1
|
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
|
2
|
Chen L, Xiong L, Guo H, Feng X, Zhu X, Xiong WC. Osteoclastic ATP6AP2 maintains β-catenin levels to prevent hyper-osteoclastic activation and trabecular bone-loss. J Bone Miner Res 2024; 39:1821-1834. [PMID: 39400061 DOI: 10.1093/jbmr/zjae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/07/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Osteoclast (OC) formation and bone resorption are regulated by several factors, including V-ATPase, Wnt/β-catenin, and RANKL/RANK signaling. ATP6AP2, also known as the prorenin receptor (PRR), is an accessory subunit of V-ATPase and a regulator of Wnt/β-catenin signaling. While the V-ATPase subunit ATP6AP1 is essential for OC formation and function, the role of ATP6AP2 in OC-lineage cells is less clear. Here, we provide evidence that ATP6AP2 plays a negative role in osteoclastogenesis and function, contrasting with the positive role of ATP6AP1. Mice with conditional KO (cKO) of ATP6AP2 in OCs (Atp6ap2LysM) exhibit trabecular bone loss, likely due to the increased osteoclastogenesis and activity, since bone formation rates (BFRs) are comparable to control mice. In vitro assays using bone marrow macrophages (BMMs) show that Atp6ap2LysM cultures have more RANKL-induced TRAP+ OC-like cells and increased bone resorptive activity. Further studies reveal that while RANKL signaling and V-ATPase activity are normal, ATP6AP2 KO OCs, but not BMMs, have reduced basal levels of Wnt/β-catenin pathway proteins, such as LRP5/6 and β-catenin, compared to controls. Wnt3A treatment induces β-catenin and suppresses OC formation in both control and ATP6AP2 KO OC-lineage cells, indicating that Wnt/β-catenin signaling negatively regulates OC-formation and operates independently of ATP6AP2. Overall, these results suggest that ATP6AP2 is critical for maintaining basal levels of LRP5/6 receptors and β-catenin in OCs, thus acting as a negative regulator of osteoclastogenesis and activation.
Collapse
Affiliation(s)
- Li Chen
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin 130022, China
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr Building, Cleveland, OH 44106, United States
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr Building, Cleveland, OH 44106, United States
- Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, United States
| | - Haohan Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr Building, Cleveland, OH 44106, United States
- Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, United States
| | - Xu Feng
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, 619 19th St S Ste P210, Birmingham, AL 35233, United States
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin 130022, China
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 2210 Circle Dr Building, Cleveland, OH 44106, United States
- Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, United States
| |
Collapse
|
3
|
Yu Y, Zhang M, Wang D, Xiang Z, Zhao Z, Cui W, Ye S, Fazhan H, Waiho K, Ikhwanuddin M, Ma H. Whole transcriptome RNA sequencing provides novel insights into the molecular dynamics of ovarian development in mud crab, Scylla paramamosain after mating. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 51:101247. [PMID: 38788625 DOI: 10.1016/j.cbd.2024.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024]
Abstract
Ovarian development in animals is a complicated biological process, requiring the simultaneous coordination among various genes and pathways. To understand the dynamic changes and molecular regulatory mechanisms of ovarian development in mud crab (Scylla paramamosain), both histological observation and whole transcriptome sequencing of ovarian tissues at different mating stages were implemented in this study. The histological results revealed that ovarian development was delayed in unmated females (60 days after courtship behavior but not mating), who exhibited an oocyte diameter of 56.38 ± 15.17 μm. Conversely, mated females exhibited accelerated the ovarian maturation process, with females reaching ovarian stage III (proliferative stage) 23 days after mating and attained an average oocyte diameter of 132.19 ± 15.07 μm. Thus, mating process is essential in promoting the rapid ovarian development in mud crab. Based on the whole transcriptome sequencing analysis, a total of 518 mRNAs, 1502 lncRNAs, 18 circRNAs and 151 miRNAs were identified to be differentially expressed between ovarian tissues at different mating stages. Notably, six differentially expressed genes (DEGs) associated with ovarian development were identified, including ovary development-related protein, red pigment concentrating hormone receptor, G2/mitotic-specific cyclin-B3-like, lutropin-chorio gonadotropic hormone receptor, renin receptor, and SoxB2. More importantly, both DEGs and targets of differentially expressed non-coding RNAs (DEncRNAs) were enriched in renin-angiotensin system, TGF-β signaling, cell adhesion molecules, MAPK signaling pathway, and ECM-receptor interaction, suggesting that these pathways may play significant roles in the ovarian development of mud crabs. Moreover, competition endogenous RNA (ceRNA) networks were constructed while mRNAs were differentially expressed between mating stages were involved in Gene Ontology (GO) biological processes such as developmental process, reproduction, and growth. These findings could provide solid foundations for the future development of female mud crab maturation enhancement strategy, and improve the understanding of the ovarian maturation process in crustaceans.
Collapse
Affiliation(s)
- Yang Yu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Higher Institute Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Mengqian Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Dahe Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Higher Institute Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Zifei Xiang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Zilin Zhao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Wenxiao Cui
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Shaopan Ye
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Hanafiah Fazhan
- International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Higher Institute Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Khor Waiho
- International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Higher Institute Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Mhd Ikhwanuddin
- International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Higher Institute Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Hongyu Ma
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; International Joint Research Center for the Development and Utilization of Important Mariculture Varieties Surrounding the South China Sea Region, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China.
| |
Collapse
|
4
|
Culver SA, Hargett SR, Balugo JLLQ, Gildea JJ, Harris TE, Siragy HM. Nephron specific ATP6AP2 knockout increases urinary excretion of fatty acids and decreases renal cortical megalin expression. Sci Rep 2024; 14:18724. [PMID: 39134597 PMCID: PMC11319469 DOI: 10.1038/s41598-024-69749-x] [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: 03/20/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
ATP6AP2 knockout in the renal nephron impairs receptor-mediated endocytosis, increasing urinary albumin and glucose excretion and impairing weight gain. Nonesterified fatty acids (NEFA) in urine are bound to albumin and reabsorbed in the proximal tubule through receptor-mediated endocytosis by the megalin-cubilin complex. We hypothesized that ATP6AP2 knockout increases urinary NEFA excretion through a reduction in megalin. Ten-week-old male C57BL/6 mice with nephron specific inducible ATP6AP2 knockout and noninduced controls were fed either normal diet (ND 12% fat) or high fat diet (HFD 45% fat) for 6 months. ATP6AP2 knockout significantly increased urine albumin:creatinine ratio in both ND and HFD fed mice while normalized urine NEFA concentration increased 489% and 259% in ND and HFD knockout mice compared to respective controls. Knockout decreased renal cortical megalin mRNA by 47% on ND and 49% on HFD while megalin protein expression decreased by 36% and 44% respectively. At the same time, markers of mTOR activity were increased while autophagy was impaired. Our results indicate that nephron specific ATP6AP2 knockout increases urinary NEFA excretion in the setting of impaired receptor-mediated endocytosis. Further investigation should determine whether ATP6AP2 contributes to obesity related ectopic lipid deposition in the proximal tubule.
Collapse
Affiliation(s)
- Silas A Culver
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA.
| | - Stefan R Hargett
- Department of Pharmacology, University of Virginia Health System, Charlottesville, USA
| | - Jamie L L Q Balugo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA
| | - John J Gildea
- Department of Pathology, University of Virginia Health System, Charlottesville, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia Health System, Charlottesville, USA
| | - Helmy M Siragy
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, P.O. Box 801409, Charlottesville, VA, 22908-1409, USA
| |
Collapse
|
5
|
Wang N, Ren L, Danser AHJ. Vacuolar H +-ATPase in Diabetes, Hypertension, and Atherosclerosis. Microcirculation 2024; 31:e12855. [PMID: 38683673 DOI: 10.1111/micc.12855] [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: 02/07/2024] [Revised: 03/29/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
Vacuolar H+-ATPase (V-ATPase) is a multisubunit protein complex which, along with its accessory proteins, resides in almost every eukaryotic cell. It acts as a proton pump and as such is responsible for regulating pH in lysosomes, endosomes, and the extracellular space. Moreover, V-ATPase has been implicated in receptor-mediated signaling. Although numerous studies have explored the role of V-ATPase in cancer, osteoporosis, and neurodegenerative diseases, research on its involvement in vascular disease remains limited. Vascular diseases pose significant challenges to human health. This review aimed to shed light on the role of V-ATPase in hypertension and atherosclerosis. Furthermore, given that vascular complications are major complications of diabetes, this review also discusses the pathways through which V-ATPase may contribute to such complications. Beginning with an overview of the structure and function of V-ATPase in hypertension, atherosclerosis, and diabetes, this review ends by exploring the pharmacological potential of targeting V-ATPase.
Collapse
Affiliation(s)
- Na Wang
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - 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
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Brazier F, Cornière N, Picard N, Chambrey R, Eladari D. Pendrin: linking acid base to blood pressure. Pflugers Arch 2024; 476:533-543. [PMID: 38110744 DOI: 10.1007/s00424-023-02897-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
Pendrin (SLC26A4) is an anion exchanger from the SLC26 transporter family which is mutated in human patients affected by Pendred syndrome, an autosomal recessive disease characterized by sensoneurinal deafness and hypothyroidism. Pendrin is also expressed in the kidney where it mediates the exchange of internal HCO3- for external Cl- at the apical surface of renal type B and non-A non-B-intercalated cells. Studies using pendrin knockout mice have first revealed that pendrin is essential for renal base excretion. However, subsequent studies have demonstrated that pendrin also controls chloride absorption by the distal nephron and that this mechanism is critical for renal NaCl balance. Furthermore, pendrin has been shown to control vascular volume and ultimately blood pressure. This review summarizes the current knowledge about how pendrin is linking renal acid-base regulation to blood pressure control.
