1
|
Costello HM, Eikenberry SA, Cheng KY, Broderick B, Joshi AS, Scott GR, McKee A, Mendez VM, Douma LG, Crislip GR, Gumz ML. Sex differences in the adrenal circadian clock: a role for BMAL1 in the regulation of urinary aldosterone excretion and renal electrolyte balance in mice. Am J Physiol Renal Physiol 2025; 328:F1-F14. [PMID: 39447118 DOI: 10.1152/ajprenal.00177.2024] [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: 06/18/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
Brain and muscle ARNT-Like 1 (BMAL1) is a circadian clock transcription factor that regulates physiological functions. Male adrenal-specific Bmal1 (ASCre/+::Bmal1) KO mice displayed blunted serum corticosterone rhythms, altered blood pressure rhythm, and altered timing of eating, but there is a lack of knowledge in females. This study investigates the role of adrenal BMAL1 in renal electrolyte handling and urinary aldosterone levels in response to low salt in male and female mice. Mice were placed in metabolic cages to measure 12-h urinary aldosterone after a standard diet and 7 days low-salt diet, as well as daily body weight, 12-h food and water intake, and renal sodium and potassium balance. Adrenal glands and kidneys were collected at ZT0 or ZT12 to measure the expression of aldosterone synthesis genes and clock genes. Compared with littermate controls, ASCre/+::Bmal1 KO male and female mice displayed increased urinary aldosterone in response to a low-salt diet, although mRNA expression of aldosterone synthesis genes was decreased. Timing of food intake was altered in ASCre/+::Bmal1 KO male and female mice, with a blunted night/day ratio. ASCre/+::Bmal1 KO female mice displayed decreases in renal sodium excretion in response to low salt, but both male and female KO mice had changes in sodium balance that were time-of-day-dependent. In addition, sex differences were found in adrenal and kidney clock gene expression. Notably, this study highlights sex differences in clock gene expression that could contribute to sex differences in physiological functions.NEW & NOTEWORTHY Our findings highlight the importance of sex as well as time-of-day in understanding the role of the circadian clock in the regulation of homeostasis. Time-of-day is a key biological variable that is often ignored in research, particularly in preclinical rodent studies. Our findings demonstrate important differences in several measures at 6 AM compared with 6 PM. Consideration of time-of-day is critical for the translation of findings in nocturnal rodent physiology to diurnal human physiology.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Sophia A Eikenberry
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Kit-Yan Cheng
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| | - Bryanna Broderick
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Advay S Joshi
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Gianna R Scott
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Annalisse McKee
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Victor M Mendez
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Lauren G Douma
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
| | - G Ryan Crislip
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States
- Research, North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida, United States
| |
Collapse
|
2
|
Schofield LG, Endacott SK, Delforce SJ, Lumbers ER, Pringle KG. Importance of the (Pro)renin Receptor in Activating the Renin-Angiotensin System During Normotensive and Preeclamptic Pregnancies. Curr Hypertens Rep 2024; 26:483-495. [PMID: 39093387 PMCID: PMC11455731 DOI: 10.1007/s11906-024-01316-1] [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] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE OF REVIEW For a healthy pregnancy to occur, a controlled interplay between the maternal circulating renin-angiotensin-aldosterone system (RAAS), placental renin-angiotensin system (RAS) and intrarenal renin-angiotensin system (iRAS) is necessary. Functionally, both the RAAS and iRAS interact to maintain blood pressure and cardiac output, as well as fluid and electrolyte balance. The placental RAS is important for placental development while also influencing the maternal circulating RAAS and iRAS. This narrative review concentrates on the (pro)renin receptor ((P)RR) and its soluble form (s(P)RR) in the context of the hypertensive pregnancy pathology, preeclampsia. RECENT FINDINGS The (P)RR and the s(P)RR have become of particular interest as not only can they activate prorenin and renin, thus influencing levels of angiotensin II (Ang II), but s(P)RR has now been shown to directly interact with and stimulate the Angiotensin II type 1 receptor (AT1R). Levels of both placental (P)RR and maternal circulating s(P)RR are elevated in patients with preeclampsia. Furthermore, s(P)RR has been shown to increase blood pressure in non-pregnant and pregnant rats and mice. In preeclamptic pregnancies, which are characterised by maternal hypertension and impaired placental development and function, we propose that there is enhanced secretion of s(P)RR from the placenta into the maternal circulation. Due to its ability to both activate prorenin and act as an AT1R agonist, excess maternal circulating s(P)RR can act on both the maternal vasculature, and the kidney, leading to RAS over-activation. This results in dysregulation of the maternal circulating RAAS and overactivation of the iRAS, contributing to maternal hypertension, renal damage, and secondary changes to neurohumoral regulation of fluid and electrolyte balance, ultimately contributing to the pathophysiology of preeclampsia.
