1
|
Luo R, Yang KT, Wang F, Zheng H, Yang T. Collecting Duct Pro(Renin) Receptor Contributes to Unilateral Ureteral Obstruction-Induced Kidney Injury via Activation of the Intrarenal RAS. Hypertension 2024; 81:2152-2161. [PMID: 39171392 PMCID: PMC11410543 DOI: 10.1161/hypertensionaha.123.21740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 05/10/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Although the concept of the intrarenal renin-angiotensin system (RAS) in renal disease is well-described in the literature, the precise pathogenic role and mechanism of this local system have not been directly assessed in the absence of confounding influence from the systemic RAS. The present study used novel mouse models of collecting duct (CD)-specific deletion of (pro)renin receptor (PRR) or renin together with pharmacological inhibition of soluble PRR production to unravel the precise contribution of the intrarenal RAS to renal injury induced by unilateral ureteral obstruction. METHODS We examined the impact of CD-specific deletion of PRR, CD-specific deletion of renin, and S1P (site-1 protease) inhibitor PF429242 treatment on renal fibrosis and inflammation and the indices of the intrarenal RAS in a mouse model of unilateral ureteral obstruction. RESULTS After 3 days of unilateral ureteral obstruction, the indices of the intrarenal RAS including the renal medullary renin content, activity and mRNA expression, and Ang (angiotensin) II content in obstructed kidneys of floxed mice were all increased. That effect was reversed with CD-specific deletion of PRR, CD-specific deletion of renin, and PF429242 treatment, accompanied by consistent improvement in renal fibrosis and inflammation. On the other hand, renal cortical renin levels were unaffected by unilateral ureteral obstruction, irrespective of the genotype. Similar results were obtained via pharmacological inhibition of S1P, the key protease for the generation of soluble PRR. CONCLUSIONS Our results reveal that PRR-dependent/soluble PRR-dependent activation of CD renin represents a key determinant of the intrarenal RAS and, thus, obstruction-induced renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Renfei Luo
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Kevin T. Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Fei Wang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Huaqing Zheng
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
Chelangarimiyandoab F, Mungara P, Batta M, Cordat E. Urinary Tract Infections: Renal Intercalated Cells Protect against Pathogens. J Am Soc Nephrol 2023; 34:1605-1614. [PMID: 37401780 PMCID: PMC10561816 DOI: 10.1681/asn.0000000000000187] [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: 01/19/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Urinary tract infections affect more than 1 in 2 women during their lifetime. Among these, more than 10% of patients carry antibiotic-resistant bacterial strains, highlighting the urgent need to identify alternative treatments. While innate defense mechanisms are well-characterized in the lower urinary tract, it is becoming evident that the collecting duct (CD), the first renal segment encountered by invading uropathogenic bacteria, also contributes to bacterial clearance. However, the role of this segment is beginning to be understood. This review summarizes the current knowledge on CD intercalated cells in urinary tract bacterial clearance. Understanding the innate protective role of the uroepithelium and of the CD offers new opportunities for alternative therapeutic strategies.
Collapse
Affiliation(s)
- Forough Chelangarimiyandoab
- Department of Physiology and Membrane Protein Disease Research Group, Faculty of Medicine & Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
3
|
Daniel EA, Sommer NA, Sharma M. Polycystic kidneys: interaction of notch and renin. Clin Sci (Lond) 2023; 137:1145-1150. [PMID: 37553961 PMCID: PMC11132639 DOI: 10.1042/cs20230023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023]
Abstract
Polycystic kidney disease (PKD) is a developmental disorder, which either manifests in early childhood or later in life, depending on the genetic mutation one harbors. The mechanisms of cyst initiation are not well understood. Increasing literature is now suggesting that Notch signaling may play a critical role in PKD. Activation of Notch signaling is important during nephrogenesis and slows down after development. Deletion of various Notch molecules in the cap mesenchyme leads to formation of cysts and early death in mice. A new study by Belyea et al. has now found that cells of renin lineage may link Notch expression and cystic kidney disease. Here, we use our understanding of Notch signaling and PKD to speculate about the significance of these interactions.