Collapse
Affiliation(s)
- François Brazier
- Centre de dépistage et de Médecine de précision des Maladies Rénales, Service de Néphrologie, Centre Hospitalier Universitaire Amiens-Picardie, Université de Picardie Jules Verne, F-80000, Amiens, France
| | - Nicolas Cornière
- Centre de dépistage et de Médecine de précision des Maladies Rénales, Service de Néphrologie, Centre Hospitalier Universitaire Amiens-Picardie, Université de Picardie Jules Verne, F-80000, Amiens, France
| | - Nicolas Picard
- Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Lyon 1, Lyon, France
| | - Régine Chambrey
- Paris Cardiovascular Research Center (PARCC), INSERM U970, F-75015, Paris, France
| | - Dominique Eladari
- Centre de dépistage et de Médecine de précision des Maladies Rénales, Service de Néphrologie, Centre Hospitalier Universitaire Amiens-Picardie, Université de Picardie Jules Verne, F-80000, Amiens, France.
- Laboratory of Tissue Biology and Therapeutic Engineering, UMR 5305 CNRS, University Lyon 1, Lyon, France.
- French Clinical Research Infrastructure Network (F-CRIN): INI-CRCT, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
7
|
Iacobini C, Vitale M, Sentinelli F, Haxhi J, Pugliese G, Menini S. Renal Expression and Localization of the Receptor for (Pro)renin and Its Ligands in Rodent Models of Diabetes, Metabolic Syndrome, and Age-Dependent Focal and Segmental Glomerulosclerosis. Int J Mol Sci 2024; 25:2217. [PMID: 38396894 PMCID: PMC10888662 DOI: 10.3390/ijms25042217] [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: 12/28/2023] [Revised: 01/27/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The (pro)renin receptor ((P)RR), a versatile protein found in various organs, including the kidney, is implicated in cardiometabolic conditions like diabetes, hypertension, and dyslipidemia, potentially contributing to organ damage. Importantly, changes in (pro)renin/(P)RR system localization during renal injury, a critical information base, remain unexplored. This study investigates the expression and topographic localization of the full length (FL)-(P)RR, its ligands (renin and prorenin), and its target cyclooxygenase-2 and found that they are upregulated in three distinct animal models of renal injury. The protein expression of these targets, initially confined to specific tubular renal cell types in control animals, increases in renal injury models, extending to glomerular cells. (P)RR gene expression correlates with protein changes in a genetic model of focal and segmental glomerulosclerosis. However, in diabetic and high-fat-fed mice, (P)RR mRNA levels contradict FL-(P)RR immunoreactivity. Research on diabetic mice kidneys and human podocytes exposed to diabetic glucose levels suggests that this inconsistency may result from disrupted intracellular (P)RR processing, likely due to increased Munc18-1 interacting protein 3. It follows that changes in FL-(P)RR cellular content mechanisms are specific to renal disease etiology, emphasizing the need for consideration in future studies exploring this receptor's involvement in renal damage of different origins.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (J.H.); (S.M.)
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (J.H.); (S.M.)
| | - Federica Sentinelli
- Department of Public Health and Infectious Diseases, “La Sapienza” University, 00189 Rome, Italy;
| | - Jonida Haxhi
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (J.H.); (S.M.)
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (J.H.); (S.M.)
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (C.I.); (M.V.); (J.H.); (S.M.)
| |
Collapse
|
8
|
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
|
9
|
Lin H, Geurts F, Hassler L, Batlle D, Mirabito Colafella KM, Denton KM, Zhuo JL, Li XC, Ramkumar N, Koizumi M, Matsusaka T, Nishiyama A, Hoogduijn MJ, Hoorn EJ, Danser AHJ. Kidney Angiotensin in Cardiovascular Disease: Formation and Drug Targeting. Pharmacol Rev 2022; 74:462-505. [PMID: 35710133 PMCID: PMC9553117 DOI: 10.1124/pharmrev.120.000236] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The concept of local formation of angiotensin II in the kidney has changed over the last 10-15 years. Local synthesis of angiotensinogen in the proximal tubule has been proposed, combined with prorenin synthesis in the collecting duct. Binding of prorenin via the so-called (pro)renin receptor has been introduced, as well as megalin-mediated uptake of filtered plasma-derived renin-angiotensin system (RAS) components. Moreover, angiotensin metabolites other than angiotensin II [notably angiotensin-(1-7)] exist, and angiotensins exert their effects via three different receptors, of which angiotensin II type 2 and Mas receptors are considered renoprotective, possibly in a sex-specific manner, whereas angiotensin II type 1 (AT1) receptors are believed to be deleterious. Additionally, internalized angiotensin II may stimulate intracellular receptors. Angiotensin-converting enzyme 2 (ACE2) not only generates angiotensin-(1-7) but also acts as coronavirus receptor. Multiple, if not all, cardiovascular diseases involve the kidney RAS, with renal AT1 receptors often being claimed to exert a crucial role. Urinary RAS component levels, depending on filtration, reabsorption, and local release, are believed to reflect renal RAS activity. Finally, both existing drugs (RAS inhibitors, cyclooxygenase inhibitors) and novel drugs (angiotensin receptor/neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors, soluble ACE2) affect renal angiotensin formation, thereby displaying cardiovascular efficacy. Particular in the case of the latter three, an important question is to what degree they induce renoprotection (e.g., in a renal RAS-dependent manner). This review provides a unifying view, explaining not only how kidney angiotensin formation occurs and how it is affected by drugs but also why drugs are renoprotective when altering the renal RAS. SIGNIFICANCE STATEMENT: Angiotensin formation in the kidney is widely accepted but little understood, and multiple, often contrasting concepts have been put forward over the last two decades. This paper offers a unifying view, simultaneously explaining how existing and novel drugs exert renoprotection by interfering with kidney angiotensin formation.
Collapse
Affiliation(s)
- Hui Lin
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Frank Geurts
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Luise Hassler
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Daniel Batlle
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Katrina M Mirabito Colafella
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Kate M Denton
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Jia L Zhuo
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Xiao C Li
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Nirupama Ramkumar
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Masahiro Koizumi
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Taiji Matsusaka
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Akira Nishiyama
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Martin J Hoogduijn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - Ewout J Hoorn
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine (H.L., A.H.J.D.) and Division of Nephrology and Transplantation (F.G., M.J.H., E.J.H.), Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands; Northwestern University Feinberg School of Medicine, Chicago, Illinois (L.H., D.B.); Monash University, Melbourne, Australia (K.M.M.C., K.M.D.); Tulane University School of Medicine, New Orleans, Louisiana (J.L.Z., X.C.L.); Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, Utah (N.R.); Division of Nephrology, Endocrinology, and Metabolism (M.K.) and Institute of Medical Sciences and Department of Basic Medicine (M.K., T.M.), Tokai University School of Medicine, Isehara, Japan; and Department of Pharmacology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Japan (A.N.)
| |
Collapse
|
10
|
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: 1.0] [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
|
11
|
Saigo S, Kino T, Uchida K, Sugawara T, Chen L, Sugiyama M, Azushima K, Wakui H, Tamura K, Ishigami T. Blood Pressure Elevation of Tubular Specific (P)RR Transgenic Mice and Lethal Tubular Degeneration due to Possible Intracellular Interactions between (P)RR and Alternative Renin Products. Int J Mol Sci 2021; 23:ijms23010302. [PMID: 35008728 PMCID: PMC8745386 DOI: 10.3390/ijms23010302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
The prorenin/renin receptor ((P)RR) is a multifunctional protein that is widely distributed in various organs. Despite intensive research for more than 20 years, this receptor has not been fully characterized. In this study, we generated mice overexpressing the tubular epithelial (P)RR gene ((P)RR-TG mice) to test the previously reported functional role of (P)RR by Ramkumar et al. in 2015 using tubular specific (P)RR KO mice. (P)RR-TG mice were maintained and analyzed in individual metabolic cages and were administered angiotensin II blocker (ARB), direct renin inhibitor (DRI), and bafilomycin, that is, vacuolar ATPase (V-ATPase) antagonist. (P)RR-TG mice were hypertensive and had alkalized urine with lower osmolality and Na+ excretion. ARB and DRI, but not bafilomycin, concurrently decreased blood pressure. Bafilomycin acidized urine of (P)RR-TG mice, or equivalently this phenomenon restored the effect of overexpressed transgene, suggesting that (P)RR functioned as a V-ATPase in renal tubules. Afterall, (P)RR-TG mice were mated with alternative renin transgenic mice (ARen2-TG), which we identified as intracellular renin previously, to generate double transgenic mice (DT-TG). Lethal renal tubular damage was observed in DT-TG mice, suggesting that intracellular renin may be a ligand for (P)RR in tubules. In summary, (P)RR did not substantially affect the tissue renin-angiotensin system (RAS) in our model of tubular specific (P)RR gene over-expression, but alternative intracellular renin may be involved in (P)RR signaling in addition to conventional V-ATPase function. Further investigations are warranted.
Collapse
Affiliation(s)
- Sae Saigo
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Tabito Kino
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kotaro Uchida
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Takuya Sugawara
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Lin Chen
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Michiko Sugiyama
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Kengo Azushima
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Hiromichi Wakui
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Kouichi Tamura
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
| | - Tomoaki Ishigami
- Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan; (S.S.); (T.K.); (K.U.); (T.S.); (L.C.); (M.S.); (K.A.); (H.W.); (K.T.)