Collapse
Affiliation(s)
- Lachlan G Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Sarah J Delforce
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia.
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia.
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia.
| |
Collapse
|
3
|
Zhu M, Yi X, Song S, Yang H, Yu J, Xu C. Principle role of the (pro)renin receptor system in cardiovascular and metabolic diseases: An update. Cell Signal 2024; 124:111417. [PMID: 39321906 DOI: 10.1016/j.cellsig.2024.111417] [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: 06/24/2024] [Revised: 09/07/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
(Pro)renin receptor (PRR), along with its soluble form, sPRR, functions not only as a crucial activator of the local renin-angiotensin system but also engages with and activates various angiotensin II-independent signaling pathways, thus playing complex and significant roles in numerous physiological and pathophysiological processes, including cardiovascular and metabolic disorders. This article reviews current knowledge on the intracellular partners of the PRR system and explores its physiological and pathophysiological impacts on cardiovascular diseases as well as conditions related to glucose and lipid metabolism, such as hypertension, heart disease, liver disease, diabetes, and diabetic complications. Targeting the PRR system could emerge as a promising therapeutic strategy for treating these conditions. Elevated levels of circulating sPRR might indicate the severity of these diseases, potentially serving as a biomarker for diagnosis and prognosis in clinical settings. A comprehensive understanding of the functions and regulatory mechanisms of the PRR system could facilitate the development of novel therapeutic approaches for the prevention and management of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Mengzhi Zhu
- College of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoli Yi
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Shanshan Song
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Huiru Yang
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jun Yu
- Center for Metabolic Disease Research and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
4
|
Zhang Y, Arzaghi H, Ma Z, Roye Y, Musah S. Epigenetics of Hypertensive Nephropathy. Biomedicines 2024; 12:2622. [PMID: 39595187 PMCID: PMC11591919 DOI: 10.3390/biomedicines12112622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertensive nephropathy (HN) is a leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD), contributing to significant morbidity, mortality, and rising healthcare costs. In this review article, we explore the role of epigenetic mechanisms in HN progression and their potential therapeutic implications. We begin by examining key epigenetic modifications-DNA methylation, histone modifications, and non-coding RNAs-observed in kidney disease. Next, we discuss the underlying pathophysiology of HN and highlight current in vitro and in vivo models used to study the condition. Finally, we compare various types of HN-induced renal injury and their associated epigenetic mechanisms with those observed in other kidney injury models, drawing inferences on potential epigenetic therapies for HN. The information gathered in this work indicate that epigenetic mechanisms can drive the progression of HN by regulating key molecular signaling pathways involved in renal damage and fibrosis. The limitations of Renin-Angiotensin-Aldosterone System (RAAS) inhibitors underscore the need for alternative treatments targeting epigenetic pathways. This review emphasizes the importance of further research into the epigenetic regulation of HN to develop more effective therapies and preventive strategies. Identifying novel epigenetic markers could provide new therapeutic opportunities for managing CKD and reducing the burden of ESRD.
Collapse
Affiliation(s)
- Yize Zhang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hamidreza Arzaghi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Zhehan Ma
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27708, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, and Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
5
|
Luo R, Yang KT, Wang F, Zheng H, Yang T. Collecting Duct Pro(Renin) Receptor Contributes to Unilateral Ureteral Obstruction-Induced Kidney Injury via Activation of the Intrarenal RAS. Hypertension 2024; 81:2152-2161. [PMID: 39171392 PMCID: PMC11410543 DOI: 10.1161/hypertensionaha.123.21740] [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: 06/26/2023] [Accepted: 05/10/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Although the concept of the intrarenal renin-angiotensin system (RAS) in renal disease is well-described in the literature, the precise pathogenic role and mechanism of this local system have not been directly assessed in the absence of confounding influence from the systemic RAS. The present study used novel mouse models of collecting duct (CD)-specific deletion of (pro)renin receptor (PRR) or renin together with pharmacological inhibition of soluble PRR production to unravel the precise contribution of the intrarenal RAS to renal injury induced by unilateral ureteral obstruction. METHODS We examined the impact of CD-specific deletion of PRR, CD-specific deletion of renin, and S1P (site-1 protease) inhibitor PF429242 treatment on renal fibrosis and inflammation and the indices of the intrarenal RAS in a mouse model of unilateral ureteral obstruction. RESULTS After 3 days of unilateral ureteral obstruction, the indices of the intrarenal RAS including the renal medullary renin content, activity and mRNA expression, and Ang (angiotensin) II content in obstructed kidneys of floxed mice were all increased. That effect was reversed with CD-specific deletion of PRR, CD-specific deletion of renin, and PF429242 treatment, accompanied by consistent improvement in renal fibrosis and inflammation. On the other hand, renal cortical renin levels were unaffected by unilateral ureteral obstruction, irrespective of the genotype. Similar results were obtained via pharmacological inhibition of S1P, the key protease for the generation of soluble PRR. CONCLUSIONS Our results reveal that PRR-dependent/soluble PRR-dependent activation of CD renin represents a key determinant of the intrarenal RAS and, thus, obstruction-induced renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Renfei Luo
