1
|
Grano de Oro A, Kumariya S, Mell B, Zubcevic J, Joe B, Osman I. Spontaneous vascular dysfunction in Dahl salt-sensitive male rats raised without a high-salt diet. Physiol Rep 2024; 12:e16165. [PMID: 39048525 PMCID: PMC11268988 DOI: 10.14814/phy2.16165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Dahl salt-sensitive (SS) rats fed a high-salt diet, but not low-salt, exhibit vascular dysfunction. Several substrains of SS rats exist that differ in their blood pressure phenotypes and salt sensitivity. The goal of this study was to investigate whether the John-Rapp-derived SS rat (SS/Jr), which exhibits spontaneous hypertension on a low-salt diet, presents with hallmarks of vascular dysfunction observed in another experimental model of hypertension independent of dietary salt, the spontaneously hypertensive rat (SHR). Endothelium-intact aortic rings and mesenteric resistance arteries were isolated from low-salt fed adult male SS/Jr rats and SHRs, or their respective controls, for isometric wire myography. Vessels were challenged with cumulative concentrations of various vasoactive substances, in the absence or presence of nitric oxide synthase or cyclooxygenase inhibitors. Despite showing some differences in their responses to various vasoactive substances, both SS/Jr rats and SHRs exhibited key features of vascular dysfunction, including endothelial dysfunction and hyperresponsiveness to vasocontractile agonists. In conclusion, this study provides evidence to support the utility of the SS/Jr rat strain maintained on a low-salt diet as a valid experimental model for vascular dysfunction, a key feature of human hypertension.
Collapse
Affiliation(s)
- Arturo Grano de Oro
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Sanjana Kumariya
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Blair Mell
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Bina Joe
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| | - Islam Osman
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized MedicineUniversity of Toledo, College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
2
|
Feng W, Guan Z, Ying WZ, Xing D, Ying KE, Sanders PW. Matrix metalloproteinase-9 regulates afferent arteriolar remodeling and function in hypertension-induced kidney disease. Kidney Int 2023; 104:740-753. [PMID: 37423509 PMCID: PMC10854403 DOI: 10.1016/j.kint.2023.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/01/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
This study tested if matrix metalloproteinase (MMP)-9 promoted microvascular pathology that initiates hypertensive (HT) kidney disease in salt-sensitive (SS) Dahl rats. SS rats lacking Mmp9 (Mmp9-/-) and littermate control SS rats were studied after one week on a normotensive 0.3% sodium chloride (Pre-HT SS and Pre-HT Mmp9-/-) or a hypertension-inducing diet containing 4.0% sodium chloride (HT SS and HT Mmp9-/-). Telemetry-monitored blood pressure of both the HT SS and HT Mmp9-/- rats increased and did not differ. Kidney microvessel transforming growth factor-beta 1 (Tgfb1) mRNA did not differ between Pre-HT SS and Pre-HT Mmp9-/- rats, but with hypertension and expression of Mmp9 and Tgfb1 increased in HT SS rats, along with phospho-Smad2 labeling of nuclei of vascular smooth muscle cells, and with peri-arteriolar fibronectin deposition. Loss of MMP-9 prevented hypertension-induced phenotypic transformation of microvascular smooth muscle cells and the expected increased microvascular expression of pro-inflammatory molecules. Loss of MMP-9 in vascular smooth muscle cells in vitro prevented cyclic strain-induced production of active TGF-β1 and phospho-Smad2/3 stimulation. Afferent arteriolar autoregulation was impaired in HT SS rats but not in HT Mmp9-/- rats or the HT SS rats treated with doxycycline, an MMP inhibitor. HT SS but not HT Mmp9-/- rats showed decreased glomerular Wilms Tumor 1 protein-positive cells (a marker of podocytes) along with increased urinary podocin and nephrin mRNA excretion, all indicative of glomerular damage. Thus, our findings support an active role for MMP-9 in a hypertension-induced kidney microvascular remodeling process that promotes glomerular epithelial cell injury in SS rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhengrong Guan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei-Zhong Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kai Er Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA.
| |
Collapse
|
3
|
Zicha J, Řezáčová L, Vaněčková I. Nitric oxide and salt resistance in Dahl rats: no role of inducible NO synthase. Physiol Res 2023; 72:123-127. [PMID: 36545880 PMCID: PMC10069813 DOI: 10.33549/physiolres.935047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Inducible NO synthase (NOS II) was proposed to play an important role in salt resistance of Dahl salt-resistant (SR/Jr) rats. Its chronic inhibition by specific inhibitors was accompanied by blood pressure (BP) elevation in animals subjected to high salt intake. The aim of our study was to evaluate 1) whether such inhibitors affect BP and/or its particular components (sympathetic tone and NO-dependent vasodilation) only under the conditions of high salt intake, and 2) whether similar BP effects are elicited after systemic or intracerebroventricular (icv) application of these inhibitors. Wistar rats fed Altromin diet (0.45 % NaCl) and SR/Jr rats fed either a low-salt (LS, 0.3 % NaCl) or a high-salt (HS, 4 % NaCl) diet were studied. Aminoguanidine (AMG) and 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (AMT) were used as NOS II inhibitors. BP and its responses to acute blockade of renin-angiotensin system (captopril), sympathetic nervous system (pentolinium) and NO synthase (L-NAME) were measured in conscious cannulated rats. There were no significant changes of BP or its components in either Wistar rats or SR/Jr rats subjected to chronic inhibition of NOS II by peroral aminoguanidine administration (50 mg/kg/day for 4 weeks). This was true for SR/Jr rats fed either LS or HS diets. Furthermore, we have studied BP effects of chronic icv administration of both NOS II inhibitors in SR/Jr rats fed HS diet, but we failed to find any BP changes elicited by such treatment. In conclusion, inducible NO synthase does not participate in the resistance of SR/Jr rats to hypertensive effects of excess salt intake.
Collapse
Affiliation(s)
- J Zicha
- Laboratory of Experimental Hypertension, Institute of Physiology of the Czech Academy of Sciences, Prague 4, Czech Republic.
| | | | | |
Collapse
|
4
|
Koch V, Gruenewald LD, Gruber-Rouh T, Eichler K, Leistner DM, Mahmoudi S, Booz C, Bernatz S, D'Angelo T, Albrecht MH, Alizadeh LS, Nour-Eldin NEA, Scholtz JE, Yel I, Vogl TJ, März W, Hardt SE, Martin SS. Homoarginine in the cardiovascular system: Pathophysiology and recent developments. Fundam Clin Pharmacol 2022; 37:519-529. [PMID: 36509694 DOI: 10.1111/fcp.12858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Upcoming experimental and epidemiological data have identified the endogenous non-proteinogenic amino acid L-homoarginine (L-hArg) not only as a novel biomarker for cardiovascular disease but also as being directly involved in the pathogenesis of cardiac dysfunction. The association of low L-hArg levels with adverse cardiovascular events and mortality has proposed the idea of nutritional supplementation to rescue pathways inversely associated with cardiovascular health. Subsequent clinical and experimental studies contributed significantly to our knowledge of potential effects on the cardiorenal axis, acting either as a biomarker or a cardiovascular active agent. In this review article, we provide a comprehensive summary of the L-hArg metabolism, pathophysiological aspects, and current developments in the field of experimental and clinical evidence in favor of protective cardiovascular effects. Establishing a reliable biomarker to identify patients at high risk to die of cardiovascular disease represents one of the main goals for tackling this disease and providing individual therapeutic guidance.
