1
|
Das UN, Hacimüftüoglu A, Akpinar E, Gul M, Abd El-Aty AM. Crosstalk between renin and arachidonic acid (and its metabolites). Lipids Health Dis 2025; 24:52. [PMID: 39962508 PMCID: PMC11831833 DOI: 10.1186/s12944-025-02463-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/02/2025] [Indexed: 02/21/2025] Open
Abstract
Renin plays a significant role in the regulation of blood pressure and fluid volume by modulating the renin‒angiotensin‒aldosterone (RAAS) system. Renin suppression reduces serum aldosterone levels and lowers blood pressure in addition to preserving renal function. However, exactly how renin synthesis and action are regulated and how renin suppression preserves renal function are not clear. We propose that arachidonic acid (AA) and its metabolites control renin synthesis, secretion, and action by virtue of its (AA) anti-inflammatory, cytoprotective actions and ability to regulate the secretion of renin. These findings suggest that direct renin suppression results in changes in AA metabolism. This proposal implies that AA and its metabolites may be developed as potential drugs to prevent and manage hypertension and preserve renal function.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle ground, WA, 98604, USA.
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey.
| | - Ahmet Hacimüftüoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Erol Akpinar
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Mustafa Gul
- Department of Physiology, Faculty of Medicine, Ataturk University, Erzurum, 25240, Turkey
| | - A M Abd El-Aty
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey
- Department of Pharmacology, Cairo University, Giza, 12211, Egypt
| |
Collapse
|
2
|
Gambaryan S, Mohagaonkar S, Nikolaev VO. Regulation of the renin-angiotensin-aldosterone system by cyclic nucleotides and phosphodiesterases. Front Endocrinol (Lausanne) 2023; 14:1239492. [PMID: 37674612 PMCID: PMC10478253 DOI: 10.3389/fendo.2023.1239492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is one of the key players in the regulation of blood volume and blood pressure. Dysfunction of this system is connected with cardiovascular and renal diseases. Regulation of RAAS is under the control of multiple intracellular mechanisms. Cyclic nucleotides and phosphodiesterases are the major regulators of this system since they control expression and activity of renin and aldosterone. In this review, we summarize known mechanisms by which cyclic nucleotides and phosphodiesterases regulate renin gene expression, secretion of renin granules from juxtaglomerular cells and aldosterone production from zona glomerulosa cells of adrenal gland. We also discuss several open questions which deserve future attention.
Collapse
Affiliation(s)
- Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Sanika Mohagaonkar
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
3
|
Zhao Q, Xiang Q, Tan Y, Xiao X, Xie H, Wang H, Yang M, Liu S. A novel CLCNKB variant in a Chinese family with classic Bartter syndrome and prenatal genetic diagnosis. Mol Genet Genomic Med 2022; 10:e2027. [PMID: 35913199 PMCID: PMC9544217 DOI: 10.1002/mgg3.2027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/10/2022] [Accepted: 07/19/2022] [Indexed: 11/18/2022] Open
Abstract
Background Type III Bartter syndrome (BS), often known as classic Bartter syndrome is caused by variants in CLCNKB gene, which encoding the basolateral chloride channel protein ClC‐Kb, and is characterized by renal salt wasting, hypokalemia, metabolic alkalosis, increased renin, and aldosterone levels. Methods A 2‐year‐old boy presented severe malnutrition, severe metabolic alkalosis and severe hypokalemia and was clinically diagnosed with BS. The trio exome sequencing (ES) was performed to discover the genetic cause of this patient, followed by validation using Sanger sequencing and quantitative polymerase chain reaction subsequently. Results The genetic analysis indicated that this patient with a compound heterozygous variants of CLCNKB gene including a novel nonsense variant c.876 T > A and a whole‐gene deletion. The two variants were inherited from his parents, respectively. Subsequently, target sequencing of CLCNKB gene was performed for next pregnancy, and prenatal genetic diagnosis was provided for the family. Conclusions The results of current study identified the compound heterozygous variants in a patient with classic BS. The novel variant expands the spectrum of CLCNKB variants in BS. Our study also indicates that ES is an alternative tool to simultaneously detect single‐nucleotide variants and copy‐number variants.
Collapse
Affiliation(s)
- Qianying Zhao
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Qinqin Xiang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yu Tan
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiao Xiao
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Hanbing Xie
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - He Wang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Mei Yang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Shanling Liu
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Das UN. Arachidonic Acid as Mechanotransducer of Renin Cell Baroreceptor. Nutrients 2022; 14:nu14040749. [PMID: 35215399 PMCID: PMC8874622 DOI: 10.3390/nu14040749] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
For normal maintenance of blood pressure and blood volume a well-balanced renin-angiotensin-aldosterone system (RAS) is necessary. For this purpose, renin is secreted as the situation demands by the juxtaglomerular cells (also called as granular cells) that are in the walls of the afferent arterioles. Juxtaglomerular cells can sense minute changes in the blood pressure and blood volume and accordingly synthesize, store, and secrete appropriate amounts of renin. Thus, when the blood pressure and blood volume are decreased JGA cells synthesize and secrete higher amounts of renin and when the blood pressure and blood volume is increased the synthesis and secretion of renin is decreased such that homeostasis is restored. To decipher this important function, JGA cells (renin cells) need to sense and transmit the extracellular physical forces to their chromatin to control renin gene expression for appropriate renin synthesis. The changes in perfusion pressure are sensed by Integrin β1 that is transmitted to the renin cell’s nucleus via lamin A/C that produces changes in the architecture of the chromatin. This results in an alteration (either increase or decrease) in renin gene expression. Cell membrane is situated in an unique location since all stimuli need to be transmitted to the cell nucleus and messages from the DNA to the cell external environment can be conveyed only through it. This implies that cell membrane structure and integrity is essential for all cellular functions. Cell membrane is composed to proteins and lipids. The lipid components of the cell membrane regulate its (cell membrane) fluidity and the way the messages are transmitted between the cell and its environment. Of all the lipids present in the membrane, arachidonic acid (AA) forms an important constituent. In response to pressure and other stimuli, cellular and nuclear shape changes occur that render nucleus to act as an elastic mechanotransducer that produces not only changes in cell shape but also in its dynamic behavior. Cell shape changes in response to external pressure(s) result(s) in the activation of cPLA2 (cytosolic phospholipase 2)-AA pathway that stretches to recruit myosin II which produces actin-myosin cytoskeleton contractility. Released AA can undergo peroxidation and peroxidized AA binds to DNA to regulate the expression of several genes. Alterations in the perfusion pressure in the afferent arterioles produces parallel changes in the renin cell membrane leading to changes in renin release. AA and its metabolic products regulate not only the release of renin but also changes in the vanilloid type 1 (TRPV1) expression in renal sensory nerves. Thus, AA and its metabolites function as intermediate/mediator molecules in transducing changes in perfusion and mechanical pressures that involves nuclear mechanotransduction mechanism. This mechanotransducer function of AA has relevance to the synthesis and release of insulin, neurotransmitters, and other soluble mediators release by specialized and non-specialized cells. Thus, AA plays a critical role in diseases such as diabetes mellitus, hypertension, atherosclerosis, coronary heart disease, sepsis, lupus, rheumatoid arthritis, and cancer.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St., Battle Ground, WA 98604, USA
| |
Collapse
|
5
|
Yang Y, Xu G. Update on Pathogenesis of Glomerular Hyperfiltration in Early Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2022; 13:872918. [PMID: 35663316 PMCID: PMC9161673 DOI: 10.3389/fendo.2022.872918] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
In the existing stages of diabetic kidney disease (DKD), the first stage of DKD is called the preclinical stage, characterized by glomerular hyperfiltration, an abnormally elevated glomerular filtration rate. Glomerular hyperfiltration is an independent risk factor for accelerated deterioration of renal function and progression of nephropathy, which is associated with a high risk for metabolic and cardiovascular disease. It is imperative to understand hyperfiltration and identify potential treatments to delay DKD progress. This paper summarizes the current mechanisms of hyperfiltration in early DKD. We pay close attention to the effect of glucose reabsorption mediated by sodium-glucose cotransporters and renal growth on hyperfiltration in DKD patients, as well as the mechanisms of nitric oxide and adenosine actions on renal afferent arterioles via tubuloglomerular feedback. Furthermore, we also focus on the contribution of the atrial natriuretic peptide, cyclooxygenase, renin-angiotensin-aldosterone system, and endothelin on hyperfiltration. Proposing potential treatments based on these mechanisms may offer new therapeutic opportunities to reduce the renal burden in this population.
Collapse
|
6
|
Wu X, Yang G, Chen S, Tang M, Jian S, Chen F, Wu X. Bartter syndrome with long-term follow-up: a case report. J Int Med Res 2020; 48:300060520947876. [PMID: 32857947 PMCID: PMC7705384 DOI: 10.1177/0300060520947876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bartter syndrome is a rare inherited disease caused by CLCNKB mutation, which results in inactivation of the chloride channel Kb protein. Bartter syndrome is characterized by extreme hypokalemia, hypochloremia, metabolic alkalosis, hyperrenin-induced angiotensinemia, hyperaldosteronemia, and normal blood pressure. We herein report a case of Bartter syndrome that manifested as vomiting, hypokalemia, metabolic alkalosis, normal blood pressure, and significant hyperrenin-induced angiotensinemia. The patient, a 5-month-old girl, carried two known heterozygous pathogenic mutations: c.88C > T (p.Arg30*), which she had inherited from her father, and c.1313G > A (p.Arg438His), which she had inherited from her mother. Treatment with indomethacin, a nonsteroidal anti-inflammatory drug, led to rapid improvement of the hypokalemia, and treatment was continued for 14 years. The indomethacin also induced a sustainable reduction in the hypokalemia and metabolic alkalosis.
Collapse
Affiliation(s)
- Xueling Wu
- Department of Respiratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Gang Yang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan, China
| | - Shiyu Chen
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, Sichuan, China
| | - Min Tang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, Sichuan, China
| | - Shan Jian
- Department of Paediatrics, Peking Union Medical College Hospital, Beijing, China
| | - Fuhui Chen
- Department of Respiratory, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiulin Wu
- Department of Geriatrics, Southwest Hospital, Military Medical University, Chongqing, China
| |
Collapse
|
7
|
The role of LNPEP and ANPEP gene polymorphisms in the pathogenesis of pre-eclampsia. Eur J Obstet Gynecol Reprod Biol 2020; 252:160-165. [PMID: 32619880 DOI: 10.1016/j.ejogrb.2020.06.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The exact role of renin angiotensin system (RAS) in the pathogenesis of pre-eclampsia has not been established. Gene polymorphisms, however, have been implicated in the pathophysiology. Therefore, the aim of this study was to investigate the association of the Angiotensin IV receptor and aminopeptidase-N in the pathogenesis of pre-eclampsia. STUDY DESIGN Stored blood samples of 637 South African women of African ancestry were utilized. The study population was divided into controls (n = 280) and pre-eclampsia (n = 357). Pre-eclampsia was sub-divided into early (n = 187) and late (n = 170) onset subtypes. DNA was extracted from whole blood and genotyped. Odds ratio and 95 % confidence intervals were used to assess the association. RESULTS The allele and genotype frequencies of the angiotensin receptor IV and aminopeptidase-N showed no significant difference between the control versus the pre-eclampsia groups. Similarly, allele and genotype distributions of the control group versus the subtypes of pre-eclampsia (early onset and late onset pre-eclampsia) showed no significant differences. CONCLUSION The single nucleotide polymorphisms of angiotensin IV receptor (rs18059) and aminopeptidase-N (rs6496603) are not associated with the pathogenesis of pre-eclampsia in women of African ancestry.