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Kevin T. Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Fei Wang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Huaqing Zheng
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
6
|
Schofield LG, Delforce SJ, Pryor JC, Endacott SK, Lumbers ER, Marshall SA, Pringle KG. The soluble (pro)renin receptor promotes a preeclampsia-like phenotype both in vitro and in vivo. Hypertens Res 2024; 47:1627-1641. [PMID: 38605139 PMCID: PMC11150152 DOI: 10.1038/s41440-024-01678-8] [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: 11/20/2023] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Preeclampsia is classified as new-onset hypertension coupled with gross endothelial dysfunction. Placental (pro)renin receptor ((P)RR) and plasma soluble (P)RR (s(P)RR) are elevated in patients with preeclampsia. Thus, we aimed to interrogate the role (P)RR may play in the pathogenesis of preeclampsia. Human uterine microvascular endothelial cells (HUtMECs, n = 4) were cultured with either; vehicle (PBS), 25-100 nM recombinant s(P)RR, or 10 ng/ml TNF-a (positive control) for 24 h. Conditioned media and cells were assessed for endothelial dysfunction markers via qPCR, ELISA, and immunoblot. Angiogenic capacity was assessed through tube formation and adhesion assays. Additionally, pregnant rats were injected with an adenovirus overexpressing s(P)RR from mid-pregnancy (day 8.5), until term (n = 6-7 dams/treatment). Maternal and fetal tissues were assessed. HUtMECs treated with recombinant s(P)RR displayed increased expression of endothelial dysfunction makers including vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and endothelin-1 mRNA expression (P = 0.003, P = 0.001, P = 0.009, respectively), along with elevated endothelin-1 protein secretion (P < 0.001) compared with controls. Recombinant s(P)RR impaired angiogenic capacity decreasing the number of branches, total branch length, and mesh area (P < 0.001, P = 0.004, and P = 0.009, respectively), while also increasing vascular adhesion (P = 0.032). +ADV rats exhibited increased systolic (P = 0.001), diastolic (P = 0.010), and mean arterial pressures (P = 0.012), compared with -ADV pregnancies. Renal arteries from +ADV-treated rats had decreased sensitivity to acetylcholine-induced relaxation (P = 0.030), compared with -ADV pregnancies. Our data show that treatment with s(P)RR caused hypertension and growth restriction in vivo and caused marked endothelial dysfunction in vitro. These findings demonstrate the significant adverse actions of s(P)RR on vascular dysfunction that is characteristic of the preeclamptic phenotype.
Collapse
Affiliation(s)
- Lachlan G Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Jennifer C Pryor
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- National Health & Medical Research Council (NHMRC) Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, The Ritchie Centre, School of Clinical Sciences, Monash University and The Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
7
|
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
|
8
|
Grote K, Schaefer AC, Soufi M, Ruppert V, Linne U, Mukund Bhagwat A, Szymanski W, Graumann J, Gercke Y, Aldudak S, Hilfiker-Kleiner D, Schieffer E, Schieffer B. Targeting the High-Density Lipoprotein Proteome for the Treatment of Post-Acute Sequelae of SARS-CoV-2. Int J Mol Sci 2024; 25:4522. [PMID: 38674105 PMCID: PMC11049911 DOI: 10.3390/ijms25084522] [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: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Here, we target the high-density lipoprotein (HDL) proteome in a case series of 16 patients with post-COVID-19 symptoms treated with HMG-Co-A reductase inhibitors (statin) plus angiotensin II type 1 receptor blockers (ARBs) for 6 weeks. Patients suffering from persistent symptoms (post-acute sequelae) after serologically confirmed SARS-CoV-2 infection (post-COVID-19 syndrome, PCS, n = 8) or following SARS-CoV-2 vaccination (PVS, n = 8) were included. Asymptomatic subjects with corresponding serological findings served as healthy controls (n = 8/8). HDL was isolated using dextran sulfate precipitation and the HDL proteome of all study participants was analyzed quantitatively by mass spectrometry. Clinical symptoms were assessed using questionnaires before and after therapy. The inflammatory potential of the patients' HDL proteome was addressed in human endothelial cells. The HDL proteome of patients with PCS and PVS showed no significant differences; however, compared to controls, the HDL from PVS/PCS patients displayed significant alterations involving hemoglobin, cytoskeletal proteins (MYL6, TLN1, PARVB, TPM4, FLNA), and amyloid precursor protein. Gene Ontology Biological Process (GOBP) enrichment analysis identified hemostasis, peptidase, and lipoprotein regulation pathways to be involved. Treatment of PVS/PCS patients with statins plus ARBs improved the patients' clinical symptoms. After therapy, three proteins were significantly increased (FAM3C, AT6AP2, ADAM10; FDR < 0.05) in the HDL proteome from patients with PVS/PCS. Exposure of human endothelial cells with the HDL proteome from treated PVS/PCS patients revealed reduced inflammatory cytokine and adhesion molecule expression. Thus, HDL proteome analysis from PVS/PCS patients enables a deeper insight into the underlying disease mechanisms, pointing to significant involvement in metabolic and signaling disturbances. Treatment with statins plus ARBs improved clinical symptoms and reduced the inflammatory potential of the HDL proteome. These observations may guide future therapeutic strategies for PVS/PCS patients.
