1
|
Yang T, Gao ZX, Mao ZH, Wu P. Soluble (pro)renin receptor as a novel regulator of renal medullary Na + reabsorption. Am J Physiol Renal Physiol 2025; 328:F239-F247. [PMID: 39508841 DOI: 10.1152/ajprenal.00156.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 01/22/2025] Open
Abstract
Epithelial sodium channel (ENaC) represents a major route of Na+ reabsorption in the aldosterone-sensitive distal nephron where the bulk of ENaC activity is considered to occur in the cortical collecting duct (CCD). Relatively, ENaC activity in the medulla, especially the inner medulla, is often neglected. (Pro)renin receptor (PRR), also termed ATP6ap2, a newly characterized member of the renin-angiotensin system, has emerged as an important regulator of ENaC in the distal nephron. The ENaC regulatory action of PRR is largely mediated by the 28 kDa soluble PRR (sPRR). Although all three subunits of ENaC are under the control of aldosterone, sPRR only mediates the upregulation of α-ENaC but not the other two subunits. Furthermore, sPRR-dependent regulation of α-ENaC only occurs in the renal inner medulla but not in the cortex. sPRR also rapidly upregulates ENaC activity via Nox4-derived H2O2. Overall, sPRR has emerged as an important regulator of renal medullary Na+ reabsorption in the context of overactivation of the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Tianxin Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Zhong-Xiuzi Gao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zi-Hui Mao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Peng Wu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
2
|
Yan Z, Yang T, Li X, Jiang Z, Jia W, Zhou J, Fang H. Apelin-13: a novel approach to suppressing renin production in RVHT. Am J Physiol Cell Physiol 2024; 326:C1683-C1696. [PMID: 38646785 DOI: 10.1152/ajpcell.00092.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Renovascular hypertension (RVHT) is characterized by renal artery stenosis and overactivated renin-angiotensin system (RAS). Apelin, known for its negative modulation of RAS, has protective effects against cardiovascular diseases. The role and mechanisms of the primary active form of apelin, apelin-13, in RVHT are unclear. In this study, male Sprague-Dawley rats were divided into control, two-kidney one-clip (2K1C) model, and 2K1C with apelin-13 treatment groups. Renin expression was analyzed using immunohistochemistry and molecular techniques. Full-length (pro)renin receptor (fPRR) and soluble PRR (sPRR) levels were assessed via Western blotting, and cAMP levels were measured using ELISA. Plasma renin content, plasma renin activity (PRA), angiotensin II (ANG II), and sPRR levels were determined by ELISA. Human Calu-6 and mouse As4.1 cells were used to investigate renin production mechanisms. The 2K1C model exhibited increased systolic blood pressure, plasma renin content, PRA, sPRR, and ANG II levels, while apelin-13 treatment reduced these elevations. Apelin-13 inhibited cAMP production, renin mRNA expression, protein synthesis, and PRR/sPRR protein expression in renal tissue. In Calu-6 cells, cAMP-induced fPRR and site-1 protease (S1P)-derived sPRR expression, which was blocked by cAMP-responsive element-binding protein (CREB) inhibition. Apelin-13 suppressed cAMP elevation, CREB phosphorylation, fPRR/sPRR protein expression, and renin production. Recombinant sPRR (sPRR-His) stimulated renin production, which was inhibited by the PRR decoy peptide PRO20 and S1P inhibitor PF429242. These findings suggest that apelin-13 inhibits plasma renin expression through the cAMP/PKA/sPRR pathway, providing a potential therapeutic approach for RVHT. Understanding the regulation of renin production is crucial for developing effective treatments.NEW & NOTEWORTHY Our research elucidated that apelin-13 inhibits renin production through the cAMP/PKA/soluble (pro)renin receptor pathway, presenting a promising therapeutic approach for renovascular hypertension (RVHT) by targeting renin expression mechanisms. These findings underscore the potential of apelin-13 as a novel strategy to address RVHT.
Collapse
Affiliation(s)
- Ziqing Yan
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Teng Yang
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Xinxuan Li
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Zipeng Jiang
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Wankun Jia
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Jin Zhou
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| | - Hui Fang
- School of PharmacyWeifang Medical University, Weifang, Shandong, China
| |
Collapse
|
3
|
Wang B, Jie H, Wang S, Dong B, Zou Y. The role of (pro)renin receptor and its soluble form in cardiovascular diseases. Front Cardiovasc Med 2023; 10:1086603. [PMID: 36824459 PMCID: PMC9941963 DOI: 10.3389/fcvm.2023.1086603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
The renin-angiotensin system (RAS) is a major classic therapeutic target for cardiovascular diseases. In addition to the circulating RAS, local tissue RAS has been identified in various tissues and plays roles in tissue inflammation and tissue fibrosis. (Pro)renin receptor (PRR) was identified as a new member of RAS in 2002. Studies have demonstrated the effects of PRR and its soluble form in local tissue RAS. Moreover, as an important part of vacuolar H+-ATPase, it also contributes to normal lysosome function and cell survival. Evidently, PRR participates in the pathogenesis of cardiovascular diseases and may be a potential therapeutic target of cardiovascular diseases. This review focuses on the effects of PRR and its soluble form on the physiological state, hypertension, myocardial ischemia reperfusion injury, heart failure, metabolic cardiomyopathy, and atherosclerosis. We aimed to investigate the possibilities and challenges of PRR and its soluble form as a new therapeutic target in cardiovascular diseases.
Collapse
Affiliation(s)
- Boyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China,Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China,Department of Cardiology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Jie
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China,Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China,Shuangxi Wang,
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China,Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China,Department of Cardiology, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Bo Dong,
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Yunzeng Zou,
| |
Collapse
|
4
|
Fu Z, Zheng H, Kaewsaro K, Lambert J, Chen Y, Yang T. Mutagenesis of the cleavage site of (pro)renin receptor abrogates aldosterone-salt-induced hypertension and renal injury in mice. Am J Physiol Renal Physiol 2023; 324:F1-F11. [PMID: 36302140 PMCID: PMC9762973 DOI: 10.1152/ajprenal.00088.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/29/2022] [Accepted: 10/17/2022] [Indexed: 02/04/2023] Open
Abstract
Soluble (pro)renin receptor (sPRR), the extracellular domain of (pro)renin receptor (PRR), is primarily generated by site-1 protease and furin. It has been reported that sPRR functions as an important regulator of intrarenal renin contributing to angiotensin II (ANG II)-induced hypertension. Relatively, less is known for the function of sPRR in ANG II-independent hypertension such as mineralocorticoid excess. In the present study, we used a novel mouse model with mutagenesis of the cleavage site in PRR (termed as PRRR279V/L282V or mutant) to examine the phenotype during aldosterone (Aldo)-salt treatment. The hypertensive response of mutant mice to Aldo-salt treatment was blunted in parallel with the attenuated response of plasma volume expansion and renal medullary α-epithelial Na+ channel expression. Moreover, Aldo-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied by blunted polydipsia and polyuria. Together, these results represent strong evidence favoring endogenous sPRR as a mediator of Aldo-salt-induced hypertension and renal injury.NEW & NOTEWORTHY We used a novel mouse model with mutagenesis of the cleavage site of PRR to support soluble PRR as an essential mediator of aldosterone-salt-induced hypertension and also as a potential therapeutic target for patients with mineralocorticoid excess. We firstly report that soluble PRR-dependent pathway medicates the Na+-retaining action of aldosterone in the distal nephron, which opens up a new area for a better understanding of the molecular basis of renal handling of Na+ balance and blood pressure.
Collapse
Affiliation(s)
- Ziwei Fu
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Huaqing Zheng
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
- Renal Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Kannaree Kaewsaro
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Jacob Lambert
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Yanting Chen
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Tianxin Yang
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
- Renal Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
5
|
Xu C, Chen Y, Ramkumar N, Zou CJ, Sigmund CD, Yang T. Collecting duct renin regulates potassium homeostasis in mice. Acta Physiol (Oxf) 2023; 237:e13899. [PMID: 36264268 PMCID: PMC10754139 DOI: 10.1111/apha.13899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 01/03/2023]
Abstract
AIM The kaliuretic action of the renin-angiotensin-aldosterone system (RAAS) is well established as highlighted by hyperkalemia side effect of RAAS inhibitors but such action is usually ascribed to systemic RAAS. The present study addresses the involvement of intrarenal RAAS in K+ homeostasis with emphasis on locally generated renin within the collecting duct (CD). METHODS Wild-type (Floxed) and CD-specific deletion of renin (CD renin KO) mice were treated for 7 days with a high K+ (HK) diet to investigate the role of CD renin in kaliuresis regulation and further define the underlying mechanism with emphasis on analysis of intrarenal aldosterone biosynthesis. RESULTS In floxed mice, renin levels were elevated in the renal medulla and urine following a 1-week HK diet, indicating activation of the intrarenal renin. CD renin KO mice had blunted HK-induced intrarenal renin response and developed impaired kaliuresis and elevated plasma K+ level (4.45 ± 0.14 vs. 3.89 ± 0.04 mM, p < 0.01). In parallel, HK-induced intrarenal aldosterone and CYP11B2 expression along with expression of renal outer medullary K+ channel (ROMK), calcium-activated potassium channel subunit alpha-1 (α-BK), α-Na+ -K+ -ATPase, and epithelial sodium channel (β-ENaC and cleaved-γ-ENaC) expression were all significantly blunted in CD renin KO mice in contrast to the unaltered responses of plasma aldosterone and adrenal CYP11B2. CONCLUSION Taken together, these results support a kaliuretic action of CD renin during HK intake.