Collapse
|
8
|
Steglich A, Hickmann L, Linkermann A, Bornstein S, Hugo C, Todorov VT. Beyond the Paradigm: Novel Functions of Renin-Producing Cells. Rev Physiol Biochem Pharmacol 2020; 177:53-81. [PMID: 32691160 DOI: 10.1007/112_2020_27] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The juxtaglomerular renin-producing cells (RPC) of the kidney are referred to as the major source of circulating renin. Renin is the limiting factor in renin-angiotensin system (RAS), which represents a proteolytic cascade in blood plasma that plays a central role in the regulation of blood pressure. Further cells disseminated in the entire organism express renin at a low level as part of tissue RASs, which are thought to locally modulate the effects of systemic RAS. In recent years, it became increasingly clear that the renal RPC are involved in developmental, physiological, and pathophysiological processes outside RAS. Based on recent experimental evidence, a novel concept emerges postulating that next to their traditional role, the RPC have non-canonical RAS-independent progenitor and renoprotective functions. Moreover, the RPC are part of a widespread renin lineage population, which may act as a global stem cell pool coordinating homeostatic, stress, and regenerative responses throughout the organism. This review focuses on the RAS-unrelated functions of RPC - a dynamic research area that increasingly attracts attention.
Collapse
Affiliation(s)
- Anne Steglich
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Linda Hickmann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Linkermann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bornstein
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Christian Hugo
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vladimir T Todorov
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|
9
|
Abstract
Bartter syndrome is an inherited renal tubular disorder caused by a defective salt reabsorption in the thick ascending limb of loop of Henle, resulting in salt wasting, hypokalemia, and metabolic alkalosis. Mutations of several genes encoding the transporters and channels involved in salt reabsorption in the thick ascending limb cause different types of Bartter syndrome. A poor phenotype-genotype relationship due to the interaction with other cotransporters and different degrees of compensation through alternative pathways is currently reported. However, phenotypic identification still remains the first step to guide the suspicion of Bartter syndrome. Given the rarity of the syndrome, and the lack of genetic characterization in most cases, limited clinical evidence for treatment is available and the therapy is based mainly on the comprehension of renal physiology and relies on the physician's personal experiences. A better understanding of the mutated channels and transporters could possibly generate targets for specific treatment in the future, also encompassing drugs aiming to correct deficiencies in folding or plasma membrane expression of the mutated proteins.
Collapse
Affiliation(s)
- Tamara da Silva Cunha
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, São Paulo, Brazil,
| | - Ita Pfeferman Heilberg
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, São Paulo, Brazil,
| |
Collapse
|
10
|
Pharmacological properties of ASP7657, a novel, potent, and selective prostaglandin EP4 receptor antagonist. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:1319-1326. [PMID: 30076448 DOI: 10.1007/s00210-018-1545-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/19/2018] [Indexed: 10/28/2022]
Abstract
We determined the pharmacologic profile of ASP7657, trans-4-[({[1-(quinolin-2-ylmethyl)-5-(trifluoromethyl)-1H-indol-7 yl] carbonyl} amino) methyl] cyclohexanecarboxylic acid methanesulfonate (1:1), a newly synthesized selective E-type prostaglandin (EP)4 receptor antagonist using several in vitro and in vivo experiments. ASP7657 exhibited high affinity for rat and human EP4 receptors, with Ki values of 6.02 nM and 2.21 nM, respectively. In addition, ASP7657 potently inhibited the PGE2-induced cyclic adenosine monophosphate (cAMP) increase in Chinese hamster ovary (CHO) cells expressing rat EP4 receptors and human lymphoblastoid T (Jurkat) cells, with IC50 values of 0.86 nM and 0.29 nM, respectively. In contrast, ASP7657 did not inhibit the PGE2-induced intracellular calcium increase in HEK293 cells expressing rat EP1 and EP3 receptors, or cAMP increase in CHO cells expressing rat EP2 receptors. ASP7657 showed good pharmacokinetic properties following oral dosing and dose-dependently antagonized the prostaglandin (PG)E2-mediated inhibition of lipopolysaccharide-induced tumor necrosis factor-α release from rat whole blood culture. In addition, 4 weeks repeated oral administration of ASP7657 dose-dependently attenuated albuminuria in type 2 diabetic mice; these effects were significant at doses of 0.01 mg/kg or higher. These results demonstrate that ASP7657 is a potent and selective EP4 receptor antagonist that may be useful in future studies to help clarify the physiological and pathophysiological roles of PG.
Collapse
|
11
|
Meurer M, Ebert K, Schweda F, Höcherl K. The renal vasodilatory effect of prostaglandins is ameliorated in isolated-perfused kidneys of endotoxemic mice. Pflugers Arch 2018; 470:1691-1703. [DOI: 10.1007/s00424-018-2183-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/29/2022]
|
12
|
Riquier-Brison ADM, Sipos A, Prókai Á, Vargas SL, Toma L, Meer EJ, Villanueva KG, Chen JCM, Gyarmati G, Yih C, Tang E, Nadim B, Pendekanti S, Garrelds IM, Nguyen G, Danser AHJ, Peti-Peterdi J. The macula densa prorenin receptor is essential in renin release and blood pressure control. Am J Physiol Renal Physiol 2018; 315:F521-F534. [PMID: 29667908 DOI: 10.1152/ajprenal.00029.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.
Collapse
Affiliation(s)
- Anne D M Riquier-Brison
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Arnold Sipos
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Ágnes Prókai
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Sarah L Vargas
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Lldikó Toma
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Elliott J Meer
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Karie G Villanueva
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Jennifer C M Chen
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Georgina Gyarmati
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Christopher Yih
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Elaine Tang
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Bahram Nadim
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Sujith Pendekanti
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Ingrid M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam , The Netherlands
| | - Genevieve Nguyen
- Centre for Interdisciplinary Research in Biology, UMR INSERM U1050, Collège de France, Paris , France
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam , The Netherlands
| | - János Peti-Peterdi
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| |
Collapse
|
13
|
Shetty K, Shetty R, Rao P, Ballal M, Kiran A, Reddy S, Pai U, Samanth J. Comparison of Plasma Levels of Renin, Vasopressin and Atrial Natriuretic Peptide in Hypertensive Amlodipine Induced Pedal Oedema, Non-Oedema and Cilnidipine Treated Patients. J Clin Diagn Res 2017; 11:FC05-FC08. [PMID: 28764190 PMCID: PMC5535383 DOI: 10.7860/jcdr/2017/25097.9958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/06/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Amlodipine is a third generation dihydropyridine group of calcium channel blocker and having an excellent antihypertensive profile. Pedal Oedema (PE) is the major drawback of amlodipine therapy and the incidence of Amlodipine Induced Pedal Oedema (AIPE) has been found significantly high. Several neurohumoral factors influence the incidence of oedema. AIM We aimed to compare the plasma levels of renin, vasopressin and atrial natriuretic peptide in hypertensive AIPE, non-oedema and cilnidipine treated patients. MATERIALS AND METHODS The present prospective, interventional study was conducted on 104 mild to moderate hypertensive patients (52 patients in each group), after due consideration of eligibility criteria. Plasma Renin (PR), Vasopressin (VAS), and the Atrial Natriuretic Peptide (ANP) was estimated by ELISA test and compared between the AIPE, Amlodipine Treated Non-Oedema (ATNE) in Phase I, and AIPE and Cilnidipine Treated (CT) Groups in Phase II. RESULTS The clinical and demographic parameters were matched. PR was significantly high in AIPE group than the ATNE, and it was significantly reduced after one month follow up with the substitution of cilnidipine. The median (IQR) value of PR was 4.87 (3.58, 6.63), 3.50 (1.44, 5.47) and 2.66 (1.02, 5.66) ng/ml in AIPE, ATNE, CT group respectively. VAS was significantly high in AIPE group than ATNE, and it significantly reduced after one month follow up with CT group. The median (IQR) value of vasopressin was 6.78 (2.55, 9.16), 2.58 (1.61, 5.73) and 2.50 (1.23, 5.00) ng/ml in AIPE, ATNE and CT groups respectively. There was no significant difference seen in plasma ANP levels between the groups. The p-value was <0.05 which is statistically significant. CONCLUSION The AIPE may not be volume overload or fluid retention; it may be due to persistent raise in adrenergic activity followed chronic amlodipine therapy. Cilnidipine relatively suppresses the sympathetic activity, and completely resolves the AIPE by significantly reducing PR and VAS levels. ANP did not show a difference between groups. Cilnidipine is the suitable alternative antihypertensive drug for AIPE patients.
Collapse
Affiliation(s)
- Kiran Shetty
- PhD Scholar, Department of Cardiology, Kasturba Medical College, Manipal, Karnataka, India
| | - Ranjan Shetty
- Professor, Department of Cardiology, Kasturba Medical College, Manipal, Karnataka, India
| | - Pragna Rao
- Professor, Department of Biochemistry, Kasturba Medical College, Manipal, Karnataka, India
| | - Mamatha Ballal
- Professor, Department of Microbiology, Kasturba Medical College, Manipal, Karnataka, India
| | - Amruth Kiran
- Lecturer, Department of Microbiology, Kasturba Medical College, Manipal, Karnataka, India
| | - Sravan Reddy
- Registrar, Department of Cardiology, Kasturba Medical College, Manipal, Karnataka, India
| | - Umesh Pai
- Lecturer, Department of Cardiovascular Technology Course, School of Allied Health Science, Manipal, Karnataka, India
| | - Jyothi Samanth
- Lecturer, Department of Cardiovascular Technology Course, School of Allied Health Science, Manipal, Karnataka, India
| |
Collapse
|
14
|
Soy Protein Alleviates Hypertension and Fish Oil Improves Diastolic Heart Function in the Han:SPRD-Cy Rat Model of Cystic Kidney Disease. Lipids 2015; 51:635-42. [PMID: 26626478 DOI: 10.1007/s11745-015-4095-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/05/2015] [Indexed: 12/20/2022]
Abstract
Abnormalities in cardiac structure and function are very common among people with chronic kidney disease, in whom cardiovascular disease is the major cause of death. Dietary soy protein and fish oil reduce kidney disease progression in the Han:SPRD-Cy model of cystic renal disease. However, the effects of these dietary interventions in preventing alterations in cardiac structure and function due to kidney disease (reno-cardiac syndrome) in a cystic kidney disease model are not known. Therefore, weanling Han:SPRD-Cy diseased (Cy/+) and normal (+/+) rats were given diets containing either casein or soy protein, and either soy or fish oil in a three-way design for 8 weeks. Diseased rats had larger hearts, augmented left ventricular mass, and higher systolic and mean arterial blood pressure compared to the normal rats. Assessment of cardiac function using two-dimensional guided M-mode and pulse-wave Doppler echocardiography revealed that isovolumic relaxation time was prolonged in the diseased compared to normal rats, reflecting a diastolic heart dysfunction, and fish oil prevented this elevation. Soy protein resulted in a small improvement in systolic and mean arterial pressure but did not improve diastolic heart function, while fish oil prevented diastolic heart dysfunction in this model of cystic kidney disease.