- Correspondence: or
| |
Collapse
|
12
|
Culver SA, Akhtar S, Rountree-Jablin C, Keller SR, Cathro HP, Gildea JJ, Siragy HM. Knockout of Nephron ATP6AP2 Impairs Proximal Tubule Function and Prevents High-Fat Diet-Induced Obesity in Male Mice. Endocrinology 2021; 162:bqab200. [PMID: 34534267 PMCID: PMC8489432 DOI: 10.1210/endocr/bqab200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 12/24/2022]
Abstract
ATP6AP2 expression is increased in the nephron during high-fat diet (HFD) and its knockout (ATP6AP2 KO) reduces body weight (WT) in mice. We evaluated the contribution of ATP6AP2 to urinary glucose (UG) and albumin (Ualb) handling during HFD. We hypothesized that nephron ATP6AP2 KO increases UG and Ualb and minimizes HFD-induced obesity. Eight-week-old male C57BL/6J mice with inducible nephron-specific ATP6AP2 KO and noninduced controls were fed either normal diet (ND, 12% kcal fat) or HFD (45% kcal fat) for 6 months. ATP6AP2 KO mice on ND had 20% (P < 0.01) lower WT compared with controls. HFD-fed mice had 41% (P < 0.05) greater WT than ND-fed control mice. In contrast, ATP6AP2 KO abrogated the increase in WT induced by HFD by 40% (P < 0.05). Mice on HFD had less caloric intake compared with ND controls (P < 0.01). There were no significant differences in metabolic rate between all groups. UG and Ualb was significantly increased in ATP6AP2 KO mice on both ND and HFD. ATP6AP2 KO showed greater levels of proximal tubule apoptosis and histologic evidence of proximal tubule injury. In conclusion, our results demonstrate that nephron-specific ATP6AP2 KO is associated with glucosuria and albuminuria, most likely secondary to renal proximal tubule injury and/or dysfunction. Urinary loss of nutrients may have contributed to the reduced WT of knockout mice on ND and lack of WT gain in response to HFD. Future investigation should elucidate the mechanisms by which loss of renal ATP6AP2 causes proximal tubule injury and dysfunction.
Collapse
Affiliation(s)
- Silas A Culver
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Safia Akhtar
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Callie Rountree-Jablin
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Susanna R Keller
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Helen P Cathro
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - John J Gildea
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Helmy M Siragy
- Division of Endocrinology, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
13
|
Kashio-Yokota Y, Sato S, Hirose T, Watanabe T, Endo A, Watanabe F, Endo M, Ohba K, Mori T, Takahashi K. Elevated (Pro)renin Receptor Expression by Anti-Cancer Drugs, Carboplatin and Paclitaxel, in Cultured Cancer Cells: Possible Involvement of Apoptosis and Autophagy. TOHOKU J EXP MED 2021; 255:91-104. [PMID: 34645770 DOI: 10.1620/tjem.255.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
(Pro)renin receptor [(P)RR] is a component of the renin-angiotensin system and plays an essential role in the activity of vacuolar H+-ATPase and autophagy. (P)RR is expressed in cancer cells. However, the relationship among (P)RR, apoptosis and autophagy in the treatment of anti-cancer drugs has not been clarified. The aim of this study was to clarify the effects of anti-cancer drugs with autophagy-promoting activity on (P)RR expression in cancer cells. MCF-7 breast cancer cells and A549 lung cancer cells were treated with carboplatin or paclitaxel, and the expression of (P)RR, apoptosis markers and autophagy markers were assessed by RT-qPCR, western blot analysis and immunocytochemistry. Expression levels of (P)RR mRNA and soluble (P)RR protein were increased by carboplatin or paclitaxel in a dose-dependent manner. Immunofluorescence staining of (P)RR was increased in both MCF-7 and A549 cells treated by carboplatin or paclitaxel. Apoptosis induction was shown by elevated BAX/BCL2 mRNA levels and increased active caspase3-positive cells. Moreover, autophagy induction was confirmed by increased levels of autophagy-associated mRNAs and LC3B-II proteins. (P)RR knockdown by (P)RR-specific siRNA suppressed the cell viability in MCF-7 cells and A549 cells under the treatment of carboplatin or paclitaxel, suggesting that (P)RR deficiency inhibits the proliferation of cancer cells in a pathway different from carboplatin or paclitaxel. The present study showed that the expression of (P)RR mRNA and soluble (P)RR was increased by anti-cancer drugs with autophagy-promoting activity. Upregulated (P)RR and autophagy may constitute a stress adaptation that protects cancer cells from apoptosis.
Collapse
Affiliation(s)
- Yurina Kashio-Yokota
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Shigemitsu Sato
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Takuo Hirose
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine.,Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University.,Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Tomoki Watanabe
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Akari Endo
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine.,Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Fumihiko Watanabe
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Moe Endo
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Koji Ohba
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University.,Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| |
Collapse
|
14
|
Hoffmann N, Peters J. Functions of the (pro)renin receptor (Atp6ap2) at molecular and system levels: pathological implications in hypertension, renal and brain development, inflammation, and fibrosis. Pharmacol Res 2021; 173:105922. [PMID: 34607004 DOI: 10.1016/j.phrs.2021.105922] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/16/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
The (pro)renin receptor [(P)RR, Atp6ap2] was initially discovered as a membrane-bound binding partner of prorenin and renin. A soluble (P)RR has additional paracrine effects and is involved in metabolic syndrome and kidney damage. Meanwhile it is clear that most of the effects of the (P)RR are independent of prorenin. In the kidney, (P)RR plays an important role in renal dysfunction by activating proinflammatory and profibrotic molecules. In the brain, (P)RR is expressed in cardiovascular regulatory nuclei and is linked to hypertension. (P)RR is known to be an essential component of the v-ATPase as a key accessory protein and plays an important role in kidney, brain and heart via regulating the pH of the extracellular space and intracellular compartments. V-ATPase and (P)RR together act on WNT and mTOR signalling pathways, which are responsible for cellular homeostasis and autophagy. (P)RR through its role in v-ATPase assembly and function is also important for fast recycling endocytosis by megalin. In the kidney, megalin together with v-ATPase and (P)RR is crucial for endocytic uptake of components of the RAS and their intracellular processing. In the brain, (P)RR, v-ATPases and megalin are important regulators both during development and in the adult. All three proteins are associated with diseases such as XLMR, XMRE, X-linked parkinsonism and epilepsy, cognitive disorders with Parkinsonism, spasticity, intellectual disability, and Alzheimer's Disease which are characterized by impaired neuronal function and/or neuronal loss. The present review focusses on the relevant effects of Atp6ap2 without assigning them necessarily to the RAS. Mechanistically, many effects can be well explained by the role of Atp6ap2 for v-ATPase assembly and function. Furthermore, application of a soluble (P)RR analogue as new therapeutic option is discussed.
Collapse
Affiliation(s)
- Nadin Hoffmann
- Institute of Physiology, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Str. 15A, 17475, Greifswald, Germany
| | - Jörg Peters
- Institute of Physiology, University Medicine Greifswald, Friedrich-Ludwig-Jahn-Str. 15A, 17475, Greifswald, Germany.
| |
Collapse
|
15
|
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: 11] [Impact Index Per Article: 2.8] [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
|
16
|
Figueiredo M, Daryadel A, Sihn G, Müller DN, Popova E, Rouselle A, Nguyen G, Bader M, Wagner CA. The (pro)renin receptor (ATP6ap2) facilitates receptor-mediated endocytosis and lysosomal function in the renal proximal tubule. Pflugers Arch 2021; 473:1229-1246. [PMID: 34228176 PMCID: PMC8302575 DOI: 10.1007/s00424-021-02598-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
The ATP6ap2 (Pro)renin receptor protein associates with H+-ATPases which regulate organellar, cellular, and systemic acid-base homeostasis. In the kidney, ATP6ap2 colocalizes with H+-ATPases in various cell types including the cells of the proximal tubule. There, H+-ATPases are involved in receptor-mediated endocytosis of low molecular weight proteins via the megalin/cubilin receptors. To study ATP6ap2 function in the proximal tubule, we used an inducible shRNA Atp6ap2 knockdown rat model (Kd) and an inducible kidney-specific Atp6ap2 knockout mouse model. Both animal lines showed higher proteinuria with elevated albumin, vitamin D binding protein, and procathepsin B in urine. Endocytosis of an injected fluid-phase marker (FITC- dextran, 10 kDa) was normal whereas processing of recombinant transferrin, a marker for receptor-mediated endocytosis, to lysosomes was delayed. While megalin and cubilin expression was unchanged, abundance of several subunits of the H+-ATPase involved in receptor-mediated endocytosis was reduced. Lysosomal integrity and H+-ATPase function are associated with mTOR signaling. In ATP6ap2, KO mice mTOR and phospho-mTOR appeared normal but increased abundance of the LC3-B subunit of the autophagosome was observed suggesting a more generalized impairment of lysosomal function in the absence of ATP6ap2. Hence, our data suggests a role for ATP6ap2 for proximal tubule function in the kidney with a defect in receptor-mediated endocytosis in mice and rats.
Collapse
Affiliation(s)
- Marta Figueiredo
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Arezoo Daryadel
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Gabin Sihn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Dominik N Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Elena Popova
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Anthony Rouselle
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | | | - Michael Bader
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Charite University Medicine Berlin, Berlin, Germany.
- Institute for Biology, University of Lübeck, Lübeck, Germany.