Collapse
Affiliation(s)
- Chuanming Xu
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, Utah, UT 84132
| | - Yanting Chen
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, Utah, UT 84132
| | - Nirupama Ramkumar
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, UT 84132
| | - Chang-Jiang Zou
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, Utah, UT 84132
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, Utah, UT 84132
| |
Collapse
|
6
|
Devaux CA, Camoin-Jau L. An update on angiotensin-converting enzyme 2 structure/functions, polymorphism, and duplicitous nature in the pathophysiology of coronavirus disease 2019: Implications for vascular and coagulation disease associated with severe acute respiratory syndrome coronavirus infection. Front Microbiol 2022; 13:1042200. [PMID: 36519165 PMCID: PMC9742611 DOI: 10.3389/fmicb.2022.1042200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 08/01/2023] Open
Abstract
It has been known for many years that the angiotensin-converting enzyme 2 (ACE2) is a cell surface enzyme involved in the regulation of blood pressure. More recently, it was proven that the severe acute respiratory syndrome coronavirus (SARS-CoV-2) interacts with ACE2 to enter susceptible human cells. This functional duality of ACE2 tends to explain why this molecule plays such an important role in the clinical manifestations of coronavirus disease 2019 (COVID-19). At the very start of the pandemic, a publication from our Institute (entitled "ACE2 receptor polymorphism: susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome"), was one of the first reviews linking COVID-19 to the duplicitous nature of ACE2. However, even given that COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin system (RAS), we were still far from understanding the complexity of the mechanisms which are controlled by ACE2 in different cell types. To gain insight into the physiopathology of SARS-CoV-2 infection, it is essential to consider the polymorphism and expression levels of the ACE2 gene (including its alternative isoforms). Over the past 2 years, an impressive amount of new results have come to shed light on the role of ACE2 in the pathophysiology of COVID-19, requiring us to update our analysis. Genetic linkage studies have been reported that highlight a relationship between ACE2 genetic variants and the risk of developing hypertension. Currently, many research efforts are being undertaken to understand the links between ACE2 polymorphism and the severity of COVID-19. In this review, we update the state of knowledge on the polymorphism of ACE2 and its consequences on the susceptibility of individuals to SARS-CoV-2. We also discuss the link between the increase of angiotensin II levels among SARS-CoV-2-infected patients and the development of a cytokine storm associated microvascular injury and obstructive thrombo-inflammatory syndrome, which represent the primary causes of severe forms of COVID-19 and lethality. Finally, we summarize the therapeutic strategies aimed at preventing the severe forms of COVID-19 that target ACE2. Changing paradigms may help improve patients' therapy.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Center National de la Recherche Scientifique, Marseille, France
| | - Laurence Camoin-Jau
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Laboratoire d’Hématologie, Hôpital de La Timone, APHM, Boulevard Jean-Moulin, Marseille, France
| |
Collapse
|
7
|
Munday JS, Odom T, Dittmer KE, Wetzel S, Hillmer K, Tan ST. Multimodal Blockade of the Renin-Angiotensin System Is Safe and Is a Potential Cancer Treatment for Cats. Vet Sci 2022; 9:vetsci9080411. [PMID: 36006326 PMCID: PMC9413835 DOI: 10.3390/vetsci9080411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/21/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary As activation of the renin-angiotensin system (RAS) promotes cancer cell growth, medications that inhibit RAS activation could reduce cancer progression. However, studies in people in which RAS has been inhibited by a single treatment have not been consistently beneficial, possibly as RAS can be activated by many different cellular pathways. Multiple treatments have been used to more consistently block RAS in people, but such multimodal treatments have never previously been evaluated in veterinary species. In the present study, the safety of multimodal RAS inhibition using a combination of five treatments was assessed in six cats with cancer. Cats were treated for 8 weeks and none of the cats developed low blood pressure, evidence of kidney or liver disease, or significant adverse effects. Of the six cats enrolled in the study, one cat was withdrawn from the study due to difficulties administering the medications and another cat died of an unrelated cause. Two cats were euthanatized due to cancer progression during the study period while two cats completed the 8-week treatment period. The study showed that a multimodal blockade of RAS has the potential to be a safe and cost-effective treatment for cancer in cats. Abstract The role of the renin-angiotensin system (RAS) in cancer growth and progression is well recognized in humans. However, studies on RAS inhibition with a single agent have not shown consistent anticancer effects, potentially due to the neoplastic cells utilizing alternative pathways for RAS activation. To achieve more complete RAS inhibition, multimodal therapy with several medications that simultaneously block multiple steps in the RAS has been developed for use in humans. In the present study, the safety of multimodal RAS inhibition using atenolol, benazepril, metformin, curcumin, and meloxicam was assessed in six cats with squamous cell carcinomas. Cats were treated for 8 weeks, with blood pressure measured and blood sampled five times during the treatment period. None of the cats developed hypotension, azotemia, or increased serum liver enzyme concentrations. The packed cell volume of one cat decreased to just below the reference range during treatment. One cat was reported to have increased vomiting, although this occurred infrequently. One cat was withdrawn from the study due to difficulties administering the medications, and another cat died of an unrelated cause. Two cats were euthanatized during the study period due to cancer progression. Two cats completed the 8-week study period. One was subsequently euthanized due to cancer progression while the other cat is still alive 32 weeks after entering the study and is still receiving the multimodal blockade of the RAS. This is the first evaluation of multimodal blockade of the RAS in veterinary species. The study showed that the treatment is safe, with only mild adverse effects observed in two treated cats. Due to the small number of cats, the efficacy of treatment could not be evaluated. However, evidence from human studies suggests that a multimodal blockade of RAS could be a safe and cost-effective treatment option for cancer in cats.
Collapse
Affiliation(s)
- John S. Munday
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand
- Correspondence:
| | - Thomas Odom
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand
| | - Keren E. Dittmer
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand
| | - Sarah Wetzel
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand
| | | | - Swee T. Tan
- Gillies McIndoe Research Institute, Wellington 7184, New Zealand
| |
Collapse
|
8
|
Lin H, Geurts F, Hassler L, Batlle D, Mirabito Colafella KM, Denton KM, Zhuo JL, Li XC, Ramkumar N, Koizumi M, Matsusaka T, Nishiyama A, Hoogduijn MJ, Hoorn EJ, Danser AHJ. Kidney Angiotensin in Cardiovascular Disease: Formation and Drug Targeting. Pharmacol Rev 2022; 74:462-505. [PMID: 35710133 PMCID: PMC9553117 DOI: 10.1124/pharmrev.120.000236] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.
Collapse
Affiliation(s)
- Hui Lin
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Frank Geurts
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Luise Hassler
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Daniel Batlle
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Katrina M Mirabito Colafella
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Kate M Denton
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Jia L Zhuo
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Xiao C Li
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Nirupama Ramkumar
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Masahiro Koizumi
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Taiji Matsusaka
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Akira Nishiyama
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Martin J Hoogduijn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Ewout J Hoorn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| |
Collapse
|
9
|
Yang T. Revisiting the relationship between (Pro)Renin receptor and the intrarenal RAS: focus on the soluble receptor. Curr Opin Nephrol Hypertens 2022; 31:351-357. [PMID: 35703290 PMCID: PMC9286065 DOI: 10.1097/mnh.0000000000000806] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The (pro)renin receptor (PRR), also termed as ATPase H+ transporting accessory protein 2 (ATP6AP2), was originally cloned as a specific receptor for prorenin and renin [together called (pro)renin]. Given the wide tissue distribution of PRR, PRR was further postulated to act as a regulator of tissue renin. However, assigning a physiological role of PRR within the renin-angiotensin system (RAS) has been challenging largely due to its pleotropic functions in regulation of embryogenesis, autophagy, and H+ transport. The current review will summarize recent advances in understanding the roles of sPPR within the intrarenal RAS as well as those outside this local system. RECENT FINDINGS Site-1 protease (S1P) is a predominant source of sPPR at least in the kidney. So far most of the known physiological functions of PRR including renal handling of electrolytes and fluid and blood pressure are mediated by sPRR. In particular, sPRR serves as a positive regulator of collecting duct renin to activate the intrarenal RAS during water deprivation or angiotensin-II (AngII) infusion. However, PRR/sPRR can act in renin-independent manner under other circumstances. SUMMARY S1P-derived sPRR has emerged as a key regulator of kidney function and blood pressure and its relationship with the intrarenal RAS depends on the physiological context.
Collapse
Affiliation(s)
- Tianxin Yang
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
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: 7] [Impact Index Per Article: 2.3] [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
|
11
|
Souza LA, Earley YF. (Pro)renin Receptor and Blood Pressure Regulation: A Focus on the Central Nervous System. Curr Hypertens Rev 2022; 18:101-116. [PMID: 35086455 PMCID: PMC9662243 DOI: 10.2174/1570162x20666220127105655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/02/2021] [Accepted: 12/06/2021] [Indexed: 01/27/2023]
Abstract
The renin-angiotensin system (RAS) is classically described as a hormonal system in which angiotensin II (Ang II) is one of the main active peptides. The action of circulating Ang II on its cognate Ang II type-1 receptor (AT1R) in circumventricular organs has important roles in regulating the autonomic nervous system, blood pressure (BP) and body fluid homeostasis, and has more recently been implicated in cardiovascular metabolism. The presence of a local or tissue RAS in various tissues, including the central nervous system (CNS), is well established. However, because the level of renin, the rate-limiting enzyme in the systemic RAS, is very low in the brain, how endogenous angiotensin peptides are generated in the CNS-the focus of this review-has been the subject of considerable debate. Notable in this context is the identification of the (pro)renin receptor (PRR) as a key component of the brain RAS in the production of Ang II in the CNS. In this review, we highlight cellular and anatomical locations of the PRR in the CNS. We also summarize studies using gain- and loss-of function approaches to elucidate the functional importance of brain PRR-mediated Ang II formation and brain RAS activation, as well as PRR-mediated Ang II-independent signaling pathways, in regulating BP. We further discuss recent developments in PRR involvement in cardiovascular and metabolic diseases and present perspectives for future directions.