Collapse
Affiliation(s)
- Emily A Daniel
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| | - Nicole A Sommer
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| | - Madhulika Sharma
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| |
Collapse
|
4
|
Lara LS, Gonzalez AA, Hennrikus MT, Prieto MC. Hormone-Dependent Regulation of Renin and Effects on Prorenin Receptor Signaling in the Collecting Duct. Curr Hypertens Rev 2022; 18:91-100. [PMID: 35170417 PMCID: PMC10132771 DOI: 10.2174/1573402118666220216105357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/22/2021] [Accepted: 12/13/2021] [Indexed: 01/27/2023]
Abstract
The production of renin by the principal cells of the collecting duct has widened our understanding of the regulation of intrarenal angiotensin II (Ang II) generation and blood pressure. In the collecting duct, Ang II increases the synthesis and secretion of renin by mechanisms involving the activation of Ang II type 1 receptor (AT1R) via stimulation of the PKCα, Ca2+, and cAMP/PKA/CREB pathways. Additionally, paracrine mediators, including vasopressin (AVP), prostaglandins, bradykinin (BK), and atrial natriuretic peptide (ANP), regulate renin in principal cells. During Ang II-dependent hypertension, despite plasma renin activity suppression, renin and prorenin receptor (RPR) are upregulated in the collecting duct and promote de novo formation of intratubular Ang II. Furthermore, activation of PRR by its natural agonists, prorenin and renin, may contribute to the stimulation of profibrotic factors independent of Ang II. Thus, the interactions of RAS components with paracrine hormones within the collecting duct enable tubular compartmentalization of the RAS to orchestrate complex mechanisms that increase intrarenal Ang II, Na+ reabsorption, and blood pressure.
Collapse
Affiliation(s)
- Lucienne S Lara
- Instituto de Ciencias Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Matthew T Hennrikus
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane Renal and Hypertension Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
5
|
Yang T. Revisiting the relationship between (Pro)Renin receptor and the intrarenal RAS: focus on the soluble receptor. Curr Opin Nephrol Hypertens 2022; 31:351-357. [PMID: 35703290 PMCID: PMC9286065 DOI: 10.1097/mnh.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The (pro)renin receptor (PRR), also termed as ATPase H+ transporting accessory protein 2 (ATP6AP2), was originally cloned as a specific receptor for prorenin and renin [together called (pro)renin]. Given the wide tissue distribution of PRR, PRR was further postulated to act as a regulator of tissue renin. However, assigning a physiological role of PRR within the renin-angiotensin system (RAS) has been challenging largely due to its pleotropic functions in regulation of embryogenesis, autophagy, and H+ transport. The current review will summarize recent advances in understanding the roles of sPPR within the intrarenal RAS as well as those outside this local system. RECENT FINDINGS Site-1 protease (S1P) is a predominant source of sPPR at least in the kidney. So far most of the known physiological functions of PRR including renal handling of electrolytes and fluid and blood pressure are mediated by sPRR. In particular, sPRR serves as a positive regulator of collecting duct renin to activate the intrarenal RAS during water deprivation or angiotensin-II (AngII) infusion. However, PRR/sPRR can act in renin-independent manner under other circumstances. SUMMARY S1P-derived sPRR has emerged as a key regulator of kidney function and blood pressure and its relationship with the intrarenal RAS depends on the physiological context.