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
17
|
Watanabe H, Belyea BC, Paxton RL, Li M, Dzamba BJ, DeSimone DW, Gomez RA, Sequeira-Lopez MLS. Renin Cell Baroreceptor, a Nuclear Mechanotransducer Central for Homeostasis. Circ Res 2021; 129:262-276. [PMID: 33993729 PMCID: PMC8273135 DOI: 10.1161/circresaha.120.318711] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Brian C. Belyea
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Robert L. Paxton
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Minghong Li
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Bette J. Dzamba
- Department of Cell Biology, University of Virginia, School of Medicine, Charlottesville, Virginia
| | - Douglas W. DeSimone
- Department of Cell Biology, University of Virginia, School of Medicine, Charlottesville, Virginia
| | - R. Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Maria Luisa S. Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
18
|
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: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
The intrarenal renin-angiotensin system is critical for the regulation of tubule sodium reabsorption, renal haemodynamics and blood pressure. The excretion of renin in urine can result from its increased filtration, the inhibition of renin reabsorption by megalin in the proximal tubule, or its secretion by the principal cells of the collecting duct. Modest increases in circulating or intrarenal angiotensin II (ANGII) stimulate the synthesis and secretion of angiotensinogen in the proximal tubule, which provides sufficient substrate for collecting duct-derived renin to form angiotensin I (ANGI). In models of ANGII-dependent hypertension, ANGII suppresses plasma renin, suggesting that urinary renin is not likely to be the result of increased filtered load. In the collecting duct, ANGII stimulates the synthesis and secretion of prorenin and renin through the activation of ANGII type 1 receptor (AT1R) expressed primarily by principal cells. The stimulation of collecting duct-derived renin is enhanced by paracrine factors including vasopressin, prostaglandin E2 and bradykinin. Furthermore, binding of prorenin and renin to the prorenin receptor in the collecting duct evokes a number of responses, including the non-proteolytic enzymatic activation of prorenin to produce ANGI from proximal tubule-derived angiotensinogen, which is then converted into ANGII by luminal angiotensin-converting enzyme; stimulation of the epithelial sodium channel (ENaC) in principal cells; and activation of intracellular pathways linked to the upregulation of cyclooxygenase 2 and profibrotic genes. These findings suggest that dysregulation of the renin-angiotensin system in the collecting duct contributes to the development of hypertension by enhancing sodium reabsorption and the progression of kidney injury.
Collapse
Affiliation(s)
- Minolfa C. Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA.,
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaiso, Chile
| | - Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - L. Gabriel Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
19
|
Ramkumar N, Stuart D, Peterson CS, Hu C, Wheatley W, Cho JM, Symons JD, Kohan DE. Loss of Soluble (Pro)renin Receptor Attenuates Angiotensin-II Induced Hypertension and Renal Injury. Circ Res 2021; 129:50-62. [PMID: 33890822 PMCID: PMC8225587 DOI: 10.1161/circresaha.120.317532] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| | - Caitlin S. Peterson
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| | - Chunyan Hu
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| | - William Wheatley
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| | - Jae Min Cho
- Nutrition and Integrative Physiology, University of Utah Health,Salt Lake City, UT
| | - J David Symons
- Nutrition and Integrative Physiology, University of Utah Health,Salt Lake City, UT
- Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health, Salt Lake City, UT
| |
Collapse
|
20
|
Kidney intercalated cells and the transcription factor FOXi1 drive cystogenesis in tuberous sclerosis complex. Proc Natl Acad Sci U S A 2021; 118:2020190118. [PMID: 33536341 PMCID: PMC8017711 DOI: 10.1073/pnas.2020190118] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in TSC1 or TSC2 gene and affects multiple organs, including the kidney, where it presents with angiomyolipomata and cysts that can result in kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Current studies demonstrate that kidney cyst epithelia in TSC mouse models and in humans with TSC are composed of hyperproliferating intercalated cells, along with activation of H+-ATPase and carbonic anhydrase 2. Interfering with intercalated cell proliferation completely inhibited and inactivating carbonic anhydrase 2 significantly protected against cyst formation in TSC. Targeting the acid base and/or electrolyte transporters of intercalated cells may provide a therapeutic approach for the treatment of kidney cysts in TSC. Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl−/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.
Collapse
|
21
|
D'Ambrosio V, Azzarà A, Sangiorgi E, Gurrieri F, Hess B, Gambaro G, Ferraro PM. Results of a Gene Panel Approach in a Cohort of Patients with Incomplete Distal Renal Tubular Acidosis and Nephrolithiasis. Kidney Blood Press Res 2021; 46:469-474. [PMID: 34107482 DOI: 10.1159/000516389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Distal renal tubular acidosis (dRTA) is characterized by an impairment of urinary acidification resulting in metabolic acidosis, hypokalemia, and inappropriately elevated urine pH. If not treated, this chronic condition eventually leads to nephrocalcinosis, nephrolithiasis, impaired renal function, and bone demineralization. dRTA is a well-defined entity that can be diagnosed by genetic testing of 5 genes known to be disease-causative. Incomplete dRTA (idRTA) is defined as impaired urinary acidification that does not lead to overt metabolic acidosis and therefore can be diagnosed if patients fail to adequately acidify urine after an ammonium chloride (NH4Cl) challenge or furosemide and fludrocortisone test. It is still uncertain whether idRTA represents a distinct entity or is part of the dRTA spectrum and whether it is caused by mutations in the same genes of overt dRTA. METHODS In this cross-sectional study, we investigated a group of 22 stone formers whose clinical features were suspicious of idRTA. They underwent an NH4Cl challenge and were found to have impaired urinary acidification ability. These patients were then analyzed by genetic testing with sequencing of 5 genes: SLC4A1, ATP6V1B1, ATP6V0A4, FOXI1, and WDR72. RESULTS Two unrelated individuals were found to have two different variants in SLC4A1 that had never been described before. CONCLUSIONS Our results suggest the involvement of other genes or nongenetic tubular dysfunction in the pathogenesis of idRTA in stone formers. However, genetic testing may represent a cost-effective tool to recognize, treat, and prevent complications in these patients.
Collapse
Affiliation(s)
- Viola D'Ambrosio
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessia Azzarà
- Istituto di Medicina Genomica, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eugenio Sangiorgi
- Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Medicina Genomica, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fiorella Gurrieri
- Unità di genetica Medica e Funzionale, Università Campus Bio-Medico, Rome, Italy
| | - Bernhard Hess
- Internal Medicine and Nephrology, Klinik Im Park, Zurich, and University of Zurich, Zurich, Switzerland
| | - Giovanni Gambaro
- Renal Unit, Division of Nephrology and Dialysis, Department of Medicine, University of Verona, Verona, Italy
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
22
|
Emathinger JM, Nelson JW, Gurley SB. Advances in use of mouse models to study the renin-angiotensin system. Mol Cell Endocrinol 2021; 529:111255. [PMID: 33789143 PMCID: PMC9119406 DOI: 10.1016/j.mce.2021.111255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/19/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The renin-angiotensin system (RAS) is a highly complex hormonal cascade that spans multiple organs and cell types to regulate solute and fluid balance along with cardiovascular function. Much of our current understanding of the functions of the RAS has emerged from a series of key studies in genetically-modified animals. Here, we review key findings from ground-breaking transgenic models, spanning decades of research into the RAS, with a focus on their use in studying blood pressure. We review the physiological importance of this regulatory system as evident through the examination of mouse models for several major RAS components: angiotensinogen, renin, ACE, ACE2, and the type 1 A angiotensin receptor. Both whole-animal and cell-specific knockout models have permitted critical RAS functions to be defined and demonstrate how redundancy and multiplicity within the RAS allow for compensatory adjustments to maintain homeostasis. Moreover, these models present exciting opportunities for continued discovery surrounding the role of the RAS in disease pathogenesis and treatment for cardiovascular disease and beyond.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/deficiency
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensinogen/deficiency
- Angiotensinogen/genetics
- Animals
- Blood Pressure/genetics
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Kidney/cytology
- Kidney/metabolism
- Mice
- Mice, Knockout
- Receptor, Angiotensin, Type 1/deficiency
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Renin/deficiency
- Renin/genetics
- Renin-Angiotensin System/genetics
- Signal Transduction
- Water-Electrolyte Balance/genetics
Collapse
Affiliation(s)
- Jacqueline M Emathinger
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Susan B Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
23
|
Petrillo F, Iervolino A, Angrisano T, Jelen S, Costanzo V, D’Acierno M, Cheng L, Wu Q, Guerriero I, Mazzarella MC, De Falco A, D’Angelo F, Ceccarelli M, Caraglia M, Capasso G, Fenton RA, Trepiccione F. Dysregulation of Principal Cell miRNAs Facilitates Epigenetic Regulation of AQP2 and Results in Nephrogenic Diabetes Insipidus. J Am Soc Nephrol 2021; 32:1339-1354. [PMID: 33727367 PMCID: PMC8259636 DOI: 10.1681/asn.2020010031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/02/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs), formed by cleavage of pre-microRNA by the endoribonuclease Dicer, are critical modulators of cell function by post-transcriptionally regulating gene expression. METHODS Selective ablation of Dicer in AQP2-expressing cells (DicerAQP2Cre+ mice) was used to investigate the role of miRNAs in the kidney collecting duct of mice. RESULTS The mice had severe polyuria and nephrogenic diabetes insipidus, potentially due to greatly reduced AQP2 and AQP4 levels. Although epithelial sodium channel levels were decreased in cortex and increased in inner medulla, amiloride-sensitive sodium reabsorption was equivalent in DicerAQP2Cre+ mice and controls. Small-RNA sequencing and proteomic analysis revealed 31 and 178 significantly regulated miRNAs and proteins, respectively. Integrated bioinformatic analysis of the miRNAome and proteome suggested alterations in the epigenetic machinery and various transcription factors regulating AQP2 expression in DicerAQP2Cre+ mice. The expression profile and function of three miRNAs (miR-7688-5p, miR-8114, and miR-409-3p) whose predicted targets were involved in epigenetic control (Phf2, Kdm5c, and Kdm4a) or transcriptional regulation (GATA3, GATA2, and ELF3) of AQP2 were validated. Luciferase assays could not demonstrate direct interaction of AQP2 or the three potential transcription factors with miR-7688-5p, miR-8114, and miR-409-3p. However, transfection of respective miRNA mimics reduced AQP2 expression. Chromatin immunoprecipitation assays demonstrated decreased Phf2 and significantly increased Kdm5c interactions at the Aqp2 gene promoter in DicerAQP2Cre+ mice, resulting in decreased RNA Pol II association. CONCLUSIONS Novel evidence indicates miRNA-mediated epigenetic regulation of AQP2 expression.