Collapse
|
15
|
Gonzalez AA, Cifuentes-Araneda F, Ibaceta-Gonzalez C, Gonzalez-Vergara A, Zamora L, Henriquez R, Rosales CB, Navar LG, Prieto MC. Vasopressin/V2 receptor stimulates renin synthesis in the collecting duct. Am J Physiol Renal Physiol 2015; 310:F284-93. [PMID: 26608789 DOI: 10.1152/ajprenal.00360.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/18/2015] [Indexed: 12/19/2022] Open
Abstract
Renin is synthesized in the principal cells of the collecting duct (CD), and its production is increased via cAMP in angiotensin (ANG) II-dependent hypertension, despite suppression of juxtaglomerular (JG) renin. Vasopressin, one of the effector hormones of the renin-angiotensin system (RAS) via the type 2-receptor (V2R), activates the cAMP/PKA/cAMP response element-binding protein (CREB) pathway and aquaporin-2 expression in principal cells of the CD. Accordingly, we hypothesized that activation of V2R increases renin synthesis via PKA/CREB, independently of ANG II type 1 (AT1) receptor activation in CD cells. Desmopressin (DDAVP; 10(-6) M), a selective V2R agonist, increased renin mRNA (∼3-fold), prorenin (∼1.5-fold), and renin (∼2-fold) in cell lysates and cell culture media in the M-1 CD cell line. Cotreatment with DDAVP+H89 (PKA inhibitor) or CREB short hairpin (sh) RNA prevented this response. H89 also blunted DDAVP-induced CREB phosphorylation and nuclear localization. In 48-h water-deprived (WD) mice, prorenin-renin protein levels were increased in the renal inner medulla (∼1.4- and 1.8-fold). In WD mice treated with an ACE inhibitor plus AT1 receptor blockade, renin mRNA and prorenin protein levels were still higher than controls, while renin protein content was not changed. In M-1 cells, ANG II or DDAVP increased prorenin-renin protein levels; however, there were no further increases by combined treatment. These results indicate that in the CD the activation of the V2R stimulates renin synthesis via the PKA/CREB pathway independently of RAS, suggesting a critical role for vasopressin in the regulation of renin in the CD.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile;
| | | | | | - Alex Gonzalez-Vergara
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Leonardo Zamora
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ricardo Henriquez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Carla B Rosales
- Department of Physiology Tulane University, School of Medicine, New Orleans, Louisiana; and
| | - L Gabriel Navar
- Department of Physiology Tulane University, School of Medicine, New Orleans, Louisiana; and Hypertension and Renal Center of Excellence, Tulane University, School of Medicine, New Orleans, Louisiana
| | - Minolfa C Prieto
- Department of Physiology Tulane University, School of Medicine, New Orleans, Louisiana; and Hypertension and Renal Center of Excellence, Tulane University, School of Medicine, New Orleans, Louisiana
| |
Collapse
|
16
|
Abadir PM, Siragy HM. Angiotensin type 1 receptor mediates renal production and conversion of prostaglandins E2 to F2α in conscious diabetic rats. J Renin Angiotensin Aldosterone Syst 2015. [PMID: 26195268 DOI: 10.1177/1470320315592566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Previous studies demonstrated that stimulation of angiotensin subtype 1 receptor (AT1R) led to increased renal generation of prostaglandins E2 (PGE2) and renal inflammation. In turn, PGE2 increases AT1R activity. The conversion of PGE2 to the less active metabolite prostaglandin F2α (PGF2α) via 9-ketoreductase interrupts this feedback loop. The effects of diabetes on the interface between AT1R, PGE2 and PGF2α are not well established. We hypothesized that in diabetes, an aberrant AT1R activity enhances the biosynthesis of PGE2 and impairs the activity of PGE 9-ketoreductase, leading to accumulation of PGE2. MATERIALS AND METHODS Using microdialysis technique, we monitored renal interstitial fluid levels of angiotensin II (Ang II), PGE2 and PGF2α in control and AT1R blocker, valsartan, treated diabetic rats (N=8 each). We utilized the PGF2α to PGE2 ratio as indirect measure of PGE 9-ketoreductase activity. RESULTS Diabetes increased renal interstitial fluid levels of Ang II, PGE2 and PGF2α. PGF2α/PGE2 ratio increased by the third week, but declined by the sixth week of diabetes. Valsartan reduced PGE2 and PGF2α levels and increased Ang II and the conversion of PGE2 to PGF2α. CONCLUSION Our results suggest that in diabetes, AT1R increases PGE2 generation and reduces conversion of PGE2 to PGF2α with the progression of diabetes.
Collapse
Affiliation(s)
- Peter M Abadir
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, Baltimore, USA
| | - Helmy M Siragy
- University of Virginia School of Medicine, Department of Medicine, Charlottesville, USA
| |
Collapse
|
17
|
Hao S, Hernandez A, Quiroz-Munoz M, Cespedes C, Vio CP, Ferreri NR. PGE(2) EP(3) receptor downregulates COX-2 expression in the medullary thick ascending limb induced by hypertonic NaCl. Am J Physiol Renal Physiol 2014; 307:F736-46. [PMID: 25080527 DOI: 10.1152/ajprenal.00204.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We tested the hypothesis that inhibition of EP3 receptors enhances cyclooxygenase (COX)-2 expression in the thick ascending limb (TAL) induced by hypertonic stimuli. COX-2 protein expression in the outer medulla increased approximately twofold in mice given free access to 1% NaCl in the drinking water for 3 days. The increase was associated with an approximate threefold elevation in COX-2 mRNA accumulation and an increase in PGE2 production by isolated medullary (m)TAL tubules from 77.3 ± 8.4 to 165.7 ± 10.8 pg/mg protein. Moreover, administration of NS-398 abolished the increase in PGE2 production induced by 1% NaCl. EP3 receptor mRNA levels also increased approximately twofold in the outer medulla of mice that ingested 1% NaCl. The selective EP3 receptor antagonist L-798106 increased COX-2 mRNA by twofold in mTAL tubules, and the elevation in COX-2 protein induced by 1% NaCl increased an additional 50% in mice given L-798106. COX-2 mRNA in primary mTAL cells increased twofold in response to media made hypertonic by the addition of NaCl (400 mosmol/kg H2O). L-798106 increased COX-2 mRNA twofold in isotonic media and fourfold in cells exposed to 400 mosmol/kg H2O. PGE2 production by mTAL cells increased from 79.3 ± 4.6 to 286.7 ± 6.3 pg/mg protein after challenge with 400 mosmol/kg H2O and was inhibited in cells transiently transfected with a lentivirus short hairpin RNA construct targeting exon 5 of COX-2 to silence COX-2. Collectively, the data suggest that local hypertonicity in the mTAL is associated with an increase in COX-2 expression concomitant with elevated EP3 receptor expression, which limits COX-2 activity in this segment of the nephron.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Alejandra Hernandez
- Department of Physiology, Center for Aging and Regeneration, CARE Chile UC, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Mariana Quiroz-Munoz
- Department of Physiology, Center for Aging and Regeneration, CARE Chile UC, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Carlos Cespedes
- Department of Physiology, Center for Aging and Regeneration, CARE Chile UC, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Carlos P Vio
- Department of Physiology, Center for Aging and Regeneration, CARE Chile UC, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| |
Collapse
|
18
|
Wang F, Lu X, Peng K, Du Y, Zhou SF, Zhang A, Yang T. Prostaglandin E-prostanoid4 receptor mediates angiotensin II-induced (pro)renin receptor expression in the rat renal medulla. Hypertension 2014; 64:369-77. [PMID: 24866147 DOI: 10.1161/hypertensionaha.114.03654] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiotensin II (Ang II) stimulates (pro)renin receptor (PRR) expression in the renal collecting duct, triggering the local renin response in the distal nephron. Our recent study provided evidence for involvement of cyclooxygenase-2-prostaglandin E2 pathway in Ang II-dependent stimulation of PRR expression in the collecting duct. Here, we tested the role of E-prostanoid (EP) subtypes acting downstream of cyclooxygenase-2 in this phenomenon. In primary rat inner medullary collecting duct cells, Ang II treatment for 12 hours induced a 1.8-fold increase in the full-length PRR protein expression. To assess the contribution of EP receptor, the cell was pretreated with specific EP receptor antagonists: SC-51382 (for EP1), L-798106 (for EP3), L-161982 (for EP4), and ONO-AE3-208 (ONO, a structurally distinct EP4 antagonist). The upregulation of PRR expression by Ang II was consistently abolished by L-161982 and ONO and partially suppressed by SC-51382 but was unaffected by L-798106. The PRR expression was also significantly elevated by the EP4 agonist CAY10598 in the absence of Ang II. Sprague-Dawley rats were subsequently infused for 1 or 2 weeks with vehicle, Ang II alone, or in combination with ONO. Ang II infusion induced parallel increases in renal medullary PRR protein and renal medullary and urinary renin activity and total renin content, all of which were blunted by ONO. Both tail cuff plethysmography and telemetry demonstrated attenuation of Ang II hypertension by ONO. Overall, these results have established a crucial role of the EP4 receptor in mediating the upregulation of renal medullary PRR expression and renin activity during Ang II hypertension.
Collapse
Affiliation(s)
- Fei Wang
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Xiaohan Lu
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Kexin Peng
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Yaomin Du
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Shu-Feng Zhou
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Aihua Zhang
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.)
| | - Tianxin Yang
- From the Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China (F.W., X.L., K.P., T.Y.); Department of Internal Medicine, University of Utah, Salt Lake City (F.W., X.L., T.Y.); Veterans Affairs Medical Center, Salt Lake City, UT (F.W., X.L., T.Y.); Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China (Y.D.); Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa (S.-F.Z.); and Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China (A.Z.).
| |
Collapse
|
19
|
Eberle JAM, Müller-Roth KL, Widmayer P, Chubanov V, Gudermann T, Breer H. Putative interaction of brush cells with bicarbonate secreting cells in the proximal corpus mucosa. Front Physiol 2013; 4:182. [PMID: 23874305 PMCID: PMC3711009 DOI: 10.3389/fphys.2013.00182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/26/2013] [Indexed: 01/18/2023] Open
Abstract
The gastric epithelium is protected from the highly acidic luminal content by alkaline mucus which is secreted from specialized epithelial cells. In the stomach of mice strong secretion of alkaline fluid was observed at the “gastric groove,” the border between corpus and fundus mucosa. Since this region is characterized by numerous brush cells it was proposed that these cells might secrete alkaline solution as suggested for brush cells in the bile duct. In fact, it was found that in this region multiple cells express elements which are relevant for the secretion of bicarbonate, including carbonic anhydrase (CAII), the cystic fibrosis transmembrane conductance regulator (CFTR) and the Na+/H+ exchanger (NHE1). However, this cell population was distinct from brush cells which express the TRP-channel TRPM5 and are considered as putative sensory cells. The location of both cell populations in close proximity implies the possibility for a paracrine interaction. This view was substantiated by the finding that brush cells express prostaglandin synthase-1 (COX-1) and the neighboring cells a specific receptor type for prostaglandins. The notion that brush cells may be able to sense a local acidification was supported by the observation that they express the channel PKD1L3 which contributes to the acid responsiveness of gustatory sensory cells. The results support the concept that brush cells may sense the luminal content and influence via prostaglandins the secretion of alkaline solution.
Collapse
|
20
|
Abstract
The renin-angiotensin-aldosterone-system (RAAS) plays a central role in the pathophysiology of heart failure and cardiorenal interaction. Drugs interfering in the RAAS form the pillars in treatment of heart failure and cardiorenal syndrome. Although RAAS inhibitors improve prognosis, heart failure–associated morbidity and mortality remain high, especially in the presence of kidney disease. The effect of RAAS blockade may be limited due to the loss of an inhibitory feedback of angiotensin II on renin production. The subsequent increase in prorenin and renin may activate several alternative pathways. These include the recently discovered (pro-) renin receptor, angiotensin II escape via chymase and cathepsin, and the formation of various angiotensin subforms upstream from the blockade, including angiotensin 1–7, angiotensin III, and angiotensin IV. Recently, the direct renin inhibitor aliskiren has been proven effective in reducing plasma renin activity (PRA) and appears to provide additional (tissue) RAAS blockade on top of angiotensin-converting enzyme and angiotensin receptor blockers, underscoring the important role of renin, even (or more so) under adequate RAAS blockade. Reducing PRA however occurs at the expense of an increase plasma renin concentration (PRC). PRC may exert direct effects independent of PRA through the recently discovered (pro-) renin receptor. Additional novel possibilities to interfere in the RAAS, for instance using vitamin D receptor activation, as well as the increased knowledge on alternative pathways, have revived the question on how ideal RAAS-guided therapy should be implemented. Renin and prorenin are pivotal since these are at the base of all of these pathways.