Collapse
Affiliation(s)
- Vitali Koch
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.,Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Katrin Eichler
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - David M Leistner
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Christian Booz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Simon Bernatz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tommaso D'Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University Hospital Messina, Messina, Italy
| | | | - Leona S Alizadeh
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Jan-Erik Scholtz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ibrahim Yel
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Winfried März
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon S Martin
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
5
|
Koch V, Gruenewald LD, Gruber‐Rouh T, Martin S, Eichler K, Booz C, Yel I, Vogl TJ, Buchner K, Hagenmueller M, März W, Frey N, Hardt SE, Riffel JH. Homoarginine treatment of rats improves cardiac function and remodeling in response to pressure overload. Fundam Clin Pharmacol 2022; 36:992-1004. [PMID: 35697514 DOI: 10.1111/fcp.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Vitali Koch
- Goethe University Frankfurt Frankfurt am Main Germany
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | | | | | - Simon Martin
- Goethe University Frankfurt Frankfurt am Main Germany
| | | | | | - Ibrahim Yel
- Goethe University Frankfurt Frankfurt am Main Germany
| | | | - Kristina Buchner
- Institute of Human Genetics, Section for Developmental Genetics University of Heidelberg Heidelberg Germany
| | - Marco Hagenmueller
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Winfried März
- Synlab Academy Synlab Holding Deutschland GmbH Augsburg Germany
| | - Norbert Frey
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Stefan E. Hardt
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| | - Johannes H. Riffel
- Department of Cardiology, Angiology, and Pulmonology Heidelberg University Hospital Heidelberg Germany
| |
Collapse
|
6
|
Koch V, Weber C, Riffel JH, Buchner K, Buss SJ, Hein S, Mereles D, Hagenmueller M, Erbel C, März W, Booz C, Albrecht MH, Vogl TJ, Frey N, Hardt SE, Ochs M. Impact of Homoarginine on Myocardial Function and Remodeling in a Rat Model of Chronic Renal Failure. J Cardiovasc Pharmacol Ther 2022; 27:10742484211054620. [PMID: 34994208 DOI: 10.1177/10742484211054620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE Low plasma concentrations of the amino acid homoarginine (HA) have been shown to correlate with adverse cardiovascular outcome, particularly in patients with chronic kidney disease. The present study sought to investigate the effect of HA treatment on cardiac remodeling in rats undergoing artificially induced renal insufficiency by 5/6 nephrectomy (5/6 Nx). METHODS A total of 33 male Wistar rats were randomly divided into sham and 5/6 Nx groups, receiving either placebo treatment or 400 mg·kg-1·day-1 HA over a 4-week period. RESULTS 5/6 Nx per se resulted in adverse myocardial remodeling with aggravated cardiac function and associated cardiac overload as the most obvious alteration (-23% ejection fraction, P < 0.0001), as well as increased myocardial fibrosis (+80%, P = 0.0005) compared to placebo treated sham animals. HA treatment of 5/6 Nx rats has led to an improvement of ejection fraction (+24%, P = 0.0003) and fractional shortening (+21%, P = 0.0126), as well as a decrease of collagen deposition (-32%, P = 0.0041), left ventricular weight (-14%, P = 0.0468), and myocyte cross-sectional area (-12%, P < 0.0001). These changes were accompanied by a downregulation of atrial natriuretic factor (-65% P < 0.0001) and collagen type V alpha 1 chain (-44%, P = 0.0006). Sham animals revealed no significant changes in cardiac function, myocardial fibrosis, or any of the aforementioned molecular changes after drug treatment. CONCLUSION Dietary HA supplementation appears to have the potential of preventing cardiac remodeling and improving heart function in the setting of chronic kidney disease. Our findings shed new light on HA as a possible new therapeutic agent for patients at high cardiovascular risk.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christophe Weber
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes H Riffel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Kristina Buchner
- Institute of Human Genetics, Section for Developmental Genetics, 27178University of Heidelberg, Heidelberg, Germany
| | - Sebastian J Buss
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Selina Hein
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Derliz Mereles
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Hagenmueller
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Erbel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried März
- Synlab Academy, Synlab Holding Deutschland GmbH, Augsburg, Germany
| | - Christian Booz
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Thomas J Vogl
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Ochs
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Zheng X, Zhou L, Jin Y, Zhao X, Ahmad H, OuYang Y, Chen S, Du J, Chen X, Chen L, Gao D, Yang Z, Tian Z. β-Aminoisobutyric acid supplementation attenuated salt-sensitive hypertension in Dahl salt-sensitive rats through prevention of insufficient fumarase. Amino Acids 2021; 54:169-180. [PMID: 34837556 DOI: 10.1007/s00726-021-03092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
The human Dietary Approaches to Stop Hypertension-Sodium Trial has shown that β-aminoisobutyric acid (BAIBA) may prevent the development of salt-sensitive hypertension (SSHT). However, the specific antihypertensive mechanism remains unclear in the renal tissues of salt-sensitive (SS) rats. In this study, BAIBA (100 mg/kg/day) significantly attenuated SSHT via increased nitric oxide (NO) content in the renal medulla, and it induced a significant increase in NO synthesis substrates (L-arginine and malic acid) in the renal medulla. BAIBA enhanced the activity levels of total NO synthase (NOS), inducible NOS, and constitutive NOS. BAIBA resulted in increased fumarase activity and decreased fumaric acid content in the renal medulla. The high-salt diet (HSD) decreased fumarase expression in the renal cortex, and BAIBA increased fumarase expression in the renal medulla and renal cortex. Furthermore, in the renal medulla, BAIBA increased the levels of ATP, ADP, AMP, and ADP/ATP ratio, thus further activating AMPK phosphorylation. BAIBA prevented the decrease in renal medullary antioxidative defenses induced by the HSD. In conclusion, BAIBA's antihypertensive effect was underlined by the phosphorylation of AMPK, the prevention of fumarase's activity reduction caused by the HSD, and the enhancement of NO content, which in concert attenuated SSHT in SS rats.
Collapse
Affiliation(s)
- Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Luxin Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuexin Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinrui Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yanan OuYang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Sa Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jie Du
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiangbo Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lan Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
8
|
Mokhaneli MC, Botha-Le Roux S, Fourie CMT, Böger R, Schwedhelm E, Mels CMC. L-homoarginine is associated with decreased cardiovascular- and all-cause mortality. Eur J Clin Invest 2021; 51:e13472. [PMID: 33320332 DOI: 10.1111/eci.13472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Increasing evidence suggests that L-homoarginine, an endogenous analogue of the amino acid L-arginine, may have beneficial effects on vascular homeostasis. We examined whether L-homoarginine is associated with 10-year risk of all-cause and cardiovascular mortality in a black South African population. METHODS We included 669 black South African participants (mean age 59.5 years), 143 of whom died during the 10-year follow-up period. Mortality data were acquired via verbal autopsy. Plasma L-homoarginine (and other related markers) were analysed with liquid chromatography-tandem mass spectrometry. RESULTS Survivors had higher L-homoarginine levels compared with nonsurvivors (1.25 µM vs. 0.89 µM; P < .001). Multivariable Cox regression analyses revealed that higher plasma L-homoarginine predicted a reduction in 10-year cardiovascular (hazard ratio [HR] per SD increment, 0.61; 95% CI 0.50 to 0.75) and all-cause (hazard ratio [HR] per SD increment, 0.59; 95% CI 0.41 to 0.84) mortality risk. CONCLUSION Higher L-homoarginine levels are associated with reduced risk of 10-year cardiovascular and all-cause mortality. Regulation of L-homoarginine levels as a therapeutic target in the management of cardiovascular disease should be investigated.
Collapse
Affiliation(s)
- Maserame Cleopatra Mokhaneli
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Carla Maria Theresia Fourie
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Rainer Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Catharina Martha Cornelia Mels
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
9
|
Abstract
Hypertension is a leading risk factor for disease burden worldwide. The kidneys, which have a high specific metabolic rate, play an essential role in the long-term regulation of arterial blood pressure. In this review, we discuss the emerging role of renal metabolism in the development of hypertension. Renal energy and substrate metabolism is characterized by several important and, in some cases, unique features. Recent advances suggest that alterations of renal metabolism may result from genetic abnormalities or serve initially as a physiological response to environmental stressors to support tubular transport, which may ultimately affect regulatory pathways and lead to unfavorable cellular and pathophysiological consequences that contribute to the development of hypertension.
Collapse
Affiliation(s)
- Zhongmin Tian
- grid.43169.390000 0001 0599 1243The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Mingyu Liang
- grid.30760.320000 0001 2111 8460Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI USA
| |
Collapse
|
10
|
Effects of Angiotensin-(1-7) and Angiotensin II on Acetylcholine-Induced Vascular Relaxation in Spontaneously Hypertensive Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6512485. [PMID: 31827689 PMCID: PMC6886389 DOI: 10.1155/2019/6512485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023]
Abstract
Endothelial dysfunction of small arteries occurs in patients with hypertension and in various hypertensive models. Endothelial function is usually evaluated by the degree of acetylcholine- (ACh-) induced vascular relaxation. Our previous study has found that compared to Wistar-Kyoto rats (WKY), ACh-induced vasodilatation was attenuated significantly in the mesenteric artery (MA), coronary artery (CA), and pulmonary artery (PA) of spontaneously hypertensive rats (SHR). This study investigated the influence of angiotensin- (Ang-) (1-7) and Ang II on blood pressure and ACh-induced vascular relaxation, as well as their interactive roles and downstream signal pathways in SHR and WKY. Intravenous injection of Ang II significantly increased, while Ang-(1-7) decreased the mean arterial pressure (MAP) in SHR. Ang-(1-7) improved ACh-induced relaxation in the MA, CA, and PA of SHR, while Ang II further attenuated it, which were inhibited by pretreatment with Mas receptor antagonist A-779 or AT1 receptor antagonist losartan, respectively. Ang-(1-7) decreased the basal arterial tension, and Ang II induced great vasoconstriction in SHR. Pretreatment with Ang-(1-7) inhibited the Ang II-induced pressor response, vasoconstriction, and the effects on ACh-induced relaxation in SHR. AT1 receptor expression was higher, while nitric oxide (NO), cGMP, and protein kinase G (PKG) levels of arteries were lower in SHR than in WKY. Ang II decreased, while Ang-(1-7) increased, the levels of NO, cGMP, and PKG of arteries. In addition, pretreatment with Ang-(1-7) inhibited the Ang II-induced reduction of NO, cGMP, and PKG in SHR. These results indicate that the activation of the Mas receptor by Ang-(1-7) can improve endothelial function and decrease MAP in SHR and inhibit the deteriorative effect of Ang II on endothelial function through the NO-cGMP-PKG pathway.
Collapse
|
11
|
Angiotensin-(1-7) induced vascular relaxation in spontaneously hypertensive rats. Nitric Oxide 2019; 88:1-9. [PMID: 30880106 DOI: 10.1016/j.niox.2019.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 11/20/2022]
Abstract
Enhanced vasoconstriction and decreased vasodilatation due to endothelial dysfunction contribute to the progression of hypertension. Angiotensin (Ang)-(1-7) plays important roles in regulating the cardiovascular activity. The current study aimed to investigate the roles of Ang-(1-7) in modulating blood pressure, vascular tension and its signal pathway in spontaneously hypertensive rats (SHR). The effects of intravenous injection of drugs were determined in rats with anesthesia in vivo. Mesenteric artery (MA), coronary artery (CA) and pulmonary artery (PA) were isolated from rats and isometric tension measurements in arteries were performed. Compared with Wistar-Kyoto rats (WKY), the high K+ induced vasoconstriction was enhanced and acetylcholine-induced vasodilatation were attenuated in the MA, CA and PA in SHR. Intravenous injection of Ang-(1-7) decreased, while A-779 increased mean arterial pressure and abolished the hypotensive effect of Ang-(1-7) in SHR. Ang-(1-7) caused dose-dependent relaxation in MA, CA and PA in SHR, which was inhibited by pretreatment with Mas receptor antagonist A-779, nitric oxide (NO) synthase inhibitor l-NAME, guanylate cyclase inhibitor ODQ and protein kinase G (PKG) inhibitor DT-2. The Mas receptor expression, NO, cGMP and PKG levels of the three above arteries of SHR were lower than that of WKY. Ang-(1-7) increased the NO, cGMP and PKG levels in arteries from SHR, which was blocked by A-779. Activation of the Mas receptor by Ang-(1-7) relaxes the MA, CA, and PA through the NO-cGMP-PKG pathway, which contributes to the decrease of arterial pressure in SHR.
