effects in all vascular beds, whereas L-NAME+sodium nitroprusside enhanced its effect in mesentery+pancreas only. In conclusion, vasodilator prostanoids, but not nitric oxide, counterregulate angiotensin II-mediated vasoconstriction in vivo.
Collapse
Affiliation(s)
- M P Schuijt
- Department of Pharmacology, Erasmus University Rotterdam, Room EE1418b, Dr. Molewaterplein 50, 3015 GE, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
36
|
Abstract
Renal cyclooxygenase 1 and 2 activity produces five primary prostanoids: prostaglandin E2, prostaglandin F2alpha, prostaglandin I2, thromboxane A2, and prostaglandin D2. These lipid mediators interact with a family of distinct G protein-coupled prostanoid receptors designated EP, FP, IP, TP, and DP, respectively, which exert important regulatory effects on renal function. The intrarenal distribution of these prostanoid receptors has been mapped, and the consequences of their activation have been partially characterized. FP, TP, and EP1 receptors preferentially couple to an increase in cell calcium. EP2, EP4, DP, and IP receptors stimulate cyclic AMP, whereas the EP3 receptor preferentially couples to Gi, inhibiting cyclic AMP generation. EP1 and EP3 mRNA expression predominates in the collecting duct and thick limb, respectively, where their stimulation reduces NaCl and water absorption, promoting natriuresis and diuresis. The FP receptor is highly expressed in the distal convoluted tubule, where it may have a distinct effect on renal salt transport. Although only low levels of EP2 receptor mRNA are detected in the kidney and its precise intrarenal localization is uncertain, mice with targeted disruption of the EP2 receptor exhibit salt-sensitive hypertension, suggesting that this receptor may also play an important role in salt excretion. In contrast, EP4 receptor mRNA is predominantly expressed in the glomerulus, where it may contribute to the regulation of glomerular hemodynamics and renin release. The IP receptor mRNA is highly expressed near the glomerulus, in the afferent arteriole, where it may also dilate renal arterioles and stimulate renin release. Conversely, TP receptors in the glomerulus may counteract the effects of these dilator prostanoids and increase glomerular resistance. At present there is little evidence for DP receptor expression in the kidney. These receptors act in a concerted fashion as physiological buffers, protecting the kidney from excessive functional changes during periods of physiological stress. Nonsteroidal anti-inflammatory drug (NSAID)-mediated cyclooxygenase inhibition results in the loss of these combined effects, which contributes to their renal effects. Selective prostanoid receptor antagonists may provide new therapeutic approaches for specific disease states.
Collapse
Affiliation(s)
- M D Breyer
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
37
|
Ruiz-Ortega M, Lorenzo O, Rupérez M, Blanco J, Egido J. Systemic infusion of angiotensin II into normal rats activates nuclear factor-kappaB and AP-1 in the kidney: role of AT(1) and AT(2) receptors. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:1743-56. [PMID: 11337372 PMCID: PMC1891960 DOI: 10.1016/s0002-9440(10)64130-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent studies have pointed out the implication of angiotensin II (Ang II) in various pathological settings. However, the molecular mechanisms and the AngII receptor (AT) subtypes involved are not fully identified. We investigated whether AngII elicited the in vivo activation of nuclear transcription factors that play important roles in the pathogenesis of renal and vascular injury. Systemic infusion of Ang II into normal rats increased renal nuclear factor (NF)-kappaB and AP-1 binding activity that was associated with inflammatory cell infiltration and tubular damage. Interestingly, infiltrating cells presented activated NF-kappaB complexes, suggesting the involvement of AngII in inflammatory cell activation. When rats were treated with AT(1) or AT(2) receptor antagonists different responses were observed. The AT(1) antagonist diminished NF-kappaB activity in glomerular and tubular cells and abolished AP-1 in renal cells, improved tubular damage and normalized the arterial blood pressure. The AT(2) antagonist diminished mononuclear cell infiltration and NF-kappaB activity in glomerular and inflammatory cells, without any effect on AP-1 and blood pressure. These data suggest that AT(1) mainly mediates tubular injury via AP-1/NF-kappaB, whereas AT(2) receptor participates in the inflammatory cell infiltration in the kidney by NF-kappaB. Our results provide novel information on AngII receptor signaling and support the recent view of Ang II as a proinflammatory modulator.