Collapse
|
21
|
Neubauer B, Machura K, Kettl R, Lopez MLSS, Friebe A, Kurtz A. Endothelium-derived nitric oxide supports renin cell recruitment through the nitric oxide-sensitive guanylate cyclase pathway. Hypertension 2013; 61:400-7. [PMID: 23297374 DOI: 10.1161/hypertensionaha.111.00221] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic challenge of renin-angiotensin causes recruitment of renin-producing cells in the kidney along the media layer of afferent arterioles and hypertrophy of cells in the juxtaglomerular apparatus. This study aimed to define the role of nitric oxide (NO) with regard to the recruitment pattern of renin-producing cells and to the possible pathways along which NO could act. We considered the hypothesis that endothelium-derived NO acts via NO-sensitive guanylate cyclase. Mice were treated with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks, which led to a 13-fold increase in renin expression associated with marked recruitment of renin cells in afferent arterioles and hypertrophy of the juxtaglomerular apparatus in wild-type mice. In wild-type mice additionally treated with the nonselective NO synthase inhibitor L-NAME, the recruitment of renin-expressing cells along the afferent arterioles was absent and juxtaglomerular hypertrophy was diminished. An almost identical attenuation of renin cell recruitment as with L-NAME treatment in wild-type mice was found in mice lacking the endothelial isoform of NO synthase. Treatment of mice lacking NO-sensitive guanylate cyclase in renin-expressing cells and preglomerular smooth muscle cells with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks produced juxtaglomerular hypertrophy like in wild-type mice, but no recruitment in afferent arterioles. These findings suggest that endothelium-derived NO and concomitant formation of cGMP in preglomerular renin cell precursors supports recruitment of renin-expressing cells along preglomerular vessels, but not in the juxtaglomerular apparatus.
Collapse
Affiliation(s)
- Björn Neubauer
- Institute of Physiology, University of Regensburg, Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
22
|
Pöschke A, Kern N, Maruyama T, Pavenstädt H, Narumiya S, Jensen BL, Nüsing RM. The PGE(2)-EP4 receptor is necessary for stimulation of the renin-angiotensin-aldosterone system in response to low dietary salt intake in vivo. Am J Physiol Renal Physiol 2012; 303:F1435-42. [PMID: 22993066 DOI: 10.1152/ajprenal.00512.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased cyclooxygenase-2 (COX-2) expression and PGE(2) synthesis have been shown to be prerequisites for renal renin release after Na(+) deprivation. To answer the question of whether EP4 receptor type of PGE(2) mediates renin regulation under a low-salt diet, we examined renin regulation in EP4(+/+), EP4(-/-), and in wild-type mice treated with EP4 receptor antagonist. After 2 wk of a low-salt diet (0.02% wt/wt NaCl), EP4(+/+) mice showed diminished Na(+) excretion, unchanged K(+) excretion, and reduced Ca(2+) excretion. Diuresis and plasma electrolytes remained unchanged. EP4(-/-) exhibited a similar attenuation of Na(+) excretion; however, diuresis and K(+) excretion were enhanced, and plasma Na(+) concentration was higher, whereas plasma K(+) concentration was lower compared with control diet. There were no significant differences between EP4(+/+) and EP4(-/-) mice in blood pressure, creatinine clearance, and plasma antidiuretic hormone (ADH) concentration. Following salt restriction, plasma renin and aldosterone concentrations and kidney renin mRNA level rose significantly in EP4(+/+) but not in EP4(-/-) and in wild-type mice treated with EP4 antagonist ONO-AE3-208. In the latter two groups, the low-salt diet caused a significantly greater rise in PGE(2) excretion. Furthermore, mRNA expression for COX-2 and PGE(2) synthetic activity was significantly greater in EP4(-/-) than in EP4(+/+) mice. We conclude that low dietary salt intake induces expression of COX-2 followed by enhanced renal PGE(2) synthesis, which stimulates the renin-angiotensin-aldosterone system by activation of EP4 receptor. Most likely, defects at the step of EP4 receptor block negative feedback mechanisms on the renal COX system, leading to persistently high PGE(2) levels, diuresis, and K(+) loss.
Collapse
Affiliation(s)
- Antje Pöschke
- Institute of Clinical Pharmacology, Goethe Univ., Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Schnermann J, Briggs JP. Tubular control of renin synthesis and secretion. Pflugers Arch 2012; 465:39-51. [PMID: 22665048 DOI: 10.1007/s00424-012-1115-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 01/11/2023]
Abstract
The intratubular composition of fluid at the tubulovascular contact site of the juxtaglomerular apparatus serves as regulatory input for secretion and synthesis of renin. Experimental evidence, mostly from in vitro perfused preparations, indicates an inverse relation between luminal NaCl concentration and renin secretion. The cellular transduction mechanism is initiated by concentration-dependent NaCl uptake through the Na-K-2Cl cotransporter (NKCC2) with activation of NKCC2 causing inhibition and deactivation of NKCC2 causing stimulation of renin release. Changes in NKCC2 activity are coupled to alterations in the generation of paracrine factors that interact with granular cells. Among these factors, generation of PGE2 in a COX-2-dependent fashion appears to play a dominant role in the stimulatory arm of tubular control of renin release. [NaCl] is a determinant of local PG release over an appropriate concentration range, and blockade of COX-2 activity interferes with the NaCl dependency of renin secretion. The complex array of local paracrine controls also includes nNOS-mediated synthesis of nitric oxide, with NO playing the role of a modifier of the intracellular signaling pathway. A role of adenosine may be particularly important when [NaCl] is increased, and at least some of the available evidence is consistent with an important suppressive effect of adenosine at higher salt concentrations.
Collapse
Affiliation(s)
- Jurgen Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 10, Rm 4D50, NIDDK, NIH, 10 Center Drive MSC 1370, Bethesda, MD 20892, USA.
| | | |
Collapse
|
24
|
Bayorh M, Rollins-Hairston A, Adiyiah J, Lyn D, Eatman D. Eplerenone inhibits aldosterone-induced renal expression of cyclooxygenase. J Renin Angiotensin Aldosterone Syst 2012; 13:353-9. [PMID: 22554826 DOI: 10.1177/1470320312443911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The upregulation of cyclooxygenase (COX) expression by aldosterone (ALDO) or high salt diet intake is very interesting and complex in the light of what is known about the role of COX in renal function. Thus, in this study, we hypothesize that apocynin (APC) and/or eplerenone (EPL) inhibit ALDO/salt-induced kidney damage by preventing the production of prostaglandin E₂ (PGE₂). METHODS Dahl salt-sensitive rats on either a low-salt or high-salt diet were treated with ALDO (0.2 mg pellet) in the presence of EPL (100 mg/kg/day) or APC (1.5 mM). Indirect blood pressure, prostaglandins and ALDO levels and histological changes were measured. RESULTS Cyclooxygenase-2 (COX-2) levels were upregulated in the renal tubules and peritubular vessels after high-salt intake, and APC attenuated renal tubular COX-2 protein expression induced by ALDO. Plasma PGE₂ levels were significantly reduced by ALDO in the rats fed a low-salt diet when compared to rats fed a high-salt diet. PGE₂ was blocked by EPL but increased in the presence of APC. CONCLUSIONS The beneficial effects of EPL may be associated with an inhibition of PGE₂. The mechanism underlying the protective effects of EPL is clearly distinct from that of APC and suggests that these agents can have differential roles in cardiovascular disease.
Collapse
Affiliation(s)
- Ma Bayorh
- Department of Pharmacology/Toxicology, Morehouse School of Medicine, Atlanta, Georgia 30310, USA.
| | | | | | | | | |
Collapse
|
25
|
Nilsson L, Madsen K, Topcu SO, Jensen BL, Frøkiær J, Nørregaard R. Disruption of cyclooxygenase-2 prevents downregulation of cortical AQP2 and AQP3 in response to bilateral ureteral obstruction in the mouse. Am J Physiol Renal Physiol 2012; 302:F1430-9. [PMID: 22397925 DOI: 10.1152/ajprenal.00682.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Bilateral ureteral obstruction (BUO) in rats is associated with increased cyclooxygenase type 2 (COX-2) expression, and selective COX-2 inhibition prevents downregulation of aquaporins (AQPs) in response to BUO. It was hypothesized that a murine model would display similar changes in renal COX-2 and AQPs upon BUO and that targeted disruption of COX-2 protects against BUO-induced suppression of collecting duct AQPs. COX-2(-/-) and wild-type littermates (C57BL/6) were employed to determine COX-1, -2, AQP2, and AQP3 protein abundances and localization after BUO. In a separate series, sham and BUO wild-type mice were treated with a selective COX-2 inhibitor, parecoxib. The COX-2 protein level increased in wild-type mice in response to BUO and was not detectable in COX-2(-/-). COX-1 protein abundance was increased in sham-operated and BUO mice. Total AQP2 and -3 mRNA and protein levels decreased significantly after BUO in the cortex+outer medulla (C+OM) and inner medulla (IM). The decrease in C+OM AQP2 and -3 levels was attenuated/prevented in COX-2(-/-) mice, whereas there was no change in the IM. In parallel, inhibition of COX-2 by parecoxib rescued C+OM AQP3 and IM AQP2 protein level in wild-type mice subjected to BUO. In summary, 1) In C57BL/6 mice, ureteral obstruction increases renal COX-2 expression in interstitial cells and lowers AQP2/-3 abundance and 2) inhibition of COX-2 activity by targeted disruption or pharmacological blockade attenuates obstruction-induced AQP downregulation. In conclusion, COX-2-derived prostaglandins contribute to downregulation of transcellular water transporters in the collecting duct and likely to postobstruction diureses in the mouse.
Collapse
Affiliation(s)
- Line Nilsson
- The Water and Salt Research Center, Institute of Clinical Medicine, Aarhus University. Hospital-Skejby, Brendstrupgaardsvej 100, Aarhus N, Denmark
| | | | | | | | | | | |
Collapse
|
26
|
Schnermann J, Briggs JP. Synthesis and secretion of renin in mice with induced genetic mutations. Kidney Int 2012; 81:529-38. [PMID: 22258323 DOI: 10.1038/ki.2011.451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The juxtaglomerular (JG) cell product renin is rate limiting in the generation of the bioactive octapeptide angiotensin II. Rates of synthesis and secretion of the aspartyl protease renin by JG cells are controlled by multiple afferent and efferent pathways originating in the CNS, cardiovascular system, and kidneys, and making critical contributions to the maintenance of extracellular fluid volume and arterial blood pressure. Since both excesses and deficits of angiotensin II have deleterious effects, it is not surprising that control of renin is secured by a complex system of feedforward and feedback relationships. Mice with genetic alterations have contributed to a better understanding of the networks controlling renin synthesis and secretion. Essential input for the setting of basal renin generation rates is provided by β-adrenergic receptors acting through cyclic adenosine monophosphate, the primary intracellular activation mechanism for renin mRNA generation. Other major control mechanisms include COX-2 and nNOS affecting renin through PGE2, PGI2, and nitric oxide. Angiotensin II provides strong negative feedback inhibition of renin synthesis, largely an indirect effect mediated by baroreceptor and macula densa inputs. Adenosine appears to be a dominant factor in the inhibitory arms of the baroreceptor and macula densa mechanisms. Targeted gene mutations have also shed light on a number of novel aspects related to renin processing and the regulation of renin synthesis and secretion.