Collapse
|
12
|
Tommasi S, Elliot DJ, Da Boit M, Gray SR, Lewis BC, Mangoni AA. Homoarginine and inhibition of human arginase activity: kinetic characterization and biological relevance. Sci Rep 2018; 8:3697. [PMID: 29487337 PMCID: PMC5829263 DOI: 10.1038/s41598-018-22099-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/16/2018] [Indexed: 01/27/2023] Open
Abstract
The inhibition of arginase, resulting in higher arginine (ARG) availability for nitric oxide synthesis, may account for the putative protective effect of homoarginine (HOMOARG) against atherosclerosis and cardiovascular disease. However, uncertainty exists regarding the significance of HOMOARG-induced arginase inhibition in vivo. A novel UPLC-MS method, measuring the conversion of ARG to ornithine (ORN), was developed to determine arginase 1 and arginase 2 inhibition by HOMOARG, lysine (LYS), proline (PRO), agmatine (AG), asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), and NG-Monomethyl-L-arginine (L-NMMA). Plasma HOMOARG, ARG and ORN concentrations were further measured in 50 healthy older adults >65 years (27 males and 23 females). HOMOARG inhibited arginase 1 with IC50 and Ki values of 8.14 ± 0.52 mM and 6.1 ± 0.50 mM, and arginase 2 with IC50 and Ki values of 2.52 ± 0.01 mM and 1.73 ± 0.10 mM, respectively. Both arginase isoforms retained 90% activity vs. control when physiological HOMOARG concentrations (1-10 µM) were used. In partial correlation analysis, plasma HOMOARG was not associated with ARG (P = 0.38) or ARG/ORN ratio (P = 0.73) in older adults. Our results suggest that arginase inhibition is unlikely to play a significant role in the reported cardio-protective effects of HOMOARG.
Collapse
Affiliation(s)
- S Tommasi
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - D J Elliot
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - M Da Boit
- Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - S R Gray
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - B C Lewis
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - A A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia.
| |
Collapse
|
13
|
Palygin O, Ilatovskaya DV, Levchenko V, Endres BT, Geurts AM, Staruschenko A. Nitric oxide production by glomerular podocytes. Nitric Oxide 2017; 72:24-31. [PMID: 29128399 DOI: 10.1016/j.niox.2017.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/27/2017] [Accepted: 11/07/2017] [Indexed: 01/18/2023]
Abstract
Nitric Oxide (NO), a potent vasodilator and vital signaling molecule, has been shown to contribute to the regulation of glomerular ultrafiltration. However, whether changes in NO occur in podocytes during the pathogenesis of salt-sensitive hypertension has not yet been thoroughly examined. We showed here that podocytes produce NO, and further hypothesized that hypertensive animals would exhibit reduced NO production in these cells in response to various paracrine factors, which might contribute to the damage of glomeruli filtration barrier and development of proteinuria. To test this, we isolated glomeruli from the kidneys of Dahl salt-sensitive (SS) rats fed a low salt (LS; 0.4% NaCl) or high salt (HS; 4% NaCl, 3 weeks) diets and loaded podocytes with either a combination of NO and Ca2+ fluorophores (DAF-FM and Fura Red, respectively) or DAF-FM alone. Changes in fluorescence were observed with confocal microscopy in response to adenosine triphosphate (ATP), angiotensin II (Ang II), and hydrogen peroxide (H2O2). Application of Ang II resulted in activation of both NO and intracellular calcium ([Ca2+]i) transients. In contrast, ATP promoted [Ca2+]i transients, but did not have any effects on NO production. SS rats fed a HS diet for 3 weeks demonstrated impaired NO production: the response to Ang II or H2O2 in podocytes of glomeruli isolated from SS rats fed a HS diet was significantly reduced compared to rats fed a LS diet. Therefore, glomerular podocytes from hypertensive rats showed a diminished NO release in response to Ang II or oxidative stress, suggesting that podocytic NO signaling is dysfunctional in this condition and likely contributes to the development of kidney injury.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bradley T Endres
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
14
|
Feng W, Chen B, Xing D, Li X, Fatima H, Jaimes EA, Sanders PW. Haploinsufficiency of the Transcription Factor Ets-1 Is Renoprotective in Dahl Salt-Sensitive Rats. J Am Soc Nephrol 2017; 28:3239-3250. [PMID: 28696249 DOI: 10.1681/asn.2017010085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/22/2017] [Indexed: 11/03/2022] Open
Abstract
Studies using Dahl salt-sensitive (SS) rats identified specific quantitative trait loci that predispose animals to hypertension-associated albuminuria and kidney injury. We explored the hypothesis that kidney-specific expression of the transcription factor Ets-1, located within one of these loci on chromosome 8, mediates glomerular injury in SS hypertension. During the first week on a high-salt diet, SS rats and SS rats with only one functioning Ets-1 gene (ES rats) demonstrated similar increases in BP. However, serum creatinine concentration, albuminuria, and glomerular expression of ETS-1 and two ETS-1 targets, MCP-1 and MMP2, did not increase as substantially in ES rats as in SS rats. Mean BP subsequently increased further in SS rats and remained higher than that of ES rats for the rest of the study. After 4 weeks of high-salt intake, ES rats still showed a lower mean serum creatinine concentration and less albuminuria, as well as less histologic evidence of glomerular injury and kidney fibrosis, than SS rats did. To investigate the specific contribution of renal Ets-1, we transplanted kidneys from ES or SS rats into salt-resistant SS-Chr 13BN/McwiCrl (SS-13BN) rats. Within 10 days on a high-salt diet, BP increased similarly in ES and SS allograft recipients, becoming significantly higher than the BP of control isograft recipients. However, mean serum creatinine concentration and albuminuria remained lower in ES allograft recipients than in SS allograft recipients at 2 weeks, and ES allografts showed less glomerular injury and interstitial fibrosis. In conclusion, reduced renal expression of ETS-1 prevented hypertension-associated kidney injury in SS rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine,
| | - Bo Chen
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dongqi Xing
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine
| | - Xingsheng Li
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine
| | - Huma Fatima
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Edgar A Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Paul W Sanders
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine.,Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
15
|
Hou E, Sun N, Zhang F, Zhao C, Usa K, Liang M, Tian Z. Malate and Aspartate Increase L-Arginine and Nitric Oxide and Attenuate Hypertension. Cell Rep 2017; 19:1631-1639. [DOI: 10.1016/j.celrep.2017.04.071] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/27/2016] [Accepted: 04/26/2017] [Indexed: 01/29/2023] Open
|
16
|
Abstract
Salt resistance/sensitivity refers specifically to the effect of dietary sodium chloride (salt) intake on BP. Increased dietary salt intake promotes an early and uniform expansion of extracellular fluid volume and increased cardiac output. To compensate for these hemodynamic changes and maintain constant BP in salt resistance, renal and peripheral vascular resistance falls and is associated with an increase in production of nitric oxide. In contrast, the decline in peripheral vascular resistance and the increase in nitric oxide are impaired or absent in salt sensitivity, promoting an increase in BP in these individuals. Endothelial dysfunction may pose a particularly significant risk factor in the development of salt sensitivity and subsequent hypertension. Vulnerable salt-sensitive populations may have in common underlying endothelial dysfunction due to genetic or environmental influences. These individuals may be very sensitive to the hemodynamic stress of increased effective blood volume, setting in motion untoward molecular and biochemical events that lead to overproduction of TGF-β, oxidative stress, and limited bioavailable nitric oxide. Finally, chronic high-salt ingestion produces endothelial dysfunction, even in salt-resistant subjects. Thus, the complex syndrome of salt sensitivity may be a function of the endothelium, which is integrally involved in the vascular responses to high salt intake.
Collapse
Affiliation(s)
| | - Louis J Dell'Italia
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
| | - Paul W Sanders
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
17
|
Yang G, Chu PL, Rump LC, Le TH, Stegbauer J. ACE2 and the Homolog Collectrin in the Modulation of Nitric Oxide and Oxidative Stress in Blood Pressure Homeostasis and Vascular Injury. Antioxid Redox Signal 2017; 26:645-659. [PMID: 27889958 DOI: 10.1089/ars.2016.6950] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Hypertension is the leading risk factor causing mortality and morbidity worldwide. Angiotensin (Ang) II, the most active metabolite of the renin-angiotensin system, plays an outstanding role in the pathogenesis of hypertension and vascular injury. Activation of angiotensin converting enzyme 2 (ACE2) has shown to attenuate devastating effects of Ang II in the cardiovascular system by reducing Ang II degradation and increasing Ang-(1-7) generation leading to Mas receptor activation. Recent Advances: Activation of the ACE2/Ang-(1-7)/Mas receptor axis reduces hypertension and improves vascular injury mainly through an increased nitric oxide (NO) bioavailability and decreased reactive oxygen species production. Recent studies reported that shedding of the enzymatically active ectodomain of ACE2 from the cell surface seems to regulate its activity and serves as an interorgan communicator in cardiovascular disease. In addition, collectrin, an ACE2 homolog with no catalytic activity, regulates blood pressure through an NO-dependent mechanism. CRITICAL ISSUES Large body of experimental data confirmed sustained beneficial effects of ACE2/Ang-(1-7)/Mas receptor axis activation on hypertension and vascular injury. Experimental studies also suggest that activation of collectrin might be beneficial in hypertension and endothelial dysfunction. Their role in clinical hypertension is unclear as selective and reliable activators of both axes are not yet available. FUTURE DIRECTIONS This review will highlight the results of recent research progress that illustrate the role of both ACE and collectrin in the modulation of NO and oxidative stress in blood pressure homeostasis and vascular injury, providing evidence for the potential therapeutic application of ACE2 and collectrin in hypertension and vascular disease. Antioxid. Redox Signal. 26, 645-659.
Collapse
Affiliation(s)
- Guang Yang
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| | - Pei-Lun Chu
- 2 Division of Nephrology, Department of Medicine, University of Virginia , Charlottesville, Virginia.,3 Department of Internal Medicine, Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Lars C Rump
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| | - Thu H Le
- 2 Division of Nephrology, Department of Medicine, University of Virginia , Charlottesville, Virginia
| | - Johannes Stegbauer
- 1 Department of Nephrology, Medical Faculty, Heinrich-Heine University Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
18
|
Vasdev S, Gill V, Parai S, Gadag V. Dietary Vitamin E Supplementation Attenuates Hypertension in Dahl Salt-Sensitive Rats. J Cardiovasc Pharmacol Ther 2016; 10:103-11. [PMID: 15965561 DOI: 10.1177/107424840501000204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There is strong evidence that excess dietary salt (NaCl) is a major factor contributing to the development of hypertension. Salt-sensitive humans and rats develop hypertension even on a normal-salt diet. Salt sensitivity is associated with glucose intolerance and insulin resistance in both humans and animal models, including Dahl salt-sensitive (DSS) rats. In insulin resistance, impaired glucose metabolism leads to elevated endogenous aldehydes that bind sulfhydryl groups of membrane proteins, altering calcium channels, and increasing cytosolic free calcium ([Ca2+] i) and blood pressure. Vitamin E lowers tissue aldehyde conjugates, cytosolic [Ca2+] i, and blood pressure in spontaneously hypertensive rats and fructose-induced hypertensive Wistar Kyoto rats, models of insulin resistance. This study investigated the effect of a normal-salt diet on tissue aldehyde conjugates, cytosolic [Ca2+] i, and blood pressure in DSS rats and the effect of vitamin E supplementation on blood pressure and associated biochemical changes in these animals. Seven-week-old DSS rats were divided into 3 groups of 6 animals each and treated for 6 weeks with diets as follows: low-salt (0.4% NaCl); normal-salt (0.7% NaCl) and normal salt (0.7% NaCl) plus vitamin E (34 mg/kg feed). At completion, animals in the normal-salt group had significantly elevated systolic blood pressure, cytosolic [Ca2+] i, and tissue aldehyde conjugates compared with the low-salt group. They also showed smooth muscle cell hyperplasia in small arteries and arterioles of the kidney. Dietary vitamin E supplementation significantly attenuated the increase in systolic blood pressure and associated biochemical and histopathologic changes.