Collapse
Affiliation(s)
- Tianxin Yang
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
6
|
Paterson MR, Jackson KL, Dona MSI, Farrugia GE, Visniauskas B, Watson AMD, Johnson C, Prieto MC, Evans RG, Charchar F, Pinto AR, Marques FZ, Head GA. Deficiency of MicroRNA-181a Results in Transcriptome-Wide Cell-Specific Changes in the Kidney and Increases Blood Pressure. Hypertension 2021; 78:1322-1334. [PMID: 34538100 PMCID: PMC8573069 DOI: 10.1161/hypertensionaha.121.17384] [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] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Madeleine R. Paterson
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia; Monash University, Melbourne, Australia
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Kristy L. Jackson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University Parkville, Australia
| | - Malathi S. I. Dona
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Gabriella E. Farrugia
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Bruna Visniauskas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, the USA
| | - Anna M. D. Watson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Chad Johnson
- Monash Micro Imaging, Monash University, Melbourne, Australia
| | - Minolfa C. Prieto
- Department of Physiology, School of Medicine, Tulane University, New Orleans, the USA
| | - Roger G. Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Fadi Charchar
- Health Innovation and Transformation Centre, Federation University, Ballarat, Australia
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | - Alexander R. Pinto
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University Parkville, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Francine Z. Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia; Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Geoffrey A. Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Pharmacology, Monash University, Melbourne, Australia
| |
Collapse
|
7
|
Renin-Angiotensin System Induced Secondary Hypertension: The Alteration of Kidney Function and Structure. Int J Nephrol 2021. [PMID: 31628476 PMCID: PMC8505109 DOI: 10.1155/2021/5599754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Long-term hypertension is known as a major risk factor for cardiovascular and chronic kidney disease (CKD). The Renin-angiotensin system (RAS) plays a key role in hypertension pathogenesis. Angiotensin II (Ang II) enhancement in Ang II-dependent hypertension leads to progressive CKD and kidney fibrosis. In the two-kidney one-clip model (2K1C), more renin is synthesized in the principal cells of the collecting duct than juxtaglomerular cells (JGCs). An increase of renal Ang I and Ang II levels and a decrease of renal cortical and medullary Ang 1–7 occur in both kidneys of the 2K1C hypertensive rat model. In addition, the activity of the angiotensin-converting enzyme (ACE) increases, while ACE2's activity decreases in the medullary region of both kidneys in the 2K1C hypertensive model. Also, the renal prolyl carboxypeptidase (PrCP) expression and its activity reduce in the clipped kidneys. The imbalance in the production of renal ACE, ACE2, and PrCP expression causes the progression of renal injury. Intrarenal angiotensinogen (AGT) expression and urine AGT (uAGT) excretion rates in the unclipped kidney are greater than the clipped kidney in the 2K1C hypertensive rat model. The enhancement of Ang II in the clipped kidney is related to renin secretion, while the elevation of intrarenal Ang II in the unclipped kidney is related to stimulation of AGT mRNA and protein in proximal tubule cells by a direct effect of systemic Ang II level. Ang II-dependent hypertension enhances macrophages and T-cell infiltration into the kidney which increases cytokines, and AGT synthesis in proximal tubules is stimulated via cytokines. Accumulation of inflammatory cells in the kidney aggravates hypertension and renal damage. Moreover, Ang II-dependent hypertension alters renal Ang II type 1 & 2 receptors (AT1R & AT2R) and Mas receptor (MasR) expression, and the renal interstitial fluid bradykinin, nitric oxide, and cGMP response to AT1R, AT2R, or BK B2-receptor antagonists. Based on a variety of sources including PubMed, Google Scholar, Scopus, and Science-Direct, in the current review, we will discuss the role of RAS-induced secondary hypertension on the alteration of renal function.
Collapse
|
8
|
Kessel F, Steglich A, Hickmann L, Lira-Martinez R, Gerlach M, Sequeira-Lopez ML, Gomez RA, Hugo C, Todorov VT. Patterns of differentiation of renin lineage cells during nephrogenesis. Am J Physiol Renal Physiol 2021; 321:F378-F388. [PMID: 34338032 DOI: 10.1152/ajprenal.00151.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Developmentally heterogeneous renin expressing cells serve as progenitors for mural, glomerular and tubular cells during nephrogenesis and are collectively termed renin lineage cells (RLCs). In this study, we quantified different renal vascular and tubular cell types based on specific markers, assessed proliferation, and de-novo differentiation in the RLC population. We used kidney sections of mRenCre-mT/mG mice throughout nephrogenesis. Marker positivity was evaluated in whole digitalized sections. At embryonic day 16, RLCs appeared in the developing kidney, and expression of all stained markers in RLCs was observed. The proliferation rate of RLCs did not differ from the proliferation rate of non-RLCs. The RLCs expanded mainly by de-novo differentiation (neogenesis). The fractions of RLCs originating from the stromal progenitors of the metanephric mesenchyme (renin producing cells, vascular smooth muscle cells, mesangial cells) decreased during nephrogenesis. In contrast, aquaporin 2 positive RLCs in the collecting duct system that embryonically emerges almost exclusively from the ureteric bud, expanded postpartum. The cubilin positive RLC fraction in the proximal tubule, deriving from the cap mesenchyme, remained constant. During nephrogenesis, RLCs were continuously detectable in the vascular and tubular compartments of the kidney. Therein, various patterns of RLC differentiation that depend on the embryonic origin of the cells were identified.