Collapse
Affiliation(s)
- Federica Petrillo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anna Iervolino
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Tiziana Angrisano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Sabina Jelen
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Vincenzo Costanzo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | | | - Lei Cheng
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Qi Wu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ilaria Guerriero
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | | | - Alfonso De Falco
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Fulvio D’Angelo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Michele Ceccarelli
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Electrical Engineering and Information Technology (DIETI) University of Naples “Federico II”, Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovambattista Capasso
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Francesco Trepiccione
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
24
|
Morosin SK, Lochrin AJ, Delforce SJ, Lumbers ER, Pringle KG. The (pro)renin receptor ((P)RR) and soluble (pro)renin receptor (s(P)RR) in pregnancy. Placenta 2021; 116:43-50. [PMID: 34020806 DOI: 10.1016/j.placenta.2021.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 11/25/2022]
Abstract
The (pro)renin receptor ((P)RR) is a multi-functional protein that can be proteolytically cleaved and released in a soluble form (s(P)RR). Recently, the (P)RR and s(P)RR have become of interest in pregnancy and its associated pathologies. This is because the (P)RR not only activates tissue renin angiotensin systems, but it is also an integral component of vacuolar-ATPase, activates the wingless/integrated (Wnt)/β-catenin and extracellular signal regulated kinases 1 and 2/mitogen-activated protein kinase signalling pathways, and stabilises the β subunit of pyruvate dehydrogenase. Additionally, s(P)RR is detected in plasma and urine, and maternal plasma levels are elevated in pregnancy complications including fetal growth restriction, preeclampsia and gestational diabetes mellitus. Therefore, s(P)RR has potential as a biomarker for these pregnancy pathologies. Preliminary functional findings suggest that s(P)RR may be important for regulating fluid balance, inflammation and blood pressure, all of which contribute to a successful pregnancy. The (P)RR and s(P)RR regulate pathways that are known to be important in maintaining pregnancy, however their role in the physiological context of pregnancy is poorly characterised. This review summarises the known and potential functions of the (P)RR and s(P)RR in pregnancy, and how their dysregulation may contribute to pregnancy complications. It also highlights the need for further research into the source and function of s(P)RR in pregnancy. Soluble (P)RR levels could be indicative of placental, kidney or liver dysfunction and therefore be a novel clinical biomarker, or therapeutic target, to improve the detection and treatment of pregnancy pathologies.
Collapse
Affiliation(s)
- Saije K Morosin
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Alyssa J Lochrin
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Reproductive Science, Pregnancy and Reproduction Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, 2305, New South Wales, Australia.
| |
Collapse
|
25
|
Chen Y, Xu C, Hu J, Deng M, Qiu Q, Mo S, Du Y, Yang T. Diuretic Action of Apelin-13 Mediated by Inhibiting cAMP/PKA/sPRR Pathway. Front Physiol 2021; 12:642274. [PMID: 33868005 PMCID: PMC8044521 DOI: 10.3389/fphys.2021.642274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence is showing that apelin plays an important role in regulating salt and water balance by counteracting the antidiuretic action of vasopressin (AVP). However, the underlying mechanism remains unknown. Here, we hypothesized that (pro) renin receptor (PRR)/soluble prorenin receptor (sPRR) might mediate the diuretic action of apelin in the distal nephron. During water deprivation (WD), the urine concentrating capability was impaired by an apelin peptide, apelin-13, accompanied by the suppression of the protein expression of aquaporin 2 (AQP2), NKCC2, PRR/sPRR, renin and nuclear β-catenin levels in the kidney. The upregulated expression of AQP2 or PRR/sPRR both induced by AVP and 8-Br-cAMP was blocked by apelin-13, PKA inhibitor (H89), or β-catenin inhibitor (ICG001). Interestingly, the blockage of apelin-13 on AVP-induced AQP2 protein expression was reversed by exogenous sPRR. Together, the present study has defined the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/sPRR pathway in the CD as the molecular target of the diuretic action of apelin.
Collapse
Affiliation(s)
- Yanting Chen
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China
| | - Chuanming Xu
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China.,Center for Translational Medicine, Jiangxi University of Traditional Chinese, Nanchang, China
| | - Jiajia Hu
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China
| | - Mokan Deng
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China
| | - Qixiang Qiu
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China
| | - Shiqi Mo
- Zhongshan School of Medicine, Institute of Hypertension, Sun Yat-sen University, Guangzhou, China
| | - Yanhua Du
- Department of Pharmacology, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, United States
| |
Collapse
|
26
|
Arthur G, Osborn JL, Yiannikouris FB. (Pro)renin receptor in the kidney: function and significance. Am J Physiol Regul Integr Comp Physiol 2021; 320:R377-R383. [PMID: 33470188 DOI: 10.1152/ajpregu.00259.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
(Pro)renin receptor (PRR), a 350-amino acid receptor initially thought of as a receptor for the binding of renin and prorenin, is multifunctional. In addition to its role in the renin-angiotensin system (RAS), PRR transduces several intracellular signaling molecules and is a component of the vacuolar H+-ATPase that participates in autophagy. PRR is found in the kidney and particularly in great abundance in the cortical collecting duct. In the kidney, PRR participates in water and salt balance, acid-base balance, and autophagy and plays a role in development and progression of hypertension, diabetic retinopathy, and kidney fibrosis. This review highlights the role of PRR in the development and function of the kidney, namely, the macula densa, podocyte, proximal and distal convoluted tubule, and the principal cells of the collecting duct, and focuses on PRR function in body fluid volume homeostasis, blood pressure regulation, and acid-base balance. This review also explores new advances in the molecular mechanism involving PRR in normal renal health and pathophysiological states.
Collapse
Affiliation(s)
- Gertrude Arthur
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Jeffrey L Osborn
- Department of Biology, University of Kentucky, Lexington, Kentucky
| | | |
Collapse
|
27
|
Eaton AF, Merkulova M, Brown D. The H +-ATPase (V-ATPase): from proton pump to signaling complex in health and disease. Am J Physiol Cell Physiol 2020; 320:C392-C414. [PMID: 33326313 PMCID: PMC8294626 DOI: 10.1152/ajpcell.00442.2020] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A primary function of the H+-ATPase (or V-ATPase) is to create an electrochemical proton gradient across eukaryotic cell membranes, which energizes fundamental cellular processes. Its activity allows for the acidification of intracellular vesicles and organelles, which is necessary for many essential cell biological events to occur. In addition, many specialized cell types in various organ systems such as the kidney, bone, male reproductive tract, inner ear, olfactory mucosa, and more, use plasma membrane V-ATPases to perform specific activities that depend on extracellular acidification. It is, however, increasingly apparent that V-ATPases are central players in many normal and pathophysiological processes that directly influence human health in many different and sometimes unexpected ways. These include cancer, neurodegenerative diseases, diabetes, and sensory perception, as well as energy and nutrient-sensing functions within cells. This review first covers the well-established role of the V-ATPase as a transmembrane proton pump in the plasma membrane and intracellular vesicles and outlines factors contributing to its physiological regulation in different cell types. This is followed by a discussion of the more recently emerging unconventional roles for the V-ATPase, such as its role as a protein interaction hub involved in cell signaling, and the (patho)physiological implications of these interactions. Finally, the central importance of endosomal acidification and V-ATPase activity on viral infection will be discussed in the context of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Amity F Eaton
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Maria Merkulova
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
28
|
Bovée DM, Cuevas CA, Zietse R, Danser AHJ, Mirabito Colafella KM, Hoorn EJ. Salt-sensitive hypertension in chronic kidney disease: distal tubular mechanisms. Am J Physiol Renal Physiol 2020; 319:F729-F745. [DOI: 10.1152/ajprenal.00407.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) causes salt-sensitive hypertension that is often resistant to treatment and contributes to the progression of kidney injury and cardiovascular disease. A better understanding of the mechanisms contributing to salt-sensitive hypertension in CKD is essential to improve these outcomes. This review critically explores these mechanisms by focusing on how CKD affects distal nephron Na+ reabsorption. CKD causes glomerulotubular imbalance with reduced proximal Na+ reabsorption and increased distal Na+ delivery and reabsorption. Aldosterone secretion further contributes to distal Na+ reabsorption in CKD and is not only mediated by renin and K+ but also by metabolic acidosis, endothelin-1, and vasopressin. CKD also activates the intrarenal renin-angiotensin system, generating intratubular angiotensin II to promote distal Na+ reabsorption. High dietary Na+ intake in CKD contributes to Na+ retention by aldosterone-independent activation of the mineralocorticoid receptor mediated through Rac1. High dietary Na+ also produces an inflammatory response mediated by T helper 17 cells and cytokines increasing distal Na+ transport. CKD is often accompanied by proteinuria, which contains plasmin capable of activating the epithelial Na+ channel. Thus, CKD causes both local and systemic changes that together promote distal nephron Na+ reabsorption and salt-sensitive hypertension. Future studies should address remaining knowledge gaps, including the relative contribution of each mechanism, the influence of sex, differences between stages and etiologies of CKD, and the clinical relevance of experimentally identified mechanisms. Several pathways offer opportunities for intervention, including with dietary Na+ reduction, distal diuretics, renin-angiotensin system inhibitors, mineralocorticoid receptor antagonists, and K+ or H+ binders.
