1
|
Bankir L, Guerrot D, Bichet DG. Vaptans or voluntary increased hydration to protect the kidney: how do they compare? Nephrol Dial Transplant 2023; 38:562-574. [PMID: 34586414 DOI: 10.1093/ndt/gfab278] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/13/2022] Open
Abstract
The adverse effects of vasopressin (AVP) in diverse forms of chronic kidney disease have been well described. They depend on the antidiuretic action of AVP mediated by V2 receptors (V2R). Tolvaptan, a selective V2R antagonist, is now largely used for the treatment of patients with autosomal dominant polycystic kidney disease. Another way to reduce the adverse effects of AVP is to reduce endogenous AVP secretion by a voluntary increase in fluid intake. These two approaches differ in several ways, including the level of thirst and AVP. With voluntary increased drinking, plasma osmolality will decline and so will AVP secretion. Thus, not only will V2R-mediated effects be reduced, but also those mediated by V1a and V1b receptors (V1aR and V1bR). In contrast, selective V2R antagonism will induce a loss of fluid that will stimulate AVP secretion and thus increase AVP's influence on V1a and V1b receptors. V1aR is expressed in the luminal side of the collecting duct (CD) and in inner medullary interstitial cells, and their activation induces the production of prostaglandins, mostly prostaglandin E2 (PGE2). Intrarenal PGE2 has been shown to reduce sodium and water reabsorption in the CD and increase blood flow in the renal medulla, both effects contributing to increase sodium and water excretion and reduce urine-concentrating activity. Conversely, non-steroidal anti-inflammatory drugs have been shown to induce significant water and sodium retention and potentiate the antidiuretic effects of AVP. Thus, during V2R antagonism, V1aR-mediated actions may be responsible for part of the diuresis observed with this drug. These V1aR-dependent effects do not take place with a voluntary increase in fluid intake. In summary, while both strategies may have beneficial effects, the information reviewed here leads us to assume that pharmacological V2R antagonism, with resulting stimulation of V1aR and increased PGE2 production, may provide greater benefit than voluntary high water intake. The influence of tolvaptan on the PGE2 excretion rate and the possibility to use somewhat lower tolvaptan doses than presently prescribed remain to be evaluated.
Collapse
Affiliation(s)
- Lise Bankir
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,CNRS, ERL 8228-Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Dominique Guerrot
- Départment de Néphrologie, Hôpital Universitaire de Rouen, Rouen, France.,Université de Normandie, UNIROUEN, INSERM U1096, Rouen, France
| | - Daniel G Bichet
- Université de Montréal, Montréal, Quebec, Canada.,Département de Pharmacologie, Département de Physiologie, and Département de Médecine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
2
|
Srivastava T, Garola RE, Zhou J, Boinpelly VC, Priya L, Ali MF, Rezaiekhaligh MH, Heruth DP, Novak J, Alon US, Joshi T, Jiang Y, McCarthy ET, Savin VJ, Johnson ML, Sharma R, Sharma M. Prostanoid receptors in hyperfiltration-mediated glomerular injury: Novel agonists and antagonists reveal opposing roles for EP2 and EP4 receptors. FASEB J 2022; 36:e22559. [PMID: 36125047 DOI: 10.1096/fj.202200875r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
Increased fluid-flow shear stress (FFSS) contributes to hyperfiltration-induced podocyte and glomerular injury resulting in progression of chronic kidney disease (CKD). We reported that increased FFSS in vitro and in vivo upregulates PGE2 receptor EP2 (but not EP4 expression), COX2-PGE2 -EP2 axis, and EP2-linked Akt-GSK3β-β-catenin signaling pathway in podocytes. To understand and use the disparities between PGE2 receptors, specific agonists, and antagonists of EP2 and EP4 were used to assess phosphorylation of Akt, GSK3β and β-catenin in podocytes using Western blotting, glomerular filtration barrier function using in vitro albumin permeability (Palb ) assay, and mitigation of hyperfiltration-induced injury in unilaterally nephrectomized (UNX) mice at 1 and 6 months. Results show an increase in Palb by PGE2 , EP2 agonist (EP2AGO ) and EP4 antagonist (EP4ANT ), but not by EP2 antagonist (EP2ANT ) or EP4 agonist (EP4AGO ). Pretreatment with EP2ANT blocked the effect of PGE2 or EP2AGO on Palb . Modulation of EP2 and EP4 also induced opposite effects on phosphorylation of Akt and β-Catenin. Individual agonists or antagonists of EP2 or EP4 did not induce significant improvement in albuminuria in UNX mice. However, treatment with a combination EP2ANT + EP4AGO for 1 or 6 months caused a robust decrease in albuminuria. EP2ANT + EP4AGO combination did not impact adaptive hypertrophy or increased serum creatinine. Observed differences between expression of EP2 and EP4 on the glomerular barrier highlight these receptors as potential targets for intervention. Safe and effective mitigating effect of EP2ANT + EP4AGO presents a novel opportunity to delay the progression of hyperfiltration-associated CKD as seen in transplant donors.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA.,Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Lakshmi Priya
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammed Farhan Ali
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Mohammad H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, Missouri, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.,MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Ellen T McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Virginia J Savin
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, University of Missouri at Kansas City-School of Dentistry, Kansas City, Missouri, USA
| | - Ram Sharma
- Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation (MVBRF), Kansas City, Missouri, USA.,Renal Research Laboratory, Kansas City VA Medical Center, Kansas City, Missouri, USA.,Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
3
|
Xu C, Yang G, Fu Z, Chen Y, Xie S, Wang F, Yang T. Na +-Retaining Action of COX-2 (Cyclooxygenase-2)/EP 1 Pathway in the Collecting Duct via Activation of Intrarenal Renin-Angiotensin-Aldosterone System and Epithelial Sodium Channel. Hypertension 2022; 79:1190-1202. [PMID: 35296155 DOI: 10.1161/hypertensionaha.121.17245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The collecting duct (CD) is a major site of both biosynthesis and action of prostaglandin E2 as highlighted by the predominant expression of COX-2 (cyclooxygenase-2) and some E-prostanoid (EP) subtypes at this nephron site. The purpose of this study was to determine the relevance and mechanism of CD COX-2/prostaglandin E2/EP1 signaling for the regulation of Na+ hemostasis during Na+ depletion. METHODS Mice with Aqp2Cre-driven deletion of COX-2 (COX-2fl/flAqp2Cre+) or the EP1 subtype (EP1fl/flAqp2Cre+) were generated and the Na+-wasting phenotype of these mice during low-salt (LS) intake was examined. EP subtypes responsible for prostaglandin E2-induced local renin response were analyzed in primary cultured mouse inner medullary CD cells. RESULTS Following 28-day LS intake, COX-2fl/flAqp2Cre+ mice exhibited a higher urinary Na+ excretion and lower cumulative Na+ balance, accompanied with suppressed intrarenal renin, AngII (angiotensin II), and aldosterone, expression of CYP11B2 (cytochrome P450 family 11 subfamily B member 2), and blunted expression of epithelial sodium channel subunits compared to floxed controls (COX-2fl/flAqp2Cre-), whereas no differences were observed for indices of systemic renin-angiotensin-aldosterone system. In cultured CD cells, exposure to prostaglandin E2 stimulated release of soluble (pro)renin receptor, prorenin/renin and aldosterone and the stimulation was more sensitive to antagonism of EP1 as compared other EP subtypes. Subsequently, EP1fl/flAqp2Cre+ mice largely recapitulated Na+-wasting phenotype seen in COX-2fl/flAqp2Cre+ mice. CONCLUSIONS The study for the first time reports that CD COX-2/EP1 pathway might play a key role in maintenance of Na+ homeostasis in the face of Na+ depletion, at least in part, through activation of intrarenal renin-angiotensin-aldosterone-system and epithelial sodium channel.