Collapse
Affiliation(s)
- Jurgen Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
27
|
Facemire CS, Nguyen M, Jania L, Beierwaltes WH, Kim HS, Koller BH, Coffman TM. A major role for the EP4 receptor in regulation of renin. Am J Physiol Renal Physiol 2011; 301:F1035-41. [PMID: 21835766 DOI: 10.1152/ajprenal.00054.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prostaglandins have been implicated as paracrine regulators of renin secretion, but the specific pathways and receptor(s) carrying out these functions have not been fully elucidated. To examine the contributions of prostanoid synthetic pathways and receptors to regulation of renin in the intact animal, we used a panel of mice with targeted disruption of several key genes: cyclooxygenase-2 (COX-2), microsomal PGE synthases 1 and 2 (mPGES1, mPGES2), EP2 and EP4 receptors for PGE(2), and the IP receptor for PGI(2). To activate the macula densa signal for renin stimulation, mice were treated with furosemide over 5 days and renin mRNA levels were determined by real-time RT-PCR. At baseline, there were no differences in renin mRNA levels between wild-type and the various strains of mutant mice. Furosemide caused marked stimulation of renin mRNA expression across all groups of wild-type control mice. This response was completely abrogated in the absence of COX-2, but was unaffected in mice lacking mPGES1 or mPGES2. The absence of G(s)/cAMP-linked EP2 receptors had no effect on stimulation of renin by furosemide and there was only a modest, insignificant reduction in renin responses in mice lacking the IP receptor. By contrast, renin stimulation in EP4(-/-) mice was significantly reduced by ∼70% compared with wild-type controls. These data suggest that stimulation of renin by the macula densa mechanism is mediated by PGE(2) through a pathway requiring COX-2 and the EP4 receptor, but not EP2 or IP receptors. Surprisingly, mPGES1 or mPGES2 are not required, suggesting other alternative mechanisms for generating PGE(2) in response to macula densa stimulation.
Collapse
Affiliation(s)
- Carie S Facemire
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
In the adult organism, systemically circulating renin almost exclusively originates from the juxtaglomerular cells in the afferent arterioles of the kidneys. These cells share similarities with pericytes and myofibro-blasts. They store renin in a vesicular network and granules and release it in a regulated fashion. The release mode of renin is not understood; in particular, the involvement of SNARE proteins is unknown. Renin release is acutely increased via the cAMP signaling pathway, which is triggered mainly by catecholamines and other G(s)-coupled agonists, and is inhibited by calcium-related pathways that are commonly activated by vasoconstrictors. Renin release from juxtaglomerular cells is directly modulated in an inverse fashion by the blood pressure inside the afferent arterioles and by the chloride content in the tubule fluid at the macula densa segment of the distal tubule. Renin release is stimulated by nitric oxide and by prostanoids released by neighboring endothelial and macula densa cells. Steady-state renin concentrations in the plasma are determined essentially by the number of renin-producing cells in the afferent arterioles, which changes in parallel with challenges to the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Armin Kurtz
- Physiologisches Institut der Universität, Regensburg, Germany.
| |
Collapse
|
29
|
Mendez M, Gross KW, Glenn ST, Garvin JL, Carretero OA. Vesicle-associated membrane protein-2 (VAMP2) mediates cAMP-stimulated renin release in mouse juxtaglomerular cells. J Biol Chem 2011; 286:28608-18. [PMID: 21708949 PMCID: PMC3151102 DOI: 10.1074/jbc.m111.225839] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Renin is essential for blood pressure control. Renin is stored in granules in juxtaglomerular (JG) cells, located in the pole of the renal afferent arterioles. The second messenger cAMP stimulates renin release. However, it is unclear whether fusion and exocytosis of renin-containing granules is involved. In addition, the role of the fusion proteins, SNAREs (soluble N-ethylmaleimide-sensitive factor attachment proteins), in renin release from JG cells has not been studied. The vesicle SNARE proteins VAMP2 (vesicle associated membrane protein 2) and VAMP3 mediate cAMP-stimulated exocytosis in other endocrine cells. Thus, we hypothesized that VAMP2 and/or -3 mediate cAMP-stimulated renin release from JG cells. By fluorescence-activated cell sorting, we isolated JG cells expressing green fluorescent protein and compared the relative abundance of VAMP2/3 in JG cells versus total mouse kidney mRNA by quantitative PCR. We found that VAMP2 and VAMP3 mRNA are expressed and enriched in JG cells. Confocal imaging of primary cultures of JG cells showed that VAMP2 (but not VAMP3) co-localized with renin-containing granules. Cleavage of VAMP2 and VAMP3 with tetanus toxin blocked cAMP-stimulated renin release from JG cells by ∼50% and impaired cAMP-stimulated exocytosis by ∼50%, as monitored with FM1–43. Then we specifically knocked down VAMP2 or VAMP3 by adenoviral-mediated delivery of short hairpin silencing RNA. We found that silencing VAMP2 blocked cAMP-induced renin release by ∼50%. In contrast, silencing VAMP3 had no effect on basal or cAMP-stimulated renin release. We conclude that VAMP2 and VAMP3 are expressed in JG cells, but only VAMP2 is targeted to renin-containing granules and mediates the stimulatory effect of cAMP on renin exocytosis.
Collapse
Affiliation(s)
- Mariela Mendez
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | | | |
Collapse
|
30
|
Abstract
Many reports indicate that there is an increase in almost all of the components of the renin-angiotensin system (RAS) during an uncomplicated pregnancy, but renin activity, angiotensin II, and aldosterone decrease in preeclampsia (PE) for reasons that are unclear. PE is a life-threatening disorder of late pregnancy characterized by hypertension, proteinuria, increased soluble fms-like tyrosine kinase-1, as well as renal and placental morphologic abnormalities. Although a leading cause of maternal and perinatal morbidity and mortality, the pathogenic mechanisms of PE remain largely undefined. Immunologic mechanisms and aberrations of the RAS have been long considered contributors to the disorder. Bridging these two concepts, numerous studies report the presence of the angiotensin II type I receptor agonistic autoantibody (AT(1)-AA) found circulating in preeclamptic women. This autoantibody induces many key features of the disorder through AT(1) receptor signaling, and has been implicated in the pathogenesis of PE. Here we review the functions of the RAS during normal pregnancy and PE, and highlight the role of AT(1)-AA in both animal models and in the human disorder.
Collapse
Affiliation(s)
- Roxanna A Irani
- Department of Biochemistry & Molecular Biology, University of Texas at Houston Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | | |
Collapse
|
31
|
Aldehni F, Tang T, Madsen K, Plattner M, Schreiber A, Friis UG, Hammond HK, Han PL, Schweda F. Stimulation of renin secretion by catecholamines is dependent on adenylyl cyclases 5 and 6. Hypertension 2011; 57:460-8. [PMID: 21282557 DOI: 10.1161/hypertensionaha.110.167130] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The sympathetic nervous system stimulates renin release from juxtaglomerular cells via the β-adrenoreceptor-cAMP pathway. Recent in vitro studies have suggested that the calcium-inhibited adenylyl cyclases (ACs) 5 and 6 possess key roles in the control of renin exocytosis. To investigate the relative contribution of AC5 and AC6 to the regulation of renin release in vivo we performed experiments using AC5 and AC6 knockout mice. Male AC5(-/-) mice exhibited normal plasma renin concentrations, renal renin synthesis (mRNA and renin content), urinary volume, and systolic blood pressure. In male AC6(-/-) mice, plasma renin concentration (AC6(-/-): 732 ± 119; AC6 (+/+): 436 ± 78 ng of angiotensin I per hour*mL(-1); P<0.05), and renin synthesis were stimulated associated with an increased excretion of dilute urine (1.55-fold; P<0.05) and reduced blood pressure (-10.6 mm Hg; P<0.001). Stimulation of plasma renin concentration by a single injection of the β-adrenoreceptor agonist isoproterenol (10 mg/kg IP) was significantly attenuated in AC5(-/-) (male: -20%; female: -33%) compared with wild-type mice in vivo. The mitigation of the plasma renin concentration response to isoproterenol was even more pronounced in AC6(-/-) (male: -63%; female: -50% versus AC6(+/+)). Similarly, the effects of isoproterenol, prostaglandin E2, and pituitary adenylyl cyclase-activating polypeptide on renin release from isolated perfused kidneys were attenuated to a higher extent in AC6(-/-) (-51% to -98% versus AC6(+/+)) than in AC5(-/-) (-31% to 46% versus AC5(+/+)). In conclusion, both AC5 and AC6 are involved in the stimulation of renin secretion in vivo, and AC6 is the dominant isoforms in this process.
Collapse
Affiliation(s)
- Fadi Aldehni
- Institute of Physiology, University of Regensburg, 93040 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Castrop H, Höcherl K, Kurtz A, Schweda F, Todorov V, Wagner C. Physiology of Kidney Renin. Physiol Rev 2010; 90:607-73. [PMID: 20393195 DOI: 10.1152/physrev.00011.2009] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The protease renin is the key enzyme of the renin-angiotensin-aldosterone cascade, which is relevant under both physiological and pathophysiological settings. The kidney is the only organ capable of releasing enzymatically active renin. Although the characteristic juxtaglomerular position is the best known site of renin generation, renin-producing cells in the kidney can vary in number and localization. (Pro)renin gene transcription in these cells is controlled by a number of transcription factors, among which CREB is the best characterized. Pro-renin is stored in vesicles, activated to renin, and then released upon demand. The release of renin is under the control of the cAMP (stimulatory) and Ca2+(inhibitory) signaling pathways. Meanwhile, a great number of intrarenally generated or systemically acting factors have been identified that control the renin secretion directly at the level of renin-producing cells, by activating either of the signaling pathways mentioned above. The broad spectrum of biological actions of (pro)renin is mediated by receptors for (pro)renin, angiotensin II and angiotensin-( 1 – 7 ).
Collapse
Affiliation(s)
- Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Klaus Höcherl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Vladimir Todorov
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
33
|
Facemire CS, Griffiths R, Audoly LP, Koller BH, Coffman TM. The impact of microsomal prostaglandin e synthase 1 on blood pressure is determined by genetic background. Hypertension 2010; 55:531-8. [PMID: 20065147 DOI: 10.1161/hypertensionaha.109.145631] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostaglandin (PG)E(2) has multiple actions that may affect blood pressure. It is synthesized from arachidonic acid by the sequential actions of phospholipases, cyclooxygenases, and PGE synthases. Although microsomal PGE synthase (mPGES)1 is the only genetically verified PGE synthase, results of previous studies examining the consequences of mPGES1 deficiency on blood pressure (BP) are conflicting. To determine whether genetic background modifies the impact of mPGES1 on BP, we generated mPGES1(-/-) mice on 2 distinct inbred backgrounds, DBA/1lacJ and 129/SvEv. On the DBA/1 background, baseline BP was similar between wild-type (WT) and mPGES1(-/-) mice. By contrast, on the 129 background, baseline BPs were significantly higher in mPGES1(-/-) animals than WT controls. During angiotensin II infusion, the DBA/1 mPGES1(-/-) and WT mice developed mild hypertension of similar magnitude, whereas 129-mPGES1(-/-) mice developed more severe hypertension than WT controls. DBA/1 animals developed only minimal albuminuria in response to angiotensin II infusion. By contrast, WT 129 mice had significantly higher levels of albumin excretion than WT DBA/1 and the extent of albuminuria was further augmented in 129 mPGES1(-/-) animals. In WT mice of both strains, the increase in urinary excretion of PGE(2) with angiotensin II was attenuated in mPGES1(-/-) animals. Urinary excretion of thromboxane was unaffected by angiotensin II in the DBA/1 lines but increased more than 4-fold in 129 mPGES1(-/-) mice. These data indicate that genetic background significantly modifies the BP response to mPGES1 deficiency. Exaggerated production of thromboxane may contribute to the robust hypertension and albuminuria in 129 mPGES1-deficient mice.