Collapse
Affiliation(s)
- Lucas A.C. Souza
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, NV, USA
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, NV, USA
| |
Collapse
|
12
|
Wang F, Chen Y, Zou CJ, Luo R, Yang T. Mutagenesis of the Cleavage Site of Pro Renin Receptor Abrogates Angiotensin II-Induced Hypertension in Mice. Hypertension 2021; 78:115-127. [PMID: 34024121 PMCID: PMC9212214 DOI: 10.1161/hypertensionaha.121.16770] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Yanting Chen
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Chang-jiang Zou
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
13
|
Kilmister EJ, Hansen L, Davis PF, Hall SRR, Tan ST. Cell Populations Expressing Stemness-Associated Markers in Vascular Anomalies. Front Surg 2021; 7:610758. [PMID: 33634164 PMCID: PMC7900499 DOI: 10.3389/fsurg.2020.610758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/31/2020] [Indexed: 12/31/2022] Open
Abstract
Treatment of vascular anomalies (VAs) is mostly empirical and, in many instances unsatisfactory, as the pathogeneses of these heterogeneous conditions remain largely unknown. There is emerging evidence of the presence of cell populations expressing stemness-associated markers within many types of vascular tumors and vascular malformations. The presence of these populations in VAs is supported, in part, by the observed clinical effect of the mTOR inhibitor, sirolimus, that regulates differentiation of embryonic stem cells (ESCs). The discovery of the central role of the renin-angiotensin system (RAS) in regulating stem cells in infantile hemangioma (IH) provides a plausible explanation for its spontaneous and accelerated involution induced by β-blockers and ACE inhibitors. Recent work on targeting IH stem cells by inhibiting the transcription factor SOX18 using the stereoisomer R(+) propranolol, independent of β-adrenergic blockade, opens up exciting opportunities for novel treatment of IH without the β-adrenergic blockade-related side effects. Gene mutations have been identified in several VAs, involving mainly the PI3K/AKT/mTOR and/or the Ras/RAF/MEK/ERK pathways. Existing cancer therapies that target these pathways engenders the exciting possibility of repurposing these agents for challenging VAs, with early results demonstrating clinical efficacy. However, there are several shortcomings with this approach, including the treatment cost, side effects, emergence of treatment resistance and unknown long-term effects in young patients. The presence of populations expressing stemness-associated markers, including transcription factors involved in the generation of induced pluripotent stem cells (iPSCs), in different types of VAs, suggests the possible role of stem cell pathways in their pathogenesis. Components of the RAS are expressed by cell populations expressing stemness-associated markers in different types of VAs. The gene mutations affecting the PI3K/AKT/mTOR and/or the Ras/RAF/MEK/ERK pathways interact with different components of the RAS, which may influence cell populations expressing stemness-associated markers within VAs. The potential of targeting these populations by manipulating the RAS using repurposed, low-cost and commonly available oral medications, warrants further investigation. This review presents the accumulating evidence demonstrating the presence of stemness-associated markers in VAs, their expression of the RAS, and their interaction with gene mutations affecting the PI3K/AKT/mTOR and/or the Ras/RAF/MEK/ERK pathways, in the pathogenesis of VAs.
Collapse
Affiliation(s)
| | - Lauren Hansen
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Paul F. Davis
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | | | - Swee T. Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand
- Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Wellington, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Feng Y, Peng K, Luo R, Wang F, Yang T. Site-1 Protease-Derived Soluble (Pro)Renin Receptor Contributes to Angiotensin II-Induced Hypertension in Mice. Hypertension 2021; 77:405-416. [PMID: 33280408 PMCID: PMC7803453 DOI: 10.1161/hypertensionaha.120.15100] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of PRR ([pro]renin receptor) contributes to enhancement of intrarenal RAS and renal medullary α-ENaC and thus elevated blood pressure during Ang II (angiotensin II) infusion. The goal of the present study was to test whether such action of PRR was mediated by sPRR (soluble PRR), generated by S1P (site-1 protease), a newly identified PRR cleavage protease. F1 B6129SF1/J mice were infused for 6 days with control or Ang II at 300 ng/kg per day alone or in combination with S1P inhibitor PF-429242 (PF), and blood pressure was monitored by radiotelemetry. S1P inhibition significantly attenuated Ang II-induced hypertension accompanied with suppressed urinary and renal medullary renin levels and expression of renal medullary but not renal cortical α-ENaC expression. The effects of S1P inhibition were all reversed by supplement with histidine-tagged sPRR termed as sPRR-His. Ussing chamber technique was performed to determine amiloride-sensitive short-circuit current, an index of ENaC activity in confluent mouse cortical collecting duct cell line cells exposed for 24 hours to Ang II, Ang II + PF, or Ang II + PF + sPRR-His. Ang II-induced ENaC activity was blocked by PF, which was reversed by sPRR-His. Together, these results support that S1P-derived sPRR mediates Ang II-induced hypertension through enhancement of intrarenal renin level and activation of ENaC.
Collapse
Affiliation(s)
- Ye Feng
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Kexin Peng
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Renfei Luo
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Fei Wang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Tianxin Yang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
15
|
Monteonofrio L, Florio MC, AlGhatrif M, Lakatta EG, Capogrossi MC. Aging- and gender-related modulation of RAAS: potential implications in COVID-19 disease. VASCULAR BIOLOGY 2020; 3:R1-R14. [PMID: 33537555 PMCID: PMC7849461 DOI: 10.1530/vb-20-0014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a new infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is frequently characterized by a marked inflammatory response with severe pneumonia and respiratory failure associated with multiorgan involvement. Some risk factors predispose patients to develop a more severe infection and to an increased mortality; among them, advanced age and male gender have been identified as major and independent risk factors for COVID-19 poor outcome. The renin-angiotensin-aldosterone system (RAAS) is strictly involved in COVID-19 because angiotensin converting enzyme 2 (ACE2) is the host receptor for SARS-CoV-2 and also converts pro-inflammatory angiotensin (Ang) II into anti-inflammatory Ang(1–7). In this review, we have addressed the effect of aging and gender on RAAS with emphasis on ACE2, pro-inflammatory Ang II/Ang II receptor 1 axis and anti-inflammatory Ang(1–7)/Mas receptor axis.
Collapse
Affiliation(s)
- Laura Monteonofrio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maria Cristina Florio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Majd AlGhatrif
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maurizio C Capogrossi
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Wang F, Sun Y, Luo R, Lu X, Yang B, Yang T. COX-2-independent activation of renal (pro)renin receptor contributes to DOCA-salt hypertension in rats. Am J Physiol Renal Physiol 2020; 319:F647-F653. [PMID: 32799674 PMCID: PMC7642891 DOI: 10.1152/ajprenal.00112.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022] Open
Abstract
It has been shown that cyclooxygenase (COX)-2-dependent activation of renal (pro)renin receptor (PRR) contributes to angiotensin II (ANG II)-induced hypertension. However, less is known about the involvement of this mechanism in ANG II-independent hypertension. The goal of the present study was to test whether or not COX-2-dependent upregulation of PRR serves as a universal mechanism contributing to ANG II-dependent and -independent hypertension. Here, we examined the association between renal COX-2 and PRR during deoxycorticosterone acetate (DOCA)-salt hypertension in rats. By immunoblot analysis and immunofluorescence, renal protein expression of PRR was remarkably upregulated by DOCA-salt treatment. Surprisingly, this upregulation of renal PRR expression was unaffected by a COX-2 inhibitor, celecoxib. To address the role of renal PRR to the pathogenesis of DOCA-salt hypertension, a decoy PRR inhibitor, PRO20, was infused to the renal medulla of uninephrectomized Sprague-Dawley rats for 14 days. Radiotelemetry demonstrated effective attenuation of DOCA-salt hypertension by intramedullary infusion of a PRR inhibitor, PRO20. In parallel, DOCA-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied with blunted polydipsia and polyuria. In contrast, intravenous infusion of PRO20 was less effective in attenuating DOCA-salt hypertension and cardiorenal injury. Together, these results suggest that COX-2-independent activation of renal PRR contributes to DOCA-salt hypertension.
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Ying Sun
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Xiaohan Lu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
17
|
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: 0.8] [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
|
18
|
Abstract
The (pro)renin receptor ((P)RR) was first identified as a single-transmembrane receptor in human kidneys and initially attracted attention owing to its potential role as a regulator of the tissue renin-angiotensin system (RAS). Subsequent studies found that the (P)RR is widely distributed in organs throughout the body, including the kidneys, heart, brain, eyes, placenta and the immune system, and has multifaceted functions in vivo. The (P)RR has roles in various physiological processes, such as the cell cycle, autophagy, acid-base balance, energy metabolism, embryonic development, T cell homeostasis, water balance, blood pressure regulation, cardiac remodelling and maintenance of podocyte structure. These roles of the (P)RR are mediated by its effects on important biological systems and pathways including the tissue RAS, vacuolar H+-ATPase, Wnt, partitioning defective homologue (Par) and tyrosine phosphorylation. In addition, the (P)RR has been reported to contribute to the pathogenesis of diseases such as fibrosis, hypertension, pre-eclampsia, diabetic microangiopathy, acute kidney injury, cardiovascular disease, cancer and obesity. Current evidence suggests that the (P)RR has key roles in the normal development and maintenance of vital organs and that dysfunction of the (P)RR is associated with diseases that are characterized by a disruption of the homeostasis of physiological functions.
Collapse
|
19
|
Tan DC, Roth IM, Wickremesekera AC, Davis PF, Kaye AH, Mantamadiotis T, Stylli SS, Tan ST. Therapeutic Targeting of Cancer Stem Cells in Human Glioblastoma by Manipulating the Renin-Angiotensin System. Cells 2019; 8:cells8111364. [PMID: 31683669 PMCID: PMC6912312 DOI: 10.3390/cells8111364] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with glioblastoma (GB), a highly aggressive brain tumor, have a median survival of 14.6 months following neurosurgical resection and adjuvant chemoradiotherapy. Quiescent GB cancer stem cells (CSCs) invariably cause local recurrence. These GB CSCs can be identified by embryonic stem cell markers, express components of the renin-angiotensin system (RAS) and are associated with circulating CSCs. Despite the presence of circulating CSCs, GB patients rarely develop distant metastasis outside the central nervous system. This paper reviews the current literature on GB growth inhibition in relation to CSCs, circulating CSCs, the RAS and the novel therapeutic approach by repurposing drugs that target the RAS to improve overall symptom-free survival and maintain quality of life.