Collapse
Affiliation(s)
- Chuanming Xu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Guangrui Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Ziwei Fu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Yanting Chen
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Shiying Xie
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
4
|
Wang L, Wu Y, Jia Z, Yu J, Huang S. Roles of EP Receptors in the Regulation of Fluid Balance and Blood Pressure. Front Endocrinol (Lausanne) 2022; 13:875425. [PMID: 35813612 PMCID: PMC9262144 DOI: 10.3389/fendo.2022.875425] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
Prostaglandin E2 (PGE2) is an important prostanoid expressing throughout the kidney and cardiovascular system. Despite the diverse effects on fluid metabolism and blood pressure, PGE2 is implicated in sustaining volume and hemodynamics homeostasis. PGE2 works through four distinct E-prostanoid (EP) receptors which are G protein-coupled receptors. To date, pharmacological specific antagonists and agonists of all four subtypes of EP receptors and genetic targeting knockout mice for each subtype have helped in uncoupling the diverse functions of PGE2 and discriminating the respective characteristics of each receptor. In this review, we summarized the functions of individual EP receptor subtypes in the renal and blood vessels and the molecular mechanism of PGE2-induced fluid metabolism and blood pressure homeostasis.
Collapse
Affiliation(s)
- Lu Wang
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Hematology and Oncology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yiqian Wu
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Yu
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Songming Huang, ; Jing Yu,
| | - Songming Huang
- Jiangsu Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Songming Huang, ; Jing Yu,
| |
Collapse
|
5
|
Prieto MC, Gonzalez AA, Visniauskas B, Navar LG. The evolving complexity of the collecting duct renin-angiotensin system in hypertension. Nat Rev Nephrol 2021; 17:481-492. [PMID: 33824491 PMCID: PMC8443079 DOI: 10.1038/s41581-021-00414-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
The intrarenal renin-angiotensin system is critical for the regulation of tubule sodium reabsorption, renal haemodynamics and blood pressure. The excretion of renin in urine can result from its increased filtration, the inhibition of renin reabsorption by megalin in the proximal tubule, or its secretion by the principal cells of the collecting duct. Modest increases in circulating or intrarenal angiotensin II (ANGII) stimulate the synthesis and secretion of angiotensinogen in the proximal tubule, which provides sufficient substrate for collecting duct-derived renin to form angiotensin I (ANGI). In models of ANGII-dependent hypertension, ANGII suppresses plasma renin, suggesting that urinary renin is not likely to be the result of increased filtered load. In the collecting duct, ANGII stimulates the synthesis and secretion of prorenin and renin through the activation of ANGII type 1 receptor (AT1R) expressed primarily by principal cells. The stimulation of collecting duct-derived renin is enhanced by paracrine factors including vasopressin, prostaglandin E2 and bradykinin. Furthermore, binding of prorenin and renin to the prorenin receptor in the collecting duct evokes a number of responses, including the non-proteolytic enzymatic activation of prorenin to produce ANGI from proximal tubule-derived angiotensinogen, which is then converted into ANGII by luminal angiotensin-converting enzyme; stimulation of the epithelial sodium channel (ENaC) in principal cells; and activation of intracellular pathways linked to the upregulation of cyclooxygenase 2 and profibrotic genes. These findings suggest that dysregulation of the renin-angiotensin system in the collecting duct contributes to the development of hypertension by enhancing sodium reabsorption and the progression of kidney injury.
Collapse
Affiliation(s)
- Minolfa C. Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA.,
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaiso, Chile
| | - Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - L. Gabriel Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
6
|
Bamgbola OF, Ahmed Y. Differential diagnosis of perinatal Bartter, Bartter and Gitelman syndromes. Clin Kidney J 2020; 14:36-48. [PMID: 33564404 PMCID: PMC7857843 DOI: 10.1093/ckj/sfaa172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The common finding of hypokalemic alkalosis in several unrelated disorders may confound the early diagnosis of salt-losing tubulopathy (SLT). Antenatal Bartter syndrome (BS) must be considered in idiopathic early-onset polyhydramnios. Fetal megabladder in BS may allow its distinction from third-trimester polyhydramnios that occurs in congenital chloride diarrhea (CCD). Fetal megacolon occurs in CCD while fecal chloride >90 mEq/L in infants is diagnostic. Failure-to-thrive, polydipsia and polyuria in early childhood are the hallmarks of classic BS. Unlike BS, there is low urinary chloride in hypokalemic alkalosis of intractable emesis and cystic fibrosis. Rarely, renal salt wasting may result from cystinosis, Dent disease, disorders of paracellular claudin-10b and Kir4.1 potassium-channel deficiency. Acquired BS may result from calcimimetic up-regulation of a calcium-sensing receptor or autoantibody inactivation of sodium chloride co-transporters in Sjögren syndrome. A relatively common event of heterozygous gene mutations for Gitelman syndrome increases the likelihood of its random occurrence in certain diseases of adult onset. Finally, diuretic abuse is the most common differential diagnosis of SLT. Unlike the persistent elevation in BS, urinary chloride concentration losses waxes and wanes on day-to-day assessment in patients with diuretic misuse.
Collapse
Affiliation(s)
- Oluwatoyin Fatai Bamgbola
- Department of Pediatrics, Division of Pediatric Nephrology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Youssef Ahmed
- Department of Pediatrics, Kings County Hospital, Brooklyn, NY, USA
| |
Collapse
|
7
|
Wilcox CS, Testani JM, Pitt B. Pathophysiology of Diuretic Resistance and Its Implications for the Management of Chronic Heart Failure. Hypertension 2020; 76:1045-1054. [PMID: 32829662 PMCID: PMC10683075 DOI: 10.1161/hypertensionaha.120.15205] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diuretic resistance implies a failure to increase fluid and sodium (Na+) output sufficiently to relieve volume overload, edema, or congestion, despite escalating doses of a loop diuretic to a ceiling level (80 mg of furosemide once or twice daily or greater in those with reduced glomerular filtration rate or heart failure). It is a major cause of recurrent hospitalizations in patients with chronic heart failure and predicts death but is difficult to diagnose unequivocally. Pharmacokinetic mechanisms include the low and variable bioavailability of furosemide and the short duration of all loop diuretics that provides time for the kidneys to restore diuretic-induced Na+ losses between doses. Pathophysiological mechanisms of diuretic resistance include an inappropriately high daily salt intake that exceeds the acute diuretic-induced salt loss, hyponatremia or hypokalemic, hypochloremic metabolic alkalosis, and reflex activation of the renal nerves. Nephron mechanisms include tubular tolerance that can develop even during the time that the renal tubules are exposed to a single dose of diuretic, or enhanced reabsorption in the proximal tubule that limits delivery to the loop, or an adaptive increase in reabsorption in the downstream distal tubule and collecting ducts that offsets ongoing blockade of Na+ reabsorption in the loop of Henle. These provide rationales for novel strategies including the concurrent use of diuretics that block these nephron segments and even sequential nephron blockade with multiple diuretics and aquaretics combined in severely diuretic-resistant patients with heart failure.