Collapse
Affiliation(s)
- Carie S Facemire
- Department of Medicine, Division of Nephrology, Duke University and Durham Veterans Affairs Medical Centers, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
34
|
Prostaglandin F2alpha elevates blood pressure and promotes atherosclerosis. Proc Natl Acad Sci U S A 2009; 106:7985-90. [PMID: 19416858 DOI: 10.1073/pnas.0811834106] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Little is known about prostaglandin F(2alpha) in cardiovascular homeostasis. Prostaglandin F(2alpha) dose-dependently elevates blood pressure in WT mice via activation of the F prostanoid (FP) receptor. The FP is expressed in preglomerular arterioles, renal collecting ducts, and the hypothalamus. Deletion of the FP reduces blood pressure, coincident with a reduction in plasma renin concentration, angiotensin, and aldosterone, despite a compensatory up-regulation of AT1 receptors and an augmented hypertensive response to infused angiotensin II. Plasma and urinary osmolality are decreased in FP KOs that exhibit mild polyuria and polydipsia. Atherogenesis is retarded by deletion of the FP, despite the absence of detectable receptor expression in aorta or in atherosclerotic lesions in Ldlr KOs. Although vascular TNF(alpha), inducible nitric oxide enzyme and TGF(beta) are reduced and lesional macrophages are depleted in the FP/Ldlr double KOs, this result reflects the reduction in lesion burden, as the FP is not expressed on macrophages and its deletion does not alter macrophage cytokine generation. Blockade of the FP offers an approach to the treatment of hypertension and its attendant systemic vascular disease.
Collapse
|
35
|
Vargas SL, Toma I, Kang JJ, Meer EJ, Peti-Peterdi J. Activation of the succinate receptor GPR91 in macula densa cells causes renin release. J Am Soc Nephrol 2009; 20:1002-11. [PMID: 19389848 DOI: 10.1681/asn.2008070740] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Macula densa (MD) cells of the juxtaglomerular apparatus (JGA) are salt sensors and generate paracrine signals that control renal blood flow, glomerular filtration, and release of the prohypertensive hormone renin. We hypothesized that the recently identified succinate receptor GPR91 is present in MD cells and regulates renin release. Using immunohistochemistry, we identified GPR91 in the apical plasma membrane of MD cells. Treatment of MD cells with succinate activated mitogen-activated protein kinases (MAPKs; p38 and extracellular signal-regulated kinases 1/2) and cyclooxygenase 2 (COX-2) and induced the synthesis and release of prostaglandin E(2), a potent vasodilator and classic paracrine mediator of renin release. Using microperfused JGA and real-time confocal fluorescence imaging of quinacrine-labeled renin granules, we detected significant renin release in response to tubular succinate (EC(50) 350 microM). Genetic deletion of GPR91 (GPR91(-/-) mice) or pharmacologic inhibition of MAPK or COX-2 blocked succinate-induced renin release. Streptozotocin-induced diabetes caused GPR91-dependent upregulation of renal cortical phospho-p38, extracellular signal-regulated kinases 1/2, COX-2, and renin content. Salt depletion for 1 wk increased plasma renin activity seven-fold in wild-type mice but only 3.4-fold in GPR91(-/-) mice. In summary, MD cells can sense alterations in local tissue metabolism via accumulation of tubular succinate and GPR91 signaling, which involves the activation of MAPKs, COX-2, and the release of prostaglandin E(2). This mechanism may be integral in the regulation of renin release and activation of the renin-angiotensin system in health and disease.
Collapse
Affiliation(s)
- Sarah Laurin Vargas
- Keck School of Medicine, University of Southern California, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
36
|
Irani RA, Xia Y. The functional role of the renin-angiotensin system in pregnancy and preeclampsia. Placenta 2008; 29:763-71. [PMID: 18687466 DOI: 10.1016/j.placenta.2008.06.011] [Citation(s) in RCA: 212] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 06/22/2008] [Accepted: 06/23/2008] [Indexed: 01/23/2023]
Abstract
During normal pregnancy, the renin-angiotensin system (RAS) plays a vitally important role in salt balance and subsequent well-being of mother and fetus. In this balance, one must consider not only the classical renal RAS but also that of the uteroplacental unit, where both maternal and fetal tissues contribute to the signaling cascade. Many studies have shown that in normal pregnancy there is an increase in almost all of the components of the RAS. In derangements of pregnancy this delicate equilibrium can become unbalanced. Preeclampsia is one such case. It is a disorder of pregnancy characterized by hypertension, proteinuria and placental abnormalities associated with shallow trophoblast invasion and impaired spiral artery remodeling. Despite being a leading cause of maternal death and a major contributor to maternal and perinatal morbidity, the mechanisms responsible for the pathogenesis of preeclampsia are poorly understood. Immunological mechanisms and the RAS have been long considered to be involved in the development of preeclampsia. Numerous recent studies demonstrate the presence of the angiotensin II type I receptor agonistic autoantibody (AT1-AA). This autoantibody can induce many key features of the disorder and upregulate molecules involved in the pathogenesis of preeclampsia. Here we review the functional role of the RAS during pregnancy and the impact of AT1-AA on preeclampsia.
Collapse
Affiliation(s)
- R A Irani
- University of Texas - Houston Health Science Center, Department of Biochemistry and Molecular Biology, 6431 Fannin Street, MSB 6.200, Houston, TX 77030, USA
| | | |
Collapse
|
37
|
Abstract
Cyclooxygenase-derived prostanoids exert complex and diverse functions within the kidney. The biological effect of each prostanoid is controlled at multiple levels, including (a) enzymatic reactions catalyzed sequentially by cyclooxygenase and prostanoid synthase for the synthesis of bioactive prostanoid and (b) the interaction with its receptors that mediate its functions. Cyclooxygenase-derived prostanoids act in an autocrine or a paracrine fashion and can serve as physiological buffers, protecting the kidney from excessive functional changes during physiological stress. Through these actions, prostanoids play important roles in maintaining renal function, body fluid homeostasis, and blood pressure. Renal cortical COX2-derived prostanoids, particularly PGI2 and PGE2, play critical roles in maintaining blood pressure and renal function in volume-contracted states. Renal medullary COX2-derived prostanoids appear to have an antihypertensive effect in individuals challenged with a high-salt diet. Loss of EP2 or IP receptor is associated with salt-sensitive hypertension. COX2 also plays a role in maintaining renal medullary interstitial cell viability in the hypertonic environment of the medulla. Cyclooxygenase-derived prostanoids also are involved in certain pathological processes. The cortical COX2-derived PGI2 participates in the pathogenesis of renal vascular hypertension through stimulating renal renin synthesis and release. COX-derived prostanoids also appear to be involved in the pathogenesis of diabetic nephropathy. COXs, prostanoid synthases, and prostanoid receptors should provide fruitful targets for intervention in the pharmacological treatment of renal disease.
Collapse
Affiliation(s)
- Chuan-Ming Hao
- Division of Nephrology, Department of Medicine, Vanderbilt University, and Veterans Affair Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
38
|
Wagner C, de Wit C, Gerl M, Kurtz A, Höcherl K. Increased expression of cyclooxygenase 2 contributes to aberrant renin production in connexin 40-deficient kidneys. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1781-6. [PMID: 17855490 DOI: 10.1152/ajpregu.00439.2007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously found that deletion of connexin 40 (Cx40) causes a misdirection of renin-expressing cells from the media layer of afferent arterioles to the perivascular tissue, extraglomerular mesangium, and periglomerular and peritubular interstitium. The mechanisms underlying this aberrant renin expression are unknown. Here, we questioned the relevance of cyclooxygenase-2 (COX-2) activity for aberrant renin expression in Cx40-deficient kidneys. We found that COX-2 mRNA levels were increased three-fold in the renal cortex of Cx40-deficient kidneys relative to wild-type (wt) kidneys. In wt kidneys, COX-2 immunoreactivity was minimally detected in the juxtaglomerular region, but renin expression was frequently associated with COX-2 immunoreactivity in Cx40-deficient kidneys. Treatment with COX-2 inhibitors for 1 wk lowered renin mRNA levels in wt kidneys by about 40%. In Cx40-deficient kidneys, basal renin mRNA levels were increased two-fold relative to wt kidneys, and these elevated mRNA levels were reduced to levels of untreated wt mice by COX-2 inhibitors. In parallel, renin immunoreactive areas were clearly reduced by COX-2 inhibitors such that renin expression vanished and decreased significantly in the periglomerular and peritubular extensions. Notably, COX-2 inhibitor treatment lowered plasma renin concentration (PRC) in wt kidneys by about 40% but did not affect the highly elevated PRC levels in Cx40-deficient mice. These findings suggest that aberrant renin-producing cells in Cx40-deficient kidneys express significant amounts of COX-2, which contribute to renin expression in these cells, in particular, those in the periglomerular and peritubular position. Apparently, these disseminated cells do not contribute to the enhanced renin secretion rates of Cx40-deficient kidneys.
Collapse
Affiliation(s)
- Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany.
| | | | | | | | | |
Collapse
|
39
|
Abstract
The aspartyl-protease renin is the key regulator of the renin-angiotensin-aldosterone system, which is critically involved in salt, volume, and blood pressure homeostasis of the body. Renin is mainly produced and released into circulation by the so-called juxtaglomerular epithelioid cells, located in the walls of renal afferent arterioles at the entrance of the glomerular capillary network. It has been known for a long time that renin synthesis and secretion are stimulated by the sympathetic nerves and the prostaglandins and are inhibited in negative feedback loops by angiotensin II, high blood pressure, salt, and volume overload. In contrast, the events controlling the function of renin-secreting cells at the organ and cellular level are markedly less clear and remain mysterious in certain aspects. The unravelling of these mysteries has led to new and interesting insights into the process of renin release.
Collapse
Affiliation(s)
- Frank Schweda
- Institute of Physiology, University of Regensburg, Germany; and
| | - Ulla Friis
- Department of Physiology and Pharmacology, Southern Denmark University at Odense, Odense, Denmark
| | | | - Ole Skott
- Department of Physiology and Pharmacology, Southern Denmark University at Odense, Odense, Denmark
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Germany; and
| |
Collapse
|
40
|
Matzdorf C, Kurtz A, Höcherl K. COX-2 activity determines the level of renin expression but is dispensable for acute upregulation of renin expression in rat kidneys. Am J Physiol Renal Physiol 2007; 292:F1782-90. [PMID: 17376760 DOI: 10.1152/ajprenal.00513.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of cyclooxygenase 2 (COX-2) in the control of renin is still a matter of debate, since studies with COX-2-deficient mice or with COX-2 inhibitors produced conflicting findings. Therefore, we studied the effect of the COX-2 inhibitor SC-58236 on the regulation of the renin system in adult rat kidneys. Renocortical tissue levels and urinary excretion of PGE(2) were reduced to 65 and 40% of control values, respectively, after a single gavage of SC-58236 and did not further decrease on prolonged treatment. Plasma renin activity (PRA) and renin mRNA levels began to decrease after 3 days and reached a constant level of approximately 60% of control values after 5 days of treatment. Isoproterenol or left renal artery clipping for 2 days increased PRA and renin mRNA to similar levels in both vehicle- and SC-58236-treated rats after 2 days. Pretreatment with SC-58236 for 5 days, however, reduced the absolute increase in PRA and renin mRNA levels. Notably, the relative increases were not different between vehicle- and SC-58236-treated rats. Similar findings were observed for the stimulation of the renin system by angiotensin II inhibition and low salt intake. These findings indicate that COX-2 inhibition attenuates renin secretion and renin gene expression stimulated by a variety of parameters in proportion to the lowering of basal renin activity, while it does not interfere with the different stimulatory mechanism per se. As a consequence, it appears as if COX-2 activity relevantly determines the set point of the activity of the renin system in rat kidneys.