Collapse
Affiliation(s)
- Sudesh Vasdev
- Department of Medicine and Laboratory Medicine, Health Sciences Centre, Memorial University of Newfoundland, St. John's, Newfoundland, Canada.
| | | | | | | |
Collapse
|
19
|
Dalmasso C, Maranon R, Patil C, Moulana M, Romero DG, Reckelhoff JF. 20-HETE and CYP4A2 ω-hydroxylase contribute to the elevated blood pressure in hyperandrogenemic female rats. Am J Physiol Renal Physiol 2016; 311:F71-7. [PMID: 27194719 DOI: 10.1152/ajprenal.00458.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/17/2016] [Indexed: 01/08/2023] Open
Abstract
In male rats, androgen supplements increase 20-hydroxyeicosatetraenoic acid (20-HETE) via cytochrome P-450 (CYP)4A ω-hydroxylase and cause an increase in blood pressure (BP). In the present study, we determined the roles of 20-HETE and CYP4A2 on the elevated BP in hyperandrogenemic female rats. Chronic dihydrotestosterone (DHT) increased mean arterial pressure (MAP) in female Sprague-Dawley rats (96 ± 2 vs. 108 ± 2 mmHg, P < 0.05) and was associated with increased renal microvascular CYP4A2 mRNA expression (15-fold), endogenous renal 20-HETE (5-fold), and ω-hydroxylase activity (3-fold). Chronic DHT also increased MAP in low salt-fed Dahl salt-resistant female rats (81 ± 4 vs. 95 ± 1 mmHg, P < 0.05) but had no effect on MAP in Dahl salt-sensitive female rats (154 ± 3 vs. 153 ± 3 mmHg), which are known to be 20-HETE deficient. To test the role of CYP4A2, female CYP4A2(-/-) and SS.5(Bn) (wild type) rats were treated with DHT. DHT increased MAP in SS.5(Bn) female rats (104 ± 1 vs. 128 ± 1 mmHg, P < 0.05) but had no effect in CYP4A2(-/-) female rats (118 ± 1 vs. 120 ± 1 mmHg). Renal microvascular 20-HETE was reduced in control CYP4A2(-/-) female rats and was increased with DHT in SS.5(Bn) female rats (6-fold) but not CYP4A2(-/-) female rats. ω-Hydroxylase activity was 40% lower in control CYP4A2(-/-) female rats than in SS.5(Bn) female rats, and DHT decreased ω-hydroxylase activity in SS.5(Bn) female rats (by 50%) but significantly increased ω-hydroxylase activity in CYP4A2(-/-) female rats (3-fold). These data suggest that 20-HETE via CYP4A2 contributes to the elevation in BP in hyperandrogenemic female rats. The data also suggest that 20-HETE synthesis inhibition may be effective in treating the elevated BP in women with hyperandrogenemia, such as women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- Carolina Dalmasso
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rodrigo Maranon
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Department of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Chetan Patil
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mohadetheh Moulana
- Department of Psychiatry, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi; and Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jane F Reckelhoff
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
20
|
Feng W, Ying WZ, Aaron KJ, Sanders PW. Transforming growth factor-β mediates endothelial dysfunction in rats during high salt intake. Am J Physiol Renal Physiol 2015; 309:F1018-25. [PMID: 26447221 DOI: 10.1152/ajprenal.00328.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/06/2015] [Indexed: 01/08/2023] Open
Abstract
Endothelial dysfunction has been shown to be predictive of subsequent cardiovascular events and death. Through a mechanism that is incompletely understood, increased dietary salt intake promotes endothelial dysfunction in healthy, salt-resistant humans. The present study tested the hypothesis that dietary salt-induced transforming growth factor (TGF)-β promoted endothelial dysfunction and salt-dependent changes in blood pressure (BP). Sprague-Dawley rats that received diets containing 0.3% NaCl [low salt (LS)] or 8.0% NaCl [high salt (HS)] were treated with vehicle or SB-525334, a specific inhibitor of TGF-β receptor I/activin receptor-like kinase 5, beginning on day 5. BP was monitored using radiotelemetry in four groups of rats (LS, LS + SB-525334, HS, and HS + SB-525334) for up to 14 days. By day 14 of the study, mean daytime systolic BP and mean pulse pressure of the HS group treated with vehicle was greater than those in the other three groups; mean daytime systolic BP and pulse pressure of the HS + SB-525334 group did not differ from the LS and LS + SB-525334-treated groups. Whereas mean systolic BP, mean diastolic BP, and mean arterial pressure did not differ among the groups on the seventh day of the study, endothelium-dependent vasorelaxation was impaired specifically in the HS group; treatment with the activin receptor-like kinase 5 inhibitor prevented the dietary HS intake-induced increases in phospho-Smad2 (Ser(465/467)) and NADPH oxidase-4 in endothelial lysates and normalized endothelial function. These findings suggest that HS-induced endothelial dysfunction and the development of salt-dependent increases in BP were related to endothelial TGF-β signaling.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Zhong Ying
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kristal J Aaron
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
21
|
|
22
|
Papageorgiou N, Androulakis E, Papaioannou S, Antoniades C, Tousoulis D. Homoarginine in the shadow of asymmetric dimethylarginine: from nitric oxide to cardiovascular disease. Amino Acids 2015; 47:1741-50. [PMID: 26123985 DOI: 10.1007/s00726-015-2017-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/25/2015] [Indexed: 11/30/2022]
|
23
|
Abstract
Collectrin (Tmem27) is a transmembrane glycoprotein that is highly expressed in the kidney and vascular endothelium. It is a homologue of the angiotensin-converting enzyme 2 (ACE2) but harbors no catalytic domain. In the extravascular tissues of the kidney, collectrin is localized to the proximal tubule and collecting duct. Collectrin-deficient mice are featured with hypertension and exaggerated salt sensitivity. These phenotypes are associated with impaired uptake of the nitric oxide precursor L-arginine and the expression of its amino acid transporters, CAT-1 and y(+)LAT1, in endothelial cells. In addition, collectrin-deficient mice display decreased dimerization of nitric oxide synthase and decreased nitric oxide synthesis, but enhanced superoxide generation, suggesting that deletion of collectrin leads to a state of nitric oxide synthase uncoupling. These findings suggest that collectrin plays a protective role against hypertension. The collectrin knockout mouse represents a unique model for hypertension research. Furthermore, collectrin may serve as a novel therapeutic target in the treatment of hypertension.
Collapse
|
24
|
Homoarginine in the renal and cardiovascular systems. Amino Acids 2015; 47:1703-13. [DOI: 10.1007/s00726-015-1993-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/18/2015] [Indexed: 10/23/2022]
|
25
|
Gillis EE, Williams JM, Garrett MR, Mooney JN, Sasser JM. The Dahl salt-sensitive rat is a spontaneous model of superimposed preeclampsia. Am J Physiol Regul Integr Comp Physiol 2015; 309:R62-70. [PMID: 25904684 DOI: 10.1152/ajpregu.00377.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/09/2015] [Indexed: 12/24/2022]
Abstract
The mechanisms of the pathogenesis of preeclampsia, a leading cause of maternal morbidity and death worldwide, are poorly understood in part due to a lack of spontaneous animal models of the disease. We hypothesized that the Dahl salt-sensitive (S) rat, a genetic model of hypertension and kidney disease, is a spontaneous model of superimposed preeclampsia. The Dahl S was compared with the Sprague-Dawley (SD) rat, a strain with a well-characterized normal pregnancy, and the spontaneously hypertensive rat (SHR), a genetic model of hypertension that does not experience a preeclamptic phenotype despite preexisting hypertension. Mean arterial pressure (MAP, measured via telemetry) was elevated in the Dahl S and SHR before pregnancy, but hypertension was exacerbated during pregnancy only in Dahl S. In contrast, SD and SHR exhibited significant reductions in MAP consistent with normal pregnancy. Dahl S rats exhibited a severe increase in urinary protein excretion, glomerulomegaly, increased placental hypoxia, increased plasma soluble fms-like tyrosine kinase-1 (sFlt-1), and increased placental production of tumor necrosis factor-α (TNF-α). The Dahl S did not exhibit the expected decrease in uterine artery resistance during late pregnancy in contrast to the SD and SHR. Dahl S pups and litter sizes were smaller than in the SD. The Dahl S phenotype is consistent with many of the characteristics observed in human superimposed preeclampsia, and we propose that the Dahl S should be considered further as a spontaneous model to improve our understanding of the pathogenesis of superimposed preeclampsia and to identify and test new therapeutic targets for its treatment.