Collapse
Affiliation(s)
- M Ruiz-Ortega
- Laboratory of Vascular and Renal Pathology, Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain.
| | | | | | | | | |
Collapse
|
38
|
Igarashi R, Takenaga M, Takeuchi J, Kitagawa A, Matsumoto K, Mizushima Y. Marked hypotensive and blood flow-increasing effects of a new lipo-PGE(1) (lipo-AS013) due to vascular wall targeting. J Control Release 2001; 71:157-64. [PMID: 11274747 DOI: 10.1016/s0168-3659(00)00373-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipo-AS013 is being developed as an improved formulation of lipo-PGE(1), which is widely used in clinical practice in Japan and some Asian countries. We have previously reported that lipo-AS013, which is a lipid microsphere (LM) preparation of a chemically stable and lipophilic PGE(1) prodrug (AS013, Fig. 1), slowly releases small amounts of the active ingredient (AS013) in human plasma. In the present study, to estimate the vascular wall targeting ability and efficacy of lipo-AS013, we determined the hypotensive and blood flow-increasing effects of lipo-AS013, lipo-PGE(1), PGE(1)CD, and AS013. Lipo-AS013 was found to have longer-lasting hypotensive and blood flow-increasing effects than the other agents. The two LM preparations, lipo-PGE(1) and lipo-AS013, had a markedly stronger effect than PGE(1)CD and AS013 alone, demonstrating the benefit of drug delivery using LM. In spontaneously hypertensive rats (SHR), lipo-AS013 also had a significant hypotensive effect. To confirm vascular wall targeting by lipo-AS013, the localization of PGE(1) in the aorta and neovascular capillaries of rat was investigated by immunostaining. The results indicated that lipo-AS013 was more efficient at delivering the active ingredient (AS013) to the vessel wall. In conclusion, lipo-AS013 could supersede lipo-PGE(1) and PGE(1)CD in clinical use.
Collapse
Affiliation(s)
- R Igarashi
- The Second Department of the Institute of Medical Science, St Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, 216-8512, Kawasaki, Japan.
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
In both diabetic and nondiabetic renal disease, reducing blood pressure with antihypertensive therapy has beneficial effects on renal function. The key role of the renin-angiotensin system in blood pressure and volume homeostasis has long been established, but its importance for the overall normal functioning of the kidney itself is also increasingly being recognized. Angiotensin-converting enzyme (ACE) inhibitors, widely and successfully used in the treatment of hypertension, may also provide renal protection independent of blood pressure reduction; however, their relatively nonspecific mode of action in blocking an early metabolic step entails major clinical disadvantages, such as accumulation of bradykinin and substance P, that may cause the characteristic ACE-inhibitor side effects of persistent dry cough and, more rarely, angioneurotic edema. Angiotensin II antagonists or receptor blockers, a new class of antihypertensive agent, selectively antagonize the AT1 receptor subtype and, because of greater specificity, do not give rise to the side effects associated with ACE inhibitors. More important, these new drugs may have mechanistic advantages over other antihypertensives, including ACE inhibitors.
Collapse
Affiliation(s)
- L M Ruilope
- Hospital 12 de Octubre, Nephrology Service, Madrid, Spain
| |
Collapse
|
40
|
Brede M, Hein L. Transgenic mouse models of angiotensin receptor subtype function in the cardiovascular system. REGULATORY PEPTIDES 2001; 96:125-32. [PMID: 11111018 DOI: 10.1016/s0167-0115(00)00168-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Angiotensin II mediates is biological actions via different subtypes of G protein-coupled receptors, termed AT(1) and AT(2) receptors. In rodents, two AT(1) receptors have been identified, AT(1A) and AT(1B), whereas in humans a single AT(1) receptor exists. Recently, a number of transgenic animal models have been generated which overexpress or lack functional angiotensin II receptor subtypes. This review focuses on the physiological significance of angiotensin II receptor subtype diversity in the cardiovascular system. In the mouse, AT(1A) receptors are the major regulators of cardiovascular homeostasis by determining vascular tone and natriuresis. In addition, AT(1A) receptors mediate growth-stimulating signals in vascular and cardiac myocytes. AT(1B) receptors participate in blood pressure regulation, and their functions become apparent when the AT(1A) receptor gene is deleted. Deletion of the mouse gene for the AT(2) receptor subtype led to hypersensitivity to pressor and antinatriuretic effects of angiotensin II in vivo, suggesting that the AT(2) receptor subtype counteracts some of the biological effects of AT(1) receptor signalling.