Collapse
Affiliation(s)
- Karsten Grote
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Ann-Christin Schaefer
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Muhidien Soufi
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Volker Ruppert
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Uwe Linne
- Mass Spectrometry Facility, Department of Chemistry, Philipps University Marburg, 35043 Marburg, Germany;
| | - Aditya Mukund Bhagwat
- Institute of Translational Proteomics & Core Facility Translational Proteomics, Philipps University Marburg, 35043 Marburg, Germany (W.S.)
| | - Witold Szymanski
- Institute of Translational Proteomics & Core Facility Translational Proteomics, Philipps University Marburg, 35043 Marburg, Germany (W.S.)
| | - Johannes Graumann
- Institute of Translational Proteomics & Core Facility Translational Proteomics, Philipps University Marburg, 35043 Marburg, Germany (W.S.)
| | - Yana Gercke
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Sümeya Aldudak
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Denise Hilfiker-Kleiner
- Institute Cardiovascular Complications in Pregnancy and Oncologic Therapies, Philipps University Marburg, 35043 Marburg, Germany;
| | - Elisabeth Schieffer
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| | - Bernhard Schieffer
- Department of Cardiology, Angiology, and Intensive Care, Philipps University Marburg, 35043 Marburg, Germany; (K.G.); (A.-C.S.); (M.S.); (V.R.); (S.A.); (E.S.)
| |
Collapse
|
9
|
Arthur G, Poupeau A, Biel K, Osborn JL, Gong M, Hinds TD, Lindner V, Loria AS. Human soluble prorenin receptor expressed in mouse renal collecting duct shows sex-specific effect on cardiorenal function. Am J Physiol Renal Physiol 2024; 326:F611-F621. [PMID: 38385173 PMCID: PMC11208026 DOI: 10.1152/ajprenal.00375.2023] [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: 11/27/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024] Open
Abstract
Soluble prorenin receptor (sPRR), a component of the renin-angiotensin system (RAS), has been identified as a plasma biomarker for hypertension and cardiovascular diseases in humans. Despite studies showing that sPRR in the kidney is produced by tubular cells in the renal collecting duct (CD), its biological actions modulating cardiorenal function in physiological conditions remain unknown. Therefore, the objective of our study was to investigate whether CD-derived human sPRR (HsPRR) expression influences cardiorenal function and examine sex and circadian differences. Thus, we investigated the status of the intrarenal RAS, water and electrolyte balance, renal filtration capacity, and blood pressure (BP) regulation in CD-HsPRR and control (CTL) mice. CD-HsPRR mice were generated by breeding human sPRR-Myc-tag mice with Hoxb7/Cre mice. Renal sPRR expression increased in CD-HsPRR mice, but circulating sPRR and RAS levels were unchanged compared with CTL mice. Only female littermates expressing CD-HsPRR showed 1) increased 24-h BP, 2) an impaired BP response to an acute dose of losartan and attenuated angiotensin II (ANG II)-induced hypertension, 3) reduced angiotensin-converting enzyme activity and ANG II content in the renal cortex, and 4) decreased glomerular filtration rate, with no changes in natriuresis and kaliuresis despite upregulation of the β-subunit of the epithelial Na+ channel in the renal cortex. These cardiorenal alterations were displayed only during the active phase of the day. Taken together, these data suggest that HsPRR could interact with ANG II type 1 receptors mediating sex-specific, ANG II-independent renal dysfunction and a prohypertensive phenotype in a sex-specific manner.NEW & NOTEWORTHY We successfully generated a humanized mouse model that expresses human sPRR in the collecting duct. Collecting duct-derived human sPRR did not change circulating sPRR and RAS levels but increased daytime BP in female mice while showing an attenuated angiotensin II-dependent pressor response. These findings may aid in elucidating the mechanisms by which women show uncontrolled BP in response to antihypertensive treatments targeting the RAS, improving approaches to reduce uncontrolled BP and chronic kidney disease incidences in women.
