1
|
Cruz-López EO, Tan L, Stolk DG, van den Bogaerdt AJ, Verdonk K, Danser AHJ. Endothelin-1- and acetylcholine-mediated effects in human and rat vessels: impact of perivascular adipose tissue, diabetes, angiotensin II, and chemerin. Blood Press 2024; 33:2414072. [PMID: 39387176 DOI: 10.1080/08037051.2024.2414072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
OBJECTIVE To study the role of perivascular adipose tissue (PVAT) in the reactivity of rat and human vessels. METHODS Iliac and mesenteric arteries were obtained from normotensive Sprague-Dawley rats, hypertensive transgenic (mRen2)27 rats overexpressing mouse renin, and (mRen2)27 rats made diabetic with streptozotocin. Human coronary arteries were obtained from donors. Concentration-response curves were constructed to endothelin-1 and acetylcholine with and without PVAT. The contribution of NO and endothelium-dependent hyperpolarization (EDH) were determined making use of the NO synthase inhibitor L-NAME and the EDH inhibitors apamin + TRAM-34. The endothelin type A and type B (ETA, ETB) receptor blockers BQ123 and BQ788, the chemerin inhibitors α-NETA and pravastatin, and the angiotensin receptor blocker losartan were also used. RESULTS In rat iliac arteries, PVAT diminished endothelin-induced constriction, while the opposite was true in human coronaries. Coronary effects were unaltered by α-NETA, pravastatin, or losartan. ETB receptor-mediated relaxation in iliac arteries occurred only with PVAT, and BQ123 blocked endothelin-1-induced constriction. Diabetes upregulated the anticontractile effects of PVAT. In rat mesenteric arteries, acetylcholine-induced relaxation with PVAT relied on NO, and on NO + EDH without PVAT. Diabetes upregulated the EDH component exclusively with PVAT. CONCLUSION PVAT modulates ET-1-induced constriction in a vessel type-dependent manner. Its enhancing effects in coronaries involved neither chemerin nor angiotensin II. Its anticontractile effects in rat iliac arteries involved ETB receptor-mediated relaxation. Diabetes upregulated PVAT's anticontractile effects. In mesenteric arteries, PVAT counterbalanced the EDH component of the relaxant effect of acetylcholine. Diabetes reversed this effect by upregulating the EDH component.
Collapse
Affiliation(s)
- Edwyn O Cruz-López
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lunbo Tan
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniël G Stolk
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Koen Verdonk
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Mishima S, Mitsui T, Tani K, Ooba H, Mitoma T, Ohira A, Maki J, Kirino S, Eto E, Hayata K, Masuyama H. Endothelin-1 production via placental (pro)renin receptor in a mouse model of preeclampsia. Placenta 2023; 138:44-50. [PMID: 37167782 DOI: 10.1016/j.placenta.2023.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
INTRODUCTION Preeclampsia (PE) pathogenesis is explained by the two-stage disorder theory. However, mechanisms underlying hypertension and proteinuria in PE remain unclear. The role of (pro)renin receptor (PRR) in PE pathology has received special attention. We examined endothelin-1 (ET-1) production via placental PRR in a PE mouse model. METHODS At 14.5 day-post-coitum (DPC), we performed a reduced uterine perfusion pressure (RUPP) operation, ligating the uterine arteriovenous vessels in female mice. We also infused these mice with a PRR inhibitor, decoy peptide in the handle region of prorenin (HRP) for mice (NH2-RIPLKKMPSV-COOH). At 18.5 DPC, blood, urine, and placenta were collected; fetus and placenta were weighed. We evaluated placental hypoxia using quantitative polymerase chain reaction (PCR), with hypoxia-inducible factor-1α (HIF-1α) as index. We also evaluated PRR, transforming growth factor-β1 (TGF-β1), and ET-1 expression in the placenta using quantitative PCR and western blotting. ET-1 concentration in blood plasma was assessed using enzyme-linked immunosorbent assay. RESULTS Blood pressure and proteinuria significantly increased, and fetal and placental weights decreased in RUPP mice. HIF-1α, PRR, TGF-β1, and ET-1 expressions considerably increased in RUPP mice placentas. ET-1 concentration in RUPP mice blood plasma was markedly increased. PRR inhibitor suppressed these changes. DISCUSSION In PE model mice that underwent RUPP treatment, placental hypoxia increased PRR and ET-1 expression suggesting a causative relationship between ET-1 and intracellular PRR signaling. RUPP treatment, when combined with HRP, reversed the effect of elevated ET-1 levels in the model. This study may help to elucidate the pathogenesis of PE considering PRR and ET-1.
Collapse
Affiliation(s)
- Sakurako Mishima
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Takashi Mitsui
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Kazumasa Tani
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Hikaru Ooba
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Tomohiro Mitoma
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Akiko Ohira
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Jota Maki
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Satoe Kirino
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Eriko Eto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Kei Hayata
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata, Okayama, 700-8558, Japan.