Collapse
Affiliation(s)
- David Ch Tan
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
| | - Imogen M Roth
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Agadha C Wickremesekera
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem 91120, Israel.
| | - Theo Mantamadiotis
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt 5040, New Zealand.
| |
Collapse
|
20
|
Zhang L, Deng M, Lu A, Chen Y, Chen Y, Wu C, Tan Z, Boini KM, Yang T, Zhu Q, Wang L. Sodium butyrate attenuates angiotensin II-induced cardiac hypertrophy by inhibiting COX2/PGE2 pathway via a HDAC5/HDAC6-dependent mechanism. J Cell Mol Med 2019; 23:8139-8150. [PMID: 31565858 PMCID: PMC6850921 DOI: 10.1111/jcmm.14684] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/13/2019] [Accepted: 04/22/2019] [Indexed: 12/24/2022] Open
Abstract
Sodium butyrate (NaBu) is reported to play important roles in a number of chronic diseases. The present work is aimed to investigate the effect of NaBu on angiotensin II (Ang II)-induced cardiac hypertrophy and the underlying mechanism in in vivo and in vitro models. Sprague Dawley rats were infused with vehicle or Ang II (200 ng/kg/min) and orally administrated with or without NaBu (1 g/kg/d) for two weeks. Cardiac hypertrophy parameters and COX2/PGE2 pathway were analysed by real-time PCR, ELISA, immunostaining and Western blot. The cardiomyocytes H9C2 cells were used as in vitro model to investigate the role of NaBu (2 mmol/L) in inhibition of Ang II-induced cardiac hypertrophy. NaBu significantly attenuated Ang II-induced increase in the mean arterial pressure. Ang II treatment remarkably increased cardiac hypertrophy as indicated by increased ratio of heart weight/body weight and enlarged cardiomyocyte size, extensive fibrosis and inflammation, as well as enhanced expression of hypertrophic markers, whereas hearts from NaBu-treated rats exhibited a significant reduction in these hypertrophic responses. Mechanistically, NaBu inhibited the expression of COX2/PGE2 along with production of ANP and phosphorylated ERK (pERK) stimulated by Ang II in in vivo and in vitro, which was accompanied by the suppression of HDAC5 and HDAC6 activities. Additionally, knocking down the expression of HDAC5 and HDAC6 via gene-editing strategy dramatically blocked Ang II-induced hypertrophic responses through COX2/PGE2 pathway. These results provide solid evidence that NaBu attenuates Ang II-induced cardiac hypertrophy by inhibiting the activation of COX2/PGE2 pathway in a HDAC5/HDAC6-dependent manner.
Collapse
Affiliation(s)
- Linlin Zhang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Mokan Deng
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Aihua Lu
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Yanting Chen
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Yang Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunying Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi Tan
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Krishna M Boini
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tianxin Yang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Qing Zhu
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Wang
- Institue of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 714] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
22
|
Granado M, Amor S, Fernández N, Carreño-Tarragona G, Iglesias-Cruz MC, Martín-Carro B, Monge L, García-Villalón AL. Effects of early overnutrition on the renal response to Ang II and expression of RAAS components in rat renal tissue. Nutr Metab Cardiovasc Dis 2017; 27:930-937. [PMID: 28958692 DOI: 10.1016/j.numecd.2017.06.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/16/2017] [Accepted: 06/29/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS The aim of this study was to analyze the effects of early overnutrition (EON) on the expression of the renin angiotensin aldosterone system (RAAS) components in renal cortex, renal arteries and renal perivascular adipose tissue (PVAT), as well as the vascular response of renal arteries to Angiotensin II (Ang II). METHODS AND RESULTS On birth day litters were adjusted to twelve (L12-control) or three (L3-overfed) pups per mother. Half of the animals were sacrificed at weaning (21 days old) and the other half at 5 months of age. Ang II-induced vasoconstriction of renal artery segments increased in young overfed rats and decreased in adult overfed rats. EON decreased the gene expression of angiotensinogen (Agt), Ang II receptors AT1 and AT2 and eNOS in renal arteries of young rats, while it increased the mRNA levels of AT-2 and ET-1 in adult rats. In renal PVAT EON up-regulated the gene expression of COX-2 and TNF-α in young rats and the mRNA levels of renin receptor both in young and in adult rats. On the contrary, Ang II receptors mRNA levels were downregulated at both ages. Renal cortex of overfed rats showed increased gene expression of Agt in adult rats and of AT1 in young rats. However the mRNA levels of AT1 were decreased in the renal cortex of overfed adult rats. CONCLUSION EON is associated with alterations in the vascular response of renal arteries to Ang II and changes in the gene expression of RAAS components in renal tissue.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Adipose Tissue/physiopathology
- Age Factors
- Angiotensin II/pharmacology
- Animal Nutritional Physiological Phenomena
- Animals
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Kidney/blood supply
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Nutritional Status
- Overnutrition/genetics
- Overnutrition/metabolism
- Overnutrition/physiopathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Renal Artery/drug effects
- Renal Artery/metabolism
- Renal Artery/physiopathology
- Renin-Angiotensin System/drug effects
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- M Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.
| | - S Amor
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - N Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - G Carreño-Tarragona
- Servicio de Hematología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M C Iglesias-Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - B Martín-Carro
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - L Monge
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - A L García-Villalón
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
23
|
Alexanian A, Sorokin A. Cyclooxygenase 2: protein-protein interactions and posttranslational modifications. Physiol Genomics 2017; 49:667-681. [PMID: 28939645 DOI: 10.1152/physiolgenomics.00086.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Numerous studies implicate the cyclooxygenase 2 (COX2) enzyme and COX2-derived prostanoids in various human diseases, and thus, much effort has been made to uncover the regulatory mechanisms of this enzyme. COX2 has been shown to be regulated at both the transcriptional and posttranscriptional levels, leading to the development of nonsteroidal anti-inflammatory drugs (NSAIDs) and selective COX2 inhibitors (COXIBs), which inhibit the COX2 enzyme through direct targeting. Recently, evidence of posttranslational regulation of COX2 enzymatic activity by s-nitrosylation, glycosylation, and phosphorylation has also been presented. Additionally, posttranslational regulators that actively downregulate COX2 expression by facilitating increased proteasome degradation of this enzyme have also been reported. Moreover, recent data identified proteins, located in close proximity to COX2 enzyme, that serve as posttranslational modulators of COX2 function, upregulating its enzymatic activity. While the precise mechanisms of the protein-protein interaction between COX2 and these regulatory proteins still need to be addressed, it is likely these interactions could regulate COX2 activity either as a result of conformational changes of the enzyme or by impacting subcellular localization of COX2 and thus affecting its interactions with regulatory proteins, which further modulate its activity. It is possible that posttranslational regulation of COX2 enzyme by such proteins could contribute to manifestation of different diseases. The uncovering of posttranslational regulation of COX2 enzyme will promote the development of more efficient therapeutic strategies of indirectly targeting the COX2 enzyme, as well as provide the basis for the generation of novel diagnostic tools as biomarkers of disease.
Collapse
Affiliation(s)
- Anna Alexanian
- Cardiovascular Center and Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Andrey Sorokin
- Cardiovascular Center and Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
24
|
Fang H, Xu C, Lu A, Zou CJ, Xie S, Chen Y, Zhou L, Liu M, Wang L, Wang W, Yang T. (Pro)renin receptor mediates albumin-induced cellular responses: role of site-1 protease-derived soluble (pro)renin receptor in renal epithelial cells. Am J Physiol Cell Physiol 2017; 313:C632-C643. [PMID: 28903918 DOI: 10.1152/ajpcell.00006.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Proteinuria is a characteristic of chronic kidney disease and also a causative factor that promotes the disease progression, in part, via activation of the intrarenal renin-angiotensin system (RAS). (Pro)renin receptor (PRR), a newly discovered component of the RAS, binds renin and (pro)renin to promote angiotensin I generation. The present study was performed to test the role of soluble PRR (sPRR) in albumin overload-induced responses in cultured human renal proximal tubular cell line human kidney 2 (HK-2) cells. Bovine serum albmuin (BSA) treatment for 24 h at 20 mg/ml induced renin activity and inflammation, both of which were attenuated by a PRR decoy inhibitor PRO20. BSA treatment induced a more than fivefold increase in medium sPRR due to enhanced cleavage of PRR. Surprisingly, this cleavage event was unaffected by inhibition of furin or a disintegrin and metalloproteinase 19. Screening for a novel cleavage enzyme led to the identification of site-1 protease (S1P). Inhibition of S1P with PF-429242 or siRNA remarkably suppressed BSA-induced sPRR production, renin activity, and inflammatory response. Administration of a recombinant sPRR, termed sPRR-His, reversed the effects of S1P inhibition. In HK-2 cells overexpressing PRR, mutagenesis of the S1P, but not furin cleavage site, reduced sPRR levels. Together, these results suggest that PRR mediates albumin-induced cellular responses through S1P-derived sPRR.
Collapse
Affiliation(s)
- Hui Fang
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Chuanming Xu
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China.,Department of Internal Medicine, University of Utah School of Medicine and Veterans Affairs Medical Center , Salt Lake City, Utah
| | - Aihua Lu
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Chang-Jiang Zou
- Department of Internal Medicine, University of Utah School of Medicine and Veterans Affairs Medical Center , Salt Lake City, Utah
| | - Shiying Xie
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Yanting Chen
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Li Zhou
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Mi Liu
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Lei Wang
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Weidong Wang
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine , Guangzhou , China.,Department of Internal Medicine, University of Utah School of Medicine and Veterans Affairs Medical Center , Salt Lake City, Utah
| |
Collapse
|
25
|
Wang L, Zhu Q, Lu A, Liu X, Zhang L, Xu C, Liu X, Li H, Yang T. Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin-angiotensin system. J Hypertens 2017; 35:1899-1908. [PMID: 28509726 PMCID: PMC11157961 DOI: 10.1097/hjh.0000000000001378] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Butyrate, a short-chain fatty acid, is the end product of the fermentation of complex carbohydrates by the gut microbiota. Recently, sodium butyrate (NaBu) has been found to play a protective role in a number of chronic diseases. However, it is still unclear whether NaBu has a therapeutic potential in hypertension. The present study was aimed to investigate the role of NaBu in angiotensin II (Ang II)-induced hypertension and to further explore the underlying mechanism. METHODS Ang II was infused into uninephrectomized Sprague-Dawley rats with or without intramedullary infusion of NaBu for 14 days. Mean arterial blood pressure was recorded by the telemetry system. Renal tissues, serum samples, and 24-h urine samples were collected to examine renal injury and the regulation of the (pro)renin receptor (PRR) and renin. RESULTS Intramedullary infusion of NaBu in Sprague-Dawley rats lowered the Ang II-induced mean arterial pressure from 129 ± 6 mmHg to 108 ± 4 mmHg (P < 0.01). This corresponded with an improvement in Ang II-induced renal injury, including urinary albumin, glomerulosclerosis, and renal fibrosis, as well as the expression of inflammatory mediators tumor necrosis factor α, interleukin 6. The renal expression of PRR, angiotensinogen, angiotensin I-converting enzyme and the urinary excretion of soluble PRR, renin, and angiotensinogen were all increased by Ang II infusion but decreased by NaBu treatment. In cultured innermedullary collecting duct cells, NaBu treatment attenuated Ang II-induced expression of PRR and renin. CONCLUSION These results demonstrate that NaBu exerts an antihypertensive action, likely by suppressing the PRR-mediated intrarenal renin-angiotensin system.