Collapse
Affiliation(s)
- Gertrude Arthur
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Audrey Poupeau
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Katherine Biel
- Department of Nutrition and Dietetics, University of Kentucky, Lexington, Kentucky, United States
| | - Jeffrey L Osborn
- Department of Pathophysiology, Arkansas Colleges of Health Education, Fort Smith, Arkansas, United States
| | - Ming Gong
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Volkhard Lindner
- MaineHealth Institute for Research, Scarborough, Maine, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
- SAHA Cardiovascular Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
10
|
Yang TY, Chang PJ, Ko YS, Shen SR, Chang SF. Assessment of the (Pro)renin Receptor Protein Expression in Organs. Curr Issues Mol Biol 2024; 46:1741-1753. [PMID: 38534729 DOI: 10.3390/cimb46030113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
The (pro)renin receptor ((P)RR) is an essential component of the renin-angiotensin system (RAS) as a specific single-pass transmembrane receptor for prorenin and renin and has now emerged as a multifunctional protein implicated in a wide variety of developmental and physio-pathological processes and pathways. The (P)RR may be of pathological significance in metabolic syndrome. The (P)RR has received much consideration; substantial efforts have been made to understand the localization, regulation, and function of the (P)RR at both a molecular and system level. (P)RR regulation of cell function depends on whether it is intact or cleaved into its constituent forms. Therefore, the present chapter describes immunohistochemical approaches to examine the expression of (P)RR in various organs. It was shown that different molecular forms of (P)RR could be present in different tissue compartments in almost all organs. Among them, the liver has high PRR activity. Our findings could elucidate more detailed distribution of different (P)RR molecular forms in different organs, which could provide useful information to further investigate the pathophysiological mechanisms of the development of various diseases in the future.
Collapse
Affiliation(s)
- Teng-Yao Yang
- Cardiovascular Department, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yu-Shien Ko
- Cardiovascular Division, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Siou-Ru Shen
- Cardiovascular Department, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Shun-Fu Chang
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Center for General Education, Chiayi Chang Gung University of Science and Technology, Chiayi 613, Taiwan
| |
Collapse
|
11
|
Roth L, Dogan S, Tuna BG, Aranyi T, Benitez S, Borrell-Pages M, Bozaykut P, De Meyer GRY, Duca L, Durmus N, Fonseca D, Fraenkel E, Gillery P, Giudici A, Jaisson S, Johansson M, Julve J, Lucas-Herald AK, Martinet W, Maurice P, McDonnell BJ, Ozbek EN, Pucci G, Pugh CJA, Rochfort KD, Roks AJM, Rotllan N, Shadiow J, Sohrabi Y, Spronck B, Szeri F, Terentes-Printzios D, Tunc Aydin E, Tura-Ceide O, Ucar E, Yetik-Anacak G. Pharmacological modulation of vascular ageing: A review from VascAgeNet. Ageing Res Rev 2023; 92:102122. [PMID: 37956927 DOI: 10.1016/j.arr.2023.102122] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
Vascular ageing, characterized by structural and functional changes in blood vessels of which arterial stiffness and endothelial dysfunction are key components, is associated with increased risk of cardiovascular and other age-related diseases. As the global population continues to age, understanding the underlying mechanisms and developing effective therapeutic interventions to mitigate vascular ageing becomes crucial for improving cardiovascular health outcomes. Therefore, this review provides an overview of the current knowledge on pharmacological modulation of vascular ageing, highlighting key strategies and promising therapeutic targets. Several molecular pathways have been identified as central players in vascular ageing, including oxidative stress and inflammation, the renin-angiotensin-aldosterone system, cellular senescence, macroautophagy, extracellular matrix remodelling, calcification, and gasotransmitter-related signalling. Pharmacological and dietary interventions targeting these pathways have shown potential in ameliorating age-related vascular changes. Nevertheless, the development and application of drugs targeting vascular ageing is complicated by various inherent challenges and limitations, such as certain preclinical methodological considerations, interactions with exercise training and sex/gender-related differences, which should be taken into account. Overall, pharmacological modulation of endothelial dysfunction and arterial stiffness as hallmarks of vascular ageing, holds great promise for improving cardiovascular health in the ageing population. Nonetheless, further research is needed to fully elucidate the underlying mechanisms and optimize the efficacy and safety of these interventions for clinical translation.