Collapse
Affiliation(s)
- Christopher Stuart Wilcox
- From the Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington DC (C.S.W.)
| | | | - Bertram Pitt
- Division of Cardiology, University of Michigan, Ann Arbor, MI (B.P.)
| |
Collapse
|
8
|
Chen Y, Xu C. The interaction partners of (pro)renin receptor in the distal nephron. FASEB J 2020; 34:14136-14149. [PMID: 32975331 DOI: 10.1096/fj.202001711r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/11/2022]
Abstract
The (pro)renin receptor (PRR), a key regulator of intrarenal renin-angiotensin system (RAS), is predominantly presented in podocytes, proximal tubules, distal convoluted tubules, and the apical membrane of collecting duct A-type intercalated cells, and plays a crucial role in hypertension, cardiovascular disease, kidney disease, and fluid homeostasis. In addition to its well-known renin-regulatory function, increasing evidence suggests PRR can also act in a variety of intracellular signaling cascades independently of RAS in the renal medulla, including Wnt/β-catenin signaling, cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2 ) signaling, and the apelinergic system, and work as a component of the vacuolar H+ -ATPase. PRR and these pathways regulate the expression/activity of each other that controlling blood pressure and renal functions. In this review, we highlight recent findings regarding the antagonistic interaction between PRR and ELABELA/apelin, the mutually stimulatory relationship between PRR and COX-2/PGE2 or Wnt/β-catenin signaling in the renal medulla, and their involvement in the regulation of intrarenal RAS thereby control blood pressure, renal injury, and urine concentrating ability in health and patho-physiological conditions. We also highlight the latest progress in the involvement of PRR for the vacuolar H+ -ATPase activity.
Collapse
Affiliation(s)
- Yanting Chen
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Chuanming Xu
- Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA.,Center for Translational Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
9
|
Li Y, Xia W, Zhao F, Wen Z, Zhang A, Huang S, Jia Z, Zhang Y. Prostaglandins in the pathogenesis of kidney diseases. Oncotarget 2018; 9:26586-26602. [PMID: 29899878 PMCID: PMC5995175 DOI: 10.18632/oncotarget.25005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
Prostaglandins (PGs) are important lipid mediators produced from arachidonic acid via the sequential catalyzation of cyclooxygenases (COXs) and specific prostaglandin synthases. There are five subtypes of PGs, namely PGE2, PGI2, PGD2, PGF2α, and thromboxane A2 (TXA2). PGs exert distinct roles by combining to a diverse family of membrane-spanning G protein-coupled prostanoid receptors. The distribution of these PGs, their specific synthases and receptors vary a lot in the kidney. This review summarized the recent findings of PGs together with the COXs and their specific synthases and receptors in regulating renal function and highlighted the insights into their roles in the pathogenesis of various kidney diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Weiwei Xia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Fei Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhaoying Wen
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
10
|
COX-2-derived PGE 2 triggers hyperplastic renin expression and hyperreninemia in aldosterone synthase-deficient mice. Pflugers Arch 2018; 470:1127-1137. [PMID: 29455241 PMCID: PMC6013527 DOI: 10.1007/s00424-018-2118-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/26/2018] [Accepted: 02/01/2018] [Indexed: 11/25/2022]
Abstract
Pharmacological inhibition or genetic loss of function defects of the renin angiotensin aldosterone system (RAAS) causes compensatory renin cell hyperplasia and hyperreninemia. The triggers for the compensatory stimulation of renin synthesis and secretion in this situation may be multimodal. Since cyclooxygenase-2 (COX-2) expression in the macula densa is frequently increased in states of a defective RAAS, we have investigated a potential role of COX-2 and its derived prostaglandins for renin expression and secretion in aldosterone synthase-deficient mice (AS−/−) as a model for a genetic defect of the RAAS. In comparison with wild-type mice (WT), AS−/− mice had 9-fold and 30-fold increases of renin mRNA and of plasma renin concentrations (PRC), respectively. Renin immunoreactivity in the kidney cortex of AS−/− mice was 10-fold higher than in WT. Macula densa COX-2 expression was 5-fold increased in AS−/− kidneys relative to WT kidneys. Treatment of AS−/− mice with the COX-2 inhibitor SC-236 for 1 week lowered both renal renin mRNA and PRC by 70%. Hyperplastic renin cells in AS−/− kidneys were found to express the prostaglandin E2 receptors EP2 and EP4. Global deletion of EP2 receptors did not alter renin mRNA nor PRC values in AS−/− mice. Renin cell-specific inducible deletion of the EP4 receptor lowered renin mRNA and PRC by 25% in AS−/− mice. Renin cell-specific inducible deletion of the EP4 receptor in combination with global deletion of the EP2 receptor lowered renin mRNA and PRC by 70–75% in AS−/− mice. Lineage tracing of renin-expressing cells revealed that deletion of EP2 and EP4 leads to a preferential downregulation of perivascular renin expression. Our findings suggest that increased macula densa COX-2 activity in AS−/− mice triggers perivascular renin expression and secretion via prostaglandin E2.
Collapse
|
11
|
Physiological and pathophysiological implications of PGE2 and the PGE2 synthases in the kidney. Prostaglandins Other Lipid Mediat 2018; 134:1-6. [DOI: 10.1016/j.prostaglandins.2017.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/09/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022]
|
12
|
Hyperfiltration-associated biomechanical forces in glomerular injury and response: Potential role for eicosanoids. Prostaglandins Other Lipid Mediat 2017; 132:59-68. [PMID: 28108282 DOI: 10.1016/j.prostaglandins.2017.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/22/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022]
Abstract
Hyperfiltration is a well-known risk factor in progressive loss of renal function in chronic kidney disease (CKD) secondary to various diseases. A reduced number of functional nephrons due to congenital or acquired cause(s) results in hyperfiltration in the remnant kidney. Hyperfiltration-associated increase in biomechanical forces, namely pressure-induced tensile stress and fluid flow-induced shear stress (FFSS) determine cellular injury and response. We believe the current treatment of CKD yields limited success because it largely attenuates pressure-induced tensile stress changes but not the effect of FFSS on podocytes. Studies on glomerular podocytes, tubular epithelial cells and bone osteocytes provide evidence for a significant role of COX-2 generated PGE2 and its receptors in response to tensile stress and FFSS. Preliminary observations show increased urinary PGE2 in children born with a solitary kidney. FFSS-induced COX2-PGE2-EP2 signaling provides an opportunity to identify targets and, for developing novel agents to complement currently available treatment.