| |
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
|
Cruz-López EO, Ren L, Uijl E, Clahsen-van Groningen MC, van Veghel R, Garrelds IM, Domenig O, Poglitsch M, Zlatev I, Rooney T, Kasper A, Nioi P, Foster D, Danser AHJ. Blood pressure-independent renoprotective effects of small interference RNA targeting liver angiotensinogen in experimental diabetes. Br J Pharmacol 2023; 180:80-93. [PMID: 36106615 PMCID: PMC10091936 DOI: 10.1111/bph.15955] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Small interfering RNA (siRNA) targeting liver angiotensinogen lowers blood pressure, but its effects in hypertensive diabetes are unknown. EXPERIMENTAL APPROACH To address this, TGR (mRen2)27 rats (angiotensin II-dependent hypertension model) were made diabetic with streptozotocin over 18 weeks and treated with either vehicle, angiotensinogen siRNA, the AT1 antagonist valsartan, the ACE inhibitor captopril, valsartan + siRNA or valsartan + captopril for the final 3 weeks. Mean arterial pressure (MAP) was measured via radiotelemetry. KEY RESULTS MAP before treatment was 153 ± 2 mmHg. Diabetes resulted in albuminuria, accompanied by glomerulosclerosis and podocyte effacement, without a change in glomerular filtration rate. All treatments lowered MAP and cardiac hypertrophy, and the largest drop in MAP was observed with siRNA + valsartan. Treatment with siRNA lowered circulating angiotensinogen by >99%, and the lowest circulating angiotensin II and aldosterone levels occurred in the dual treatment groups. Angiotensinogen siRNA did not affect renal angiotensinogen mRNA expression, confirming its liver-specificity. Furthermore, only siRNA with or without valsartan lowered renal angiotensin I. All treatments lowered renal angiotensin II and the reduction was largest (>95%) in the siRNA + valsartan group. All treatments identically lowered albuminuria, whereas only siRNA with or without valsartan restored podocyte foot processes and reduced glomerulosclerosis. CONCLUSION AND IMPLICATIONS Angiotensinogen siRNA exerts renoprotection in diabetic TGR (mRen2)27 rats and this relies, at least in part, on the suppression of renal angiotensin II formation from liver-derived angiotensinogen. Clinical trials should now address whether this is also beneficial in human diabetic kidney disease.
Collapse
Affiliation(s)
- Edwyn O Cruz-López
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Liwei Ren
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.,Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Estrellita Uijl
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.,Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marian C Clahsen-van Groningen
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.,Institute of Experimental Medicine and Systems Biology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Richard van Veghel
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Ivan Zlatev
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | - Anne Kasper
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Paul Nioi
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Don Foster
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
5
|
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
|
6
|
Prieto MC, Gonzalez AA, Visniauskas B, Navar LG. The evolving complexity of the collecting duct renin-angiotensin system in hypertension. Nat Rev Nephrol 2021; 17:481-492. [PMID: 33824491 PMCID: PMC8443079 DOI: 10.1038/s41581-021-00414-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
The intrarenal renin-angiotensin system is critical for the regulation of tubule sodium reabsorption, renal haemodynamics and blood pressure. The excretion of renin in urine can result from its increased filtration, the inhibition of renin reabsorption by megalin in the proximal tubule, or its secretion by the principal cells of the collecting duct. Modest increases in circulating or intrarenal angiotensin II (ANGII) stimulate the synthesis and secretion of angiotensinogen in the proximal tubule, which provides sufficient substrate for collecting duct-derived renin to form angiotensin I (ANGI). In models of ANGII-dependent hypertension, ANGII suppresses plasma renin, suggesting that urinary renin is not likely to be the result of increased filtered load. In the collecting duct, ANGII stimulates the synthesis and secretion of prorenin and renin through the activation of ANGII type 1 receptor (AT1R) expressed primarily by principal cells. The stimulation of collecting duct-derived renin is enhanced by paracrine factors including vasopressin, prostaglandin E2 and bradykinin. Furthermore, binding of prorenin and renin to the prorenin receptor in the collecting duct evokes a number of responses, including the non-proteolytic enzymatic activation of prorenin to produce ANGI from proximal tubule-derived angiotensinogen, which is then converted into ANGII by luminal angiotensin-converting enzyme; stimulation of the epithelial sodium channel (ENaC) in principal cells; and activation of intracellular pathways linked to the upregulation of cyclooxygenase 2 and profibrotic genes. These findings suggest that dysregulation of the renin-angiotensin system in the collecting duct contributes to the development of hypertension by enhancing sodium reabsorption and the progression of kidney injury.
Collapse
Affiliation(s)
- Minolfa C. Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA.,
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaiso, Chile
| | - Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - L. Gabriel Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
7
|
Martens N, Schepers M, Zhan N, Leijten F, Voortman G, Tiane A, Rombaut B, Poisquet J, Sande NVD, Kerksiek A, Kuipers F, Jonker JW, Liu H, Lütjohann D, Vanmierlo T, Mulder MT. 24(S)-Saringosterol Prevents Cognitive Decline in a Mouse Model for Alzheimer's Disease. Mar Drugs 2021; 19:190. [PMID: 33801706 PMCID: PMC8065937 DOI: 10.3390/md19040190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
We recently found that dietary supplementation with the seaweed Sargassum fusiforme, containing the preferential LXRβ-agonist 24(S)-saringosterol, prevented memory decline and reduced amyloid-β (Aβ) deposition in an Alzheimer's disease (AD) mouse model without inducing hepatic steatosis. Here, we examined the effects of 24(S)-saringosterol as a food additive on cognition and neuropathology in AD mice. Six-month-old male APPswePS1ΔE9 mice and wildtype C57BL/6J littermates received 24(S)-saringosterol (0.5 mg/25 g body weight/day) (APPswePS1ΔE9 n = 20; C57BL/6J n = 19) or vehicle (APPswePS1ΔE9 n = 17; C57BL/6J n = 19) for 10 weeks. Cognition was assessed using object recognition and object location tasks. Sterols were analyzed by gas chromatography/mass spectrometry, Aβ and inflammatory markers by immunohistochemistry, and gene expression by quantitative real-time PCR. Hepatic lipids were quantified after Oil-Red-O staining. Administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice without affecting the Aβ plaque load. Moreover, 24(S)-saringosterol prevented the increase in the inflammatory marker Iba1 in the cortex of APPswePS1ΔE9 mice (p < 0.001). Furthermore, 24(S)-saringosterol did not affect the expression of lipid metabolism-related LXR-response genes in the hippocampus nor the hepatic neutral lipid content. Thus, administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice independent of effects on Aβ load and without adverse effects on liver fat content. The anti-inflammatory effects of 24(S)-saringosterol may contribute to the prevention of cognitive decline.