Collapse
Affiliation(s)
- Friederike Kessel
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Anne Steglich
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Linda Hickmann
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Institute of Physiology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ricardo Lira-Martinez
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Gerlach
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Core Facility Cellular Imaging (CFCI), University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Maria Luisa Sequeira-Lopez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - R Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Christian Hugo
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vladimir T Todorov
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
9
|
High glucose induces trafficking of prorenin receptor and stimulates profibrotic factors in the collecting duct. Sci Rep 2021; 11:13815. [PMID: 34226610 PMCID: PMC8257763 DOI: 10.1038/s41598-021-93296-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Growing evidence indicates that prorenin receptor (PRR) is upregulated in collecting duct (CD) of diabetic kidney. Prorenin is secreted by the principal CD cells, and is the natural ligand of the PRR. PRR activation stimulates fibrotic factors, including fibronectin, collagen, and transforming growth factor-β (TGF-β) contributing to tubular fibrosis. However, whether high glucose (HG) contributes to this effect is unknown. We tested the hypothesis that HG increases the abundance of PRR at the plasma membrane of the CD cells, thus contributing to the stimulation of downstream fibrotic factors, including TGF-β, collagen I, and fibronectin. We used streptozotocin (STZ) male Sprague–Dawley rats to induce hyperglycemia for 7 days. At the end of the study, STZ-induced rats showed increased prorenin, renin, and angiotensin (Ang) II in the renal inner medulla and urine, along with augmented downstream fibrotic factors TGF-β, collagen I, and fibronectin. STZ rats showed upregulation of PRR in the renal medulla and preferential distribution of PRR on the apical aspect of the CD cells. Cultured CD M-1 cells treated with HG (25 mM for 1 h) showed increased PRR in plasma membrane fractions compared to cells treated with normal glucose (5 mM). Increased apical PRR was accompanied by upregulation of TGF-β, collagen I, and fibronectin, while PRR knockdown prevented these effects. Fluorescence resonance energy transfer experiments in M-1 cells demonstrated augmented prorenin activity during HG conditions. The data indicate HG stimulates profibrotic factors by inducing PRR translocation to the plasma membrane in CD cells, which in perspective, might be a novel mechanism underlying the development of tubulointerstitial fibrosis in diabetes mellitus.
Collapse
|
10
|
Wang F, Chen Y, Zou CJ, Luo R, Yang T. Mutagenesis of the Cleavage Site of Pro Renin Receptor Abrogates Angiotensin II-Induced Hypertension in Mice. Hypertension 2021; 78:115-127. [PMID: 34024121 PMCID: PMC9212214 DOI: 10.1161/hypertensionaha.121.16770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Yanting Chen
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Chang-jiang Zou
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Feng Y, Peng K, Luo R, Wang F, Yang T. Site-1 Protease-Derived Soluble (Pro)Renin Receptor Contributes to Angiotensin II-Induced Hypertension in Mice. Hypertension 2021; 77:405-416. [PMID: 33280408 PMCID: PMC7803453 DOI: 10.1161/hypertensionaha.120.15100] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of PRR ([pro]renin receptor) contributes to enhancement of intrarenal RAS and renal medullary α-ENaC and thus elevated blood pressure during Ang II (angiotensin II) infusion. The goal of the present study was to test whether such action of PRR was mediated by sPRR (soluble PRR), generated by S1P (site-1 protease), a newly identified PRR cleavage protease. F1 B6129SF1/J mice were infused for 6 days with control or Ang II at 300 ng/kg per day alone or in combination with S1P inhibitor PF-429242 (PF), and blood pressure was monitored by radiotelemetry. S1P inhibition significantly attenuated Ang II-induced hypertension accompanied with suppressed urinary and renal medullary renin levels and expression of renal medullary but not renal cortical α-ENaC expression. The effects of S1P inhibition were all reversed by supplement with histidine-tagged sPRR termed as sPRR-His. Ussing chamber technique was performed to determine amiloride-sensitive short-circuit current, an index of ENaC activity in confluent mouse cortical collecting duct cell line cells exposed for 24 hours to Ang II, Ang II + PF, or Ang II + PF + sPRR-His. Ang II-induced ENaC activity was blocked by PF, which was reversed by sPRR-His. Together, these results support that S1P-derived sPRR mediates Ang II-induced hypertension through enhancement of intrarenal renin level and activation of ENaC.