Collapse
|
13
|
Angiotensin II-AT1-receptor signaling is necessary for cyclooxygenase-2-dependent postnatal nephron generation. Kidney Int 2016; 91:818-829. [PMID: 28040266 DOI: 10.1016/j.kint.2016.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 10/25/2016] [Accepted: 11/03/2016] [Indexed: 11/23/2022]
Abstract
Deletion of cyclooxygenase-2 (COX-2) causes impairment of postnatal kidney development. Here we tested whether the renin angiotensin system contributes to COX-2-dependent nephrogenesis in mice after birth and whether a rescue of impaired renal development and function in COX-2-/- mice was achievable. Plasma renin concentration in mouse pups showed a birth peak and a second peak around day P8 during the first 10 days post birth. Administration of the angiotensin II receptor AT1 antagonist telmisartan from day P1 to P3 did not result in cortical damage. However, telmisartan treatment from day P3 to P8, the critical time frame of renal COX-2 expression, led to hypoplastic glomeruli, a thinned subcapsular cortex and maturational arrest of superficial glomeruli quite similar to that observed in COX-2-/- mice. In contrast, AT2 receptor antagonist PD123319 was without any effect on renal development. Inhibition of the renin angiotensin system by aliskiren and enalapril caused similar glomerular defects as telmisartan. Administration of the AT1 receptor agonist L162313 to COX-2-/- pups improved kidney growth, ameliorated renal defects, but had no beneficial effect on reduced cortical mass. L162313 rescued impaired renal function by reducing serum urea and creatinine and mitigated pathologic albumin excretion. Moreover, glomerulosclerosis in the kidneys of COX-2-/- mice was reduced. Thus, angiotensin II-AT1-receptor signaling is necessary for COX-2-dependent normal postnatal nephrogenesis and maturation.
Collapse
|
14
|
Soleimani M, Barone S, Xu J, Alshahrani S, Brooks M, McCormack FX, Smith RD, Zahedi K. Prostaglandin-E2 Mediated Increase in Calcium and Phosphate Excretion in a Mouse Model of Distal Nephron Salt Wasting. PLoS One 2016; 11:e0159804. [PMID: 27442254 PMCID: PMC4956050 DOI: 10.1371/journal.pone.0159804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/10/2016] [Indexed: 01/29/2023] Open
Abstract
Contribution of salt wasting and volume depletion to the pathogenesis of hypercalciuria and hyperphosphaturia is poorly understood. Pendrin/NCC double KO (pendrin/NCC-dKO) mice display severe salt wasting under basal conditions and develop profound volume depletion, prerenal renal failure, and metabolic alkalosis and are growth retarded. Microscopic examination of the kidneys of pendrin/NCC-dKO mice revealed the presence of calcium phosphate deposits in the medullary collecting ducts, along with increased urinary calcium and phosphate excretion. Confirmatory studies revealed decreases in the expression levels of sodium phosphate transporter-2 isoforms a and c, increases in the expression of cytochrome p450 family 4a isotypes 12 a and b, as well as prostaglandin E synthase 1, and cyclooxygenases 1 and 2. Pendrin/NCC-dKO animals also had a significant increase in urinary prostaglandin E2 (PGE-2) and renal content of 20-hydroxyeicosatetraenoic acid (20-HETE) levels. Pendrin/NCC-dKO animals exhibit reduced expression levels of the sodium/potassium/2chloride co-transporter 2 (NKCC2) in their medullary thick ascending limb. Further assessment of the renal expression of NKCC2 isoforms by quantitative real time PCR (qRT-PCR) reveled that compared to WT mice, the expression of NKCC2 isotype F was significantly reduced in pendrin/NCC-dKO mice. Provision of a high salt diet to rectify volume depletion or inhibition of PGE-2 synthesis by indomethacin, but not inhibition of 20-HETE generation by HET0016, significantly improved hypercalciuria and salt wasting in pendrin/NCC dKO mice. Both high salt diet and indomethacin treatment also corrected the alterations in NKCC2 isotype expression in pendrin/NCC-dKO mice. We propose that severe salt wasting and volume depletion, irrespective of the primary originating nephron segment, can secondarily impair the reabsorption of salt and calcium in the thick ascending limb of Henle and/or proximal tubule, and reabsorption of sodium and phosphate in the proximal tubule via processes that are mediated by PGE-2.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
- * E-mail:
| | - Sharon Barone
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
| | - Jie Xu
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Saeed Alshahrani
- Department of Pharmacology and Cell Biophysics and, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Marybeth Brooks
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Francis X. McCormack
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Roger D. Smith
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Kamyar Zahedi
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
15
|
Jensen BL. Prorenin Receptor, a Necessary Component in Urine Concentration Mechanism. J Am Soc Nephrol 2016; 27:2919-2921. [PMID: 27098238 DOI: 10.1681/asn.2016030344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
16
|
Clark JL, Rech L, Chaity N, Sihag J, Taylor CG, Aliani M. Possible deleterious hormonal changes associated with low-sodium diets. Nutr Rev 2015; 73:22-35. [PMID: 26024055 DOI: 10.1093/nutrit/nuu003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The average salt intake of people in Canada, the United States, and Europe is about 3,400 mg of sodium per day, which exceeds the recommended intake levels set by various health organizations. The World Health Organization recommends a worldwide reduction of sodium intake to less than 2,000 mg per day. Most research to date has focused on the negative effects of high-sodium intake; however, little information is available on the metabolic effects of low-sodium intakes. This review focuses on the hormonal changes associated with low-sodium diets, especially the hormones involved in metabolism and cardiovascular and renal function. Based largely on rodent studies, low-sodium diets have been associated with changes in glycemic control, energy metabolism, cardiovascular disease risk, cholesterol concentrations, inflammation, and functioning of the renin-angiotensin-aldosterone system. Overall, research has revealed mixed results regarding the impact of dietary sodium intake on various hormones. Further research is required to assess the effects of sodium reduction on hormones and their associated pathways in order to determine the likelihood of any unintended effects.
Collapse
Affiliation(s)
- Jaime L Clark
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Leslie Rech
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nazia Chaity
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jyoti Sihag
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Carla G Taylor
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michel Aliani
- J.L. Clark, L. Rech, N. Chaity, J. Sihag, C.G. Taylor, and M. Aliani are with the Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. J.L. Clark, L. Rech, C.G. Taylor, and M. Aliani are with the Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Research Centre, Winnipeg, Manitoba, Canada. C.G. Taylor is with the Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
17
|
Nasrallah R, Hassouneh R, Hébert RL. PGE2, Kidney Disease, and Cardiovascular Risk: Beyond Hypertension and Diabetes. J Am Soc Nephrol 2015; 27:666-76. [PMID: 26319242 DOI: 10.1681/asn.2015050528] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
An important measure of cardiovascular health is obtained by evaluating the global cardiovascular risk, which comprises a number of factors, including hypertension and type 2 diabetes, the leading causes of illness and death in the world, as well as the metabolic syndrome. Altered immunity, inflammation, and oxidative stress underlie many of the changes associated with cardiovascular disease, diabetes, and the metabolic syndrome, and recent efforts have begun to elucidate the contribution of PGE2 in these events. This review summarizes the role of PGE2 in kidney disease outcomes that accelerate cardiovascular disease, highlights the role of cyclooxygenase-2/microsomal PGE synthase 1/PGE2 signaling in hypertension and diabetes, and outlines the contribution of PGE2 to other aspects of the metabolic syndrome, particularly abdominal adiposity, dyslipidemia, and atherogenesis. A clearer understanding of the role of PGE2 could lead to new avenues to improve therapeutic options and disease management strategies.