Collapse
Affiliation(s)
- Lei Wang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Qing Zhu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Aihua Lu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Xiaofen Liu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Linlin Zhang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Chuanming Xu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Xiyang Liu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Haobo Li
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
- Veterans Affairs Medical Center, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
26
|
Prostaglandin E 2 Induces Prorenin-Dependent Activation of (Pro)renin Receptor and Upregulation of Cyclooxygenase-2 in Collecting Duct Cells. Am J Med Sci 2017; 354:310-318. [PMID: 28918839 DOI: 10.1016/j.amjms.2017.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prostaglandin E2 (PGE2) regulates renin expression in renal juxtaglomerular cells. PGE2 acts through E-prostanoid (EP) receptors in the renal collecting duct (CD) to regulate sodium and water balance. CD cells express EP1 and EP4, which are linked to protein kinase C (PKC) and PKA downstream pathways, respectively. Previous studies showed that the presence of renin in the CD, and that of PKC and PKA pathways, activate its expression. The (pro)renin receptor (PRR) is also expressed in CD cells, and its activation enhances cyclooxygenase-2 (COX-2) through extracellular signal-regulated kinase (ERK). We hypothesized that PGE2 stimulates prorenin and renin synthesis leading to subsequent activation of PRR and upregulation of COX-2. METHODS We used a mouse M-1 CD cell line that expresses EP1, EP3 and EP4 but not EP2. RESULTS PGE2 (10-6M) treatment increased prorenin and renin protein levels at 4 and 8 hours. No differences were found at 12-hour after PGE2 treatment. Phospho-ERK was significantly augmented after 12 hours. COX-2 expression was decreased after 4 hours of PGE2 treatment, but increased after 12 hours. Interestingly, the full-length form of the PRR was upregulated only at 12 hours. PGE2-mediated phospho-ERK and COX-2 upregulation was suppressed by PRR silencing. CONCLUSIONS Our results suggest that PGE2 induces biphasic regulation of COX-2 through renin-dependent PRR activation via EP1 and EP4 receptors. PRR-mediated increases in COX-2 expression may enhance PGE2 synthesis in CD cells serving as a buffer mechanism in conditions of activated renin-angiotensin system.
Collapse
|
27
|
Yu X, Yang Y, Yuan H, Wu M, Li S, Gong W, Yu J, Xia W, Zhang Y, Ding G, Huang S, Jia Z, Zhang A. Inhibition of COX-2/PGE2 cascade ameliorates cisplatin-induced mesangial cell apoptosis. Am J Transl Res 2017; 9:1222-1229. [PMID: 28386348 PMCID: PMC5376013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/07/2017] [Indexed: 06/07/2023]
Abstract
Cisplatin is one of the most potent cytotoxic drug for the treatment of many types of cancer. However, the side effects on normal tissues, particularly on the kidney, greatly limited its use in clinic. Emerging evidence demonstrated that cisplatin could directly cause mesangial cell apoptosis, while the potential mechanism is still elusive. Here we examined the contribution of COX-2 in cisplatin-induced mesangial cell apoptosis. Firstly, we found cisplatin induced cell apoptosis in mesangial cells shown by increased number of apoptotic cells in parallel with the upregulation of Bax and the downregulation of Bcl-2. Interestingly, cisplatin-induced cell apoptosis was accompanied by an upregulation of COX-2 at both mRNA and protein levels in dose- and time-dependent manners. Importantly, inhibition of COX-2 via a specific COX-2 inhibitor celecoxib markedly blocked cisplatin-induced mesangial cell apoptosis as evidenced by the decreased number of apoptotic cells, blocked increments of cleaved caspase-3 and Bax, and reversed Bcl-2 downregulation. Meanwhile, cisplatin-induced PGE2 production was markedly blocked by the treatment of celecoxib. In conclusion, this study indicated that COX-2/PGE2 cascade activation mediated cisplatin-induced mesangial cell apoptosis. The findings not only offered new insights into the understanding of cisplatin nephrotoxicity but also provided the therapeutic potential by targeting COX-2/PGE2 cascade in treating cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Xiaowen Yu
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Yunwen Yang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Hui Yuan
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Meng Wu
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Shuzhen Li
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Wei Gong
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Jing Yu
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Weiwei Xia
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Yue Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Guixia Ding
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical UniversityNanjing 210029, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, China
| |
Collapse
|
28
|
Affiliation(s)
- Tianxin Yang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City; and Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.
| |
Collapse
|
29
|
Yang T, Liu M. Regulation and function of renal medullary cyclooxygenase-2 during high salt loading. Front Biosci (Landmark Ed) 2017; 22:128-136. [PMID: 27814606 DOI: 10.2741/4476] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prostaglandins (PGs) are important autocrine/paracrine regulators that contribute to sodium balance and blood pressure control. Along the nephron, the highest amount of PGE2 is found in the distal nephron, an important site for fine-tuning of urinary sodium and water excretion. Cylooxygenase-2 (COX-2) is abundantly expressed in the renal medulla and its expression along with urinary PGE2 excretion is highly induced by chronic salt loading. Factors involved in high salt-induced COX-2 expression in the renal medulla include the hypertonicity, fluid shear stress (FSS), and hypoxia-inducible factor-1 alpha (HIF-1 alpha). Site-specific inhibition of COX-2 in the renal medulla of Sprague-Dawley rats causes sodium retention and salt-sensitive hypertension. Together, these results support the concept that renal medullary COX-2 functions an important natriuretic mediator that is activated by salt loading and its products promote sodium excretion and contribute to maintenance of sodium balance and blood pressure.
Collapse
Affiliation(s)
- Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah,
| | - Mi Liu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah and Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, 510080, China
| |
Collapse
|
30
|
Xu C, Lu A, Lu X, Zhang L, Fang H, Zhou L, Yang T. Activation of Renal (Pro)Renin Receptor Contributes to High Fructose-Induced Salt Sensitivity. Hypertension 2016; 69:339-348. [PMID: 27993957 DOI: 10.1161/hypertensionaha.116.08240] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/09/2016] [Accepted: 11/28/2016] [Indexed: 12/16/2022]
Abstract
A high-fructose diet is shown to induce salt-sensitive hypertension, but the underlying mechanism largely remains unknown. The major goal of the present study was to test the role of renal (pro)renin receptor (PRR) in this model. In Sprague-Dawley rats, high-fructose intake increased renal expression of full-length PRR, which were attenuated by allopurinol. High-fructose intake also upregulated renal mRNA and protein expression of sodium/hydrogen exchanger 3 and Na/K/2Cl cotransporter, as well as in vivo Na/K/2Cl cotransporter activity, all of which were nearly completely blocked by a PRR decoy inhibitor PRO20 or allopurinol treatment. Parallel changes were observed for indices of intrarenal renin-angiotensin-system including renal and urinary renin and angiotensin II levels. Radiotelemetry demonstrated that high-fructose or a high-salt diet alone did not affect mean arterial pressure, but the combination of the 2 maneuvers induced a ≈10-mm Hg increase of mean arterial pressure, which was blunted by PRO20 or allopurinol treatment. In cultured human kidney 2 cells, both fructose and uric acid increased protein expression of soluble PRR in a time- and dose-dependent manner; fructose-induced PRR upregulation was inhibited by allopurinol. Taken together, our data suggest that fructose via uric acid stimulates renal expression of PRR/soluble PRR that stimulate sodium/hydrogen exchanger 3 and Na/K/2Cl cotransporter expression and intrarenal renin-angiotensin system to induce salt-sensitive hypertension.
Collapse
Affiliation(s)
- Chuanming Xu
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Aihua Lu
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Xiaohan Lu
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Linlin Zhang
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Hui Fang
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Li Zhou
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.)
| | - Tianxin Yang
- From the Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China (C.X., A.L., X.L., L.Z., H.F., L.Z., T.Y.); Internal Medicine, University of Utah, Salt Lake City (C.X., X.L., T.Y.); and Veterans Affairs Medical Center, Salt Lake City, Utah (X.L., T.Y.).
| |
Collapse
|
31
|
Zhou L, Liu Y. Wnt/β-catenin signaling and renin-angiotensin system in chronic kidney disease. Curr Opin Nephrol Hypertens 2016; 25:100-6. [PMID: 26808707 DOI: 10.1097/mnh.0000000000000205] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Intrarenal activation of the renin-angiotensin system (RAS) plays an essential role in the pathogenesis of hypertension and chronic kidney diseases (CKD). However, how RAS genes are regulated in vivo was poorly understood until recently. This review focuses on recent findings of the transcriptional regulation of RAS components, as well as their implication in developing novel strategies to treat the patients with CKD. RECENT FINDINGS Bioinformatics analyses have uncovered the presence of putative binding sites for T-cell factor/β-catenin in the promoter region of all RAS genes. Both in-vitro and in-vivo studies confirm that Wnt/β-catenin is the master upstream regulator that controls the expression of all RAS components tested, such as angiotensinogen, renin, angiotensin converting enzyme and the angiotensin II type I receptor in the kidney. Targeted inhibition of Wnt/β-catenin, by either small molecule ICG-001 or endogenous Wnt antagonist Klotho, represses RAS activation and ameliorates proteinuria and kidney injury. Blockade of Wnt/β-catenin signaling also normalizes blood pressure in a mouse model of CKD. SUMMARY These recent studies identify Wnt/β-catenin as the master regulator that controls multiple RAS genes, and suggest that targeting this upstream signaling could be an effective strategy for the treatment of patients with hypertension and CKD.