Collapse
Affiliation(s)
- Ye Feng
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Kexin Peng
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Renfei Luo
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Fei Wang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Tianxin Yang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City
| |
Collapse
|
12
|
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
|
13
|
Steglich A, Hickmann L, Linkermann A, Bornstein S, Hugo C, Todorov VT. Beyond the Paradigm: Novel Functions of Renin-Producing Cells. Rev Physiol Biochem Pharmacol 2020; 177:53-81. [PMID: 32691160 DOI: 10.1007/112_2020_27] [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] [Indexed: 12/14/2022]
Abstract
The juxtaglomerular renin-producing cells (RPC) of the kidney are referred to as the major source of circulating renin. Renin is the limiting factor in renin-angiotensin system (RAS), which represents a proteolytic cascade in blood plasma that plays a central role in the regulation of blood pressure. Further cells disseminated in the entire organism express renin at a low level as part of tissue RASs, which are thought to locally modulate the effects of systemic RAS. In recent years, it became increasingly clear that the renal RPC are involved in developmental, physiological, and pathophysiological processes outside RAS. Based on recent experimental evidence, a novel concept emerges postulating that next to their traditional role, the RPC have non-canonical RAS-independent progenitor and renoprotective functions. Moreover, the RPC are part of a widespread renin lineage population, which may act as a global stem cell pool coordinating homeostatic, stress, and regenerative responses throughout the organism. This review focuses on the RAS-unrelated functions of RPC - a dynamic research area that increasingly attracts attention.
Collapse
Affiliation(s)
- Anne Steglich
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Linda Hickmann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Linkermann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bornstein
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Christian Hugo
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vladimir T Todorov
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|
14
|
Abstract
The epithelium of the kidney collecting duct (CD) is composed mainly of two different types of cells with distinct and complementary functions. CD principal cells traditionally have been considered to have a major role in Na+ and water regulation, while intercalated cells (ICs) were thought to largely modulate acid-base homeostasis. In recent years, our understanding of IC function has improved significantly owing to new research findings. Thus, we now have a new model for CD transport that integrates mechanisms of salt and water reabsorption, K+ homeostasis, and acid-base status between principal cells and ICs. There are three main types of ICs (type A, type B, and non-A, non-B), which first appear in the late distal convoluted tubule or in the connecting segment in a species-dependent manner. ICs can be detected in CD from cortex to the initial part of the inner medulla, although some transport proteins that are key components of ICs also are present in medullary CD, cells considered inner medullary. Of the three types of ICs, each has a distinct morphology and expresses different complements of membrane transport proteins that translate into very different functions in homeostasis and contributions to CD luminal pro-urine composition. This review includes recent discoveries in IC intracellular and paracrine signaling that contributes to acid-base regulation as well as Na+, Cl-, K+, and Ca2+ homeostasis. Thus, these new findings highlight the potential role of ICs as targets for potential hypertension treatments.
Collapse
Affiliation(s)
- Renee Rao
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Núria M Pastor-Soler
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA.