Collapse
Affiliation(s)
- Corina Matzdorf
- Institute für Pharmakologie, Universität Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
41
|
Makhanova N, Lee G, Takahashi N, Sequeira Lopez ML, Gomez RA, Kim HS, Smithies O. Kidney function in mice lacking aldosterone. Am J Physiol Renal Physiol 2005; 290:F61-9. [PMID: 16118390 DOI: 10.1152/ajprenal.00257.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To explore the effects of decreased amounts or absence of aldosterone, we have disrupted the gene coding for aldosterone synthase (AS) in mice and investigated blood pressure and kidney function in AS+/+, AS+/-, and AS-/- mice. AS+/- mice have normal blood pressures and show no abnormalities in electrolytes or kidney gene expression, but they have significantly higher than normal urine volume and lower urine osmolality. In contrast, the AS-/- mice have low blood pressure, abnormal electrolyte homeostasis (increased plasma concentrations of K+, Ca2+, and Mg2+ and decreased concentrations of HCO3(-) and Cl- but no difference in the plasma Na+ level), and disturbances in water metabolism (higher urine output, decreased urine osmolality, and impaired urine concentrating and diluting ability). Absence of aldosterone in the AS-/- mice induced several compensatory changes: an increased food intake-to-body weight ratio, an elevated plasma concentration of glucocorticoids, and strong activation of the renin-angiotensin system. Parallel with the markedly increased synthesis and release of renin, the AS-/- mice showed increased expression of cyclooxygenase-2 (COX-2) in macula densa. On salt supplementation, plasma electrolyte concentrations and kidney renin and COX-2 levels became similar to those of wild-type mice, but the lower blood pressure of the AS-/- mice was not corrected. Thus absence of aldosterone in AS-/- mice results in impairment of Na+ reabsorption in the distal nephron, decreased blood pressure, and strong renin-angiotensin system activation. Our data show the substantial correction of these abnormalities, except the low blood pressure, by high dietary salt does not depend on aldosterone.
Collapse
Affiliation(s)
- Natalia Makhanova
- Dept. of Pathology & Laboratory Medicine, Univ. of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7525, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Castrop H, Lorenz JN, Hansen P, Friis U, Mizel D, Oppermann M, Jensen B, Briggs J, Skott O, Schnermann J. Contribution of the basolateral isoform of the Na-K-2Cl- cotransporter (NKCC1/BSC2) to renin secretion. Am J Physiol Renal Physiol 2005; 289:F1185-92. [PMID: 16106034 PMCID: PMC3153878 DOI: 10.1152/ajprenal.00455.2004] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute administration of loop diuretics like furosemide leads to a stimulation of renin secretion, an effect thought to result from inhibition of Na-K-2Cl cotransporter (NKCC2)-mediated salt transport at the luminal surface of the macula densa (MD). However, loop diuretics also inhibit NKCC1, the second isoform of the Na-K-2Cl cotransporter, with similar potency. In the present study, we examined the influence of furosemide on renin secretion in NKCC1-deficient mice to distinguish between effects of the loop diuretic involving NKCC2 and, by implication, the MD pathway, and effects that might occur via inhibition of NKCC1. Baseline plasma renin concentration (PRC) was 1,212 +/- 211 in NKCC1+/+ (n = 13) and 3,851 +/- 579 ng ANG I.ml(-1).h(-1) in NKCC1-/- mice (n = 14; P = 0.00024). Acute administration of furosemide (50 mg/kg i.p.) increased PRC significantly to 9,324 +/- 1,018 ng ANG I.ml(-1).h(-1) in NKCC1+/+ (n = 13; P < 0.0001 compared with basal) and to 14,188 +/- 2,274 ng ANG I.ml(-1).h(-1) in NKCC1-/- mice [n = 14; P = 0.0002 compared with basal; P = 0.034 compared with wild-type (WT) plus furosemide]. Renin mRNA expression was about threefold higher in NKCC1-/- compared with WT mice. There was considerable recruitment of granular cells to upstream regions of afferent arterioles in NKCC1-/- mice. Patch-clamp studies in single juxtaglomerular granular (JG) cells from WT mice showed an approximately 10% increase in membrane capacitance during incubation with furosemide (10(-4) M), indicating a direct effect of the loop diuretic on renin secretion. No effect of furosemide on membrane capacitance was observed in JG cells from NKCC1-deficient mice. Furosemide (10(-3) M) significantly stimulated renin release from primary cultures of JG cells from WT mice, whereas no response was observed in NKCC1-/- mice. Our data suggest that a functional NKCC1 suppresses basal renin release, at least in part, through a direct effect on JG cells.
Collapse
Affiliation(s)
- Hayo Castrop
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | | | - Pernille Hansen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Ulla Friis
- Department of Physiology, University of Southern Denmark, Odense, Denmark
| | - Diane Mizel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Mona Oppermann
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Boye Jensen
- Department of Physiology, University of Southern Denmark, Odense, Denmark
| | - Josie Briggs
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Ole Skott
- Department of Physiology, University of Southern Denmark, Odense, Denmark
| | - Jurgen Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
43
|
Friis UG, Stubbe J, Uhrenholt TR, Svenningsen P, Nüsing RM, Skøtt O, Jensen BL. Prostaglandin E2 EP2 and EP4 receptor activation mediates cAMP-dependent hyperpolarization and exocytosis of renin in juxtaglomerular cells. Am J Physiol Renal Physiol 2005; 289:F989-97. [PMID: 15985651 DOI: 10.1152/ajprenal.00201.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PGE(2) and PGI(2) stimulate renin secretion and cAMP accumulation in juxtaglomerular granular (JG) cells. We addressed, at the single-cell level, the receptor subtypes and intracellular transduction mechanisms involved. Patch clamp was used to determine cell capacitance (C(m)), current, and membrane voltage in response to PGE(2), EP2 and EP4 receptor agonists, and an IP receptor agonist. PGE(2) (0.1 micromol/l) increased C(m) significantly, and the increase was abolished by intracellular application of the protein kinase A antagonist Rp-8-CPT-cAMPS. EP2-selective ligands butaprost (1 micromol/l), AE1-259-01 (1 nmol/l), EP4-selective agonist AE1-329 (1 nmol/l), and IP agonist iloprost (1 micromol/l) significantly increased C(m) mediated by PKA. The EP4 antagonist AE3-208 (10 nmol/l) blocked the effect of EP4 agonist but did not alter the response to PGE(2). Application of both EP4 antagonist and EP2-antagonist AH-6809 abolished the effects of PGE(2) on C(m) and current. EP2 and EP4 ligands stimulated cAMP formation in JG cells. PGE(2) rapidly stimulated renin secretion from superfused JG cells and diminished the membrane-adjacent granule pool as determined by confocal microscopy. The membrane potential hyperpolarized significantly after PGE(2), butaprost, AE1-329 and AE1-259 and outward current was augmented in a PKA-dependent fashion. PGE(2)-stimulated outward current, but not C(m) change, was abolished by the BK(Ca) channel inhibitor iberiotoxin (300 nmol/l). EP2 and EP4 mRNA was detected in sampled JG cells, and the preglomerular and glomerular vasculature was immunopositive for EP4. Thus IP, EP2, and EP4 receptors are associated with JG cells, and their activation leads to rapid PKA-mediated exocytotic fusion and release of renin granules.
Collapse
Affiliation(s)
- Ulla G Friis
- Dept. of Physiology and Pharmacology, University of Southern Denmark, DK-5000 Odense C, Denmark
| | | | | | | | | | | | | |
Collapse
|
44
|
Fujino T, Nakagawa N, Yuhki KI, Hara A, Yamada T, Takayama K, Kuriyama S, Hosoki Y, Takahata O, Taniguchi T, Fukuzawa J, Hasebe N, Kikuchi K, Narumiya S, Ushikubi F. Decreased susceptibility to renovascular hypertension in mice lacking the prostaglandin I2 receptor IP. J Clin Invest 2004; 114:805-12. [PMID: 15372104 PMCID: PMC516260 DOI: 10.1172/jci21382] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Accepted: 07/14/2004] [Indexed: 12/22/2022] Open
Abstract
Persistent reduction of renal perfusion pressure induces renovascular hypertension by activating the renin-angiotensin-aldosterone system; however, the sensing mechanism remains elusive. Here we investigated the role of PGI2 in renovascular hypertension in vivo, employing mice lacking the PGI2 receptor (IP-/- mice). In WT mice with a two-kidney, one-clip model of renovascular hypertension, the BP was significantly elevated. The increase in BP in IP-/- mice, however, was significantly lower than that in WT mice. Similarly, the increases in plasma renin activity, renal renin mRNA, and plasma aldosterone in response to renal artery stenosis were all significantly lower in IP-/- mice than in WT mice. All these parameters were measured in mice lacking the four PGE2 receptor subtypes individually, and we found that these mice had similar responses to WT mice. PGI2 is produced by COX-2 and a selective inhibitor of this enzyme, SC-58125, also significantly reduced the increases in plasma renin activity and renin mRNA expression in WT mice with renal artery stenosis, but these effects were absent in IP-/- mice. When the renin-angiotensin-aldosterone system was activated by salt depletion, SC-58125 blunted the response in WT mice but not in IP-/- mice. These results indicate that PGI2 derived from COX-2 plays a critical role in regulating the release of renin and consequently renovascular hypertension in vivo.
Collapse
MESH Headings
- Animals
- Blood Pressure/physiology
- Creatinine/blood
- Creatinine/urine
- Crosses, Genetic
- Cyclooxygenase Inhibitors/pharmacology
- Dinoprostone/physiology
- Epoprostenol/physiology
- Gene Expression Regulation
- Genetic Predisposition to Disease
- Hypertension, Renal/etiology
- Hypertension, Renal/genetics
- Hypertension, Renal/physiopathology
- Hypertension, Renal/prevention & control
- Infarction/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nephrectomy
- Prostaglandin-Endoperoxide Synthases/genetics
- Pyrazoles/pharmacology
- RNA, Messenger/genetics
- Receptors, Epoprostenol/deficiency
- Receptors, Epoprostenol/genetics
- Renal Circulation
- Renin-Angiotensin System/genetics
Collapse
Affiliation(s)
- Takayuki Fujino
- Department of Pharmacology, Asahikawa Medical College, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schweda F, Klar J, Narumiya S, Nüsing RM, Kurtz A. Stimulation of renin release by prostaglandin E2is mediated by EP2and EP4receptors in mouse kidneys. Am J Physiol Renal Physiol 2004; 287:F427-33. [PMID: 15113745 DOI: 10.1152/ajprenal.00072.2004] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PGE2is a potent stimulator of renin release. So far, the contribution of each of the four PGE2receptor subtypes (EP1–EP4) in the regulation of renin release has not been characterized. Therefore, we investigated the effects PGE2on renin secretion rates (RSR) from isolated, perfused kidneys of EP1−/−, EP2−/−, EP3−/−, EP4−/−, and wild-type mice. PGE2concentration dependently stimulated RSR from kidneys of all four knockout strains with a threshold concentration of 1 nM in EP1−/−, EP2−/−, EP3−/−, and wild-type mice, whereas the threshold concentration was shifted to 10 nM in EP4−/− mice. Moreover, the maximum stimulation of RSR by PGE2at 1 μM was significantly reduced in EP4−/− (12.8-fold of control) and EP2−/− (15.9-fold) compared with wild-type (20.7-fold), EP1−/− (23.8-fold), and EP3−/− (20.1-fold). In contrast, stimulation of RSR by either the loop diuretic bumetanide or the β-adrenoceptor agonist isoproterenol was similar in all strains. PGE2exerted a dual effect on renal vascular tone, inducing vasodilatation at low concentrations (1 nmol/) and vasoconstriction at higher concentrations (100 nmol/) in kidneys of wild-type mice. In kidneys of EP2−/− as well as EP4−/− mice, vasodilatation at low PGE2concentrations was prevented, whereas vasoconstriction at higher concentrations was augmented. In contrast, the vasodilatatory component was pronounced in kidneys of EP1and EP3knockout mice, whereas in both genotypes the vasoconstriction at higher PGE2concentrations was markedly blunted. Our data provide evidence that PGE2stimulates renin release via activation of EP2and EP4receptors, whereas EP1and EP3receptors appear to be without functional relevance in juxtaglomerular cells. In contrast, all four receptor subtypes are involved in the control of renal vascular tone, EP1and EP3receptors increasing, and EP2as well as EP4receptors, decreasing it.