Collapse
Affiliation(s)
- Nikita Martens
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Na Zhan
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Frank Leijten
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Gardi Voortman
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Janne Poisquet
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Nienke van de Sande
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Folkert Kuipers
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Tim Vanmierlo
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
8
|
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
|
9
|
Hitzerd E, Neuman RI, Broekhuizen M, Simons SHP, Schoenmakers S, Reiss IKM, Koch BCP, van den Meiracker AH, Versmissen J, Visser W, Danser AHJ. Transfer and Vascular Effect of Endothelin Receptor Antagonists in the Human Placenta. Hypertension 2019; 75:877-884. [PMID: 31884859 DOI: 10.1161/hypertensionaha.119.14183] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Increasing evidence suggests a role for the ET (endothelin) system in preeclampsia. Hence, blocking this system with endothelin receptor antagonists (ERAs) could be a therapeutic strategy. Yet, clinical studies are lacking due to possible teratogenic effects of ERAs. In this study, we investigated the placental transfer of ERAs and their effect on ET-1-mediated vasoconstriction. Term placentas were dually perfused with the selective ETAR (ET type A receptor) antagonists sitaxentan and ambrisentan or the nonselective ETAR/ETBR antagonist macitentan and subsequently exposed to ET-1 in the fetal circulation. ET-1 concentration-response curves after incubation with sitaxentan, ambrisentan, macitentan, or the selective ETBR antagonist BQ-788 were also constructed in isolated chorionic plate arteries using wire-myography, and gene expression of the ET-system was quantified in healthy and early onset preeclamptic placentas. At steady state, the mean fetal-to-maternal transfer ratios were 0.32±0.05 for sitaxentan, 0.21±0.02 for ambrisentan, and 0.05±0.01 for macitentan. Except for BQ-788, all ERAs lowered the response to ET-1, both in the perfused cotyledon and isolated chorionic plate arteries. Placental gene expression of ECE-1, ETAR, and ETBR were comparable in healthy and preeclamptic placentas, while ET-1 expression was higher in preeclampsia. Our study is the first to show direct transfer of ERAs across the term human placenta. Furthermore, ETAR exclusively mediates ET-1-induced constriction in the fetoplacental vasculature. Given its limited transfer, macitentan could be considered as potential preeclampsia therapy. Extending knowledge on placental transfer to placentas of preeclamptic pregnancies is required to determine whether ERAs might be applied safely in preeclampsia.
Collapse
Affiliation(s)
- Emilie Hitzerd
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Pediatrics, division of Neonatology (E.H., M.B., S.H.P.S., I.K.M.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Rugina I Neuman
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Obstetrics and Gynecology, Division of Obstetrics and Prenatal Medicine (R.I.N., S.S., W.V.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Michelle Broekhuizen
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Pediatrics, division of Neonatology (E.H., M.B., S.H.P.S., I.K.M.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Department of Cardiology, Division of Experimental Cardiology (M.B.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Sinno H P Simons
- Department of Pediatrics, division of Neonatology (E.H., M.B., S.H.P.S., I.K.M.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynecology, Division of Obstetrics and Prenatal Medicine (R.I.N., S.S., W.V.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, division of Neonatology (E.H., M.B., S.H.P.S., I.K.M.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Birgit C P Koch
- Department of Pharmacy (B.C.P.K.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Anton H van den Meiracker
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jorie Versmissen
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Willy Visser
- Department of Obstetrics and Gynecology, Division of Obstetrics and Prenatal Medicine (R.I.N., S.S., W.V.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - A H Jan Danser
- From the Department of Internal Medicine, Division of Pharmacology and Vascular Medicine (E.H., R.I.N., M.B., A.H.v.d.M., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Ramkumar N, Kohan DE. The (pro)renin receptor: an emerging player in hypertension and metabolic syndrome. Kidney Int 2019; 95:1041-1052. [PMID: 30819554 DOI: 10.1016/j.kint.2018.10.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022]
Abstract
The (pro)renin receptor (PRR) is a multifunctional protein that is expressed in multiple organs. Binding of prorenin/renin to the PRR activates angiotensin II-dependent and angiotensin II-independent pathways. The PRR is also involved in autophagy and Wnt/ß catenin signaling, functions that are not contingent on prorenin binding. Emerging evidence suggests that the PRR plays an important role in blood pressure regulation and glucose and lipid metabolism. Herein, we review PRR function in health and disease, with particular emphasis on hypertension and the metabolic syndrome.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA; Salt Lake Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Moes AD, Severs D, Verdonk K, van der Lubbe N, Zietse R, Danser AHJ, Hoorn EJ. Mycophenolate Mofetil Attenuates DOCA-Salt Hypertension: Effects on Vascular Tone. Front Physiol 2018; 9:578. [PMID: 29867591 PMCID: PMC5968119 DOI: 10.3389/fphys.2018.00578] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/01/2018] [Indexed: 12/28/2022] Open
Abstract
Inflammation is increasingly recognized as a driver of hypertension. Both genetic and pharmacological inhibition of B and T cells attenuates most forms of experimental hypertension. Accordingly, the immunosuppressive drug mycophenolate mofetil (MMF) reduces blood pressure in the deoxycorticosterone acetate (DOCA-) salt model. However, the mechanisms by which MMF prevent hypertension in the DOCA-salt model remain unclear. Recent studies indicate that immunosuppression can inhibit sodium transporter activity in the kidney, but its effect on vascular tone is not well characterized. Therefore, the aim of the present study was to analyze the vascular and renal tubular effects of MMF in the DOCA-salt model in rats (4 weeks without uninephrectomy). Co-treatment with MMF attenuated the rise in blood pressure from day 11 onward resulting in a significantly lower telemetric mean arterial pressure after 4 weeks of treatment (108 ± 7 vs. 130 ± 9 mmHg, P < 0.001 by two-way analysis of variance). MMF significantly reduced the number of CD3+ cells in kidney cortex and inner medulla, but not in outer medulla. In addition, MMF significantly reduced urinary interferon-γ excretion. Vascular tone was studied ex vivo using wire myographs. An angiotensin II type 2 (AT2) receptor antagonist blocked the effects of angiotensin II (Ang II) only in the vehicle group. Conversely, L-NAME significantly increased the Ang II response only in the MMF group. An endothelin A receptor blocker prevented vasoconstriction by endothelin-1 in the MMF but not in the vehicle group. MMF did not reduce the abundances of the kidney sodium transporters NHE3, NKCC2, NCC, or ENaC. Together, our ex vivo results suggest that DOCA-salt induces AT2 receptor-mediated vasoconstriction. MMF prevents this response and increases nitric oxide availability. These data provide insight in the antihypertensive mechanism of MMF and the role of inflammation in dysregulating vascular tone.