Collapse
Affiliation(s)
- Rania Nasrallah
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramzi Hassouneh
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Yang Y, Gomez JA, Herrera M, Perez-Marco R, Repenning P, Zhang Z, Payne A, Pratt RE, Koller B, Beierwaltes WH, Coffman T, Mirotsou M, Dzau VJ. Salt restriction leads to activation of adult renal mesenchymal stromal cell-like cells via prostaglandin E2 and E-prostanoid receptor 4. Hypertension 2015; 65:1047-54. [PMID: 25776075 DOI: 10.1161/hypertensionaha.114.04611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/09/2015] [Indexed: 12/16/2022]
Abstract
Despite the importance of juxtaglomerular cell recruitment in the pathophysiology of cardiovascular diseases, the mechanisms that underlie renin production under conditions of chronic stimulation remain elusive. We have previously shown that CD44+ mesenchymal-like cells (CD44+ cells) exist in the adult kidney. Under chronic sodium deprivation, these cells are recruited to the juxtaglomerular area and differentiate to new renin-expressing cells. Given the proximity of macula densa to the juxtaglomerular area and the importance of macula densa released prostanoids in renin synthesis and release, we hypothesized that chronic sodium deprivation induces macula densa release of prostanoids, stimulating renal CD44+ cell activation and differentiation. CD44+ cells were isolated from adult kidneys and cocultured with the macula densa cell line, MMDD1, in normal or low-sodium medium. Low sodium stimulated prostaglandin E2 production by MMDD1 and induced migration of CD44+ cells. These effects were inhibited by addition of a cyclooxygenase 2 inhibitor (NS398) or an E-prostanoid receptor 4 antagonist (AH23848) to MMDD1 or CD44+ cells, respectively. Addition of prostaglandin E2 to CD44+ cells increased cell migration and induced renin expression. In vivo activation of renal CD44+ cells during juxtaglomerular recruitment was attenuated in wild-type mice subjected to salt restriction in the presence of cyclooxygenase 2 inhibitor rofecoxib. Similar results were observed in E-prostanoid receptor 4 knockout mice subjected to salt restriction. These results show that the prostaglandin E2/E-prostanoid receptor 4 pathway plays a key role in the activation of renal CD44+ mesenchymal stromal cell-like cells during conditions of juxtaglomerular recruitment; highlighting the importance of this pathway as a key regulatory mechanism of juxtaglomerular recruitment.
Collapse
Affiliation(s)
- Yanqiang Yang
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Jose A Gomez
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Marcela Herrera
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Romelia Perez-Marco
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Peter Repenning
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Zhiping Zhang
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Alan Payne
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Richard E Pratt
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Beverly Koller
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - William H Beierwaltes
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Thomas Coffman
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Maria Mirotsou
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.)
| | - Victor J Dzau
- From the Mandel Center for Hypertension and Atherosclerosis Research, and the Cardiovascular Research Center (Y.Y., J.A.G., R.P.-M., Z.Z., A.P., R.E.P., M.M., V.J.D.) and Division of Nephrology, Department of Medicine (M.H., T.C.), Duke University Medical Center, Durham, NC; Department of Genetics, University of North Carolina at Chapel Hill (P.R., B.K.); and Henry Ford Hospital, Detroit, MI (W.H.B.).
| |
Collapse
|
19
|
Yang T. Crosstalk between (Pro)renin receptor and COX-2 in the renal medulla during angiotensin II-induced hypertension. Curr Opin Pharmacol 2015; 21:89-94. [PMID: 25681793 DOI: 10.1016/j.coph.2014.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/16/2014] [Accepted: 12/21/2014] [Indexed: 01/13/2023]
Abstract
Angiotensin II (AngII) is an octapeptide hormone that plays a central role in regulation of sodium balance, plasma volume, and blood pressure. Its role in the pathogenesis of hypertension is highlighted by the wide use of inhibitors of the renin-angiotensin system (RAS) as the first-line antihypertensive therapy. However, despite intensive investigation, the mechanism of AngII-induced hypertension is still incompletely understood. Although diverse pathways are likely involved, increasing evidence suggests that the activation of intrarenal RAS may represent a dominant mechanism of AngII-induced hypertension. (Pro)renin receptor (PRR), a potential regulator of intrarenal RAS, is expressed in the intercalated cells of the collecting duct (CD) and induced by AngII, in parallel with increased renin in the principal cells of the CD. Activation of PRR elevated PGE2 release and COX-2 expression in renal inner medullary cells whereas COX-2-derived PGE2via the EP4 receptor mediates the upregulation of PRR during AngII infusion, thus forming a vicious cycle. The mutually stimulatory relationship between PRR and COX-2 in the distal nephron may play an important role in mediating AngII-induced hypertension.
Collapse
Affiliation(s)
- Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, United States.
| |
Collapse
|
20
|
Nasrallah R, Hassouneh R, Hébert RL. Chronic kidney disease: targeting prostaglandin E2 receptors. Am J Physiol Renal Physiol 2014; 307:F243-50. [PMID: 24966087 DOI: 10.1152/ajprenal.00224.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease is a leading cause of morbidity and mortality in the world. A better understanding of disease mechanisms has been gained in recent years, but the current management strategies are ineffective at preventing disease progression. A widespread focus of research is placed on elucidating the specific processes implicated to find more effective therapeutic options. PGE2, acting on its four EP receptors, regulates many renal disease processes; thus EP receptors could prove to be important targets for kidney disease intervention strategies. This review summarizes the major pathogenic mechanisms contributing to initiation and progression of chronic kidney disease, emphasizing the role of hyperglycemia, hypertension, inflammation, and oxidative stress. We have long recognized the multifaceted role of PGs in both the initiation and progression of chronic kidney disease, yet studies are only now seriously contemplating specific EP receptors as targets for therapy. Given the plethora of renal complications attributed to PG involvement in the kidney, this review highlights these pathogenic events and emphasizes the PGE2 receptor targets as options available to complement current therapeutic strategies.
Collapse
Affiliation(s)
- Rania Nasrallah
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramzi Hassouneh
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
21
|
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: 5.4] [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
|
22
|
Abstract
In the mammalian kidney, prostaglandins (PGs) are important mediators of physiologic processes, including modulation of vascular tone and salt and water. PGs arise from enzymatic metabolism of free arachidonic acid (AA), which is cleaved from membrane phospholipids by phospholipase A2 activity. The cyclooxygenase (COX) enzyme system is a major pathway for metabolism of AA in the kidney. COX are the enzymes responsible for the initial conversion of AA to PGG2 and subsequently to PGH2, which serves as the precursor for subsequent metabolism by PG and thromboxane synthases. In addition to high levels of expression of the "constitutive" rate-limiting enzyme responsible for prostanoid production, COX-1, the "inducible" isoform of cyclooxygenase, COX-2, is also constitutively expressed in the kidney and is highly regulated in response to alterations in intravascular volume. PGs and thromboxane A2 exert their biological functions predominantly through activation of specific 7-transmembrane G-protein-coupled receptors. COX metabolites have been shown to exert important physiologic functions in maintenance of renal blood flow, mediation of renin release and regulation of sodium excretion. In addition to physiologic regulation of prostanoid production in the kidney, increases in prostanoid production are also seen in a variety of inflammatory renal injuries, and COX metabolites may serve as mediators of inflammatory injury in renal disease.