Collapse
Affiliation(s)
- Dominique M. Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catharina A. Cuevas
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katrina M. Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Wang F, Sun Y, Luo R, Lu X, Yang B, Yang T. COX-2-independent activation of renal (pro)renin receptor contributes to DOCA-salt hypertension in rats. Am J Physiol Renal Physiol 2020; 319:F647-F653. [PMID: 32799674 PMCID: PMC7642891 DOI: 10.1152/ajprenal.00112.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022] Open
Abstract
It has been shown that cyclooxygenase (COX)-2-dependent activation of renal (pro)renin receptor (PRR) contributes to angiotensin II (ANG II)-induced hypertension. However, less is known about the involvement of this mechanism in ANG II-independent hypertension. The goal of the present study was to test whether or not COX-2-dependent upregulation of PRR serves as a universal mechanism contributing to ANG II-dependent and -independent hypertension. Here, we examined the association between renal COX-2 and PRR during deoxycorticosterone acetate (DOCA)-salt hypertension in rats. By immunoblot analysis and immunofluorescence, renal protein expression of PRR was remarkably upregulated by DOCA-salt treatment. Surprisingly, this upregulation of renal PRR expression was unaffected by a COX-2 inhibitor, celecoxib. To address the role of renal PRR to the pathogenesis of DOCA-salt hypertension, a decoy PRR inhibitor, PRO20, was infused to the renal medulla of uninephrectomized Sprague-Dawley rats for 14 days. Radiotelemetry demonstrated effective attenuation of DOCA-salt hypertension by intramedullary infusion of a PRR inhibitor, PRO20. In parallel, DOCA-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied with blunted polydipsia and polyuria. In contrast, intravenous infusion of PRO20 was less effective in attenuating DOCA-salt hypertension and cardiorenal injury. Together, these results suggest that COX-2-independent activation of renal PRR contributes to DOCA-salt hypertension.
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Ying Sun
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Xiaohan Lu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
30
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Oh ES, Bennett DA, Walston JD, Abadir PM. Brain Renin-Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front Neurosci 2020; 14:586314. [PMID: 33117127 PMCID: PMC7561440 DOI: 10.3389/fnins.2020.586314] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The renin–angiotensin system (RAS) was initially considered to be part of the endocrine system regulating water and electrolyte balance, systemic vascular resistance, blood pressure, and cardiovascular homeostasis. It was later discovered that intracrine and local forms of RAS exist in the brain apart from the endocrine RAS. This brain-specific RAS plays essential roles in brain homeostasis by acting mainly through four angiotensin receptor subtypes; AT1R, AT2R, MasR, and AT4R. These receptors have opposing effects; AT1R promotes vasoconstriction, proliferation, inflammation, and oxidative stress while AT2R and MasR counteract the effects of AT1R. AT4R is critical for dopamine and acetylcholine release and mediates learning and memory consolidation. Consequently, aging-associated dysregulation of the angiotensin receptor subtypes may lead to adverse clinical outcomes such as Alzheimer’s disease and frailty via excessive oxidative stress, neuroinflammation, endothelial dysfunction, microglial polarization, and alterations in neurotransmitter secretion. In this article, we review the brain RAS from this standpoint. After discussing the functions of individual brain RAS components and their intracellular and intracranial locations, we focus on the relationships among brain RAS, aging, frailty, and specific neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and vascular cognitive impairment, through oxidative stress, neuroinflammation, and vascular dysfunction. Finally, we discuss the effects of RAS-modulating drugs on the brain RAS and their use in novel treatment approaches.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatrics, Department of Internal Medicine, Ankara University School of Medicine, Ankara, Turkey.,Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Claudene J George
- Division of Geriatrics, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
31
|
Chen Y, Xu C. The interaction partners of (pro)renin receptor in the distal nephron. FASEB J 2020; 34:14136-14149. [PMID: 32975331 DOI: 10.1096/fj.202001711r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/11/2022]
Abstract
The (pro)renin receptor (PRR), a key regulator of intrarenal renin-angiotensin system (RAS), is predominantly presented in podocytes, proximal tubules, distal convoluted tubules, and the apical membrane of collecting duct A-type intercalated cells, and plays a crucial role in hypertension, cardiovascular disease, kidney disease, and fluid homeostasis. In addition to its well-known renin-regulatory function, increasing evidence suggests PRR can also act in a variety of intracellular signaling cascades independently of RAS in the renal medulla, including Wnt/β-catenin signaling, cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2 ) signaling, and the apelinergic system, and work as a component of the vacuolar H+ -ATPase. PRR and these pathways regulate the expression/activity of each other that controlling blood pressure and renal functions. In this review, we highlight recent findings regarding the antagonistic interaction between PRR and ELABELA/apelin, the mutually stimulatory relationship between PRR and COX-2/PGE2 or Wnt/β-catenin signaling in the renal medulla, and their involvement in the regulation of intrarenal RAS thereby control blood pressure, renal injury, and urine concentrating ability in health and patho-physiological conditions. We also highlight the latest progress in the involvement of PRR for the vacuolar H+ -ATPase activity.
Collapse
Affiliation(s)
- Yanting Chen
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Chuanming Xu
- Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA.,Center for Translational Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
32
|
Wang R, Long T, Hassan A, Wang J, Sun Y, Xie XS, Li X. Cryo-EM structures of intact V-ATPase from bovine brain. Nat Commun 2020; 11:3921. [PMID: 32764564 PMCID: PMC7414150 DOI: 10.1038/s41467-020-17762-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
The vacuolar-type H+-ATPases (V-ATPase) hydrolyze ATP to pump protons across the plasma or intracellular membrane, secreting acids to the lumen or acidifying intracellular compartments. It has been implicated in tumor metastasis, renal tubular acidosis, and osteoporosis. Here, we report two cryo-EM structures of the intact V-ATPase from bovine brain with all the subunits including the subunit H, which is essential for ATPase activity. Two type-I transmembrane proteins, Ac45 and (pro)renin receptor, along with subunit c", constitute the core of the c-ring. Three different conformations of A/B heterodimers suggest a mechanism for ATP hydrolysis that triggers a rotation of subunits DF, inducing spinning of subunit d with respect to the entire c-ring. Moreover, many lipid molecules have been observed in the Vo domain to mediate the interactions between subunit c, c", (pro)renin receptor, and Ac45. These two structures reveal unique features of mammalian V-ATPase and suggest a mechanism of V1-Vo torque transmission.
Collapse
Affiliation(s)
- Rong Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tao Long
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Abdirahman Hassan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jin Wang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yingyuan Sun
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiao-Song Xie
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
33
|
Cho HM, Kim I. Maternal high-fructose intake induces hypertension through activating histone codes on the (pro)renin receptor promoter. Biochem Biophys Res Commun 2020; 527:596-602. [PMID: 32423811 DOI: 10.1016/j.bbrc.2020.04.081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 12/29/2022]
Abstract
High-fructose intake induces hypertension via the renal expression of (pro)renin receptor (PRR) that stimulates the expression of sodium/hydrogen exchanger 3, Na/K/2Cl cotransporter 2, and genes of the intrarenal renin-angiotensin system. We hypothesize that maternal high-fructose intake induces hypertension in subsequent generation offspring through activating histone codes on the PRR promoter. Mice dams were offered 20% fructose solution during pregnancy and lactation, while the subsequent 1st to 4th generation offspring were raised without fructose. Blood pressure was measured via tail-cuff method. The mRNA and protein expression were determined using quantitative real-time polymerase chain reaction and western blotting, respectively. Histone modification was evaluated using a chromatin immunoprecipitation assay. Maternal high-fructose intake statistically significantly increased blood pressure in the 1st and 2nd generations of offspring compared to the control group. Expression levels of sodium transporters and PRR were increased in the kidneys of the 1st to 3rd generation offspring. Increased enrichment of active histone codes such as H3Ac and H3K4me2 but decreased enrichment of repressive histone codes such as H3K9me3 and H3K27me3 on the PRR promoter were observed in the 1st to 3rd not the 4th generation. Moreover, there was increased the mRNA expression for histone acetyltransferase and methyl transferases for H3K4 in the 1st and 2nd generation offspring compared to the control group. This study implicates that maternal high-fructose intake induces hypertension in multigenerational offspring through activating histone codes on the PRR promoter.
Collapse
Affiliation(s)
- Hyun Min Cho
- Department of Pharmacology, Daegu, Republic of Korea; Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - InKyeom Kim
- Department of Pharmacology, Daegu, Republic of Korea; Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
34
|
|
35
|
Sun Y, Goes Martini A, Janssen MJ, Garrelds IM, Masereeuw R, Lu X, Danser AHJ. Megalin: A Novel Endocytic Receptor for Prorenin and Renin. Hypertension 2020; 75:1242-1250. [PMID: 32200675 DOI: 10.1161/hypertensionaha.120.14845] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Megalin is an endocytic receptor contributing to protein reabsorption. Impaired expression or trafficking of megalin increases urinary renin and allowed the detection of prorenin, which normally is absent in urine. Here, we investigated (pro)renin uptake by megalin, using both conditionally immortalized proximal tubule epithelial cells and Brown Norway Rat yolk sac cells (BN16). To distinguish binding and internalization, cells were incubated with recombinant human (pro)renin at 4°C and 37°C, respectively. (Pro)renin levels were assessed by immunoradiometric assay. At 4°C, BN16 cells bound 3× more prorenin than renin, suggestive for a higher affinity of prorenin. Similarly, at 37°C, prorenin accumulated at 3- to 4-fold higher levels than renin in BN16 cells. Consequently, depletion of medium prorenin (but not renin) content occurred after 24 hours. No such differences were observed in conditionally immortalized proximal tubule epithelial cells, and M6P (mannose-6-phosphate) greatly reduced conditionally immortalized proximal tubule epithelial cells (pro)renin uptake, suggesting that these cells accumulate (pro)renin largely via M6P receptors. M6P did not affect (pro)renin uptake in BN16 cells. Yet, inhibiting megalin expression with siRNA greatly reduced (pro)renin binding and internalization by BN16 cells. Furthermore, treating BN16 cells with albumin, an endogenous ligand of megalin, also decreased binding and internalization of (pro)renin, while deleting the (pro)renin receptor affected the latter only. Exposing prorenin's prosegment with the renin inhibitor aliskiren dramatically increased prorenin binding, while after prosegment cleavage with trypsin prorenin binding was identical to that of renin. In conclusion, megalin might function as an endocytic receptor for (pro)renin and displays a preference for prorenin. Megalin-mediated endocytosis requires the (pro)renin receptor.