Collapse
Affiliation(s)
- Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Tamas Aranyi
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Sonia Benitez
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Biochemistry, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Perinur Bozaykut
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkiye
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Nergiz Durmus
- Department of Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkiye
| | - Diogo Fonseca
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Košice, Slovakia
| | - Philippe Gillery
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | - Alessandro Giudici
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, the Netherlands
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | | | - Josep Julve
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Endocrinology, Diabetes and Nutrition group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Barry J McDonnell
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Emine Nur Ozbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye
| | - Giacomo Pucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christopher J A Pugh
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Dublin, Ireland
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands
| | - Noemi Rotllan
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Pathophysiology of lipid-related diseases, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Molecular Cardiology, Dept. of Cardiology I - Coronary and Peripheral Vascular Disease, University Hospital Münster, Westfälische Wilhelms-Universität, 48149 Münster, Germany; Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czechia
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Flora Szeri
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elif Tunc Aydin
- Department of Cardiology, Hospital of Ataturk Training and Research Hospital, Katip Celebi University, Izmir, Turkiye
| | - Olga Tura-Ceide
- Biomedical Research Institute-IDIBGI, Girona, Spain; Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Eda Ucar
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye; Department of Pharmacology, Faculty of Pharmacy, Acıbadem Mehmet Aydinlar University, Istanbul, Turkiye.
| |
Collapse
|
12
|
Funke-Kaiser H, Unger T. The (pro)renin receptor as a pharmacological target in cardiorenal diseaes. Hypertens Res 2023; 46:2527-2534. [PMID: 37667044 DOI: 10.1038/s41440-023-01424-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023]
Abstract
The (pro)renin receptor ((P)RR) is not only a member of the renin-angiotensin system (RAS) but also exerts several RAS-independent functions due to its multiple signal transductions pathways. In this mini-review, we shortly discuss the molecular functions of this receptor and its pathophysiological significance with a focus on cardiorenal diseases. Finally, we provide a short summary regarding a drug discovery and drug development program on small molecule-based renin/ prorenin receptor blockers (RERBs).
Collapse
Affiliation(s)
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
13
|
Visniauskas B, Reverte V, Abshire CM, Ogola BO, Rosales CB, Galeas-Pena M, Sure VN, Sakamuri SSVP, Harris NR, Kilanowski-Doroh I, Mcnally AB, Horton AC, Zimmerman M, Katakam PVG, Lindsey SH, Prieto MC. High-plasma soluble prorenin receptor is associated with vascular damage in male, but not female, mice fed a high-fat diet. Am J Physiol Heart Circ Physiol 2023; 324:H762-H775. [PMID: 36930656 PMCID: PMC10151046 DOI: 10.1152/ajpheart.00638.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Plasma soluble prorenin receptor (sPRR) displays sexual dimorphism and is higher in women with type 2 diabetes mellitus (T2DM). However, the contribution of plasma sPRR to the development of vascular complications in T2DM remains unclear. We investigated if plasma sPRR contributes to sex differences in the activation of the systemic renin-angiotensin-aldosterone system (RAAS) and vascular damage in a model of high-fat diet (HFD)-induced T2DM. Male and female C57BL/6J mice were fed either a normal fat diet (NFD) or an HFD for 28 wk to assess changes in blood pressure, cardiometabolic phenotype, plasma prorenin/renin, sPRR, and ANG II. After completing dietary protocols, tissues were collected from males to assess vascular reactivity and aortic reactive oxygen species (ROS). A cohort of male mice was used to determine the direct contribution of increased systemic sPRR by infusion. To investigate the role of ovarian hormones, ovariectomy (OVX) was performed at 32 wk in females fed either an NFD or HFD. Significant sex differences were found after 28 wk of HFD, where only males developed T2DM and increased plasma prorenin/renin, sPRR, and ANG II. T2DM in males was accompanied by nondipping hypertension, carotid artery stiffening, and aortic ROS. sPRR infusion in males induced vascular thickening instead of material stiffening caused by HFD-induced T2DM. While intact females were less prone to T2DM, OVX increased plasma prorenin/renin, sPRR, and systolic blood pressure. These data suggest that sPRR is a novel indicator of systemic RAAS activation and reflects the onset of vascular complications during T2DM regulated by sex.NEW & NOTEWORTHY High-fat diet (HFD) for 28 wk leads to type 2 diabetes mellitus (T2DM) phenotype, concomitant with increased plasma soluble prorenin receptor (sPRR), nondipping blood pressure, and vascular stiffness in male mice. HFD-fed female mice exhibiting a preserved cardiometabolic phenotype until ovariectomy revealed increased plasma sPRR and blood pressure. Plasma sPRR may indicate the status of systemic renin-angiotensin-aldosterone system (RAAS) activation and the onset of vascular complications during T2DM in a sex-dependent manner.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