| |
Collapse
|
15
|
Leite APO, Aragão DS, Nogueira MD, Pereira RO, Jara ZP, Fiorino P, Casarini DE, Farah V. Modulation of renin angiotensin system components by high glucose levels in the culture of collecting duct cells. J Cell Physiol 2019; 234:22809-22818. [DOI: 10.1002/jcp.28845] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 11/11/2022]
Affiliation(s)
- A. P. O. Leite
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Danielle S. Aragão
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Marie D. Nogueira
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Renata O. Pereira
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Zaira P. Jara
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Department of Molecular Cardiology Lerner Research Institute—Cleveland Clinic Cleveland Ohio
| | - Patricia Fiorino
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Dulce E. Casarini
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Vera Farah
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| |
Collapse
|
16
|
Nephroprotective effects of nebivolol in 2K1C rats through regulation of the kidney ROS-ADMA-NO pathway. Pharmacol Rep 2018; 70:917-929. [DOI: 10.1016/j.pharep.2018.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/25/2018] [Accepted: 04/12/2018] [Indexed: 01/20/2023]
|
17
|
Torres Fernandez ED, Adams KV, Syed M, Maranon RO, Romero DG, Yanes Cardozo LL. Long-Lasting Androgen-Induced Cardiometabolic Effects in Polycystic Ovary Syndrome. J Endocr Soc 2018; 2:949-964. [PMID: 30087950 PMCID: PMC6065488 DOI: 10.1210/js.2018-00131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/05/2018] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrine disorder in women of reproductive age, is characterized by androgen excess and ovarian dysfunction and presents with increased cardiometabolic risk factors such as obesity, insulin resistance, and elevated blood pressure (BP). We previously reported that administration of dihydrotestosterone (DHT) to female rats elicits cardiometabolic derangements similar to those found in women with PCOS. In this study, we tested the hypothesis that the DHT-mediated cardiometabolic derangements observed in PCOS are long lasting despite DHT withdrawal. Four-week-old female Sprague Dawley rats were treated with DHT (7.5 mg/90 days) or placebo for 6 months. DHT was discontinued (ex-DHT), and rats were followed for 6 additional months. After 6 months of DHT withdrawal, food intake, body weight, fat and lean mass, fasting plasma insulin, leptin, and adiponectin were elevated in ex-DHT rats. BP remained significantly elevated, and enalapril, an angiotensin-converting enzyme (ACE) inhibitor, normalized BP in ex-DHT rats. Expression of components of the intrarenal renin-angiotensin system was increased in ex-DHT rats. The cardiometabolic features found in ex-DHT rats were associated with lower plasma androgen levels but increased expression of renal and adipose tissue androgen receptors. In summary, androgen-induced cardiometabolic effects persisted after DHT withdrawal in a PCOS experimental model. Activation of intrarenal renin-angiotensin system plays a major role in the androgen-mediated increase in BP in ex-DHT. Upregulation of the renal and adipose tissue androgen receptor may explain the long-lasting effects of androgens. In clinical scenarios characterized by hyperandrogenemia in women, prompt normalization of androgen levels may be necessary to prevent their long-lasting cardiometabolic effects.
Collapse
Affiliation(s)
- Edgar D Torres Fernandez
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kristen V Adams
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Maryam Syed
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rodrigo O Maranon
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
18
|
Szczepanska-Sadowska E, Czarzasta K, Cudnoch-Jedrzejewska A. Dysregulation of the Renin-Angiotensin System and the Vasopressinergic System Interactions in Cardiovascular Disorders. Curr Hypertens Rep 2018; 20:19. [PMID: 29556787 PMCID: PMC5859051 DOI: 10.1007/s11906-018-0823-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Purpose of Review In many instances, the renin-angiotensin system (RAS) and the vasopressinergic system (VPS) are jointly activated by the same stimuli and engaged in the regulation of the same processes. Recent Findings Angiotensin II (Ang II) and arginine vasopressin (AVP), which are the main active compounds of the RAS and the VPS, interact at several levels. Firstly, Ang II, acting on AT1 receptors (AT1R), plays a significant role in the release of AVP from vasopressinergic neurons and AVP, stimulating V1a receptors (V1aR), regulates the release of renin in the kidney. Secondly, Ang II and AVP, acting on AT1R and V1aR, respectively, exert vasoconstriction, increase cardiac contractility, stimulate the sympathoadrenal system, and elevate blood pressure. At the same time, they act antagonistically in the regulation of blood pressure by baroreflex. Thirdly, the cooperative action of Ang II acting on AT1R and AVP stimulating both V1aR and V2 receptors in the kidney is necessary for the appropriate regulation of renal blood flow and the efficient resorption of sodium and water. Furthermore, both peptides enhance the release of aldosterone and potentiate its action in the renal tubules. Summary In this review, we (1) point attention to the role of the cooperative action of Ang II and AVP for the regulation of blood pressure and the water-electrolyte balance under physiological conditions, (2) present the subcellular mechanisms underlying interactions of these two peptides, and (3) provide evidence that dysregulation of the cooperative action of Ang II and AVP significantly contributes to the development of disturbances in the regulation of blood pressure and the water-electrolyte balance in cardiovascular diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Katarzyna Czarzasta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