Collapse
Affiliation(s)
- Lili Zhou
- aState Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China bDepartment of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
32
|
(Pro)renin receptor contributes to regulation of renal epithelial sodium channel. J Hypertens 2016; 34:486-94; discussion 494. [PMID: 26771338 DOI: 10.1097/hjh.0000000000000825] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Recent studies reported increased (Pro)renin receptor (PRR) expression during low-salt intake. We hypothesized that PRR plays a role in regulation of renal epithelial sodium channel (ENaC) through serum and glucocorticoid-inducible kinase isoform 1 (SGK-1)-neural precursor cell expressed, developmentally downregulated 4-2 (Nedd4-2) signaling pathway. METHOD Male Sprague-Dawley rats on normal-sodium diet and mouse renal inner medullary collecting duct cells treated with NaCl at 130 mmol/l (normal salt), or 63 mmol/l (low salt) were studied. PRR and α-ENaC expressions were evaluated 1 week after right uninephrectomy and left renal interstitial administration of 5% dextrose, scramble shRNA, or PRR shRNA (n = 6 each treatment). RESULTS In-vivo PRR shRNA significantly reduced expressions of PRR throughout the kidney and α-ENaC subunits in the renal medulla. In inner medullary collecting duct cells, low salt or angiotensin II (Ang II) augmented the mRNA and protein expressions of PRR (P < 0.05), SGK-1 (P < 0.05), and α-ENaC (P < 0.05). Low salt or Ang II increased the phosphorylation of Nedd4-2. In cells treated with low salt or Ang II, PRR siRNA significantly downregulated the mRNA and protein expressions of PRR (P < 0.05), SGK-1 (P < 0.05), and α-ENaC expression (P < 0.05). CONCLUSION We conclude that PRR contributes to the regulation of α-ENaC via SGK-1-Nedd4-2 signaling pathway.
Collapse
|
33
|
Ramkumar N, Kohan DE. Role of the Collecting Duct Renin Angiotensin System in Regulation of Blood Pressure and Renal Function. Curr Hypertens Rep 2016; 18:29. [PMID: 26951246 DOI: 10.1007/s11906-016-0638-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Recent evidence suggests that the renal tubular renin angiotensin system regulates urinary Na(+) and water excretion and blood pressure. Three key components of the tubular renin angiotensin system, namely renin, prorenin receptor, and angiotensin-II type 1 receptor, are localized to the collecting duct. This system may modulate collecting duct Na(+) and water reabsorption via angiotensin-II-dependent and angiotensin-II-independent pathways. Further, the system may be of greatest relevance in hypertensive states and particularly those characterized by high circulating angiotensin-II. In this review, we summarize the current knowledge on the synthesis, regulation, and function of collecting duct-derived renin angiotensin system components and examine recent developments with regard to regulation of blood pressure and renal fluid and Na(+) excretion.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology, University of Utah Health Sciences Center, 30 N 1900 E SOM 4R312, Salt Lake City, UT, 84132, USA
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, 30 N 1900 E SOM 4R312, Salt Lake City, UT, 84132, USA. .,Salt Lake Veterans Affairs Medical Center, Salt Lake City, UT, USA.
| |
Collapse
|
34
|
Kang JH, Baik HW, Yoo SM, Kim JH, Cheong HI, Park CG, Kang HG, Ha IS. Aliskiren Regulates Neonatal Fc Receptor and IgG Metabolism with Attenuation of Anti-GBM Glomerulonephritis in Mice. Nephron Clin Pract 2016; 134:272-282. [DOI: 10.1159/000448789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022] Open
|
35
|
Xu C, Lu A, Wang H, Fang H, Zhou L, Sun P, Yang T. (Pro)Renin receptor regulates potassium homeostasis through a local mechanism. Am J Physiol Renal Physiol 2016; 313:F641-F656. [PMID: 27440776 DOI: 10.1152/ajprenal.00043.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 07/12/2016] [Accepted: 07/18/2016] [Indexed: 11/22/2022] Open
Abstract
(Pro)renin receptor (PRR) is highly expressed in the distal nephron, but it has an unclear functional implication. The present study was conducted to explore a potential role of renal PRR during high K+ (HK) loading. In normal Sprague-Dawley rats, a 1-wk HK intake increased renal expression of full-length PRR and urinary excretion of soluble PRR (sPRR). Administration of PRO20, a decoy peptide antagonist of PRR, in K+-loaded animals elevated plasma K+ level and decreased urinary K+ excretion, accompanied with suppressed urinary aldosterone excretion and intrarenal aldosterone levels. HK downregulated Na+-Cl- cotransporter (NCC) expression but upregulated CYP11B2 (cytochrome P-450, family 11, subfamily B, polypeptide 2), renal outer medullary K+ channel (ROMK), calcium-activated potassium channel subunit α1 (α-BK), α-Na+-K+-ATPase (α-NKA), and epithelial Na+ channel subunit β (β-ENaC), all of which were blunted by PRO20. After HK loading was completed, urinary, but not plasma renin, was upregulated, which was blunted by PRO20. The same experiments that were performed using adrenalectomized (ADX) rats yielded similar results. Interestingly, spironolactone treatment in HK-loaded ADX rats attenuated kaliuresis but promoted natriuresis, which was associated with the suppressed responses of β-ENaC, α-NKA, ROMK, and α-BK protein expression. Taken together, we discovered a novel role of renal PRR in regulation of K+ homeostasis through a local mechanism involving intrarenal renin-angiotensin-aldosterone system and coordinated regulation of membrane Na+- and K+-transporting proteins.
Collapse
Affiliation(s)
- Chuanming Xu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Aihua Lu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Hong Wang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Hui Fang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Li Zhou
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Peng Sun
- Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China; and
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; .,Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
36
|
Siljee S, Keane E, Marsh R, Brasch HD, Tan ST, Itinteang T. Expression of the Components of the Renin-Angiotensin System in Venous Malformation. Front Surg 2016; 3:24. [PMID: 27200356 PMCID: PMC4853390 DOI: 10.3389/fsurg.2016.00024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/11/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Venous malformation (VM) is the most common form of vascular malformation, consisting of a network of thin-walled ectatic venous channels with deficient or absent media. This study investigated the expression of the components of the renin-angiotensin system (RAS), namely, (pro)renin receptor (PRR), angiotensin-converting enzyme (ACE), angiotensin II receptor 1 (ATIIR1), and angiotensin II receptor 2 (AIITR2) in subcutaneous (SC) and intramuscular (IM) VM. MATERIALS AND METHODS SC (n = 7) and IM (n = 7) VM were analyzed for the expression of PRR, ACE, ATIIR1, and ATIIR2 using 3,3-diaminobenzidine and immunofluorescent (IF) immunohistochemical (IHC) staining and NanoString gene expression analysis. RESULTS IHC staining showed expression of PRR, ACE, and ATIIR1, and faint expression of ATIIR2 in the endothelium of SC and IM VM. Furthermore, ATIIR2 was expressed by cells away from the endothelium in both SC and IM VM lesions examined. NanoString analysis demonstrated the presence of PRR, ACE, and ATIIR1 but not ATIIR2. CONCLUSION The presence of PRR, ACE, ATIIR1, and potentially ATIIR2, in both SC and IM VM, suggests a role for the RAS in the biology of VM. This novel finding may lead to a mechanism-based therapy for VM.
Collapse
Affiliation(s)
- Sam Siljee
- Gillies McIndoe Research Institute , Wellington , New Zealand
| | - Emily Keane
- Gillies McIndoe Research Institute , Wellington , New Zealand
| | - Reginald Marsh
- Gillies McIndoe Research Institute, Wellington, New Zealand; University of Auckland, Auckland, New Zealand
| | - Helen D Brasch
- Gillies McIndoe Research Institute, Wellington, New Zealand; Department of Pathology, Hutt Hospital, Wellington, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand; Centre for the Study and Treatment of Vascular Birthmarks, Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Wellington, New Zealand
| | - Tinte Itinteang
- Gillies McIndoe Research Institute , Wellington , New Zealand
| |
Collapse
|
37
|
Peng K, Lu X, Wang F, Nau A, Chen R, Zhou SF, Yang T. Collecting duct (pro)renin receptor targets ENaC to mediate angiotensin II-induced hypertension. Am J Physiol Renal Physiol 2016; 312:F245-F253. [PMID: 27122543 DOI: 10.1152/ajprenal.00178.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/21/2016] [Indexed: 01/09/2023] Open
Abstract
The (pro)renin receptor (PRR) is abundantly expressed in the collecting duct (CD) and the expression is further induced by angiotensin II (ANG II). The present study was conducted to investigate the role of CD PRR during ANG II-induced hypertension and to further explore the underlying mechanism. Radiotelemetry demonstrated that a 1-wk ANG II infusion gradually and significantly induced hypertensive response in floxed mice and this response was significantly attenuated in mice lacking PRR in the CD (termed CD PRR KO). ANG II infusion in floxed mice increased urinary renin activity and selectively induced renal medullary α-epithelial sodium channel (α-ENaC) mRNA and protein expression, all of which were blunted in the null mice. In cultured mpkCCD cells grown in Transwells, transepithelial Na+ transport as measured by using a volt-ohmmeter was transiently stimulated by acute ANG II treatment, which was abolished by a PRR antagonist, PRO20. In a chronic setting, ANG II treatment induced α-ENaC mRNA expression in mpkCCD cells, which was similarly blocked by PRO20. Chronic intramedullary infusion of an ENaC inhibitor amiloride in rats significantly attenuated ANG II-induced hypertension. Overall, the present study suggests that CD PRR contributes to ANG II-induced hypertension at least partially via activation of renal medullary ENaC.