Collapse
Affiliation(s)
| | | | - Michael R Garrett
- Department of Pharmacology and Toxicology, and Department of Medicine University of Mississippi Medical Center, Jackson, Mississippi
| | | | | |
Collapse
|
26
|
Das S, Mattson DL. Exogenous L-arginine attenuates the effects of angiotensin II on renal hemodynamics and the pressure natriuresis-diuresis relationship. Clin Exp Pharmacol Physiol 2014; 41:270-8. [PMID: 24472006 DOI: 10.1111/1440-1681.12212] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 11/22/2013] [Accepted: 12/14/2013] [Indexed: 02/05/2023]
Abstract
Administration of exogenous L-arginine (L-Arg) attenuates angiotensin-II (AngII)-mediated hypertension and kidney disease in rats. The present study assessed renal hemodynamics and pressure diuresis-natriuresis in anaesthetized rats infused with vehicle, AngII (20 ng/kg per min i.v.) or AngII + L-Arg (300 μg/kg per min i.v.). Experiments in isolated aortic rings were carried out to assess L-Arg effects on the vasculature. Increasing renal perfusion pressure (RPP) from ~100 to 140 mmHg resulted in a nine- to tenfold increase in urine flow and sodium excretion rate in control animals. In comparison, AngII infusion significantly reduced renal blood flow (RBF) and glomerular filtration rate (GFR) by 40-42%, and blunted the pressure-dependent increase in urine flow and sodium excretion rate by 54-58% at elevated RPP. Supplementation of L-Arg reversed the vasoconstrictor effects of AngII and restored pressure-dependent diuresis to levels not significantly different from control rats. Dose-dependent contraction to AngII (10(-10) mol/L to 10(-7) mol/L) was observed with a maximal force equal to 27 ± 3% of the response to 10(-5) mol/L phenylephrine. Contraction to 10(-7) mol/L AngII was blunted by 75 ± 3% with 10(-4) mol/L L-Arg. The influence of L-Arg to blunt AngII-mediated contraction was eliminated by endothelial denudation or incubation with nitric oxide synthase inhibitors. Furthermore, the addition of 10(-3) mol/L cationic or neutral amino acids, which compete with L-Arg for cellular uptake, blocked the effect of L-Arg. Anionic amino acids did not influence the effects of L-Arg on AngII-mediated contraction. These studies show that L-Arg blunts AngII-mediated vascular contraction by an endothelial- and nitric oxide synthase-dependent mechanism involving cellular uptake of L-Arg.
Collapse
Affiliation(s)
- Satarupa Das
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
27
|
Pilz S, Teerlink T, Scheffer PG, Meinitzer A, Rutters F, Tomaschitz A, Drechsler C, Kienreich K, Nijpels G, Stehouwer CDA, März W, Dekker JM. Homoarginine and mortality in an older population: the Hoorn study. Eur J Clin Invest 2014; 44:200-8. [PMID: 24251815 DOI: 10.1111/eci.12208] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 11/15/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Homoarginine is an amino acid that may be involved in nitric oxide and energy metabolism. Previous studies in patient populations showed that low homoarginine levels indicate an increased risk of mortality and cardiovascular disease. We evaluated whether low plasma levels of homoarginine are associated with elevated, overall and cause-specific mortality. MATERIALS AND METHODS The Hoorn study is a population-based study among older men and women. We calculated Cox proportional hazard ratios (HRs) for overall and cause-specific mortality according to sex-specific homoarginine quartiles. RESULTS We included 606 study participants (51·3% women; 70·0 ± 6·6 years). Homoarginine concentrations were higher in men (1·63 ± 0·51 μM), compared with women (1·30 ± 0·44 μM; P < 0·001). After a median follow-up time of 7·8 years, 112 study participants died, including 31 deaths due to cardiovascular diseases and 30 due to cancer. Associations between homoarginine levels and mortality showed a threshold effect with a significant risk increase from the second to the first quartile. Compared with the upper three quartiles, the age-, sex- and BMI-adjusted HR (with 95% CI) in the first quartile was 2·26 (1·52-3·32) for overall mortality, 4·20 (2·03-8·69) for cardiovascular mortality and 1·25 (0·55-2·85) for cancer mortality. These associations remained materially unchanged after multivariate adjustments. CONCLUSIONS Low plasma concentrations of homoarginine are a risk marker for overall mortality and especially for cardiovascular mortality in the older general population. Further studies are warranted to elucidate the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Stefan Pilz
- Department of Epidemiology and Biostatistics, EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, the Netherlands; Division of Endocrinology and Metabolism, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kanbay M, Bayram Y, Solak Y, Sanders PW. Dietary potassium: a key mediator of the cardiovascular response to dietary sodium chloride. JOURNAL OF THE AMERICAN SOCIETY OF HYPERTENSION : JASH 2013; 7:395-400. [PMID: 23735420 PMCID: PMC4083820 DOI: 10.1016/j.jash.2013.04.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023]
Abstract
Potassium and sodium share a yin/yang relationship in the regulation of blood pressure (BP). BP is directly associated with the total body sodium and negatively correlated with the total body potassium. Epidemiologic, experimental, and clinical studies have shown that potassium is a significant regulator of BP and further improves cardiovascular outcomes. Hypertensive cardiovascular damage, stroke, and stroke-related death are accelerated by salt intake but might be curbed by increasing dietary potassium intake. The antihypertensive effect of potassium supplementation appears to occur through several mechanisms that include regulation of vascular sensitivity to catecholamines, promotion of natriuresis, limiting plasma renin activity, and improving endothelial function. In the absence of chronic kidney disease, the combined evidence suggests that a diet rich in potassium content serves a vasculoprotective function, particularly in the setting of salt-sensitive hypertension and prehypertension.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Istanbul Medeniyet University School of Medicine, Istanbul, Turkey.
| | | | | | | |
Collapse
|
29
|
Zicha J, Dobešová Z, Vokurková M, Rauchová H, Hojná S, Kadlecová M, Behuliak M, Vaněčková I, Kuneš J. Age-dependent salt hypertension in Dahl rats: fifty years of research. Physiol Res 2013; 61:S35-S87. [PMID: 22827876 DOI: 10.33549/physiolres.932363] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fifty years ago, Lewis K. Dahl has presented a new model of salt hypertension - salt-sensitive and salt-resistant Dahl rats. Twenty years later, John P. Rapp has published the first and so far the only comprehensive review on this rat model covering numerous aspects of pathophysiology and genetics of salt hypertension. When we summarized 25 years of our own research on Dahl/Rapp rats, we have realized the need to outline principal abnormalities of this model, to show their interactions at different levels of the organism and to highlight the ontogenetic aspects of salt hypertension development. Our attention was focused on some cellular aspects (cell membrane function, ion transport, cell calcium handling), intra- and extrarenal factors affecting renal function and/or renal injury, local and systemic effects of renin-angiotensin-aldosterone system, endothelial and smooth muscle changes responsible for abnormal vascular contraction or relaxation, altered balance between various vasoconstrictor and vasodilator systems in blood pressure maintenance as well as on the central nervous and peripheral mechanisms involved in the regulation of circulatory homeostasis. We also searched for the age-dependent impact of environmental and pharmacological interventions, which modify the development of high blood pressure and/or organ damage, if they influence the salt-sensitive organism in particular critical periods of development (developmental windows). Thus, severe self-sustaining salt hypertension in young Dahl rats is characterized by pronounced dysbalance between augmented sympathetic hyperactivity and relative nitric oxide deficiency, attenuated baroreflex as well as by a major increase of residual blood pressure indicating profound remodeling of resistance vessels. Salt hypertension development in young but not in adult Dahl rats can be attenuated by preventive increase of potassium or calcium intake. On the contrary, moderate salt hypertension in adult Dahl rats is attenuated by superoxide scavenging or endothelin-A receptor blockade which do not affect salt hypertension development in young animals.
Collapse
Affiliation(s)
- J Zicha
- Centre for Cardiovascular Research, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
L-Homoarginine and L-arginine are antagonistically related to blood pressure in an elderly population. J Hypertens 2013; 31:1114-23. [DOI: 10.1097/hjh.0b013e32836037fb] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
31
|
Drechsler C, Kollerits B, Meinitzer A, März W, Ritz E, König P, Neyer U, Pilz S, Wanner C, Kronenberg F. Homoarginine and progression of chronic kidney disease: results from the Mild to Moderate Kidney Disease Study. PLoS One 2013; 8:e63560. [PMID: 23691067 PMCID: PMC3655120 DOI: 10.1371/journal.pone.0063560] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/04/2013] [Indexed: 11/19/2022] Open
Abstract
Background Homoarginine is an amino acid derivative mainly synthesized in the kidney. It is suggested to increase nitric oxide availability, enhance endothelial function and to protect against cardiovascular diseases. We aimed to investigate the relation between homoarginine, kidney function and progression of chronic kidney disease (CKD). Methods We measured plasma homoarginine concentrations in baseline samples of the Mild to Moderate Kidney Disease (MMKD) Study, a prospective cohort study of 227 patients with CKD in Europe. Homoarginine concentrations were available in 182 of the baseline samples and in 139 of the prospectively-followed patients. We correlated homoarginine concentrations to parameters of kidney function. The association between homoarginine and progression of CKD was assessed during a follow-up of up to seven years (median 4.45 years, interquartile range 2.54–5.19) using Cox regression analysis. Progression of CKD was defined as doubling of baseline serum creatinine and/or end-stage renal disease. Results Study participants were at baseline on average 47±13 years old and 65% were male. Mean±standard deviation of homoarginine concentrations were 2.5±1.1 µmol/L and concentrations were incrementally lower at lower levels of GFR with mean concentrations of 2.90±1.02 µmol/L (GFR>90 ml/min), 2.64±1.06 µmol/L (GFR 60–90 ml/min), 2.52±1.24 µmol/L (GFR 30–60 ml/min) and 2.05±0.78 µmol/L (GFR<30 ml/min), respectively (p = 0.002). The age- and sex-adjusted risk to reach the renal endpoint was significantly higher by 62% with each decrease by one standard deviation (1.1 µmol/L) of homoarginine (HR 1.62, 95% CI 1.16–2.27, p = 0.005). This association was independent of proteinuria (HR 1.56, 95% CI 1.11–2.20, p = 0.01), and was slightly attenuated when adjusting for GFR (HR 1.40 (95% CI 0.98–1.98, p = 0.06). Conclusions Homoarginine concentrations are directly correlated with kidney function and are significantly associated with the progression of CKD. Low homoarginine concentrations might be an early indicator of kidney failure and a potential target for the prevention of disease progression which needs further investigations.