Collapse
Affiliation(s)
- Yuan Sun
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands (Y.S., A.G.M., I.M.G., A.H.J.D.).,Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (Y.S., X.L.).,Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China (Y.S.)
| | - Alexandre Goes Martini
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands (Y.S., A.G.M., I.M.G., A.H.J.D.)
| | - Manoe J Janssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands (M.J.J., R.M.)
| | - Ingrid M Garrelds
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands (Y.S., A.G.M., I.M.G., A.H.J.D.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands (M.J.J., R.M.)
| | - Xifeng Lu
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, China (Y.S., X.L.)
| | - A H Jan Danser
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands (Y.S., A.G.M., I.M.G., A.H.J.D.)
| |
Collapse
|
36
|
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
|
37
|
Abstract
Purpose of the Review The main goal of this article is to discuss how the development of state-of-the-art technology has made it possible to address fundamental questions related to how the renin-angiotensin system (RAS) operates within the brain from the neurophysiological and molecular perspective. Recent Findings The existence of the brain RAS remains surprisingly controversial. New sensitive in situ hybridization techniques and novel transgenic animals expressing reporter genes have provided pivotal information of the expression of RAS genes within the brain. We discuss studies using genetically engineered animals combined with targeted viral microinjections to study molecular mechanisms implicated in the regulation of the brain RAS. We also discuss novel drugs targeting the brain RAS that have shown promising results in clinical studies and trials. Summary Over the last 50 years, several new physiological roles of the brain RAS have been identified. In the coming years, efforts to incorporate cutting-edge technologies such as optogenetics, chemogenetics, and single-cell RNA sequencing will lead to dramatic advances in our full understanding of how the brain RAS operates at molecular and neurophysiological levels.
Collapse
|
38
|
Yoshida N, Endo J, Kinouchi K, Kitakata H, Moriyama H, Kataoka M, Yamamoto T, Shirakawa K, Morimoto S, Nishiyama A, Hashiguchi A, Higuchi I, Fukuda K, Ichihara A, Sano M. (Pro)renin receptor accelerates development of sarcopenia via activation of Wnt/YAP signaling axis. Aging Cell 2019; 18:e12991. [PMID: 31282603 PMCID: PMC6718617 DOI: 10.1111/acel.12991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/13/2019] [Accepted: 05/26/2019] [Indexed: 01/19/2023] Open
Abstract
To extend life expectancy and ensure healthy aging, it is crucial to prevent and minimize age‐induced skeletal muscle atrophy, also known as sarcopenia. However, the disease's molecular mechanism remains unclear. The age‐related Wnt/β‐catenin signaling pathway has been recently shown to be activated by the (pro)renin receptor ((P)RR). We report here that (P)RR expression was increased in the atrophied skeletal muscles of aged mice and humans. Therefore, we developed a gain‐of‐function model of age‐related sarcopenia via transgenic expression of (P)RR under control of the CAG promoter. Consistent with our hypothesis, (P)RR‐Tg mice died early and exhibited muscle atrophy with histological features of sarcopenia. Moreover, Wnt/β‐catenin signaling was activated and the regenerative capacity of muscle progenitor cells after cardiotoxin injury was impaired due to cell fusion failure in (P)RR‐Tg mice. In vitro forced expression of (P)RR protein in C2C12 myoblast cells suppressed myotube formation by activating Wnt/β‐catenin signaling. Administration of Dickkopf‐related protein 1, an inhibitor of Wnt/β‐catenin signaling, and anti‐(P)RR neutralizing antibody, which inhibits binding of (P)RR to the Wnt receptor, significantly improved sarcopenia in (P)RR‐Tg mice. Furthermore, the use of anti‐(P)RR neutralizing antibodies significantly improved the regenerative ability of skeletal muscle in aged mice. Finally, we show that Yes‐associated protein (YAP) signaling, which is coordinately regulated by Wnt/β‐catenin, contributed to the development of (P)RR‐induced sarcopenia. The present study demonstrates the use of (P)RR‐Tg mice as a novel sarcopenia model, and shows that (P)RR‐Wnt‐YAP signaling plays a pivotal role in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Naohiro Yoshida
- Department of Endocrinology and Hypertension Tokyo Women’s Medical University Tokyo Japan
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Jin Endo
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Kenichiro Kinouchi
- Department of Internal Medicine, School of Medicine Keio University Tokyo Japan
| | - Hiroki Kitakata
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Hidenori Moriyama
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Masaharu Kataoka
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Tsunehisa Yamamoto
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Kohsuke Shirakawa
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension Tokyo Women’s Medical University Tokyo Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine Kagawa University Kagawa Japan
| | - Akihiro Hashiguchi
- Department of Neurology and Geriatrics, Graduate School of Medical and Dental Sciences Kagoshima University Kagoshima Japan
| | - Itsuro Higuchi
- School of Health Sciences, Faculty of Medicine Kagoshima University Kagoshima Japan
| | - Keiichi Fukuda
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension Tokyo Women’s Medical University Tokyo Japan
| | - Motoaki Sano
- Department of Cardiology, School of Medicine Keio University Tokyo Japan
| |
Collapse
|
39
|
Atp6ap2 deletion causes extensive vacuolation that consumes the insulin content of pancreatic β cells. Proc Natl Acad Sci U S A 2019; 116:19983-19988. [PMID: 31527264 DOI: 10.1073/pnas.1903678116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pancreatic β cells store insulin within secretory granules which undergo exocytosis upon elevation of blood glucose levels. Crinophagy and autophagy are instead responsible to deliver damaged or old granules to acidic lysosomes for intracellular degradation. However, excessive consumption of insulin granules can impair β cell function and cause diabetes. Atp6ap2 is an essential accessory component of the vacuolar ATPase required for lysosomal degradative functions and autophagy. Here, we show that Cre recombinase-mediated conditional deletion of Atp6ap2 in mouse β cells causes a dramatic accumulation of large, multigranular vacuoles in the cytoplasm, with reduction of insulin content and compromised glucose homeostasis. Loss of insulin stores and gigantic vacuoles were also observed in cultured insulinoma INS-1 cells upon CRISPR/Cas9-mediated removal of Atp6ap2. Remarkably, these phenotypic alterations could not be attributed to a deficiency in autophagy or acidification of lysosomes. Together, these data indicate that Atp6ap2 is critical for regulating the stored insulin pool and that a balanced regulation of granule turnover is key to maintaining β cell function and diabetes prevention.
Collapse
|
40
|
Trepiccione F. ERA-EDTA fellowship, a 'bonne opportunité': the scientific and human experience of a fellow. Clin Kidney J 2019; 12:465-467. [PMID: 31384435 PMCID: PMC6671323 DOI: 10.1093/ckj/sfy123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Indexed: 12/02/2022] Open
Abstract
As a fellow of the ERA-EDTA long-term fellowship programme, I spent two fantastic years as a post-doc in Prof. D. Eladari’s laboratory at Inserm U970, Paris-Cardiovascular Research Centre. It was a highly formative and productive scientific experience. On a personal level, immersion into the French society and the international environment of the laboratory were added bonuses that enriched my experience. I am honoured to report my experience here from the ERA-EDTA fellowship programme. I hope this will inspire young fellows to apply for such a programme and pursue their career in science. Good mentorship, a passion for scientific investigation and determination are required.
Collapse
Affiliation(s)
- Francesco Trepiccione
- Biogem Scarl, Istituto di Ricerche Gaetano Salvatore, Ariano Irpino, Italy.,Department of Medical Translational Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| |
Collapse
|
41
|
Renin Activity in Heart Failure with Reduced Systolic Function-New Insights. Int J Mol Sci 2019; 20:ijms20133182. [PMID: 31261774 PMCID: PMC6651297 DOI: 10.3390/ijms20133182] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Regardless of the cause, symptomatic heart failure (HF) with reduced ejection fraction (rEF) is characterized by pathological activation of the renin–angiotensin–aldosterone system (RAAS) with sodium retention and extracellular fluid expansion (edema). Here, we review the role of active renin, a crucial, upstream enzymatic regulator of the RAAS, as a prognostic and diagnostic plasma biomarker of heart failure with reduced ejection fraction (HFrEF) progression; we also discuss its potential as a pharmacological bio-target in HF therapy. Clinical and experimental studies indicate that plasma renin activity is elevated with symptomatic HFrEF with edema in patients, as well as in companion animals and experimental models of HF. Plasma renin activity levels are also reported to be elevated in patients and animals with rEF before the development of symptomatic HF. Modulation of renin activity in experimental HF significantly reduces edema formation and the progression of systolic dysfunction and improves survival. Thus, specific assessment and targeting of elevated renin activity may enhance diagnostic and therapeutic precision to improve outcomes in appropriate patients with HFrEF.