Collapse
Affiliation(s)
- Kexin Peng
- 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, Utah
| | - Xiaohan Lu
- 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, Utah
| | - Fei Wang
- 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, Utah
| | - Adam Nau
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China
| | - Ren Chen
- Department of Internal Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China; and
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - 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, Utah
| |
Collapse
|
38
|
Xu Q, Jensen DD, Peng H, Feng Y. The critical role of the central nervous system (pro)renin receptor in regulating systemic blood pressure. Pharmacol Ther 2016; 164:126-34. [PMID: 27113409 DOI: 10.1016/j.pharmthera.2016.04.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Indexed: 01/24/2023]
Abstract
The systemic renin-angiotensin system (RAS) has long been recognized as a critically important system in blood pressure (BP) regulation. However, extensive evidence has shown that a majority of RAS components are also present in many tissues and play indispensable roles in BP regulation. Here, we review evidence that RAS components, notably including the newly identified (pro)renin receptor (PRR), are present in the brain and are essential for the central regulation of BP. Binding of the PRR to its ligand, prorenin or renin, increases BP and promotes progression of cardiovascular diseases in an angiotensin II-dependent and -independent manner, establishing the PRR a promising antihypertensive drug target. We also review the existing PRR blockers, including handle region peptide and PRO20, and propose a rationale for blocking prorenin/PRR activation as a therapeutic approach that does not affect the actions of the PRR in vacuolar H(+)-ATPase and development. Finally, we summarize categories of currently available antihypertensive drugs and consider future perspectives.
Collapse
Affiliation(s)
- Quanbin Xu
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA; Department of Physiology & Cell Biology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Dane D Jensen
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA; Department of Physiology & Cell Biology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Hua Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huangzhong University of Sciences and Technology, Wuhan, China
| | - Yumei Feng
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA; Department of Physiology & Cell Biology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA.
| |
Collapse
|
39
|
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
|
40
|
Ramkumar N, Stuart D, Mironova E, Bugay V, Wang S, Abraham N, Ichihara A, Stockand JD, Kohan DE. Renal tubular epithelial cell prorenin receptor regulates blood pressure and sodium transport. Am J Physiol Renal Physiol 2016; 311:F186-94. [PMID: 27053687 DOI: 10.1152/ajprenal.00088.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/31/2016] [Indexed: 12/31/2022] Open
Abstract
The physiological significance of the renal tubular prorenin receptor (PRR) has been difficult to elucidate due to developmental abnormalities associated with global or renal-specific PRR knockout (KO). We recently developed an inducible renal tubule-wide PRR KO using the Pax8/LC1 transgenes and demonstrated that disruption of renal tubular PRR at 1 mo of age caused no renal histological abnormalities. Here, we examined the role of renal tubular PRR in blood pressure (BP) regulation and Na(+) excretion and investigated the signaling mechanisms by which PRR regulates Na(+) balance. No detectable differences in BP were observed between control and PRR KO mice fed normal- or low-Na(+) diets. However, compared with controls, PRR KO mice had elevated plasma renin concentration and lower cumulative Na(+) balance with normal- and low-Na(+) intake. PRR KO mice had an attenuated hypertensive response and reduced Na(+) retention following angiotensin II (ANG II) infusion. Furthermore, PRR KO mice had significantly lower epithelial Na(+) channel (ENaC-α) expression. Treatment with mouse prorenin increased, while PRR antagonism decreased, ENaC activity in isolated split-open collecting ducts (CD). The prorenin effect was prevented by protein kinase A and Akt inhibition, but unaffected by blockade of AT1, ERK1/2, or p38 MAPK pathways. Taken together, these data indicate that renal tubular PRR, likely via direct prorenin/renin stimulation of PKA/Akt-dependent pathways, stimulates CD ENaC activity. Absence of renal tubular PRR promotes Na(+) wasting and reduces the hypertensive response to ANG II.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah;
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Elena Mironova
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas; and
| | - Vladislav Bugay
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas; and
| | - Shuping Wang
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Nikita Abraham
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Atsuhiro Ichihara
- Department of Medicine II, Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - James D Stockand
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas; and
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
41
|
Wang F, Lu X, Peng K, Fang H, Zhou L, Su J, Nau A, Yang KT, Ichihara A, Lu A, Zhou SF, Yang T. Antidiuretic Action of Collecting Duct (Pro)Renin Receptor Downstream of Vasopressin and PGE2 Receptor EP4. J Am Soc Nephrol 2016; 27:3022-3034. [PMID: 27000064 DOI: 10.1681/asn.2015050592] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 01/24/2016] [Indexed: 12/30/2022] Open
Abstract
Within the kidney, the (pro)renin receptor (PRR) is predominantly expressed in the collecting duct (CD), particularly in intercalated cells, and it is regulated by the PGE2 receptor EP4 Notably, EP4 also controls urinary concentration through regulation of aquaporin 2 (AQP2). Here, we tested the hypothesis that sequential activation of EP4 and PRR determines AQP2 expression in the CD, thus mediating the antidiuretic action of vasopressin (AVP). Water deprivation (WD) elevated renal PRR expression and urinary soluble PRR excretion in rats. Intrarenal infusion of a PRR decoy peptide, PRO20, or an EP4 antagonist partially prevented the decrease in urine volume and the increase in urine osmolality and AQP2 expression induced by 48-hour WD. In primary cultures of rat inner medullary CD cells, AQP2 expression induced by AVP treatment for 24 hours depended on sequential activation of the EP4 receptor and PRR. Additionally, mice lacking PRR in the CD exhibited increased urine volume and decreased urine osmolality under basal conditions and impaired urine concentrating capability accompanied by severe volume loss and a dangerous level of plasma hyperosmolality after WD. Together, these results suggest a previously undescribed linear AVP/PGE2/EP4/PRR pathway in the CD for regulation of AQP2 expression and urine concentrating capability.
Collapse
Affiliation(s)
- Fei Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Xiaohan Lu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Kexin Peng
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Hui Fang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Li Zhou
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Su
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Adam Nau
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Kevin T Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Atsuhiro Ichihara
- Department of Medicine II, Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan; and
| | - Aihua Lu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Tianxin Yang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah;
| |
Collapse
|
42
|
Lu X, Wang F, Liu M, Yang KT, Nau A, Kohan DE, Reese V, Richardson RS, Yang T. Activation of ENaC in collecting duct cells by prorenin and its receptor PRR: involvement of Nox4-derived hydrogen peroxide. Am J Physiol Renal Physiol 2015; 310:F1243-50. [PMID: 26697985 DOI: 10.1152/ajprenal.00492.2015] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/15/2015] [Indexed: 11/22/2022] Open
Abstract
The collecting duct (CD) has been recognized as an important source of prorenin/renin, and it also expresses (pro)renin receptor (PRR). The goal of this study was to examine the hypothesis that prorenin or renin via PRR regulates epithelial Na(+) channel (ENaC) activity in mpkCCD cells. Transepithelial Na(+) transport was measured by using a conventional epithelial volt-ohmmeter and was expressed as the calculated equivalent current (Ieq). Amiloride-inhibitable Ieq was used as a reflection of ENaC activity. Administration of prorenin in the nanomolar range induced a significant increase in Ieq that was detectable as early as 1 min, peaked at 5 min, and gradually returned to baseline within 15 min. These changes in Ieq were completely prevented by a newly developed PRR decoy inhibitor, PRO20. Prorenin-induced Ieq was inhibitable by amiloride. Compared with prorenin, renin was less effective in stimulating Ieq Prorenin-induced Ieq was attenuated by apocynin but enhanced by tempol, the latter effect being prevented by catalase. In response to prorenin treatment, the levels of total reactive oxygen species and H2O2 were both increased, as detected by spin-trap analysis and reactive oxygen species (ROS)-Glo H2O2 assay, respectively. Both siRNA-mediated Nox4 knockdown and the dual Nox1/4 inhibitor GKT137892 attenuated prorenin-induced Ieq Overall, our results demonstrate that activation of PRR by prorenin stimulates ENaC activity in CD cells via Nox4-derived H2O2.
Collapse
Affiliation(s)
- Xiaohan Lu
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Fei Wang
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Mi Liu
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Kevin T Yang
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and
| | - Adam Nau
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and
| | - Donald E Kohan
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and
| | - Van Reese
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and
| | - Russell S Richardson
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; and Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
43
|
Wang F, Lu X, Liu M, Feng Y, Zhou SF, Yang T. Renal medullary (pro)renin receptor contributes to angiotensin II-induced hypertension in rats via activation of the local renin-angiotensin system. BMC Med 2015; 13:278. [PMID: 26554902 PMCID: PMC4641338 DOI: 10.1186/s12916-015-0514-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/14/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND (Pro)renin receptor (PRR) is a new component of the renin-angiotensin system and regulates renin activity in vitro. Within the kidney, PRR is highly expressed in the renal medulla where its expression is induced by angiotensin II infusion. The objective of the present study was to test a potential role of renal medullary PRR during angiotensin II-induced hypertension. METHODS A rat AngII infusion model (100 ng/kg/min) combined with renal intramedullary infusion of PRO20, a specific inhibitor of PRR, was builded. And the intravenous PRO20 infusion serve as control. Mean arterial pressure was recorded by radiotelemetry for one week. Further analysis of kidney injury, inflammation, biochemical indices and protein localization were performed in vivo or in vitro. RESULTS Radiotelemetry demonstrated that AngII infusion elevated the mean arteria pressure from 108 ± 5.8 to 164.7 ± 6.2 mmHg. Mean arterial pressure decreased to 128.6 ± 5.8 mmHg (P < 0.05) after intramedullary infusion of PRO20, but was only modestly affected by intravenous PRO20 infusion. Indices of kidney injury, including proteinuria, glomerulosclerosis, and interstitial fibrosis, inflammation, and increased renal medullary and urinary renin activity following angiotensin II infusion were all remarkably attenuated by intramedullary PRO20 infusion. Following one week of angiotensin II infusion, increased PRR immunoreactivity was found in vascular smooth muscle cells. In cultured rat vascular smooth muscle cells, angiotensin II induced parallel increases in soluble PRR and renin activity, and the latter was significantly reduced by PRO20. CONCLUSION Renal medullary PRR mediates angiotensin II-induced hypertension, likely by amplifying the local renin response.