Collapse
Affiliation(s)
- Raymond C Harris
- George M. O'Brien Kidney and Urologic Diseases Center and Division of Nephrology, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee, USA.
| | | |
Collapse
|
23
|
Rajagopal M, Thomas SV, Kathpalia PP, Chen Y, Pao AC. Prostaglandin E2 induces chloride secretion through crosstalk between cAMP and calcium signaling in mouse inner medullary collecting duct cells. Am J Physiol Cell Physiol 2013; 306:C263-78. [PMID: 24284792 DOI: 10.1152/ajpcell.00381.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Under conditions of high dietary salt intake, prostaglandin E2 (PGE2) production is increased in the collecting duct and promotes urinary sodium chloride (NaCl) excretion; however, the molecular mechanisms by which PGE2 increases NaCl excretion in this context have not been clearly defined. We used the mouse inner medullary collecting duct (mIMCD)-K2 cell line to characterize mechanisms underlying PGE2-regulated NaCl transport. When epithelial Na(+) channels were inhibited, PGE2 exclusively stimulated basolateral EP4 receptors to increase short-circuit current (Isc(PGE2)). We found that Isc(PGE2) was sensitive to inhibition by H-89 and CFTR-172, indicating that EP4 receptors signal through protein kinase A to induce Cl(-) secretion via cystic fibrosis transmembrane conductance regulator (CFTR). Unexpectedly, we also found that Isc(PGE2) was sensitive to inhibition by BAPTA-AM (Ca(2+) chelator), 2-aminoethoxydiphenyl borate (2-APB) (inositol triphosphate receptor blocker), and flufenamic acid (FFA) [Ca(2+)-activated Cl(-) channel (CACC) inhibitor], suggesting that EP4 receptors also signal through Ca(2+) to induce Cl(-) secretion via CACC. Additionally, we observed that PGE2 stimulated an increase in Isc through crosstalk between cAMP and Ca(2+) signaling; BAPTA-AM or 2-APB inhibited a component of Isc(PGE2) that was sensitive to CFTR-172 inhibition; H-89 inhibited a component of Isc(PGE2) that was sensitive to FFA inhibition. Together, our findings indicate that PGE2 activates basolateral EP4 receptors and signals through both cAMP and Ca(2+) to stimulate Cl(-) secretion in IMCD-K2 cells. We propose that these signaling pathways, and the crosstalk between them, may provide a concerted mechanism for enhancing urinary NaCl excretion under conditions of high dietary NaCl intake.
Collapse
Affiliation(s)
- Madhumitha Rajagopal
- Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California; and
| | | | | | | | | |
Collapse
|
24
|
Konya V, Marsche G, Schuligoi R, Heinemann A. E-type prostanoid receptor 4 (EP4) in disease and therapy. Pharmacol Ther 2013; 138:485-502. [PMID: 23523686 PMCID: PMC3661976 DOI: 10.1016/j.pharmthera.2013.03.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 01/06/2023]
Abstract
The large variety of biological functions governed by prostaglandin (PG) E2 is mediated by signaling through four distinct E-type prostanoid (EP) receptors. The availability of mouse strains with genetic ablation of each EP receptor subtype and the development of selective EP agonists and antagonists have tremendously advanced our understanding of PGE2 as a physiologically and clinically relevant mediator. Moreover, studies using disease models revealed numerous conditions in which distinct EP receptors might be exploited therapeutically. In this context, the EP4 receptor is currently emerging as most versatile and promising among PGE2 receptors. Anti-inflammatory, anti-thrombotic and vasoprotective effects have been proposed for the EP4 receptor, along with its recently described unfavorable tumor-promoting and pro-angiogenic roles. A possible explanation for the diverse biological functions of EP4 might be the multiple signaling pathways switched on upon EP4 activation. The present review attempts to summarize the EP4 receptor-triggered signaling modules and the possible therapeutic applications of EP4-selective agonists and antagonists.
Collapse
Key Words
- ampk, amp-activated protein kinase
- camp, cyclic adenylyl monophosphate
- cftr, cystic fibrosis transmembrane conductance regulator
- clc, chloride channel
- cox, cyclooxygenase
- creb, camp-response element-binding protein
- dp, d-type prostanoid receptor
- dss, dextran sodium sulfate
- egfr, epidermal growth factor receptor
- enos, endothelial nitric oxide synthase
- ep, e-type prostanoid receptor
- epac, exchange protein activated by camp
- eprap, ep4 receptor-associated protein
- erk, extracellular signal-regulated kinase
- fem1a, feminization 1 homolog a
- fp, f-type prostanoid receptor
- grk, g protein-coupled receptor kinase
- 5-hete, 5-hydroxyeicosatetraenoic acid
- icer, inducible camp early repressor
- icam-1, intercellular adhesion molecule-1
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ip, i-type prostanoid receptor
- lps, lipopolysaccharide
- map, mitogen-activated protein kinase
- mcp, monocyte chemoattractant protein
- mek, map kinase kinase
- nf-κb, nuclear factor kappa-light-chain-enhancer of activated b cells
- nsaid, non-steroidal anti-inflammatory drug
- pg, prostaglandin
- pi3k, phosphatidyl insositol 3-kinase
- pk, protein kinase
- tp, t-type prostanoid receptor
- tx, thromboxane receptor
- prostaglandins
- inflammation
- vascular disease
- cancerogenesis
- renal function
- osteoporosis
Collapse
Affiliation(s)
| | | | | | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Austria
| |
Collapse
|
25
|
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: 2.1] [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
|
26
|
Vennemann A, Gerstner A, Kern N, Ferreiros Bouzas N, Narumiya S, Maruyama T, Nüsing RM. PTGS-2-PTGER2/4 signaling pathway partially protects from diabetogenic toxicity of streptozotocin in mice. Diabetes 2012; 61:1879-87. [PMID: 22522619 PMCID: PMC3379658 DOI: 10.2337/db11-1396] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostanoids are suggested to participate in diabetes pathology, but their roles are controversially discussed. The purpose of the current study was to examine the role of cyclooxygenase (prostaglandin synthase [PTGS]) enzymes and prostaglandin (PG) E(2) signaling pathways in streptozotocin (STZ)-induced type 1 diabetes. Blood glucose, insulin, and survival rate were studied in mice with targeted disruption of the genes for PTGS and PGE receptors (PTGERs). PGE(2) was found as the main prostanoid formed by the pancreas. Contrarily to PTGS-1, deficiency of PTGS-2 activity significantly amplified STZ effect, causing dramatic loss of insulin production and rise in blood glucose and death rate. STZ metabolism was unaffected by PTGS deficiency. Diabetogenicity of STZ in PTGER1(-/-), PTGER2(-/-), PTGER3(-/-), and PTGER4(-/-) mice was comparable to control mice. In striking contrast, combined knockout of PTGER2 and PTGER4 by blocking PTGER4 in PTGER2(-/-) mice strongly enhanced STZ pathology. Treatment of PTGS-2(-/-) and wild-type mice with PTGER2/PTGER4 agonists partially protected against STZ-induced diabetes and restored β-cell function. Our data uncover a previously unrecognized protective role of PTGS-2-derived PGE(2) in STZ-induced diabetes mediated by the receptor types PTGER2 and PTGER4. These findings offer the possibility to intervene in early progression of type 1 diabetes by using PTGER-selective agonists.