Collapse
|
42
|
Foco L, Pattaro C. Genetics of Blood Pressure Regulation: Possible Paths in the Labyrinth. Am J Kidney Dis 2019; 74:421-424. [PMID: 31221530 DOI: 10.1053/j.ajkd.2019.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/03/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Luisa Foco
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | | |
Collapse
|
43
|
Zhao L, Zhao Y, Tang FL, Xiong L, Su C, Mei L, Zhu XJ, Xiong WC. pHluorin-BACE1-mCherry Acts as a Reporter for the Intracellular Distribution of Active BACE1 In Vitro and In Vivo. Cells 2019; 8:E474. [PMID: 31108937 PMCID: PMC6562731 DOI: 10.3390/cells8050474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022] Open
Abstract
β-site APP-cleaving enzyme 1 (BACE1) initiates amyloid precursor protein (APP) cleavage and β-amyloid (Aβ) production, a critical step in the pathogenesis of Alzheimer's disease (AD). It is thus of considerable interest to investigate how BACE1 activity is regulated. BACE1 has its maximal activity at acidic pH and GFP variant-pHluorin-displays pH dependence. In light of these observations, we generated three tandem fluorescence-tagged BACE1 fusion proteins, named pHluorin-BACE1-mCherry, BACE1-mCherry-pHluorin and BACE1-mCherry-EGFP. Comparing the fluorescence characteristics of these proteins in response to intracellular pH changes induced by chloroquine or bafilomycin A1, we found that pHluorin-BACE1-mCherry is a better pH sensor for BACE1 because its fluorescence intensity responds to pH changes more dramatically and more quickly. Additionally, we found that (pro)renin receptor (PRR), a subunit of the v-ATPase complex, which is critical for maintaining vesicular pH, regulates pHluorin's fluorescence and BACE1 activity in pHluorin-BACE1-mCherry expressing cells. Finally, we found that the expression of Swedish mutant APP (APPswe) suppresses pHluorin fluorescence in pHluorin-BACE1-mCherry expressing cells in culture and in vivo, implicating APPswe not only as a substrate but also as an activator of BACE1. Taken together, these results suggest that the pHluorin-BACE1-mCherry fusion protein may serve as a useful tool for visualizing active/inactive BACE1 in culture and in vivo.
Collapse
Affiliation(s)
- Lu Zhao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Yang Zhao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Fu-Lei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Ce Su
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Xiao-Juan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
44
|
Maternal High Fat Diet and in-Utero Metformin Exposure Significantly Impact upon the Fetal Renal Proteome of Male Mice. J Clin Med 2019; 8:jcm8050663. [PMID: 31083566 PMCID: PMC6571731 DOI: 10.3390/jcm8050663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
There is accumulating evidence for fetal programming of later kidney disease by maternal obesity or associated conditions. We performed a hypothesis-generating study to identify potentially underlying mechanisms. Female mice were randomly split in two groups and fed either a standard diet (SD) or high fat diet (HFD) from weaning until mating and during pregnancy. Half of the dams from both groups were treated with metformin ((M), 380 mg/kg), resulting in four experimental groups (SD, SD-M, HFD, HFD-M). Caesarean section was performed on gestational day 18.5. Fetal kidney tissue was isolated from cryo-slices using laser microdissection methods and a proteomic screen was performed. For single proteins, a fold change ≥1.5 and q-value <0.05 were considered to be statistically significant. Interestingly, HFD versus SD had a larger effect on the proteome of fetal kidneys (56 proteins affected; interaction clusters shown for proteins concerning transcription/translation, mitochondrial processes, eicosanoid metabolism, H2S-synthesis and membrane remodeling) than metformin exposure in either SD (29 proteins affected; clusters shown for proteins involved in transcription/translation) or HFD (6 proteins affected; no cluster). By further analysis, ATP6V1G1, THY1, PRKCA and NDUFB3 were identified as the most promising candidates potentially mediating reprogramming effects of metformin in a maternal high fat diet.
Collapse
|
45
|
Abstract
Purpose of Review Although an independent brain renin-angiotensin system is often assumed to exist, evidence for this concept is weak. Most importantly, renin is lacking in the brain, and both brain angiotensinogen and angiotensin (Ang) II levels are exceptionally low. In fact, brain Ang II levels may well represent uptake of circulating Ang II via Ang II type 1 (AT1) receptors. Recent Findings Nevertheless, novel drugs are now aimed at the brain RAS, i.e., aminopeptidase A inhibitors should block Ang III formation from Ang II, and hence diminish AT1 receptor stimulation by Ang III, while AT2 and Mas receptor agonists are reported to induce neuroprotection after stroke. The endogenous agonists of these receptors and their origin remain unknown. Summary This review addresses the questions whether independent angiotensin generation truly occurs in the brain, what its relationship with the kidney is, and how centrally acting RAS blockers/agonists might work.
Collapse
Affiliation(s)
- Liwei Ren
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Xifeng Lu
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
46
|
Wang F, Xu C, Luo R, Peng K, Ramkumar N, Xie S, Lu X, Zhao L, Zuo CJ, Kohan DE, Yang T. Site-1 protease-derived soluble (pro)renin receptor targets vasopressin receptor 2 to enhance urine concentrating capability. JCI Insight 2019; 4:124174. [PMID: 30944256 PMCID: PMC6483716 DOI: 10.1172/jci.insight.124174] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
The antidiuretic hormone vasopressin (AVP), acting through its type 2 receptor (V2R) in the collecting duct (CD), critically controls urine concentrating capability. Here, we report that site-1 protease-derived (S1P-derived) soluble (pro)renin receptor (sPRR) participates in regulation of fluid homeostasis via targeting V2R. In cultured inner medullary collecting duct (IMCD) cells, AVP-induced V2R expression was blunted by a PRR antagonist, PRO20; a PRR-neutralizing antibody; or a S1P inhibitor, PF-429242. In parallel, sPRR release was increased by AVP and reduced by PF-429242. Administration of histidine-tagged sPRR, sPRR-His, stimulated V2R expression and also reversed the inhibitory effect of PF-429242 on the expression induced by AVP. PF-429242 treatment in C57/BL6 mice impaired urine concentrating capability, which was rescued by sPRR-His. This observation was recapitulated in mice with renal tubule-specific deletion of S1P. During the pharmacological or genetic manipulation of S1P alone or in combination with sPRR-His, the changes in urine concentration were paralleled with renal expression of V2R and aquaporin-2 (AQP2). Together, these results support that S1P-derived sPRR exerts a key role in determining renal V2R expression and, thus, urine concentrating capability.
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chuanming Xu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kexin Peng
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Nirupama Ramkumar
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Shiying Xie
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaohan Lu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Long Zhao
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Chang-Jiang Zuo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Donald E. Kohan
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension and Renal Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Han L, Li M, Wang H, Lu G, Li P. Mutation of ATP6V0A4 Gene Leads to Acid-base Disturbance and
Inferred in Kidney Stone Formation. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.377.384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
48
|
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: 36] [Impact Index Per Article: 6.0] [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
|
49
|
Electrolyte transport in the renal collecting duct and its regulation by the renin-angiotensin-aldosterone system. Clin Sci (Lond) 2019; 133:75-82. [PMID: 30622159 DOI: 10.1042/cs20180194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 01/13/2023]
Abstract
Distal nephron of the kidney plays key roles in fluid volume and electrolyte homeostasis by tightly regulating reabsorption and excretion of Na+, K+, and Cl- Studies to date demonstrate the detailed electrolyte transport mechanisms in principal cells of the cortical collecting duct, and their regulation by renin-angiotensin-aldosterone system (RAAS). In recent years, however, accumulating data indicate that intercalated cells, another cell type that is present in the cortical collecting duct, also play active roles in the regulation of blood pressure. Notably, pendrin in β-intercalated cells not only controls acid/base homeostasis, but is also one of the key components controlling salt and K+ transport in distal nephron. We have recently shown that pendrin is regulated by the co-ordinated action of angiotensin II (AngII) and aldosterone, and at the downstream of AngII, mammalian target of rapamycin (mTOR) signaling regulates pendrin through inhibiting the kinase unc51-like-kinase 1 and promoting dephosphorylation of mineralocorticoid receptor (MR). In this review, we summarize recent advances in the current knowledge on the salt transport mechanisms in the cortical collecting duct, and their regulation by the RAAS.
Collapse
|
50
|
Hoffmann S, Mullins L, Buckley C, Rider S, Mullins J. Investigating the RAS can be a fishy business: interdisciplinary opportunities using Zebrafish. Clin Sci (Lond) 2018; 132:2469-2481. [PMID: 30518571 PMCID: PMC6279434 DOI: 10.1042/cs20180721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/02/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023]
Abstract
The renin-angiotensin system (RAS) is highly conserved, and components of the RAS are present in all vertebrates to some degree. Although the RAS has been studied since the discovery of renin, its biological role continues to broaden with the identification and characterization of new peptides. The evolutionarily distant zebrafish is a remarkable model for studying the kidney due to its genetic tractability and accessibility for in vivo imaging. The zebrafish pronephros is an especially useful kidney model due to its structural simplicity yet complex functionality, including capacity for glomerular and tubular filtration. Both the pronephros and mesonephros contain renin-expressing perivascular cells, which respond to RAS inhibition, making the zebrafish an excellent model for studying the RAS. This review summarizes the physiological and genetic tools currently available for studying the zebrafish kidney with regards to functionality of the RAS, using novel imaging techniques such as SPIM microscopy coupled with targeted single cell ablation and synthesis of vasoactive RAS peptides.
Collapse
Affiliation(s)
- Scott Hoffmann
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Linda Mullins
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Charlotte Buckley
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Sebastien Rider
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - John Mullins
- University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, U.K.
| |
Collapse
|