Collapse
Affiliation(s)
- M Brede
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
| | | |
Collapse
|
41
|
Al-Qattan KK, Khan I, Alnaqeeb MA, Ali M. Thromboxane-B2, prostaglandin-E2 and hypertension in the rat 2-kidney 1-clip model: a possible mechanism of the garlic induced hypotension. Prostaglandins Leukot Essent Fatty Acids 2001; 64:5-10. [PMID: 11161580 DOI: 10.1054/plef.2000.0232] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Serum collected from unilaterally clipped and unclipped rats before and after treatment with water, garlic or cilazapril and subsequent to measuring blood pressure was assayed for thromboxane-B2 and prostaglandin-E2. The unclipped rats' thromboxane-B2 and prostaglandin-E2 levels were about 23 ng/ml and 2 ng/ml, respectively, and blood pressure was 126+/-3 mmHg. These values were not affected by either water or garlic administration. The clipped rats' thromboxane-B2 and prostaglandin-E2 concentrations were close to 34 ng/ml and 4 ng/ml, respectively, and declined only in response to garlic (by 15 ng/ml and 3 ng/ml) and cilazapril (by 12 ng/ml and 1.5 ng/ml). The blood pressure of these rats was 196+/-7 mmHg and again was reduced only by garlic to 169+/-14 mmHg and cilazapril to 137+/-5 mmHg. The no-treatment and water-treatment readings were significantly higher in the clipped rats. The data suggest that prostanoid system activity in the 2-kidney 1-clip rat is enhanced and mostly toward maintaining the hypertension. Furthermore, the blood pressure lowering effects of garlic and cilazapril might have been induced partially by a greater reduction in the synthesis of vasoconstrictor prostanoids.
Collapse
Affiliation(s)
- K K Al-Qattan
- Department of Biological Sciences, Faculty of Science, Kuwait University, P.O. Box 5969, Al-Safat 13060, Kuwait.
| | | | | | | |
Collapse
|
42
|
Siragy HM, Carey RM. Angiotensin type 2 receptors: potential importance in the regulation of blood pressure. Curr Opin Nephrol Hypertens 2001; 10:99-103. [PMID: 11195059 DOI: 10.1097/00041552-200101000-00015] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The angiotensin type 2 receptor is one of two major angiotensin II receptors that has been identified, cloned and sequenced. The other major receptor, the angiotensin type 1 receptor, is thought to mediate most of the biological responses to the peptide. The angiotensin type 2 receptor is expressed heavily in fetal tissues, but only at a low level in the adult. Documented angiotensin type 2 receptor expression sites in the adult include kidney, heart and mesenteric blood vessels. The function of the angiotensin type 2 receptor is just beginning to be explored. Most of the evidence suggests that the angiotensin type 2 receptor mediates a vasodilator signalling cascade that includes bradykinin, nitric oxide and cyclic guanosine 5-monophosphate. At least some of the beneficial actions of angiotensin type 1 receptor blockade, such as hypotension, are mediated by stimulation of the angiotensin type 2 receptor. Several recent papers suggest that angiotensin type 2 receptors, presumably located in systemic blood vessels, mediate vasodilation and hypotension. The angiotensin type 2 receptor may be a new therapeutic target and candidate gene for the pathophysiology of hypertension.
Collapse
Affiliation(s)
- H M Siragy
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, USA.
| | | |
Collapse
|
43
|
Angiotensin II subtype-2 receptor influences kininogenase activity. Am J Kidney Dis 2000; 36:1071-4. [PMID: 11054369 DOI: 10.1053/ajkd.2000.19111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Audoly LP, Oliverio MI, Coffman TM. Insights into the functions of type 1 (AT1) angiotensin II receptors provided by gene targeting. Trends Endocrinol Metab 2000; 11:263-9. [PMID: 10920382 DOI: 10.1016/s1043-2760(00)00279-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The renin-angiotensin system (RAS) has a wide range of actions in biological processes ranging from development and reproduction to cardiovascular and renal functions. Most of these actions are mediated by the octapeptide hormone angiotensin II. The identified family of angiotensin II receptors is divided into two pharmacological classes: type 1 (AT1) and type 2 (AT2). The classically recognized actions of the RAS are primarily mediated by the AT1 subtype of angiotensin receptors, and these receptors are the targets of a new class of anti-hypertensive agents. In recent years, our understanding of the physiological functions of AT1 receptors has been advanced through the use of gene-targeting technology. In this review, we will summarize the emerging picture of AT1 receptor functions that has been provided by gene-targeting experiments.