19
|
Bernhem K, Krishnan K, Bondar A, Brismar H, Aperia A, Scott L. AT 1-receptor response to non-saturating Ang-II concentrations is amplified by calcium channel blockers. BMC Cardiovasc Disord 2017; 17:126. [PMID: 28514967 PMCID: PMC5436436 DOI: 10.1186/s12872-017-0562-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/09/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Blockers of angiotensin II type 1 receptor (AT1R) and the voltage gated calcium channel 1.2 (CaV1.2) are commonly used for treatment of hypertension. Yet there is little information about the effect of physiological concentrations of angiotensin II (AngII) on AT1R signaling and whether there is a reciprocal regulation of AT1R signaling by CaV1.2. METHODS To elucidate these questions, we have studied the Ca2+ signaling response to physiological and pharmacological AngII doses in HEK293a cells, vascular smooth muscle cells and cardiomyocytes using a Ca2+ sensitive dye as the principal sensor. Intra-cellular calcium recordings were performed in presence and absence of CaV1.2 blockers. Semi-quantitative imaging methods were used to assess the plasma membrane expression of AT1R and G-protein activation. RESULTS Repeated exposure to pharmacological (100 nM) concentrations of AngII caused, as expected, a down-regulation of the Ca2+ response. In contrast, repeated exposure to physiological (1 nM) AngII concentration resulted in an enhancement of the Ca2+ response. The up-regulation of the Ca2+ response to repeated 1 nM AngII doses and the down-regulation of the Ca2+ response to repeated 100 nM Angll doses were not accompanied by a parallel change of the AT1R plasma membrane expression. The Ca2+ response to 1 nM of AngII was amplified in the presence of therapeutic concentrations of the CaV1.2 blockers, nifedipine and verapamil, in vascular smooth muscle cells, cardiomyocytes and HEK293a cells. Amplification of the AT1R response was also observed following inhibition of the calcium permeable transient receptor potential cation channels, suggesting that the activity of AT1R is sensitive to calcium influx. CONCLUSIONS Our findings have implications for the understanding of hyperactivity of the angiotensin system and for use of Ca2+ channel blockers as mono-therapy in hypertension.
Collapse
Affiliation(s)
- Kristoffer Bernhem
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Kalaiselvan Krishnan
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Alexander Bondar
- Institute of Chemical Biology and Fundamental Medicine, 630090, Novosibirsk, Russia
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, PO Box 1031, 17121, Solna, Sweden.,Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Anita Aperia
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, PO Box 1031, 17121, Solna, Sweden.
| | - Lena Scott
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, PO Box 1031, 17121, Solna, Sweden
| |
Collapse
|
20
|
Affiliation(s)
- Tianxin Yang
- From the Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City; and Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.
| |
Collapse
|
21
|
Giani JF, Eriguchi M, Bernstein EA, Katsumata M, Shen XZ, Li L, McDonough AA, Fuchs S, Bernstein KE, Gonzalez-Villalobos RA. Renal tubular angiotensin converting enzyme is responsible for nitro-L-arginine methyl ester (L-NAME)-induced salt sensitivity. Kidney Int 2016; 91:856-867. [PMID: 27988209 DOI: 10.1016/j.kint.2016.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/29/2016] [Accepted: 10/06/2016] [Indexed: 01/13/2023]
Abstract
Renal parenchymal injury predisposes to salt-sensitive hypertension, but how this occurs is not known. Here we tested whether renal tubular angiotensin converting enzyme (ACE), the main site of kidney ACE expression, is central to the development of salt sensitivity in this setting. Two mouse models were used: it-ACE mice in which ACE expression is selectively eliminated from renal tubular epithelial cells; and ACE 3/9 mice, a compound heterozygous mouse model that makes ACE only in renal tubular epithelium from the ACE 9 allele, and in liver hepatocytes from the ACE 3 allele. Salt sensitivity was induced using a post L-NAME salt challenge. While both wild-type and ACE 3/9 mice developed arterial hypertension following three weeks of high salt administration, it-ACE mice remained normotensive with low levels of renal angiotensin II. These mice displayed increased sodium excretion, lower sodium accumulation, and an exaggerated reduction in distal sodium transporters. Thus, in mice with renal injury induced by L-NAME pretreatment, renal tubular epithelial ACE, and not ACE expression by renal endothelium, lung, brain, or plasma, is essential for renal angiotensin II accumulation and salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Masahiro Eriguchi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Makoto Katsumata
- Cedars-Sinai Animal Models Core, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Xiao Z Shen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Liang Li
- Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sebastien Fuchs
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Romer A Gonzalez-Villalobos
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; CVMET Research Unit, Pfizer, Inc., Cambridge, Massachusetts, USA.