Collapse
Affiliation(s)
- Arthur D Moes
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - David Severs
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Koen Verdonk
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Nils van der Lubbe
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - A H J Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
12
|
Roksnoer LCW, Uijl E, de Vries R, Garrelds IM, Jan Danser AH. Neprilysin inhibition and endothelin-1 elevation: Focus on the kidney. Eur J Pharmacol 2018; 824:128-132. [PMID: 29432709 DOI: 10.1016/j.ejphar.2018.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/23/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
Increasing the degree of renin-angiotensin system (RAS) blockade by combining ≥2 RAS blockers marginally increases efficacy, but results in more side effects. Hence, interference with other systems is currently being investigated, like potentiation of natriuretic peptides with neprilysin inhibitors. However, the neprilysin inhibitor thiorphan was recently found to increase endothelin-1 when administered to TGR(mREN2)27 (Ren2) rats on top of RAS blockade. Here we investigated whether this effect is thiorphan-specific, by comparing the neprilysin inhibitors thiorphan and sacubitril, administered by osmotic minipumps at a low or high dose for 7 days, in Ren2 rats. Plasma and urinary levels of endothelin-1, atrial and brain natriuretic peptide (ANP, BNP) and their second messenger cyclic guanosine 3'5' monophosphate (cGMP) were monitored. No significant differences were found in the plasma concentrations of endothelin-1, cGMP, ANP and BNP after treatment, although plasma ANP tended to be higher in the high-dose thiorphan treatment group and the low- and high-dose sacubitril treatment groups, compared with vehicle. Urinary endothelin-1 increased in the low-dose thiorphan and high-dose sacubitril groups, compared with baseline, although significance was reached for the former only. Urinary cGMP rose significantly in the high-dose sacubitril treatment group compared with baseline. Both urinary endothelin-1 and cGMP were significantly higher in the high-dose sacubitril group compared with the low-dose sacubitril group. In conclusion, endothelin-1 upregulation occurs with both thiorphan and sacubitril, and is particularly apparent in neprilysin-rich organs like the kidney. High renal neprilysin levels most likely also explain why sacubitril increased cGMP in urine only.
Collapse
Affiliation(s)
- Lodi C W Roksnoer
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Estrellita Uijl
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - René de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Hennrikus M, Gonzalez AA, Prieto MC. The prorenin receptor in the cardiovascular system and beyond. Am J Physiol Heart Circ Physiol 2018; 314:H139-H145. [PMID: 29101170 PMCID: PMC5867650 DOI: 10.1152/ajpheart.00373.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 01/24/2023]
Abstract
Since the prorenin receptor (PRR) was first reported, its physiological role in many cellular processes has been under intense scrutiny. The PRR is currently recognized as a multifunctional receptor with major roles as an accessory protein of the vacuolar-type H+-ATPase and as an intermediary in the Wnt signaling pathway. As a member of the renin-angiotensin system (RAS), the PRR has demonstrated to be of relevance in cardiovascular diseases (CVD) because it can activate prorenin and enhance the enzymatic activity of renin, thus promoting angiotensin II formation. Indeed, there is an association between PRR gene polymorphisms and CVD. Independent of angiotensin II, the activation of the PRR further stimulates intracellular signals linked to fibrosis. Studies using tissues and cells from a variety of organs and systems have supported its roles in multiple functions, although some remain controversial. In the brain, the PRR appears to be involved in the central regulation of blood pressure via activation of RAS- and non-RAS-dependent mechanisms. In the heart, the PRR promotes atrial structural and electrical remodeling. Nonetheless, animals overexpressing the PRR do not exhibit cardiac injury. In the kidney, the PRR is involved in the development of ureteric bud branching, urine concentration, and regulation of blood pressure. There is great interest in the PRR contributions to T cell homeostasis and to the development of visceral and brown fat. In this mini-review, we discuss the evidence for the pathophysiological roles of the PRR with emphasis in CVD.