Collapse
MESH Headings
- Animals
- Blood Glucose/analysis
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Dinoprostone/biosynthesis
- Gene Deletion
- Insulin/blood
- Male
- Mice
- Mice, Inbred C57BL
- Pancreas/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/agonists
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/agonists
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Antje Vennemann
- Department of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Niklas Kern
- Department of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Shuh Narumiya
- Department of Pharmacology, Kyoto University, Kyoto, Japan
| | | | - Rolf M. Nüsing
- Department of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Corresponding author: Rolf M. Nüsing,
| |
Collapse
|
27
|
Regulation of renin secretion by renal juxtaglomerular cells. Pflugers Arch 2012; 465:25-37. [DOI: 10.1007/s00424-012-1126-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 06/02/2012] [Accepted: 06/06/2012] [Indexed: 01/06/2023]
|
28
|
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: 1.7] [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
|
29
|
Popp L, Häussler A, Olliges A, Nüsing R, Narumiya S, Geisslinger G, Tegeder I. Comparison of nociceptive behavior in prostaglandin E, F, D, prostacyclin and thromboxane receptor knockout mice. Eur J Pain 2012; 13:691-703. [DOI: 10.1016/j.ejpain.2008.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 08/08/2008] [Accepted: 09/07/2008] [Indexed: 11/30/2022]
|
30
|
Kawada N, Moriyama T, Kitamura H, Yamamoto R, Furumatsu Y, Matsui I, Takabatake Y, Nagasawa Y, Imai E, Wilcox CS, Rakugi H, Isaka Y. Towards developing new strategies to reduce the adverse side-effects of nonsteroidal anti-inflammatory drugs. Clin Exp Nephrol 2011; 16:25-9. [DOI: 10.1007/s10157-011-0492-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 01/11/2011] [Indexed: 11/24/2022]
|
31
|
Yoshioka W, Peterson RE, Tohyama C. Molecular targets that link dioxin exposure to toxicity phenotypes. J Steroid Biochem Mol Biol 2011; 127:96-101. [PMID: 21168493 PMCID: PMC3433800 DOI: 10.1016/j.jsbmb.2010.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 12/06/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
Many toxicology studies have elucidated health effects associated with exposure to various chemicals, but few have identified the molecular targets that cause specific endpoints of toxicity. Our understanding of the toxicity of dioxins, a group of chemicals capable of causing toxicity at environmentally relevant levels of exposure, is no exception. Dioxins are unique compared to most chemicals that we are exposed to in the environment because they activate a high affinity receptor, aryl hydrocarbon receptor (AhR), that was identified more than three decades ago. In recent years, several lines of experimental evidence have provided clues for opening the "black box" that contains the molecular mechanisms of dioxin action. These clues have emerged by toxicologists beginning to identify the molecular targets that link AhR signaling to tissue-specific toxicity phenotypes. Endpoints of dioxin toxicity for which downstream molecular targets have begun to be elucidated are observed in developmental or tissue regeneration processes, and include impaired prostate development and hydronephrosis in mouse fetuses and pups, reduced midbrain blood flow and jaw malformation in zebrafish embryos, and impaired fin regeneration in larval and adult zebrafish. Significant progress in identifying molecular targets for dioxin-induced hepatotoxicity in adult mice also has occurred. Misregulation of AhR downstream pathways, such as conversion of arachidonic acid to prostanoids via cyclooxygenase-2, and altered Wnt/β-catenin signaling downregulating Sox9, and signaling by receptors for inflammatory cytokines have been implicated in tissue-specific endpoints of dioxin toxicity. These findings may not only begin to clarify the molecular targets of dioxin action but shed light on new molecular events associated with development and disease.
Collapse
Affiliation(s)
- Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Bunkyo-ku, Tokyo 113-0033, Japan
| | - Richard E. Peterson
- Molecular and Environmental Toxicology Center and Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Bunkyo-ku, Tokyo 113-0033, Japan
- Corresponding author. Tel.: +81 3 5841 1431; fax: +81 3 5841 1434. (C. Tohyama)
| |
Collapse
|
32
|
Seyberth HW, Schlingmann KP. Bartter- and Gitelman-like syndromes: salt-losing tubulopathies with loop or DCT defects. Pediatr Nephrol 2011; 26:1789-802. [PMID: 21503667 PMCID: PMC3163795 DOI: 10.1007/s00467-011-1871-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 03/09/2011] [Accepted: 03/09/2011] [Indexed: 12/11/2022]
Abstract
Salt-losing tubulopathies with secondary hyperaldosteronism (SLT) comprise a set of well-defined inherited tubular disorders. Two segments along the distal nephron are primarily involved in the pathogenesis of SLTs: the thick ascending limb of Henle's loop, and the distal convoluted tubule (DCT). The functions of these pre- and postmacula densa segments are quite distinct, and this has a major impact on the clinical presentation of loop and DCT disorders - the Bartter- and Gitelman-like syndromes. Defects in the water-impermeable thick ascending limb, with its greater salt reabsorption capacity, lead to major salt and water losses similar to the effect of loop diuretics. In contrast, defects in the DCT, with its minor capacity of salt reabsorption and its crucial role in fine-tuning of urinary calcium and magnesium excretion, provoke more chronic solute imbalances similar to the effects of chronic treatment with thiazides. The most severe disorder is a combination of a loop and DCT disorder similar to the enhanced diuretic effect of a co-medication of loop diuretics with thiazides. Besides salt and water supplementation, prostaglandin E2-synthase inhibition is the most effective therapeutic option in polyuric loop disorders (e.g., pure furosemide and mixed furosemide-amiloride type), especially in preterm infants with severe volume depletion. In DCT disorders (e.g., pure thiazide and mixed thiazide-furosemide type), renin-angiotensin-aldosterone system (RAAS) blockers might be indicated after salt, potassium, and magnesium supplementation are deemed insufficient. It appears that in most patients with SLT, a combination of solute supplementation with some drug treatment (e.g., indomethacin) is needed for a lifetime.