Collapse
MESH Headings
- Animals
- Dinoprostone/pharmacology
- In Vitro Techniques
- Juxtaglomerular Apparatus/metabolism
- Kidney/blood supply
- Kidney/drug effects
- Kidney/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E/metabolism
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP3 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
- Renal Circulation
- Renin/metabolism
- Vascular Resistance
Collapse
Affiliation(s)
- Frank Schweda
- Institute for Physiology, University of Regensburg, 93042 Regensburg, Germany.
| | | | | | | | | |
Collapse
|
46
|
Schweda F, Kammerl M, Wagner C, Krämer BK, Kurtz A. Upregulation of macula densa cyclooxygenase-2 expression is not dependent on glomerular filtration. Am J Physiol Renal Physiol 2004; 287:F95-101. [PMID: 15180925 DOI: 10.1152/ajprenal.00404.2003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although the regulation of cyclooxygenase-2 (COX-2) expression in the kidney cortex has been extensively characterized, the physiological control mechanisms of COX-2 expression at the level of the kidney and at the level of the tubular cells are not well understood. Based on the current hypothesis that tubular salt transport might be a crucial regulator of COX-2 expression, this study aimed to determine the impact of salt delivery to the tubules (glomerular filtration) for the regulation of COX-2 in the kidney cortex in vivo. To this end, glomerular filtration of the right kidney was abrogated by the ligation of the right ureter of male Sprague-Dawley rats. After 1 wk of ligation, the animals were treated with subcutaneous infusions of furosemide (12 mg·kg−1·day−1) or with a low-salt or a high-salt diet (0.02% wt/wt; 8% wt/wt), and COX-2 as well as renin mRNA expression were determined in the ligated and the nonligated contralateral kidney. During ureteral ligation, hydronephrosis developed with a reduction of medullary mass, while the cortex was preserved. Expressions of the Na-K-2Cl cotransporter isoforms A and B were both reduced in the hydronephrotic cortex to 70 and 35% of the corresponding contralateral intact kidney. Despite the abrogation of glomerular filtration, detected by inulin clearance measurements, renocortical COX-2 mRNA abundance was stimulated by furosemide treatment (3.2-fold) or low-salt diet (2.9-fold) to similar degrees compared with the intact contralateral kidney (2.7-fold for both treatments), whereas a high-salt diet did not significantly suppress COX-2 mRNA in the macula densa region of either kidney. Renin mRNA expression was regulated strictly in parallel in both kidneys, a low-salt diet or furosemide treatment stimulating and a high-salt diet suppressing it. We conclude from these findings that salt delivery to the tubules is not an essential requirement for the upregulation of COX-2 by salt deficiency or by loop diuretics in the rat kidney cortex nor is it for chronic stimulation of renin mRNA expression.
Collapse
Affiliation(s)
- Frank Schweda
- Institut für Physiologie, Universität Regensburg, 93042 Regensburg, Germany.
| | | | | | | | | |
Collapse
|
47
|
Mertz HL, Liu J, Valego NK, Stallings SP, Figueroa JP, Rose JC. Inhibition of cyclooxygenase-2: effects on renin secretion and expression in fetal lambs. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1012-8. [PMID: 12456386 DOI: 10.1152/ajpregu.00523.2002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The importance of prostaglandins in the regulation of the renin-angiotensin system during development is not known. These experiments were conducted to examine the effects of prostaglandin synthesis inhibitors on basal and isoproterenol-induced plasma renin concentration and renin gene expression in the late-gestation fetal lamb. Eighteen lamb fetuses ranging in gestational age from 129 to 138 days underwent surgical insertion of femoral arterial and venous catheters under general endotracheal anesthesia. After a period of recovery, animals underwent an infusion of isoproterenol after administration of a saline bolus (control experiments); 24-48 h later a second study was performed after administration of NS-398, a cyclooxygenase (COX)-2 inhibitor, or saline for a second control study. Administration of COX-2 inhibitor significantly reduced baseline plasma renin levels and attenuated responses in fetal renin secretion to isoproterenol infusions. Renal cortical cells from animals receiving COX-2 inhibitor had significantly lower levels of renin mRNA compared with animals receiving only saline. Renal cortical cells in culture from animals receiving only saline exhibited increased levels of renin mRNA when treated with isoproterenol, forskolin, or IBMX. Only forskolin increased renin mRNA levels in renal cortical cells in culture from animals receiving COX-2 inhibitor. We conclude that prostaglandins play a stimulatory role in the regulation of the renin-angiotensin system and are necessary for beta-adrenergic stimulation of renin secretion and gene expression in the late-gestation fetal lamb.
Collapse
Affiliation(s)
- Heather L Mertz
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | |
Collapse
|
48
|
Hartner A, Cordasic N, Goppelt-Struebe M, Veelken R, Hilgers KF. Role of macula densa cyclooxygenase-2 in renovascular hypertension. Am J Physiol Renal Physiol 2003; 284:F498-502. [PMID: 12429553 DOI: 10.1152/ajprenal.00136.2002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Upregulation of the inducible cyclooxygenase (COX-2) in the macula densa accompanies the activation of the juxtaglomerular apparatus in many high-renin conditions. The functional role of COX-2 in these disease states is poorly understood. We tested whether COX-2 is required to increase renin in renovascular hypertension. Rats with established two-kidney, one-clip (2K1C) hypertension were treated for 2 wk with two different inhibitors of COX-2, NS-398 and rofecoxib, respectively. Hypertension in 2K1C rats was not affected or slightly enhanced by COX-2 inhibition, as measured intra-arterially in conscious animals. The increase in plasma renin activity was also unchanged by both rofecoxib and NS-398. The number of glomeruli with a renin-positive juxtaglomerular apparatus was elevated in clipped kidneys and decreased in contralateral kidneys of 2K1C rats. This pattern was unaltered by COX-2 inhibition. To test the effects of COX-2 blockade on a primarily macula densa-mediated stimulus, we studied salt depletion for comparison. A low-salt diet induced a significant increase in plasma renin activity, which was partially inhibited by treatment with NS-398. We conclude that inhibition of COX-2 in established renovascular hypertension does not affect renin synthesis or release. Thus either COX-2 is not necessary for the macula densa mechanism or the macula densa is not important for maintaining high renin in renovascular hypertension.
Collapse
Affiliation(s)
- Andrea Hartner
- Department of Medicine IV, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | | |
Collapse
|
49
|
Todorov V, Müller M, Schweda F, Kurtz A. Tumor necrosis factor-alpha inhibits renin gene expression. Am J Physiol Regul Integr Comp Physiol 2002; 283:R1046-51. [PMID: 12376397 DOI: 10.1152/ajpregu.00142.2002] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Renin, produced in renal juxtaglomerular (JG) cells, is a fundamental regulator of blood pressure. Accumulating evidence suggests that cytokines may directly influence renin production in the JG cells. TNF-alpha, which is one of the key mediators in immunity and inflammation, is known to participate in the control of vascular proliferation and contraction and hence in the pathogenesis of cardiovascular diseases. Thus TNF-alpha may exert its effects on the cardiovascular system through modulation of renal renin synthesis. Therefore we have tested the effect of TNF-alpha on renin transcription in As4.1 cells, which represent transformed mouse JG cells, and in native mouse JG cells in culture. Renin gene expression was also determined in mice lacking the gene for TNF-alpha (TNF-alpha knockout mice). TNF-alpha inhibited renin gene expression via an inhibition of the transcriptional activity, targeting the proximal 4.1 kb of the renin promoter in As4.1 cells. TNF-alpha also attenuated forskolin-stimulated renin gene expression in primary cultures of mouse JG cells. Mice lacking the TNF-alpha gene had almost threefold higher basal renal renin mRNA abundance relative to the control strain. The general physiological regulation of renin expression by salt was not disturbed in TNF-alpha knockout mice. Our data suggest that TNF-alpha inhibits renin gene transcription at the cellular level and thus may act as a modulator of renin synthesis in (physio)pathological situations.
Collapse
Affiliation(s)
- Vladimir Todorov
- Institut für Physiologie I, Universität Regensburg, D-93040 Regensburg, Germany.
| | | | | | | |
Collapse
|
50
|
Imig JD, Breyer MD, Breyer RM. Contribution of prostaglandin EP(2) receptors to renal microvascular reactivity in mice. Am J Physiol Renal Physiol 2002; 283:F415-22. [PMID: 12167591 DOI: 10.1152/ajprenal.00351.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present studies were performed to determine the contribution of EP(2) receptors to renal hemodynamics by examining afferent arteriolar responses to PGE(2), butaprost, sulprostone, and endothelin-1 in EP(2) receptor-deficient male mice (EP(2)-/-). Afferent arteriolar diameters averaged 17.8 +/- 0.8 microm in wild-type (EP(2)+/+) mice and 16.7 +/- 0.7 microm in EP(2)-/- mice at a renal perfusion pressure of 100 mmHg. Vessels from both groups of mice responded to norepinephrine (0.5 microM) with similar 17-19% decreases in diameter. Diameters of norepinephrine-preconstricted afferent arterioles increased by 7 +/- 2 and 20 +/- 6% in EP(2)+/+ mice in response to 1 microM PGE(2) and 1 microM butaprost, respectively. In contrast, afferent arteriolar diameter of EP(2)-/- mice decreased by 13 +/- 3 and 16 +/- 6% in response to PGE(2) and butaprost. The afferent arteriolar vasoconstriction to butaprost in EP(2)-/- mice was eliminated by angiotensin-converting enzyme inhibition. Sulprostone, an EP(1) and EP(3) receptor ligand, decreased afferent arteriolar diameter in both groups; however, the vasoconstriction in the EP(2)-/- mice was greater than in the EP(2)+/+ mice. Endothelin-1-mediated afferent arteriolar diameter responses were enhanced in EP(2)-/- mice. Afferent arteriolar diameter decreased by 29 +/- 7% in EP(2)-/- and 12 +/- 7% in EP(2)+/+ mice after administration of 1 nM endothelin-1. These results demonstrate that the EP(2) receptor mediates a portion of the PGE(2) afferent arteriolar vasodilation and buffers the renal vasoconstrictor responses elicited by EP(1) and EP(3) receptor activation as well as endothelin-1.
Collapse
Affiliation(s)
- John D Imig
- Vascular Biology Center, Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | |
Collapse
|