Collapse
Affiliation(s)
- Christiane Drechsler
- Division of Nephrology, Department of Medicine, University of Würzburg, Würzburg, Germany
| | - Barbara Kollerits
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Winfried März
- Department of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Eberhard Ritz
- Department of Internal Medicine, Division of Nephrology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Paul König
- Innsbruck University Hospital, Department of Clinical Nephrology, Innsbruck, Austria
| | - Ulrich Neyer
- Department of Nephrology and Dialysis, Academic Teaching Hospital, Feldkirch, Austria
| | - Stefan Pilz
- Department of Epidemiology and Biostatistics, EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria
| | - Christoph Wanner
- Division of Nephrology, Department of Medicine, University of Würzburg, Würzburg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
- * E-mail:
| | | |
Collapse
|
32
|
Abstract
Dietary sodium chloride (salt) has long been considered injurious to the kidney by promoting the development of glomerular and tubulointerstitial fibrosis. Endothelial cells throughout the vasculature and glomeruli respond to increased dietary salt intake with increased production of transforming growth factor-β (TGF-β) and nitric oxide. High-salt intake activates large-conductance, voltage- and calcium-activated potassium (BK(Ca)) channels in endothelial cells. Activation of BK(Ca) channels promotes signaling through proline-rich tyrosine kinase-2, cellular-sarcoma (c-Src), Akt (also known as protein kinase B), and mitogen-activated protein kinase pathways that lead to endothelial production of TGF-β and nitric oxide. TGF-β signaling is broadly accepted as a strong stimulator of renal fibrosis. The classic description of TGF-β signaling pathology in renal disease involves signaling through Smad proteins resulting in extracellular matrix deposition and fibrosis. Active TGF-β1 also causes fibrosis by inducing epithelial-mesenchymal transition and apoptosis. By enhancing TGF-β signaling, increased dietary salt intake leads to progressive renal failure from nephron loss and glomerular and tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Michael B Hovater
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
33
|
Drechsler C, Meinitzer A, Pilz S, Krane V, Tomaschitz A, Ritz E, März W, Wanner C. Homoarginine, heart failure, and sudden cardiac death in haemodialysis patients. Eur J Heart Fail 2012; 13:852-9. [PMID: 21791541 PMCID: PMC3143829 DOI: 10.1093/eurjhf/hfr056] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIMS Sudden cardiac death (SCD) is a major contributor to the excess mortality of patients on maintenance dialysis. Homoarginine deficiency may lead to decreased nitric oxide availability and endothelial dysfunction. Based on this rationale we assessed whether homoarginine deficiency is a risk factor for SCD in dialysis patients. METHODS AND RESULTS This study examined the association of homoarginine with cardiovascular outcomes in 1255 diabetic haemodialysis patients from the German diabetes and dialysis study. During a median of 4 years of follow-up, hazard ratios (HR) (95% CI) for reaching the following pre-specified, adjudicated endpoints were determined: SCD, myocardial infarction, stroke, death due to heart failure, and combined cardiovascular events. There was a strong association of low homoarginine concentrations with the presence of congestive heart failure and left ventricular hypertrophy as well as increased levels of brain natriuretic peptide. Per unit decrease in homoarginine, the risk of SCD increased three-fold (HR 3.1, 95% CI 2.0-4.9), attenuating slightly in multivariate models (HR 2.4; 95% CI 1.5-3.9). Patients in the lowest homoarginine quintile experienced a more than two-fold increased risk of SCD, and more than three-fold increased risk of heart failure death than patients in the highest quintile, which accounted for the high incidence of combined cardiovascular events. Low homoarginine showed a trend towards increased risk of stroke, however, myocardial infarction was not meaningfully affected. CONCLUSION Low homoarginine is a strong risk factor for SCD and death due to heart failure in haemodialysis patients. Further studies are needed to elucidate the underlying mechanisms, offering the potential to develop new interventional strategies.
Collapse
Affiliation(s)
- Christiane Drechsler
- Department of Internal Medicine 1, Division of Nephrology, University of Würzburg, Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Veeramani C, Al-Numair KS, Chandramohan G, Alsaif MA, Pugalendi KV. Protective effect of Melothria maderaspatana leaf fraction on electrolytes, catecholamines, endothelial nitric oxide synthase and endothelin-1 peptide in uninephrectomized deoxycorticosterone acetate-salt hypertensive rats. J Nat Med 2012; 66:535-43. [PMID: 22246664 DOI: 10.1007/s11418-011-0621-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/19/2011] [Indexed: 01/09/2023]
Abstract
This study was designed to investigate the protective effect of ethyl acetate fraction of Melothria maderaspatana (EAFM) leaf on electrolytes, catecholamines, endothelial nitric oxide synthase (eNOS) and endothelin-1 (ET-1) peptide in uninephrectomized deoxycorticosterone acetate (DOCA)-salt hypertensive rats. Administration of DOCA-salt significantly increased the systolic and diastolic blood pressure and treatment with EAFM significantly lowered the blood pressure. In DOCA-salt rats, the levels of sodium and chloride increased significantly while potassium level decreased and administration of EAFM brought these parameters to normality. The levels of epinephrine and norepinephrine increased significantly in DOCA-salt rats and administration of EAFM significantly decreased these parameters to normality. DOCA-salt hypertensive rats exhibited significantly decreased L: -arginine and nitrite + nitrate levels and administration of EAFM brought these parameters to normality. DOA-salt hypertensive rats showed down-regulation of eNOS and up-regulation of ET-1 protein expressions in heart and kidney, and treatment with EAFM prevented down-regulation of eNOS and significantly down-regulated the ET-1 protein expressions. In conclusion, EAFM provides good blood pressure control by enhancing potassium and decreasing sodium levels, decreasing levels of epinephrine and norepinephrine, and preventing down-regulation of eNOS and significantly down-regulating ET-1 protein expression.
Collapse
Affiliation(s)
- Chinnadurai Veeramani
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, PO Box 10219, Riyadh 11433, Saudi Arabia
| | | | | | | | | |
Collapse
|
35
|
Rajapakse NW, Mattson DL. Role of L-arginine uptake mechanisms in renal blood flow responses to angiotensin II in rats. Acta Physiol (Oxf) 2011; 203:391-400. [PMID: 21649863 DOI: 10.1111/j.1748-1716.2011.02330.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIM To examine whether reduced renal arginine transport increases the responsiveness of the renal circulation to angiotensin II in salt sensitivity, renal perfusion responses to angiotensin II were examined in the presence of L-arginine transport inhibitor, L-lysine and subsequent L-arginine in Sprague Dawley (SD) and Dahl salt-sensitive (Dahl S) rats. METHODS Laser Doppler probes and a transonic flow probe were used to measure regional renal perfusion and total renal perfusion respectively. Renal perfusion responses to intravenous (i.v.) angiotensin II were sequentially examined under control conditions and during i.v. infusion of L-lysine, L-arginine or nitric oxide synthase inhibitor, N(G)-nitro-L-arginine. RESULTS Angiotensin II (10 and 100 ng kg(-1) min(-1) , i.v.) reduced total renal (-10 ± 3 and -36 ± 5%) and cortical (-10 ± 2 and -28 ± 4%) but not medullary perfusion in SD rats. In these rats L-lysine enhanced the renal perfusion response (P = 0.003), whereas subsequent L-arginine reversed this effect (P = 0.04). Angiotensin II reduced total renal, cortical and medullary perfusion in Dahl S rats. In Dahl S rats fed high salt, L-lysine did not affect renal perfusion responses to angiotensin II, but subsequent L-arginine blunted the renal blood flow response (P = 0.01) and increased the medullary perfusion during angiotensin II infusion (P = 0.006). CONCLUSION Intact renal L-arginine transport attenuates the vasoconstrictor effects of circulating angiotensin II in the renal cortex in SD rats. L-arginine also plays an important role in protecting the renal medullary circulation from the ischemic effects of angiotensin II in Dahl S rats.
Collapse
Affiliation(s)
- N W Rajapakse
- Department of Physiology, Medical College of Wisconsin, Milwaukee, USA.
| | | |
Collapse
|
36
|
Low Serum Homoarginine Is a Novel Risk Factor for Fatal Strokes in Patients Undergoing Coronary Angiography. Stroke 2011; 42:1132-4. [DOI: 10.1161/strokeaha.110.603035] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Low serum concentrations of the amino acid homoarginine have been associated with endothelial dysfunction and an increased risk of all-cause and cardiovascular mortality. We aimed to investigate whether homoarginine levels are also associated with fatal strokes and a history of nonfatal cerebrovascular disease.
Methods—
Serum homoarginine was measured in 3305 participants of the Ludwigshafen Risk and Cardiovascular Health (LURIC) study who were referred to coronary angiography at baseline (1997 to 2000) and were followed up with respect to mortality.
Results—
During a median follow-up time of 9.9 years, 991 patients died including 61 fatal (ischemic and hemorrhagic) strokes. In a binary logistic regression analysis, the odds ratio (with 95% CI) for fatal stroke per SD of homoarginine was 0.52 (0.37 to 0.73;
P
<0.001) and remained significant after multivariable adjustments (0.62 [0.42 to 0.91];
P
=0.014). For previous cerebrovascular disease events, the multivariable adjusted OR per SD of homoarginine was 0.82 (0.70 to 0.96;
P
=0.014).
Conclusions—
Low homoarginine levels are a novel risk factor for fatal strokes and are reduced in patients with a history of cerebrovascular disease. Further studies are needed to explore the significance of homoarginine to risk stratification and therapeutic approaches in the prevention of strokes.
Collapse
|
37
|
Kanbay M, Chen Y, Solak Y, Sanders PW. Mechanisms and consequences of salt sensitivity and dietary salt intake. Curr Opin Nephrol Hypertens 2011; 20:37-43. [PMID: 21088577 PMCID: PMC3089903 DOI: 10.1097/mnh.0b013e32834122f1] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Investigation into the underlying mechanisms of salt sensitivity has made important advances in recent years. This review examines in particular the effects of sodium and potassium on vascular function. RECENT FINDINGS Sodium chloride (salt) intake promotes cutaneous lymphangiogenesis mediated through tissue macrophages and directly alters endothelial cell function, promoting increased production of transforming growth factor-β (TGF-β) and nitric oxide. In the setting of endothelial dysfunction, such as occurs with aging, diminished nitric oxide production exacerbates the vascular effects of TGF-β, promoting decreased arterial compliance and hypertension. Dietary potassium intake may serve as an important countervailing influence on the effects of salt in the vasculature. SUMMARY There is growing appreciation that, independently of alterations in blood pressure, dietary intake of sodium and potassium promotes functional changes in the vasculature and lymphatic system. These changes may protect against development of salt-sensitive hypertension. While salt sensitivity cannot be ascribed exclusively to these factors, perturbation of these processes promotes hypertension during high-salt intake. These studies add to the list of genetic and environmental factors that are associated with salt sensitivity, but in particular provide insight into adaptive mechanisms during high salt intake.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Internal Medicine, Fatih University School of Medicine, Ankara, Turkey
| | | | | | | |
Collapse
|
38
|
Meinitzer A, Drechsler C, Tomaschitz A, Pilz S, Krane V, Wanner C, März W. Homoarginin: ein neuer kardiovaskulärer Risikomarker bei Dialysepatienten / Homoarginine: a new cardiovascular risk marker in hemodialysis patients. ACTA ACUST UNITED AC 2011. [DOI: 10.1515/jlm.2011.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
39
|
März W, Meinitzer A, Drechsler C, Pilz S, Krane V, Kleber ME, Fischer J, Winkelmann BR, Böhm BO, Ritz E, Wanner C. Homoarginine, Cardiovascular Risk, and Mortality. Circulation 2010; 122:967-75. [PMID: 20733103 DOI: 10.1161/circulationaha.109.908988] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Homoarginine is an amino acid derivative that may increase nitric oxide availability and enhance endothelial function. The effect of the level of homoarginine on cardiovascular outcome and mortality is unknown.