Collapse
Affiliation(s)
- Hannsjörg W. Seyberth
- Department of Pediatrics and Adolescent Medicine, Philipps University, Marburg, Germany ,Lazarettgarten 23, 76829 Landau, Germany
| | - Karl P. Schlingmann
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| |
Collapse
|
33
|
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: 2.8] [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
|
34
|
Affiliation(s)
- Takako Hirata
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Shuh Narumiya
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
35
|
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: 189] [Impact Index Per Article: 13.5] [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
|
36
|
Seyberth HW. An improved terminology and classification of Bartter-like syndromes. ACTA ACUST UNITED AC 2008; 4:560-7. [PMID: 18695706 DOI: 10.1038/ncpneph0912] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2008] [Accepted: 07/04/2008] [Indexed: 12/15/2022]
Abstract
This Review outlines a terminology and classification of Bartter-like syndromes that is based on the underlying causes of these inherited salt-losing tubulopathies and is, therefore, more clinically relevant than the classical definition. Three major types of salt-losing tubulopathy can be defined: distal convoluted tubule dysfunction leading to hypokalemia (currently known as Gitelman or Bartter syndrome), the more-severe condition of polyuric loop dysfunction (often referred to as antenatal Bartter or hyperprostaglandin E syndrome), and the most-severe condition of combined loop and distal convoluted tubule dysfunction (antenatal Bartter or hyperprostaglandin E syndrome with sensorineural deafness). These three subtypes can each be further subdivided according to the identity of the defective ion transporter or channel: the sodium-chloride cotransporter NCCT or the chloride channel ClC-Kb in distal convoluted tubule dysfunction; the sodium-potassium-chloride cotransporter NKCC2 or the renal outer medullary potassium channel in loop dysfunction; and the chloride channels ClC-Ka and ClC-Kb or their beta-subunit Barttin in combined distal convoluted tubule and loop dysfunction. This new classification should help clinicians to better understand the pathophysiology of these syndromes and choose the most appropriate treatment for affected patients, while avoiding potentially harmful diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hannsjörg W Seyberth
- Center for Pediatric and Adolescent Medicine, Philipps University, Marburg, Germany.
| |
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
|
Clark P, Rowland SE, Denis D, Mathieu MC, Stocco R, Poirier H, Burch J, Han Y, Audoly L, Therien AG, Xu D. MF498 [N-{[4-(5,9-Diethoxy-6-oxo-6,8-dihydro-7H-pyrrolo[3,4-g]quinolin-7-yl)-3-methylbenzyl]sulfonyl}-2-(2-methoxyphenyl)acetamide], a selective E prostanoid receptor 4 antagonist, relieves joint inflammation and pain in rodent models of rheumatoid and osteoarthritis. J Pharmacol Exp Ther 2008; 325:425-34. [PMID: 18287210 DOI: 10.1124/jpet.107.134510] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Previous evidence has implicated E prostanoid receptor 4 (EP4) in mechanical hyperalgesia induced by subplantar inflammation. However, its role in chronic arthritis remains to be further defined because previous attempts have generated two conflicting lines of evidence, with one showing a marked reduction of arthritis induced by a collagen antibody in mice lacking EP4, but not EP1-EP3, and the other showing no impact of EP4 antagonism on arthritis induced by collagen. Here, we assessed the effect of a novel and selective EP4 antagonist MF498 [N-{[4-(5,9-diethoxy-6-oxo-6,8-dihydro-7H-pyrrolo[3,4-g]quinolin-7-yl)-3-methylbenzyl]sulfonyl}-2-(2-methoxyphenyl)acetamide] on inflammation in adjuvant-induced arthritis (AIA), a rat model for rheumatoid arthritis (RA), and joint pain in a guinea pig model of iodoacetate-induced osteoarthritis (OA). In the AIA model, MF498, but not the antagonist for EP1, MF266-1 [1-(5-{3-[2-(benzyloxy)-5-chlorophenyl]-2-thienyl}pyridin-3-yl)-2,2,2-trifluoroethane-1,1-diol] or EP3 MF266-3 [(2E)-N-[(5-bromo-2-methoxyphenyl)sulfonyl]-3-[5-chloro-2-(2-naphthylmethyl)phenyl]acrylamide], inhibited inflammation, with a similar efficacy as a selective cyclooxygenase 2 (COX-2) inhibitor MF-tricyclic. In addition, MF498 was as effective as an nonsteroidal anti-inflammatory drug, diclofenac, or a selective microsomal prostaglandin E synthase-1 inhibitor, MF63 [2-(6-chloro-1H-phenanthro[9,10-d]imidazol-2-yl)isophthalonitrile], in relieving OA-like pain in guinea pigs. When tested in rat models of gastrointestinal toxicity, the EP4 antagonist was well tolerated, causing no mucosal leakage or erosions. Lastly, we evaluated the renal effect of MF498 in a furosemide-induced diuresis model and demonstrated that the compound displayed a similar renal effect as MF-tricyclic [3-(3,4-difluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone], reducing furosemide-induced natriuresis by approximately 50%. These results not only suggest that EP4 is the major EP receptor in both RA and OA but also provide a proof of principle to the concept that antagonism of EP4 may be useful for treatment of arthritis.
Collapse
Affiliation(s)
- Patsy Clark
- Department of Pharmacology, Merck Frosst Centre for Therapeutic Research, 16711 Trans Canada Hwy, Kirkland, QC H9H 3L1, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Abstract
Prostaglandin E (PGE)(2) is a major arachidonic acid metabolite in a wide variety of tissues and is implicated in the control of inflammatory as well as physiological responses. At least three major forms of PGE synthase (PGES) have recently been cloned and characterized: membrane-associated PGES (mPGES)-1, mPGES-2, and cytosolic PGES (cPGES). Among them, mPGES-1 is highly inducible by cytokine and is critically involved in pain and inflammatory responses. Emerging evidence suggests that mPGES-1 may also participate in blood pressure (BP) regulation through an impact on renal and vascular functions. Within the kidney, mPGES-1 predominates in the distal nephron where its expression is highly inducible by salt loading. Mice lacking mPGES-1 exhibit blunted natriuretic response paralleled with remarkably suppressed nitric oxide production, leading to salt-sensitive hypertension. These mice also exhibit an exaggerated hypertensive response to angiotensin II infusion. Together, these results suggest that mPGES-1 may be an important physiological regulator of BP.
Collapse
Affiliation(s)
- T Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah 84132, USA.
| |
Collapse
|
41
|
Ariceta G, Rodríguez-Soriano J. Inherited Renal Tubulopathies Associated With Metabolic Alkalosis: Effects on Blood Pressure. Semin Nephrol 2006; 26:422-33. [PMID: 17275579 DOI: 10.1016/j.semnephrol.2006.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inherited tubular disorders associated with metabolic alkalosis are caused by several gene mutations encoding different tubular transporters responsible for NaCl renal handling. Body volume and renin-angiotensin-aldosterone system status are determined by NaCl reabsorption in the distal nephron. Two common hallmarks in affected individuals: hypokalemia and normal / high blood pressure, support the differential diagnosis. Bartter's syndrome, characterized by hypokalemia and normal blood pressure, is a heterogenic disease caused by the loss of function of SLC12A1 (type 1), KCNJ1 (type 2), CLCNKB (type 3), or BSND genes (type 4). As a result, patients present with renal salt wasting and hypercalciuria. Gitelman's syndrome is caused by the loss of funcion of the SLC12A3 gene and may resemble Bartter's syndrome, though is associated with the very low urinary calcium. Liddle's syndrome, also with similar phenotype but with hypertension, is produced by the gain of function of the SNCC1B or SNCC1G genes, and must be distinguished from other entities of inherited hypertension such as Apparently Mineralocorticoid Excess, of glucocorticoid remediable hypertension.
Collapse
Affiliation(s)
- Gema Ariceta
- Division of Pediatric Nephrology, Department of Pediatrics, Hospital de Cruces and Basque University School of Medicine, Bilbao, Spain.
| | | |
Collapse
|