Collapse
Affiliation(s)
- Matthew Hennrikus
- Department of Physiology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine , New Orleans, Louisiana
- Tulane University Renal and Hypertension Center of Excellence , New Orleans, Louisiana
| |
Collapse
|
14
|
|
15
|
Sun Y, Danser AHJ, Lu X. (Pro)renin receptor as a therapeutic target for the treatment of cardiovascular diseases? Pharmacol Res 2017; 125:48-56. [PMID: 28532817 DOI: 10.1016/j.phrs.2017.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 02/08/2023]
Abstract
The discovery of the (pro)renin receptor [(P)RR] 15years ago stimulated ideas on prorenin being more than renin's inactive precursor. Indeed, binding of prorenin to the (P)RR induces a conformational change in the prorenin molecule, allowing it to display angiotensin-generating activity, and additionally results in intracellular signaling in an angiotensin-independent manner. However, the prorenin levels required to observe these angiotensin-dependent and -independent effects of the (P)RR are many orders above its in vivo concentrations, both under normal and pathological conditions. Given this requirement, the idea that the (P)RR has a function within the renin-angiotensin system (RAS) is now being abandoned. Instead, research is now focused on the (P)RR as an accessory protein of vacuolar H+-ATPase (V-ATPase), potentially determining its integrity. Acting as an adaptor between Frizzled co-receptor LRP6 and V-ATPase, the (P)RR appears to be indispensable for Wnt/β-catenin signaling, thus explaining why (P)RR deletion (unlike renin deletion) is lethal even when restricted to specific cells, such as cardiomyocytes, podocytes and smooth muscle cells. Furthermore, recent studies suggest that the (P)RR may play important roles in lipoprotein metabolism and overall energy metabolism. In this review, we summarize the controversial RAS-related effects of the (P)RR, and critically review the novel non-RAS-related functions of the (P)RR, ending with a discussion on the potential of targeting the (P)RR to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Yuan Sun
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China; Erasmus Medical Center, Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Rotterdam, The Netherlands
| | - A H Jan Danser
- Erasmus Medical Center, Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Rotterdam, The Netherlands
| | - Xifeng Lu
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
16
|
Ramkumar N, Kohan DE. The nephron (pro)renin receptor: function and significance. Am J Physiol Renal Physiol 2016; 311:F1145-F1148. [DOI: 10.1152/ajprenal.00476.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/21/2016] [Indexed: 12/16/2022] Open
Abstract
The (pro)renin receptor (PRR) is a multifunctional protein that is part of the renin-angiotensin system and is an important accessory molecule for the vacuolar H+-ATPase. The PRR is widely expressed in the kidney with relatively high abundance in the distal nephron. Determining the physiological relevance of the PRR has been challenging due to early lethality in whole animal and cell-specific PRR knockout models. Recently, viable renal cell-specific PRR knockout mice have been developed; these studies suggest that PRR in the nephron can modulate renal function via angiotensin II (ANG II)-dependent and -independent cell signaling pathways. In this mini-review, we highlight new developments in nephron PRR function in health and in pathophysiological conditions.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Donald E. Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
- Salt Lake Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
17
|
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
|
18
|
Zhu T, Miller AG, Deliyanti D, Berka DR, Agrotis A, Campbell DJ, Wilkinson-Berka JL. Prorenin stimulates a pro-angiogenic and pro-inflammatory response in retinal endothelial cells and an M1 phenotype in retinal microglia. Clin Exp Pharmacol Physiol 2016; 42:537-48. [PMID: 25707593 DOI: 10.1111/1440-1681.12376] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 01/29/2015] [Accepted: 02/12/2015] [Indexed: 01/25/2023]
Abstract
Angiogenesis and inflammation are causative factors in the development of neovascular retinopathies. These processes involve the retinal endothelium and the retinal immune cells, microglia. The renin-angiotensin system contributes to retinal injury via the actions of the type 1 angiotensin receptor (AT1R). However, it has been suggested that prorenin, the initiator of the renin-angiotensin system cascade, influences retinal injury independently from the AT1R. We evaluated whether prorenin induced a pro-angiogenic and pro-inflammatory response in retinal endothelial cells and a pro-inflammatory phenotype in retinal microglia. Primary cultures of retinal endothelial cells and microglia were studied. Rat recombinant prorenin (2 nmol/L) stimulated the proliferation and tubulogenesis of retinal endothelial cells; it increased the levels of pro-angiogenic factors, vascular endothelial growth factor, angiopoietin-1, and tyrosine kinase with immunoglobulin and epidermal growth factor homology domains, and pro-inflammatory factors, intercellular adhesion molecule-1 and monocyte chemoattractant protein-1, relative to the controls. The messenger RNA levels of the (pro)renin receptor were also increased. These effects occurred in the presence of the AT1R blocker candesartan (10 μmol/L) and the renin inhibitor aliskiren (10 μmol/L). Microglia, which express the (pro)renin receptor, elicited an activated phenotype when exposed to prorenin, which was characterized by increased levels of intercellular adhesion molecule-1, monocyte chemoattractant protein-1, tumour necrosis factor-α, interleukin-6, and interleukin-1β and by decreased levels of interleukin-10 and arginase-1 relative to controls. Candesartan did not influence the effects of prorenin on retinal microglia. In conclusion, prorenin has distinct pro-angiogenic and pro-inflammatory effects on retinal cells that are independent of the AT1R, indicating the potential importance of prorenin in retinopathy.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Immunology and Pathology, Monash University, Melbourne, Vic., Australia
| | | | | | | | | | | | | |
Collapse
|
19
|
Danser AHJ. The Role of the (Pro)renin Receptor in Hypertensive Disease. Am J Hypertens 2015; 28:1187-96. [PMID: 25890829 DOI: 10.1093/ajh/hpv045] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/15/2015] [Indexed: 12/16/2022] Open
Abstract
Tissue angiotensin generation depends on the uptake of circulating (kidney-derived) renin and/or its precursor prorenin (together denoted as (pro)renin). Since tissue renin levels are usually higher than expected based upon the amount of (renin-containing) blood in tissue, an active uptake mechanism has been proposed. The (pro)renin receptor ((P)RR), discovered in 2002, appeared a promising candidate, although its nanomolar affinity for renin/prorenin is many orders of magnitude above their levels in blood. This review discusses (P)RR-related research since its discovery. First, encouraging in vitro findings supported detrimental effects of (pro)renin-(P)RR interaction, even resulting in angiotensin-independent signaling. Moreover, the putative (P)RR blocker "handle region peptide" (HRP) yielded beneficial effects in various cardiovascular animal models. Then doubt arose whether such interaction truly occurs in vivo, and (P)RR deletion unexpectedly turned out to be lethal. Moreover, HRP results could not be confirmed. Finally, it was discovered that the (P)RR actually is a component of vacuolar-type H(+)-ATPase, a multisubunit protein found in virtually every cell type which is essential for vesicle trafficking, protein degradation, and coupled transport. Nevertheless, selective (P)RR blockade in the brain with the putative antagonist PRO20 (corresponding with the first 20 amino acids of prorenin's prosegment) reduced blood pressure in the deoxycorticosteroneacetate (DOCA)-salt model, and (P)RR gene single nucleotide polymorphisms associate with hypertension. To what degree this relates to (pro)renin remains uncertain. The concept of (P)RR blockade in hypertension, if pursued, requires rigorous testing of any newly designed antagonist, and may not hold promise given the early death of tissue-specific (P)RR knockout animals.