| | - Virginia Reverte
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Caleb M Abshire
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Carla B Rosales
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Michelle Galeas-Pena
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Nicholas R Harris
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Isabella Kilanowski-Doroh
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Margaret Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
- Tulane Center for Sex-Based Biology and Medicine, New Orleans, Louisiana, United States
- Tulane Hypertension and Renal Center of Excellence, New Orleans, Louisiana, United States
| |
Collapse
|
14
|
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: 2] [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
|
15
|
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: 0] [Impact Index Per Article: 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
|
16
|
Nichols K, Yiannikouris F. The Role of (Pro)Renin Receptor in the Metabolic Syndrome. Curr Hypertens Rev 2022; 18:117-124. [PMID: 35170416 DOI: 10.2174/1573402118666220216104816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 01/27/2023]
Abstract
The prorenin receptor (PRR) is a complex multi-functional single transmembrane protein receptor that is ubiquitously expressed in organs and tissues throughout the body. PRR is involved in different cellular mechanisms that comprise the generation of Angiotensin II, the activation of Wnt/β-catenin signaling, the stimulation of ERK 1/2 pathway, and the proper functioning of the vacuolar H+-ATPase. Evidence supports the role of PRR and its soluble form, sPRR, in the classical features of the metabolic syndrome, including obesity, hypertension, diabetes, and disruption of lipid homeostasis. This review summarizes our current knowledge and highlights new advances in the pathophysiological function of PRR and sPRR in adipogenesis, adipocyte differentiation, lipolysis, glucose and insulin resistance, lipid homeostasis, energy metabolism, and blood pressure regulation.
Collapse
Affiliation(s)
- Kellea Nichols
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Frederique Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
17
|
Yang T. Soluble (Pro)Renin Receptor in Hypertension. Nephron Clin Pract 2022; 147:234-243. [PMID: 35871512 PMCID: PMC9867785 DOI: 10.1159/000525635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/27/2022] [Indexed: 01/26/2023] Open
Abstract
The (pro)renin receptor (PRR) was originally cloned as a specific single-transmembrane receptor for prorenin and renin and has now emerged as a multifunctional protein implicated in a wide variety of developmental and physiopathological processes. Activation of PRR in the kidney causes Na+ and water retention, contributing to elevation of blood pressure in response to various hypertensive stimuli. Part of the renal action of PRR depends on activation of intrarenal renin-angiotensin system. In recent years, accumulating evidence suggests that the prohypertensive action of renal PRR was largely mediated by production of the 28-kDa soluble (pro)renin receptor through protease-mediated cleavage of the extracellular domain of PRR. The generation of multiple isoforms of PRR due to the protease-mediated cleavage partially explains diversified actions of PRR. The current review will summarize recent advances in understanding the roles of sPPR in animal models of hypertension.
Collapse
Affiliation(s)
- Tianxin Yang
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
18
|
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: 17] [Impact Index Per Article: 8.5] [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
|
19
|
Zhang J, Hui Y, Liu F, Yang Q, Lu Y, Chang Y, Liu Q, Ding Y. Neohesperidin Protects Angiotensin II-Induced Hypertension and Vascular Remodeling. Front Pharmacol 2022; 13:890202. [PMID: 35677431 PMCID: PMC9168427 DOI: 10.3389/fphar.2022.890202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
Vascular remodeling due to hypertension is one of the major health challenges facing countries around the world. Neohesperidin, a flavonoid glycoside found in citrus fruits, is an antioxidant. Neohesperidin has been studied for a variety of diseases in addition to hypertension. In this study, angiotensin II was used to induce hypertension in mice (490 ng/kg/min, 14 days). We used H&E, Masson, immunofluorescence, dihydroethidine and qPCR to evaluate the effect of Nehesperidin (50 mg/kg/day, 16 days) on pathological hypertension in mice. Estimating the effect of Nehesperidin on human umbilical vein endothelial cells and vascular smooth muscle cells stimulated by angiotensin II. We found that neohesperidin inhibited angiotensin II-induced hypertension in mice. Neohesperidin reduced angiotensin II-induced vascular hypertrophy, fibrosis, inflammation and oxidative stress in vivo. Neohesperidin inhibited angiotensin II-induced ROS and DNA damage in human umbilical vein endothelial cells. Neohesperidin inhibited angiotensin II-induced migration of vascular smooth muscle cells. The results showed that Nehesperidin acts as an antioxidant and could significantly inhibit angiotensin II induced hypertension and vascular remodeling in vitro and in vivo.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yuanshu Hui