Methods and Results—
We assessed cardiovascular and all-cause mortality according to homoarginine levels in a cohort of 3305 subjects referred for coronary angiography from the LUdwigshafen RIsk and Cardiovascular Health (LURIC) Study. After investigating the relation of homoarginine with kidney function and markers of endothelial dysfunction, we explored its effects on adverse outcomes in a second high-risk cohort of 1244 patients with type 2 diabetes mellitus receiving maintenance hemodialysis (4D study [Die Deutsche Diabetes Dialyse Studie]). In the LURIC study, mean serum homoarginine levels were 2.6±1.1 μmol/L. During a median follow-up of 7.7 years, 766 patients died. After adjustments for age and sex, patients in the lowest quartile (<1.85 μmol/L) had a >4-fold higher rate of dying of cardiovascular disease (hazard ratio 4.1, 95% confidence interval 3.0 to 5.7) than patients in the highest quartile (>3.1 μmol/L). Lower homoarginine levels were associated with lower estimated glomerular filtration rate and higher levels of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Hemodialysed patients had lower mean homoarginine levels of 1.2±0.5 μmol/L and experienced a 5-fold increased mortality rate compared with LURIC patients (608 deaths during a median follow-up of 4 years). Homoarginine consistently affected mortality, which was 2-fold higher in 4D study patients in the lowest quartile (<0.87 μmol/L) than in patients in the highest quartile (>1.4 μmol/L).
Conclusions—
Homoarginine levels are independently associated with cardiovascular and all-cause mortality in patients referred for coronary angiography and in patients undergoing hemodialysis. Future studies are needed to elucidate the underlying pathomechanisms.
Collapse
Affiliation(s)
- Winfried März
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Andreas Meinitzer
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Christiane Drechsler
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Stefan Pilz
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Vera Krane
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Marcus E. Kleber
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Joachim Fischer
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Bernhard R. Winkelmann
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Bernhard O. Böhm
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Eberhard Ritz
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| | - Christoph Wanner
- From the Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., A.M.) and Department of Internal Medicine (S.P.), Division of Endocrinology and Nuclear Medicine, Medical University of Graz, Graz, Austria; Synlab Center of Laboratory Diagnostics (W.M.), Heidelberg, Germany; LURIC nonprofit LLC (M.E.K.), Freiburg, Germany; Mannheim Institute of Public Health (W.M., J.F.), Ruperto Carola University Heidelberg, Medical Faculty Mannheim, Germany; Department of Medicine (C.D., V.K., C.W
| |
Collapse
|
40
|
Nunn PB, Bell EA, Watson AA, Nash RJ. Toxicity of Non-protein Amino Acids to Humans and Domestic Animals. Nat Prod Commun 2010. [DOI: 10.1177/1934578x1000500329] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Non-protein amino acids are common in plants and are present in widely consumed animal feeds and human foods such as alfalfa ( Medicago sativa), which contains canavanine, and lentil ( Lens culinaris), which contains homoarginine. Some occur in wild species that are inadvertently harvested with crop species. Some nonprotein amino acids and metabolites can be toxic to humans, e.g. Lathyrus species contain a neurotoxic oxalyl-amino acid. Some potential toxins may be passed along a food chain via animal intermediates. The increased interest in herbal medicines in the Western countries will increase exposure to such compounds.
Collapse
Affiliation(s)
- Peter B. Nunn
- School of Pharmacy and Biomedical Sciences, St Michael's Building, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - E. Arthur Bell
- Formerly at the School of Biomedical Sciences, King's College London, WC2R 2LS, England, UK
| | - Alison A. Watson
- Phytoquest Limited, Aberystwyth University, Plas Gogerddan, Aberystwyth, Ceredigion, SY23 3EB, UK
| | - Robert J. Nash
- Phytoquest Limited, Aberystwyth University, Plas Gogerddan, Aberystwyth, Ceredigion, SY23 3EB, UK
| |
Collapse
|
41
|
Renal functional, not morphological, abnormalities account for salt sensitivity in Dahl rats. J Hypertens 2009; 27:587-98. [PMID: 19330919 DOI: 10.1097/hjh.0b013e32831ffec7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The kidney's role in the pathogenesis of salt-induced hypertension remains unclear. However, it has been suggested that inherited morphological renal abnormalities may cause hypertension. We hypothesized that functional, not morphological, derangements in Dahl salt-sensitive rats' kidneys cause NaCl retention that leads to hypertension accompanied by renal pathologic changes and proteinuria. METHOD We studied hemodynamic, renal morphologic, and biochemical differences in Dahl salt-resistant and Dahl salt-sensitive rats fed low (0.05-0.23% NaCl) or elevated (1% NaCl) salt diets. RESULTS We found similar hemodynamics, equal numbers of glomeruli, normal renal medullary interstitial cells and their osmiophilic granules, and cortical morphology in normotensive Dahl salt-resistant and Dahl salt-sensitive rats fed low dietary salt. Furthermore, aldosterone secretion, caused by angiotensin II infusion in normotensive rats fed 0.23% NaCl, was significantly less in Dahl salt-sensitive than Dahl salt-resistant rats. Increasing NaCl to 1% caused renal vasoconstriction without changing cyclic GMP excretion in Dahl salt-sensitive rats; in Dahl salt-resistant rats, cyclic GMP increased markedly and renal vascular resistance remained unchanged. On 1% NaCl for 9 months, Dahl salt-sensitive rats developed marked hypertension, severe renal vasoconstriction, glomerulosclerosis, tubulointerstitial abnormalities, and marked proteinuria; hypertension resulted from increased total peripheral resistance, as occurs in essential hypertensive humans. No hemodynamic or renal pathologic changes occurred in Dahl salt-resistant rats, and proteinuria was minimal. CONCLUSION We conclude that renal functional, not morphological, abnormalities cause salt sensitivity in Dahl rats.
Collapse
|
42
|
Abstract
Animal and human studies support an untoward effect of excess dietary NaCl (salt) intake on cardiovascular and renal function and life span. Recent work has promoted the concept that the endothelium, in particular, reacts to changes in dietary salt intake through a complex series of events that are independent of blood pressure and the renin-angiotensin-aldosterone axis. The cellular signaling events culminate in the intravascular production of transforming growth factor-beta (TGF-beta) and nitric oxide in response to increased salt intake. Plasticity of the endothelium is integral in the vascular remodeling consequences associated with excess salt intake, because nitric oxide serves as a negative regulator of TGF-beta production. Impairment of nitric oxide production, such as occurs with endothelial dysfunction in a variety of disease states, results in unopposed excess vascular TGF-beta production, which promotes reduced vascular compliance and augmented peripheral arterial constriction and hypertension. Persistent alterations in vascular function promote the increase in cardiovascular events and reductions in renal function that reduce life span during increased salt intake.
Collapse
Affiliation(s)
- Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, and Department of Veterans Affairs Medical Center, Birmingham, Alabama 35294-0007, USA.
| |
Collapse
|
43
|
Komori K, Tsujimura A, Takao T, Matsuoka Y, Miyagawa Y, Takada S, Nonomura N, Okuyama A. Nitric oxide synthesis leads to vascular endothelial growth factor synthesis via the NO/cyclic guanosine 3',5'-monophosphate (cGMP) pathway in human corpus cavernosal smooth muscle cells. J Sex Med 2008; 5:1623-35. [PMID: 18331270 DOI: 10.1111/j.1743-6109.2008.00772.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Vascular smooth muscle cells express endothelial nitric oxide synthase (eNOS) and produce nitric oxide (NO). Recently, increased NO production has been reported to induce the synthesis and secretion of vascular endothelial growth factor (VEGF) via the NO/cyclic guanosine 3',5'-monophosphate (cGMP) pathway. L-arginine (L-arg), the precursor of NO, and selective phosphodiesterase type 5 (PDE-5) inhibitors that increase levels of intracellular cGMP may complementarily enhance VEGF synthesis in corpus cavernosal smooth muscle cells (CCSMCs), and may consequently restore impaired endothelial function. Expression of eNOS in corpus cavernosal smooth muscle has also been reported. However, it is unclear whether CCSMCs can generate NO. AIM To elucidate whether CCSMCs can synthesize NO and whether NO synthesis enhances VEGF synthesis via the NO/cGMP pathway. METHODS Corpus cavernosal cells were cultured and characterized by immunocytochemistry and immunoblotting. CCSMCs were treated with L-arg. CCSMCs were also incubated with L-arg and with vardenafil, an inhibitor of PDE-5. MAIN OUTCOME MEASURES Release of NO from cells was confirmed by assay of NO metabolites (NOx). Intracellular cGMP concentration and VEGF concentration in the medium were measured. RESULTS Isolated cells were determined to be CCSMCs. The expression of eNOS by CCSMCs was also identified. NOx and cGMP levels in the L-arg-treated group were significantly greater than those in the control group. VEGF and cGMP levels in the L-arg-treated group were also significantly greater than those in the control group. VEGF and cGMP levels in the L-arg + vardenafil-treated group were significantly greater than those in the L-arg-treated group and the control group. CONCLUSIONS CCSMCs express eNOS and synthesize NO. NO synthesis leads to enhancement of VEGF synthesis via the NO/cGMP pathway. Combined L-arg and vardenafil treatment, which can enhance VEGF production, may provide a novel therapeutic strategy for the treatment of erectile dysfunction as well as endothelial dysfunction in general.