Collapse
Affiliation(s)
- A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Feng Y. ANG II-independent prorenin/(pro)renin receptor signaling pathways in the central nervous system. Am J Physiol Heart Circ Physiol 2015. [PMID: 26209058 DOI: 10.1152/ajpheart.00526.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yumei Feng
- Departments of Pharmacology, and Physiology and Cell Biology, Cardiovascular Research Center, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
21
|
Prokop JW, Petri V, Shimoyama ME, Watanabe IKM, Casarini DE, Leeper TC, Bilinovich SM, Jacob HJ, Santos RAS, Martins AS, Araujo FC, Reis FM, Milsted A. Structural libraries of protein models for multiple species to understand evolution of the renin-angiotensin system. Gen Comp Endocrinol 2015; 215:106-16. [PMID: 25260253 PMCID: PMC4375088 DOI: 10.1016/j.ygcen.2014.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/04/2014] [Accepted: 09/16/2014] [Indexed: 11/28/2022]
Abstract
The details of protein pathways at a structural level provides a bridge between genetics/molecular biology and physiology. The renin-angiotensin system is involved in many physiological pathways with informative structural details in multiple components. Few studies have been performed assessing structural knowledge across the system. This assessment allows use of bioinformatics tools to fill in missing structural voids. In this paper we detail known structures of the renin-angiotensin system and use computational approaches to estimate and model components that do not have their protein structures defined. With the subsequent large library of protein structures, we then created a species specific protein library for human, mouse, rat, bovine, zebrafish, and chicken for the system. The rat structural system allowed for rapid screening of genetic variants from 51 commonly used rat strains, identifying amino acid variants in angiotensinogen, ACE2, and AT1b that are in contact positions with other macromolecules. We believe the structural map will be of value for other researchers to understand their experimental data in the context of an environment for multiple proteins, providing pdb files of proteins for the renin-angiotensin system in six species. With detailed structural descriptions of each protein, it is easier to assess a species for use in translating human diseases with animal models. Additionally, as whole genome sequencing continues to decrease in cost, tools such as molecular modeling will gain use as an initial step in designing efficient hypothesis driven research, addressing potential functional outcomes of genetic variants with precompiled protein libraries aiding in rapid characterizations.
Collapse
Affiliation(s)
- Jeremy W Prokop
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Victoria Petri
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary E Shimoyama
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ingrid K M Watanabe
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Dulce E Casarini
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Thomas C Leeper
- Department of Chemistry, The University of Akron, Akron, OH, USA
| | | | - Howard J Jacob
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robson A S Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Almir S Martins
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabiano C Araujo
- National Institute of Science and Technology in Molecular Medicine - Department of Obstetrics and Gynecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernando M Reis
- National Institute of Science and Technology in Molecular Medicine - Department of Obstetrics and Gynecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Amy Milsted
- Department of Biology, The University of Akron, Akron, OH, USA
| |
Collapse
|
22
|
Chan SHH, Chan JYH. (Pro)renin receptor as a therapeutic target for the treatment of hypertension? Hypertension 2014; 65:278-9. [PMID: 25421978 DOI: 10.1161/hypertensionaha.114.04532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Samuel H H Chan
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Taiwan, Republic of China
| | - Julie Y H Chan
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Taiwan, Republic of China.