- Department of Heart Function Examination, The Second Hospital of Dalian Medical University, Dalian, China
| | - Fengyi Liu
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Qian Yang
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yi Lu
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yeting Chang
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Qinlong Liu
- Department of Hepatobiliary Pancreatic Surgery II, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yanchun Ding
- Department of Cardiology II, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
20
|
Xu C, Liu C, Xiong J, Yu J. Cardiovascular aspects of the (pro)renin receptor: Function and significance. FASEB J 2022; 36:e22237. [PMID: 35226776 DOI: 10.1096/fj.202101649rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases (CVDs), including all types of disorders related to the heart or blood vessels, are the major public health problems and the leading causes of mortality globally. (Pro)renin receptor (PRR), a single transmembrane protein, is present in cardiomyocytes, vascular smooth muscle cells, and endothelial cells. PRR plays an essential role in cardiovascular homeostasis by regulating the renin-angiotensin system and several intracellular signals such as mitogen-activated protein kinase signaling and wnt/β-catenin signaling in various cardiovascular cells. This review discusses the current evidence for the pathophysiological roles of the cardiac and vascular PRR. Activation of PRR in cardiomyocytes may contribute to myocardial ischemia/reperfusion injury, cardiac hypertrophy, diabetic or alcoholic cardiomyopathy, salt-induced heart damage, and heart failure. Activation of PRR promotes vascular smooth muscle cell proliferation, endothelial cell dysfunction, neovascularization, and the progress of vascular diseases. In addition, phenotypes of animals transgenic for PRR and the hypertensive actions of PRR in the brain and kidney and the soluble PRR are also discussed. Targeting PRR in local tissues may offer benefits for patients with CVDs, including heart injury, atherosclerosis, and hypertension.
Collapse
Affiliation(s)
- Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chunju Liu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jianhua Xiong
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jun Yu
- Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Qin M, Xu C, Yu J. The Soluble (Pro)Renin Receptor in Health and Diseases: Foe or Friend? J Pharmacol Exp Ther 2021; 378:251-261. [PMID: 34158404 DOI: 10.1124/jpet.121.000576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
The (pro)renin receptor (PRR) is a single-transmembrane protein that regulates the local renin-angiotensin system and participates in various intracellular signaling pathways, thus exhibiting a significant physiopathologic relevance in cellular homeostasis. A soluble form of PRR (sPRR) is generated through protease-mediated cleavage of the full-length PRR and secreted into extracellular spaces. Accumulating evidence indicates pivotal biologic functions of sPRR in various physiopathological processes. sPRR may be a novel biomarker for multiple diseases. SIGNIFICANCE STATEMENT: Circulating sPRR concentrations are elevated in patients and animals under various physiopathological conditions. This minireview highlights recent advances in sPRR functions in health and pathophysiological conditions. Results suggest that sPRR may be a novel biomarker for multiple diseases, but further studies are needed to determine the diagnostic value of sPRR.
Collapse
Affiliation(s)
- Manman Qin
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China (M.Q., C.X.), and Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania (J.Y.)
| | - Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China (M.Q., C.X.), and Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania (J.Y.)
| | - Jun Yu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China (M.Q., C.X.), and Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania (J.Y.)
| |
Collapse
|
22
|
Soluble (pro)renin receptor: a novel ligand for angiotensin II type 1 receptor? Clin Sci (Lond) 2021; 135:1627-1630. [PMID: 34240733 DOI: 10.1042/cs20210227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
This commentary highlights the study entitled 'Soluble (pro)renin receptor induces endothelial dysfunction and hypertension in mice with diet-induced obesity via activation of angiotensin II type 1 receptor' presented by Fu et al. published in Clinical Science (Clin Sci (Lond) (2021) 135(6), https://doi.org/10.1042/CS20201047). The authors evaluated the role of the soluble (pro)renin receptor (sPRR), a cleavage product of the prorenin receptor (PRR) by the site 1 protease, as a ligand for angiotensin II type 1 receptor (AT1R). They presented for the first time that sPRR directly interacts with AT1R, causing nuclear factor-κB activation, inflammation, apoptosis, and endothelial dysfunction in primary human umbilical vein endothelial cells (HUVECs). Furthermore, the interaction between sPRR and AT1R was responsible for endothelial dysfunction and hypertension in diet-induced obesity mice. These results provide a potential mechanism for obesity-induced endothelial dysfunction and hypertension. Thus, the sPRR/AT1R complex may be a novel therapeutic target for cardiovascular diseases associated with endothelial dysfunction.
Collapse
|