Collapse
Affiliation(s)
- Kazuhiko Komori
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Ying WZ, Aaron K, Sanders PW. Dietary salt activates an endothelial proline-rich tyrosine kinase 2/c-Src/phosphatidylinositol 3-kinase complex to promote endothelial nitric oxide synthase phosphorylation. Hypertension 2008; 52:1134-41. [PMID: 18981321 DOI: 10.1161/hypertensionaha.108.121582] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although many laboratories have shown that dietary NaCl (salt) intake increases NO production in rodents and humans, the mechanism has not been uncovered. In the present study, pharmacological and dominant-negative strategies were used to show that feeding a formulated diet containing increased amounts of salt to young male Sprague-Dawley rats induced the formation of an endothelial cell-signaling complex that contained proline-rich tyrosine kinase 2, c-Src (also known as pp60(c-src)), and phosphatidylinositol 3-kinase. In the setting of a high-salt diet, proline-rich tyrosine kinase 2 served as the scaffold for c-Src-mediated phosphatidylinositol 3-kinase activation. Phosphatidylinositol 3-kinase was the upstream activator of protein kinase B (Akt), which was responsible for phosphorylation of the rat endothelial isoform of NO synthase at S1176 and thereby promoted the increase in NO production. The combined findings illustrated the crucial role for a proline-rich tyrosine kinase 2-signaling complex in the endothelial response to salt intake.
Collapse
Affiliation(s)
- Wei-Zhong Ying
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | | | | |
Collapse
|
45
|
Rajapakse NW, De Miguel C, Das S, Mattson DL. Exogenous L-arginine ameliorates angiotensin II-induced hypertension and renal damage in rats. Hypertension 2008; 52:1084-90. [PMID: 18981330 DOI: 10.1161/hypertensionaha.108.114298] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Experiments were performed to determine whether exogenous L-arginine could ameliorate angiotensin II-induced hypertension and renal damage. Rats were instrumented with chronic indwelling femoral venous and arterial catheters for infusions of drugs and measurement of conscious arterial pressure. Arterial blood pressure significantly increased from 124+/-1 to 199+/-4 mm Hg, after 9 days of continuous infusion of angiotensin II (20 ng/kg per minute; IV; n=6 to 9). In contrast, the increase in arterial pressure after 9 days of angiotensin II infusion was significantly blunted by 45% (P=0.0003) in rats coadministered L-arginine (300 microg/kg per minute; IV; n=7 to 9). The glomerular injury index was significantly greater in rats administered angiotensin II in comparison with rats administered saline vehicle (P<0.001). Coinfusion of L-arginine significantly increased plasma nitrate/nitrite concentrations (P<0.001) and completely prevented angiotensin II-induced glomerular damage (P<0.001). Angiotensin II infusion alone and combined angiotensin II plus L-arginine infusion significantly increased urinary albumin excretion. Albuminuria in rats administered angiotensin II plus L-arginine is likely to be because of increased intraglomerular pressure. Our experiments demonstrate that L-arginine can blunt angiotensin II-induced hypertension and associated renal damage. This latter observation is most exciting because it indicates that increasing NO bioavailability, in addition to lowering arterial pressure, can greatly reduce hypertension-induced renal damage.
Collapse
|
46
|
Affiliation(s)
- Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
47
|
Valtonen P, Laitinen T, Lyyra-Laitinen T, Raitakari OT, Juonala M, Viikari JSA, Heiskanen N, Vanninen E, Punnonen K, Heinonen S. Serum L-Homoarginine Concentration is Elevated During Normal Pregnancy and is Related to Flow-Mediated Vasodilatation. Circ J 2008; 72:1879-84. [DOI: 10.1253/circj.cj-08-0240] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Pirjo Valtonen
- Laboratory Centre, Department of Clinical Chemistry, Kuopio University Hospital and University of Kuopio
| | - Tomi Laitinen
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital and University of Kuopio
| | - Tiina Lyyra-Laitinen
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital and University of Kuopio
| | - Olli T. Raitakari
- Department of Clinical Physiology, Turku University Hospital and University of Turku
| | - Markus Juonala
- Department of Internal Medicine, Turku University Hospital and University of Turku
| | - Jorma SA Viikari
- Department of Internal Medicine, Turku University Hospital and University of Turku
| | - Nonna Heiskanen
- Department of Obstetrics and Gynaecology, Kuopio University Hospital and University of Kuopio
| | - Esko Vanninen
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital and University of Kuopio
| | - Kari Punnonen
- Laboratory Centre, Department of Clinical Chemistry, Kuopio University Hospital and University of Kuopio
| | - Seppo Heinonen
- Department of Obstetrics and Gynaecology, Kuopio University Hospital and University of Kuopio
| |
Collapse
|
48
|
Esqueda MED, Craig T, Hinojosa-Laborde C. Effect of Ovariectomy on Renal Estrogen Receptor-α and Estrogen Receptor-β in Young Salt-Sensitive and -Resistant Rats. Hypertension 2007; 50:768-72. [PMID: 17698719 DOI: 10.1161/hypertensionaha.107.095265] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study evaluated the effect of ovariectomy on renal estrogen receptor (ER)-α and ERβ expression in young female Dahl salt-sensitive and salt-resistant rats. Our hypothesis was that estrogen depletion results in an imbalance in ERα and ERβ expression in salt-sensitive rats. Rats were subjected to sham surgery (intact), ovariectomy, and ovariectomy with estrogen replacement. Kidneys were harvested 8 weeks later. Western blot was used to measure ERα and ERβ expression in the cortex and medulla. In intact rats, ERα was 2.7- and 4.3-fold higher in salt-sensitive compared with salt-resistant rats in the renal cortex and medulla, respectively. In salt-sensitive rats, ovariectomy caused 42% and 52% decreases in ERα and 107% and 314% increases in ERβ in renal cortex and medulla, respectively. In salt-resistant rats, ovariectomy caused 33% and 150% increases in ERα and 107% and 100% increases in ERβ in renal cortex and medulla, respectively. Estrogen replacement did not alter ERα but restored ERβ expression levels similar to levels in intact rats in both salt-sensitive and salt-resistant rats. Thus, estrogen loss had opposite effects on ERα in salt-sensitive (downregulation) and salt-resistant rats (upregulation). We propose that the decrease in ERα expression in salt-sensitive rats after estrogen loss alters the balance of renal ERs and may play a role in accelerating the development of hypertension and renal damage.
Collapse
Affiliation(s)
- Maria Eugenia Davila Esqueda
- Department of Anesthesiology, University of Texas Health Science Center, 7703 Floyd Curl Dr, San Antonio, TX 78229-3900, USA
| | | | | |
Collapse
|
49
|
Wangensteen R, Rodríguez-Gomez I, Moreno JM, Vargas F, Alvarez-Guerra M. Chronic nitric oxide blockade modulates renal Na–K–2Cl cotransporters. J Hypertens 2006; 24:2451-8. [PMID: 17082729 DOI: 10.1097/01.hjh.0000251907.93298.44] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The Na-K-2Cl cotransporter (NKCC2 isoform) of the thick ascending limb of Henle's loop (TAL) plays an important role in renal sodium handling, and the vascular isoform (NKCC1) participates in the response to vasoconstrictors. Both isoforms appear to be regulated by nitric oxide. This study aimed to analyze the effect of chronic nitric oxide deficiency on tubular and vascular Na-K-2Cl cotransporters in kidney and their potential role in the development of N-nitro-L-arginine-methyl ester (L-NAME) hypertension. METHODS Wistar rats were given L-NAME (vehicle, 10, 35 and 80 mg/100 ml drinking water) for 4 weeks. Blood pressure was measured by the tail-cuff method. NKCC2 activity was estimated as the bumetanide-sensitive Rb influx in fresh isolated TAL tubules. NKCC1-contractile function was estimated as the bumetanide-sensitive vasocontractile response to phenylephrine in isolated perfused kidneys. Acute effects of L-NAME and endothelium removal were also evaluated. NKCC2 and NKCC1 protein expression were assessed by western blot analysis. RESULTS Chronic L-NAME administration increased, in a dose-dependent manner, both blood pressure and NKCC2 activity, and these changes significantly correlated (r2 = 0.89, P < 0.01). NKCC1-contractile activity decreased with the highest dose of L-NAME (80 mg/100 ml drinking water group) but it was not affected by acute nitric oxide blockade or endothelium removal. This 80 mg group showed increased NKCC2 expression in the renal medulla and decreased NKCC1 expression in aorta. CONCLUSIONS Chronic nitric oxide deficiency stimulates tubular Na-K-2Cl cotransporter, suggesting that NKCC2 hyperactivity contributes to the inability to excrete sodium, and hence to the development of L-NAME hypertension. In contrast, L-NAME hypertension develops independently of vascular NKCC1-contractile activity.
Collapse
|
50
|
Abstract
The vascular endothelium plays a fundamental role in the basal and dynamic regulation of the circulation. Thus, it has a crucial role in the pathogenesis of hypertension. A spectrum of vasoactive substances is synthesised in the endothelium; of these, nitric oxide (NO), prostacyclin (PGI2) and endothelin (ET)-1 are the most important. There is a continuous basal release of NO determining the tone of peripheral blood vessels. Systemic inhibition of NO synthesis or scavenging of NO through oxidative stress causes an increase in arterial blood pressure. Also, the renin-angiotensin-aldosterone system has a major role in hypertension as it has a direct vasoconstrictor effect and important interactions with oxygen free radicals and NO. Prostacyclin, in contrast to NO, does not contribute to the maintenance of basal vascular tone of conduit arteries, but its effect on platelets is most important. ET acts as the natural counterpart to endothelium-derived NO and has an arterial blood pressure-raising effect in man. Anti-hypertensive therapy lowers blood pressure and may influence these different mediators, thus influencing endothelial function. In summary, due to its position between the blood pressure and smooth muscle cells responsible for peripheral resistance, the endothelium is thought to be both victim and offender in arterial hypertension. The delicate balance of endothelium-derived factors is disturbed in hypertension. Specific anti-hypertensive and anti-oxidant treatment is able to restore this balance.
Collapse
Affiliation(s)
- L E Spieker
- Cardiovascular Centre, Cardiology, Dep. of Internal Medicine, University Hospital, 8091 Zürich, Switzerland
| | | | | |
Collapse
|