| |
Collapse
|
23
|
Li W, Sullivan MN, Zhang S, Worker CJ, Xiong Z, Speth RC, Feng Y. Intracerebroventricular infusion of the (Pro)renin receptor antagonist PRO20 attenuates deoxycorticosterone acetate-salt-induced hypertension. Hypertension 2014; 65:352-61. [PMID: 25421983 DOI: 10.1161/hypertensionaha.114.04458] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We previously reported that binding of prorenin to the (pro)renin receptor (PRR) plays a major role in brain angiotensin II formation and the development of deoxycorticosterone acetate (DOCA)-salt hypertension. Here, we designed and developed an antagonistic peptide, PRO20, to block prorenin binding to the PRR. Fluorescently labeled PRO20 bound to both mouse and human brain tissues with dissociation constants of 4.4 and 1.8 nmol/L, respectively. This binding was blocked by coincubation with prorenin and was diminished in brains of neuron-specific PRR-knockout mice, indicating specificity of PRO20 for PRR. In cultured human neuroblastoma cells, PRO20 blocked prorenin-induced calcium influx in a concentration- and AT(1) receptor-dependent manner. Intracerebroventricular infusion of PRO20 dose-dependently inhibited prorenin-induced hypertension in C57Bl6/J mice. Furthermore, acute intracerebroventricular infusion of PRO20 reduced blood pressure in both DOCA-salt and genetically hypertensive mice. Chronic intracerebroventricular infusion of PRO20 attenuated the development of hypertension and the increase in brain hypothalamic angiotensin II levels induced by DOCA-salt. In addition, chronic intracerebroventricular infusion of PRO20 improved autonomic function and spontaneous baroreflex sensitivity in mice treated with DOCA-salt. In summary, PRO20 binds to both mouse and human PRRs and decreases angiotensin II formation and hypertension induced by either prorenin or DOCA-salt. Our findings highlight the value of the novel PRR antagonist, PRO20, as a lead compound for a novel class of antihypertensive agents and as a research tool to establish the validity of brain PRR antagonism as a strategy for treating hypertension.
Collapse
Affiliation(s)
- Wencheng Li
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Michelle N Sullivan
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Sheng Zhang
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Caleb J Worker
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Zhenggang Xiong
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Robert C Speth
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.)
| | - Yumei Feng
- From the Department of Biomedical Sciences, Center for Cardiovascular Research, Colorado State University, Fort Collins (W.L., M.N.S., C.J.W., Y.F.); Department of Physiology, Tulane Hypertension and Renal Center of Excellence (S.Z.), and Department of Pathology and Laboratory Medicine (Z.X.), Tulane University School of Medicine, New Orleans, LA; and Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL (R.C.S.).
| |
Collapse
|
24
|
Tan P, Shamansurova Z, Bisotto S, Michel C, Gauthier MS, Rabasa-Lhoret R, Nguyen TMD, Schiller PW, Gutkowska J, Lavoie JL. Impact of the prorenin/renin receptor on the development of obesity and associated cardiometabolic risk factors. Obesity (Silver Spring) 2014; 22:2201-9. [PMID: 25044950 DOI: 10.1002/oby.20844] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Obesity is a worldwide epidemic and current treatments have limited success thus, novel therapies are warranted. Our objective was to determine whether the prorenin/renin receptor [(P)RR] is implicated in obesity. METHODS Mice received a normal or high-fat/high-carbohydrate diet with the handle region peptide (HRP), a (P)RR blocker, or saline for 10 weeks. Post-menopausal non-diabetic obese women were enrolled in the Complication Associated with Obesity Study and were classified as insulin-resistant (IRO) or -sensitive (ISO) using a hyperinsulinemic-euglycemic clamp. RESULTS In mice, obesity increased the (P)RR by twofold in adipose tissue. Likewise, renin increased by at least twofold. The HRP reduced weight gain in obese mice by 20% associated to a 19% decrease in visceral fat. This was accompanied by a 48% decrease in leptin mRNA in fat and 33% decrease in circulating leptin. Inflammatory markers were also decreased by the HRP treatment. HRP normalized triglyceridemia and reduced insulinemia by 34% in obese mice. Interestingly, we observed a 33% increase in (P)RR mRNA in the fat of IRO women compared to ISO. CONCLUSIONS This is the first report of a potential implication in obesity of the (P)RR which may be a novel therapeutic target.
Collapse
Affiliation(s)
- Paul Tan
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Biochemistry and Molecular Medicine of the Université de Montréal; Montreal Diabetes Research Center
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Batenburg WW, Verma A, Wang Y, Zhu P, van den Heuvel M, van Veghel R, Danser AHJ, Li Q. Combined renin inhibition/(pro)renin receptor blockade in diabetic retinopathy--a study in transgenic (mREN2)27 rats. PLoS One 2014; 9:e100954. [PMID: 24968134 PMCID: PMC4072720 DOI: 10.1371/journal.pone.0100954] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/31/2014] [Indexed: 12/20/2022] Open
Abstract
Dysfunction of renin-angiotensin system (RAS) contributes to the pathogenesis of diabetic retinopathy (DR). Prorenin, the precursor of renin is highly elevated in ocular fluid of diabetic patients with proliferative retinopathy. Prorenin may exert local effects in the eye by binding to the so-called (pro)renin receptor ((P)RR). Here we investigated the combined effects of the renin inhibitor aliskiren and the putative (P)RR blocker handle-region peptide (HRP) on diabetic retinopathy in streptozotocin (STZ)-induced diabetic transgenic (mRen2)27 rats (a model with high plasma prorenin levels) as well as prorenin stimulated cytokine expression in cultured Müller cells. Adult (mRen2)27 rats were randomly divided into the following groups: (1) non-diabetic; (2) diabetic treated with vehicle; (3) diabetic treated with aliskiren (10 mg/kg per day); and (4) diabetic treated with aliskiren+HRP (1 mg/kg per day). Age-matched non-diabetic wildtype Sprague-Dawley rats were used as control. Drugs were administered by osmotic minipumps for three weeks. Transgenic (mRen2)27 rat retinas showed increased apoptotic cell death of both inner retinal neurons and photoreceptors, increased loss of capillaries, as well as increased expression of inflammatory cytokines. These pathological changes were further exacerbated by diabetes. Aliskiren treatment of diabetic (mRen2)27 rats prevented retinal gliosis, and reduced retinal apoptotic cell death, acellular capillaries and the expression of inflammatory cytokines. HRP on top of aliskiren did not provide additional protection. In cultured Müller cells, prorenin significantly increased the expression levels of IL-1α and TNF-α, and this was completely blocked by aliskiren or HRP, their combination, (P)RR siRNA and the AT1R blocker losartan, suggesting that these effects entirely depended on Ang II generation by (P)RR-bound prorenin. In conclusion, the lack of effect of HRP on top of aliskiren, and the Ang II-dependency of the ocular effects of prorenin in vitro, argue against the combined application of (P)RR blockade and renin inhibition in diabetic retinopathy.