Collapse
Affiliation(s)
- L P Audoly
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, 508 Fulton Street, Durham NC 27710, USA
| | | | | |
Collapse
|
45
|
Abstract
All components of the renin-angiotensin system (RAS) are present in the kidneys and constitute a functioning renal RAS. Angiotensin II (Ang II) receptor subtypes AT(1) and AT(2) have been identified in the afferent and efferent arterioles, glomeruli, mesangial cells, and proximal tubules. AT(1) receptors regulate vasoconstriction and sodium and water reabsorption, as well as promote cell growth, proliferation, and collagen matrix deposition. Recent animal studies are elucidating the role of the less well understood AT(2) receptors. The AT(2) receptors appear to counterbalance the AT(1) receptors by increasing the production of bradykinin, nitric oxide, and cyclic guanosine monophosphate-mediating vasodilation and by promoting cell differentiation, antiproliferation, and apoptosis. Ang II subtype 1 receptor blockers prevent Ang II activation of the AT(1) receptor while leaving the AT(2) receptor open to Ang II stimulation.
Collapse
Affiliation(s)
- H M Siragy
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
46
|
Gross V, Milia AF, Plehm R, Inagami T, Luft FC. Long-term blood pressure telemetry in AT2 receptor-disrupted mice. J Hypertens 2000; 18:955-61. [PMID: 10930194 DOI: 10.1097/00004872-200018070-00018] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVES The hypertension in AT2 receptor knockout mice is imperfectly defined. Therefore, we investigated the influence of dietary salt loading and deoxycorticosterone (DOCA)-salt treatment on blood pressure and diurnal patterns of blood pressure in these mice by radiotelemetry. METHODS We used telemetry in AT2 receptor knockout and wild-type mice to measure blood pressure, heart rate, aortic pressure dp/dt, locomotor activity, and circadian rhythms. Salt-related effects were studied by increasing the salt in chow to 4%, adding 1% saline in drinking water, and by DOCA-salt treatment RESULTS Baseline blood pressures were higher in AT2 receptor knockout than in wild-type mice and were not affected by increasing the salt intake. The blood pressure increase was steeper and greater in AT2 receptor knockout than in wild-type mice after DOCA-salt treatment A circadian rhythm of blood pressure and heart rate, with higher values during the night, was seen in wild-type, but not in AT2 receptor knockout mice. In AT2 receptor knockout mice, this rhythm was only significant when daily salt intake was increased or when DOCA-salt hypertension was induced. The acrophase of blood pressure and heart rate was found between 2000 and 2400 h and was in accordance with the maximum physical activity. CONCLUSION These data suggest that AT2 knockout mice display slight hypertension which is not salt-sensitive. On the other hand, the susceptibility to develop DOCA-salt hypertension is increased. The study also illustrates the power of telemetry in monitoring long-term cardiovascular changes and circadian blood pressure and heart rate rhythms in genetically engineered mice.
Collapse
Affiliation(s)
- V Gross
- Franz Volhard Clinic and Max Delbrück Center for Molecular Medicine, Medical Faculty of the Charité, Humboldt University of Berlin, Germany
| | | | | | | | | |
Collapse
|
47
|
Zhang Y, Guan Y, Schneider A, Brandon S, Breyer RM, Breyer MD. Characterization of murine vasopressor and vasodepressor prostaglandin E(2) receptors. Hypertension 2000; 35:1129-34. [PMID: 10818076 DOI: 10.1161/01.hyp.35.5.1129] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Four E-prostanoid (EP) receptors, designated EP(1), EP(2), EP(3), and EP(4), mediate the cellular effects of prostaglandin E(2) (PGE(2)). The present studies pharmacologically characterize the vasopressor and vasodepressor EP receptors in wild-type mice (EP(2)(+/+) mice) and mice with targeted disruption of the EP(2) receptor (EP(2)(-/-) mice). Mean arterial pressure (MAP) was measured via a carotid artery catheter in anesthetized male mice. Intravenous infusion of PGE(2) decreased MAP in EP(2)(+/+) mice but increased MAP in EP(2)(-/-) mice. Infusion of EP(3)-selective agonists, including MB28767, SC46275, and sulprostone, increased MAP in both EP(2)(+/+) and EP(2)(-/-) mice. Pretreatment with SC46275 desensitized mice to the subsequent pressor effect of sulprostone, but the vasodepressor effect of PGE(2) in EP(2)(+/+) mice remained intact. Although PGE(2) alone increased MAP in EP(2)(-/-) mice, prior desensitization of the pressor effect with SC46275 allowed a residual vasodepressor effect of PGE(2) to be seen in the EP(2)(-/-) mice. An EP(4)-selective agonist (prostaglandin E(1)-OH) functioned also as a vasodepressor in both EP(2)(-/-) and EP(2)(+/+) mice. High levels of EP(3) receptor mRNA were detected in mouse aortas and rabbit preglomerular arterioles by nuclease protection, with lower expressions of EP(1), EP(2), and EP(4) mRNA. The findings suggest that combined vasodepressor effects of EP(2) and EP(4) receptors normally dominate, accounting for the depressor effects of PGE(2). In contrast, in EP(2)(-/-) mice, EP(4) receptor activity alone is insufficient to overcome the EP(3) vasopressor effect. These findings suggest that a balance between pressor and depressor PGE(2) receptors determines its net effect on arterial pressure and that these receptors may be important therapeutic targets.