Collapse
Affiliation(s)
- Fei Wang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, #74 Zhongshan 2nd Road, Science and Technology Building, 6th Floor, Guangzhou, 510080, P. R. China.,Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Xiaohan Lu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, #74 Zhongshan 2nd Road, Science and Technology Building, 6th Floor, Guangzhou, 510080, P. R. China
| | - Mi Liu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, #74 Zhongshan 2nd Road, Science and Technology Building, 6th Floor, Guangzhou, 510080, P. R. China
| | - Yumei Feng
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NE, USA
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, #74 Zhongshan 2nd Road, Science and Technology Building, 6th Floor, Guangzhou, 510080, P. R. China. .,Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA.
| |
Collapse
|
44
|
Gonzalez AA, Prieto MC. Roles of collecting duct renin and (pro)renin receptor in hypertension: mini review. Ther Adv Cardiovasc Dis 2015; 9:191-200. [PMID: 25780059 PMCID: PMC4560657 DOI: 10.1177/1753944715574817] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In angiotensin (Ang)-II-dependent hypertension, collecting duct renin synthesis and secretion are stimulated despite suppression of juxtaglomerular (JG) renin. This effect is mediated by Ang II type 1 (AT1) receptor independent of blood pressure. Although the regulation of JG renin is known, the mechanisms by which renin is regulated in the collecting duct are not completely understood. The presence of renin activity in the collecting duct may provide a pathway for intratubular Ang II formation since angiotensinogen substrate and angiotensin converting enzyme are present in the distal nephron. The recently named new member of the renin-angiotensin system (RAS), the (pro)renin receptor [(P)RR], is able to bind renin and the inactive prorenin, thus enhancing renin activity and fully activating prorenin. We have demonstrated that renin and (P)RR are augmented in renal tissues from rats infused with Ang II and during sodium depletion, suggesting a physiological role in intrarenal RAS activation. Importantly, (P)RR activation also causes activation of intracellular pathways associated with increased cyclooxygenase 2 expression and induction of profibrotic genes. In addition, renin and (P)RR are upregulated by Ang II in collecting duct cells. Although the mechanisms involved in their regulation are still under study, they seem to be dependent on the intrarenal RAS activation. The complexities of the mechanisms of stimulation also depend on cyclooxygenase 2 and sodium depletion. Our data suggest that renin and (P)RR can interact to increase intratubular Ang II formation and the activation of profibrotic genes in renal collecting duct cells. Both pathways may have a critical role in the development of hypertension and renal disease.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Chile
| | - Minolfa C Prieto
- Department of Physiology, Rm 4061, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| |
Collapse
|
45
|
Ramkumar N, Stuart D, Calquin M, Quadri S, Wang S, Van Hoek AN, Siragy HM, Ichihara A, Kohan DE. Nephron-specific deletion of the prorenin receptor causes a urine concentration defect. Am J Physiol Renal Physiol 2015; 309:F48-56. [PMID: 25995108 DOI: 10.1152/ajprenal.00126.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/11/2015] [Indexed: 01/27/2023] Open
Abstract
The prorenin receptor (PRR), a recently discovered component of the renin-angiotensin system, is expressed in the nephron in general and the collecting duct in particular. However, the physiological significance of nephron PRR remains unclear, partly due to developmental abnormalities associated with global or renal-specific PRR gene knockout (KO). Therefore, we developed mice with inducible nephron-wide PRR deletion using Pax8-reverse tetracycline transactivator and LC-1 transgenes and loxP flanked PRR alleles such that ablation of PRR occurs in adulthood, after induction with doxycycline. Nephron-specific PRR KO mice have normal survival to ∼1 yr of age and no renal histological defects. Compared with control mice, PRR KO mice had 65% lower medullary PRR mRNA and protein levels and markedly diminished renal PRR immunofluorescence. During both normal water intake and mild water restriction, PRR KO mice had significantly lower urine osmolality, higher water intake, and higher urine volume compared with control mice. No differences were seen in urine vasopressin excretion, urine Na(+) and K(+) excretion, plasma Na(+), or plasma osmolality between the two groups. However, PRR KO mice had reduced medullary aquaporin-2 levels and arginine vasopressin-stimulated cAMP accumulation in the isolated renal medulla compared with control mice. Taken together, these results suggest nephron PRR can potentially modulate renal water excretion.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah;
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Matias Calquin
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Syed Quadri
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; and
| | - Shuping Wang
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Alfred N Van Hoek
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Helmy M Siragy
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; and
| | - Atsuhiro Ichihara
- Department of Medicine II, Endocrinology and Hypertension, Tokyo Women's Medical University, Tokyo, Japan
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
46
|
Lv LL, Liu BC. Role of non-classical renin-angiotensin system axis in renal fibrosis. Front Physiol 2015; 6:117. [PMID: 25954204 PMCID: PMC4404823 DOI: 10.3389/fphys.2015.00117] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/27/2015] [Indexed: 12/15/2022] Open
Abstract
The renin–angiotensin system (RAS) is a major regulator of renal fibrosis. Besides the classical renin/Angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/AT1 and AT2 axis, multiple new axes have been recently described. The new members have added new dimensions to RAS, including the ACE2/Ang(1–7)/Mas receptor axis, the prorenin/(pro)renin receptor(PRR)/intracelluar pathway axis, and the Angiotensin A (Ang A), alamandine-Mas-related G protein coupled receptor D(MrgD) axis. This review summarized recent studies regarding role of the non-classical RAS axis in renal fibrosis, and its possible implications to the intervention of progression of chronic kidney disease.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Department of Affiliated Zhongda Hospital, Southeast University Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Department of Affiliated Zhongda Hospital, Southeast University Nanjing, China
| |
Collapse
|
47
|
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: 1.9] [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
|
48
|
Gonzalez AA, Green T, Luffman C, Bourgeois CRT, Gabriel Navar L, Prieto MC. Renal medullary cyclooxygenase-2 and (pro)renin receptor expression during angiotensin II-dependent hypertension. Am J Physiol Renal Physiol 2014; 307:F962-70. [PMID: 25143455 DOI: 10.1152/ajprenal.00267.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The (pro)renin receptor [(P)RR] upregulates cyclooxygenase-2 (COX-2) in inner medullary collecting duct (IMCD) cells through ERK1/2. Intrarenal COX-2 and (P)RR are upregulated during chronic ANG II infusion. However, the duration of COX-2 and (P)RR upregulation has not been determined. We hypothesized that during the early phase of ANG II-dependent hypertension, membrane-bound (P)RR and COX-2 are augmented in the renal medulla, serving to buffer the hypertensinogenic and vasoconstricting effects of ANG II. In Sprague-Dawley rats infused with ANG II (0.4 μg·min(-1)·kg(-1)), systolic blood pressure (BP) increased by day 7 (162 ± 5 vs. 114 ± 10 mmHg) and continued to increase by day 14 (198 ± 15 vs. 115 ± 13 mmHg). Membrane-bound (P)RR was augmented at day 3 coincident with phospho-ERK1/2 levels, COX-2 expression, and PGE2 in the renal medulla. In contrast, membrane-bound (P)RR was reduced and COX-2 protein levels were not different from controls by day 14. In cultured IMCD cells, ANG II increased secretion of the soluble (P)RR. In anesthetized rats, COX-2 inhibition decreased the glomerular filtration rate (GFR) and renal blood flow (RBF) during the early phase of ANG II infusion without altering BP. However, at 14 days of ANG II infusions, COX-2 inhibition decreased mean arterial BP (MABP), RBF, and GFR. Thus, during the early phase of ANG II-dependent hypertension, the increased (P)RR and COX-2 expression in the renal medulla may contribute to attenuate the vasoconstrictor effects of ANG II on renal hemodynamics. In contrast, at 14 days the reductions in RBF and GFR caused by COX-2 inhibition paralleled the reduced MABP, suggesting that vasoconstrictor COX-2 metabolites contribute to ANG II hypertension.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile; and
| | - Torrance Green
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Christina Luffman
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Camille R T Bourgeois
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Minolfa C Prieto
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
49
|
Quadri S, Siragy HM. Regulation of (pro)renin receptor expression in mIMCD via the GSK-3β-NFAT5-SIRT-1 signaling pathway. Am J Physiol Renal Physiol 2014; 307:F593-600. [PMID: 24990896 DOI: 10.1152/ajprenal.00245.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The localization and regulation of (pro)renin receptor (PRR) expression in kidney collecting duct cells are not well established. We hypothesized that low salt (LS) contributes to the regulation of PRR expression in these cells via the GSK-3β-NFAT5-sirtuin1 (SIRT-1) signaling pathway. Mouse inner medullary collecting duct (mIMCD) cells were treated with NaCl at 130 (normal salt; NS), 63 (LS), or 209 mM (high salt; HS) alone or in combination with NFAT5 scrambled small interfering (si) RNA, NFAT5 siRNA, or the SIRT-1 inhibitor EX-527. Compared with NS, LS increased the mRNA and protein expression of PRR by 71% and 69% (P < 0.05), and reduced phosphorylation of GSK-3β by 62% (P < 0.01), mRNA and protein expressions of NFAT5 by 65% and 45% (P < 0.05), and SIRT-1 by 44% and 50% (P < 0.01), respectively. LS also enhanced p65 NF-κB by 102% (P < 0.01). Treatment with HS significantly reduced the mRNA and protein expression of PRR by 32% and 23% (P < 0.05), and increased the mRNA and protein expression of NFAT5 by 39% and 45% (P < 0.05) and SIRT-1 by 51% and 56% (P < 0.05), respectively. HS+NFAT5 siRNA reduced the mRNA and protein expression of NFAT5 by 51% and 35% (P < 0.01) and increased the mRNA and protein expression of PRR by 148% and 70% (P < 0.01), respectively. HS+EX-527 significantly increased the mRNA and protein expression of PRR by 96% and 58% (P < 0.05), respectively. We conclude that expression of PRR in mIMCD cells is regulated by the GSK-3β-NFAT5- SIRT-1 signaling pathway.
Collapse
Affiliation(s)
- Syed Quadri
- Division of Endocrinology and Metabolism, University of Virginia Health System, Charlottesville, Virginia
| | - Helmy M Siragy
- Division of Endocrinology and Metabolism, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|