Collapse
Affiliation(s)
- Wendy W. Batenburg
- Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine, rasmus MC, GE Rotterdam, The Netherlands
| | - Amrisha Verma
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Yunyang Wang
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Mieke van den Heuvel
- Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine, rasmus MC, GE Rotterdam, The Netherlands
| | - Richard van Veghel
- Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine, rasmus MC, GE Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine, rasmus MC, GE Rotterdam, The Netherlands
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
26
|
te Riet L, van den Heuvel M, Peutz-Kootstra CJ, van Esch JHM, van Veghel R, Garrelds IM, Musterd-Bhaggoe U, Bouhuizen AM, Leijten FPJ, Danser AHJ, Batenburg WW. Deterioration of kidney function by the (pro)renin receptor blocker handle region peptide in aliskiren-treated diabetic transgenic (mRen2)27 rats. Am J Physiol Renal Physiol 2014; 306:F1179-89. [PMID: 24694588 DOI: 10.1152/ajprenal.00010.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Dual renin-angiotensin system (RAS) blockade in diabetic nephropathy is no longer feasible because of the profit/side effect imbalance. (Pro)renin receptor [(P)RR] blockade with handle region peptide (HRP) has been reported to exert beneficial effects in various diabetic models in a RAS-independent manner. To what degree (P)RR blockade adds benefits on top of RAS blockade is still unknown. In the present study, we treated diabetic TGR(mREN2)27 rats, a well-established nephropathy model with high prorenin levels [allowing continuous (P)RR stimulation in vivo], with HRP on top of renin inhibition with aliskiren. Aliskiren alone lowered blood pressure and exerted renoprotective effects, as evidenced by reduced glomerulosclerosis, diuresis, proteinuria, albuminuria, and urinary aldosterone levels as well as diminished renal (P)RR and ANG II type 1 receptor expression. It also suppressed plasma and tissue RAS activity and suppressed cardiac atrial natriuretic peptide and brain natriuretic peptide expression. HRP, when given on top of aliskiren, did not alter the effects of renin inhibition on blood pressure, RAS activity, or aldosterone. However, it counteracted the beneficial effects of aliskiren in the kidney, induced hyperkalemia, and increased plasma plasminogen activator-inhibitor 1, renal cyclooxygenase-2, and cardiac collagen content. All these effects have been linked to (P)RR stimulation, suggesting that HRP might, in fact, act as a partial agonist. Therefore, the use of HRP on top of RAS blockade in diabetic nephropathy is not advisable.
Collapse
Affiliation(s)
- Luuk te Riet
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Mieke van den Heuvel
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | | | - Joep H M van Esch
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Richard van Veghel
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Ingrid M Garrelds
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Usha Musterd-Bhaggoe
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Angelique M Bouhuizen
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Frank P J Leijten
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Wendy W Batenburg
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| |
Collapse
|
27
|
Liu FY, Liu XY, Zhang LJ, Cheng YP, Jiang YN. Binding of prorenin to (pro)renin receptor induces the proliferation of human umbilical artery smooth muscle cells via ROS generation and ERK1/2 activation. J Renin Angiotensin Aldosterone Syst 2014; 15:99-108. [PMID: 24591529 DOI: 10.1177/1470320314525215] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Since the discovery of the (pro)renin receptor (PRR), it has been considered as a novel bioactive molecule of the renin-angiotensin system (RAS). The activation of PRR can elicit a series of angiotensin II (AngII)-independent effects. MATERIALS AND METHODS In this study, we investigated the effects of prorenin and PRR on the proliferation of human umbilical artery smooth muscle (HUASM) cells and explored the possible mechanisms underlying these effects. RESULTS The binding of prorenin to PRR can promote proliferation and upregulate the anti-apoptotic protein Bcl-2 and downregulate the pro-apoptotic protein Bax independently of AngII in HUASM cells. In addition, the binding of prorenin to PRR can also increase the production of reactive oxygen species (ROS) and the phosphorylation of extracellular signal-regulated kinase (ERK1/2) independently of AngII. The pretreatment of HUASM cells with an NADPH oxidase inhibitor DPI decreased the production of ROS and also decreased the phosphorylation of ERK1/2. Furthermore, pretreatment of HUASM cells with DPI and the ERK1/2 inhibitor PD98059 significantly attenuated the prorenin-induced proliferation and regulation of apoptosis factors. CONCLUSION Binding of prorenin to PRR can induce HUASM cell proliferation via the ROS generation and ERK1/2 activation.
Collapse
Affiliation(s)
- Feng Y Liu
- Dalian Medical University, Dalian, PR China
| | - Xiao Y Liu
- Dalian Medical University, Dalian, PR China
| | - Li J Zhang
- Dalian Medical University, Dalian, PR China
| | - Yun P Cheng
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yi N Jiang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
28
|
Labruijere S, Compeer MG, van den Bogaerdt AJ, van den Brink AM, De Mey JG, Danser AJ, Batenburg WW. Long-lasting physiological antagonism of calcitonin gene-related peptide towards endothelin-1 in rat mesenteric arteries and human coronary arteries. Eur J Pharmacol 2013; 720:303-9. [DOI: 10.1016/j.ejphar.2013.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/01/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
|