Collapse
Affiliation(s)
- Y Zhang
- Division of Nephrology, Department of Molecular Physiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Many peptides influence renal function and structure in physiological and pathophysiological situations. Bioactive peptides that regulate renal function and structure encompass various substances including vasopeptides, growth factors, cytokines and peptide hormones. We highlight some novel concepts indicating that the vasoactive peptides angiotensin II and endothelin-1 play a major role in the progression of renal disease. These effects may be amplified by reduced concentration of counteracting natriuretic peptides. In addition, recent evidence suggests that peptides such as leptin, previously not considered to exert any renal effects, may be involved in renal pathophysiology under certain conditions. One of the most imperative tasks in nephrology is to develop innovative strategies to slow the progression of chronic renal disease. Interference with the renal action of bioactive peptides will certainly be part of this strategy.
Collapse
Affiliation(s)
- G Wolf
- Department of Medicine, University of Hamburg, Germany
| | | | | |
Collapse
|
49
|
Carey RM, Wang ZQ, Siragy HM. Update: role of the angiotensin type-2 (AT(2)) receptor in blood pressure regulation. Curr Hypertens Rep 2000; 2:198-201. [PMID: 10981149 DOI: 10.1007/s11906-000-0082-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In the past, virtually all of the physiologic actions of angiotensin II (ANG II) were thought to be mediated by the type-1 ANG II receptor. However, there is now a compelling body of evidence suggesting that the type-2 (AT2) receptor is an important regulator of renal function and blood pressure (BP). The AT2 receptor stimulates a bradykinin (BK)-nitric oxide (NO)-cyclic GMP vasodilator cascade in blood vessels and in the kidney. Recent studies have shown that absence of the AT2 receptor lends to pressor and natriuretic hypersensitivity to ANG II. Furthermore, there is now excellent evidence that the AT2 receptor mediates pressure natriuresis. The AT2 receptor also stimulates the conversion of prostaglandin E2 (PGE2) to PGF2. In addition, it is now apparent that the therapeutic reduction in BP with AT1 receptor blockade (eg, losartan, valsartan, candesartan) is mediated by ANG II stimulation of the AT2 receptor, leading to increased levels of BK, NO, and cGMP. Current evidence predicts that AT2 receptor agonists would be beneficial in the treatment of hypertension.
Collapse
Affiliation(s)
- R M Carey
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
50
|
Arima S, Ito S. Angiotensin II type 2 receptors in the kidney: evidence for endothelial-cell-mediated renal vasodilatation. Nephrol Dial Transplant 2000; 15:448-51. [PMID: 10727534 DOI: 10.1093/ndt/15.4.448] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
MESH Headings
- Angiotensin II/antagonists & inhibitors
- Angiotensin II/metabolism
- Angiotensin Receptor Antagonists
- Animals
- Antihypertensive Agents/pharmacology
- Cell Division
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Humans
- Hypertension, Renal/metabolism
- Hypertension, Renal/pathology
- Hypertension, Renal/prevention & control
- Kidney/blood supply
- Kidney/drug effects
- Kidney/metabolism
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Angiotensin/metabolism
- Renal Circulation/drug effects
- Renal Circulation/physiology
- Signal Transduction
- Vasodilation/drug effects
- Vasodilation/physiology
- Vasodilator Agents/pharmacology
Collapse
|