| |
Collapse
|
22
|
Calcineurin-inhibition Results in Upregulation of Local Renin and Subsequent Vascular Endothelial Growth Factor Production in Renal Collecting Ducts. Transplantation 2016; 100:325-333. [PMID: 26502369 DOI: 10.1097/tp.0000000000000961] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Tacrolimus (Tac) and Cyclosporine A (CyA) calcineurin inhibitors (CNIs) are 2 effective immunosuppressants which are essential to prevent allograft rejection. Calcineurin inhibitors are known to be nephrotoxic. However, the precise mechanism of nephrotoxicity is not fully understood. In this study, we investigated the in vivo effects of CNIs on the local renal renin-angiotensin system in the collecting duct (CD). METHODS Three-week-old mice were treated with either vehicle, CyA (2 mg/kg per day), Tac (0.075 mg/kg per day), CyA + Aliskiren (25 mg/kg per day), or Tac + Aliskiren for 3 weeks. Serum creatinine was measured. Renin and vascular endothelial growth factor (VEGF) contents in CD were evaluated with flow cytometry and multiphoton microscopy. The diameter of vessels was assessed with multiphoton microscopy, and the amount of renal collagen was determined by real-time polymerase chain reaction and Masson staining. RESULTS The elevated level of serum creatinine in CNI groups was abolished by Aliskiren. Flow cytometric analysis found elevated renin content in principal cells, which was prevented by Aliskiren. This result was further confirmed with multiphoton microscopy. The VEGF content in CD correlated with reduced capillary diameter and with the formation of fibrotic islands. CONCLUSIONS Calcineurin inhibitors induce production of renin in the CD that may contribute to decreased renal blood flow. In turn, CD responds with increased VEGF production, resulting in disproportional vessel growth, further worsening the local hypoxia and striped fibrosis surrounding the CDs. Aliskiren, a direct renin inhibitor blocks these effects and improves CNI-induced nephropathy by decreasing renin production in the CDs. Our data suggest that Aliskiren may be used for the prevention of CNI nephrotoxicity.
Collapse
|
23
|
Relative roles of principal and intercalated cells in the regulation of sodium balance and blood pressure. Curr Hypertens Rep 2016; 17:538. [PMID: 25794953 DOI: 10.1007/s11906-015-0538-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The kidney continuously adapts daily renal excretion of NaCl to match dietary intakes in order to maintain the NaCl content of the body, and keep vascular volume constant. Any situation that leads to NaCl retention favors a rise in blood pressure. The aldosterone-sensitive distal nephron, which contains two main types of cells, principal (PC) and intercalated (IC) cells, is an important site for the final regulation of urinary Na(+) excretion. Research over the past 20 years established a paradigm in which PCs are the exclusive site of Na(+) absorption while ICs are solely dedicated to acid-base transport. Recent studies have revealed the unexpected importance of ICs for NaCl reabsorption. Here, we review the mechanisms of Na(+) and Cl(-) transport in the aldosterone-sensitive distal nephron, with emphasis on the role of ICs in maintaining NaCl balance and normal blood pressure.
Collapse
|
24
|
Gonzalez AA, Prieto MC. Renin and the (pro)renin receptor in the renal collecting duct: Role in the pathogenesis of hypertension. Clin Exp Pharmacol Physiol 2015; 42:14-21. [PMID: 25371190 DOI: 10.1111/1440-1681.12319] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/17/2014] [Accepted: 09/30/2014] [Indexed: 12/14/2022]
Abstract
The intrarenal renin-angiotensin system (RAS) plays a critical role in the pathogenesis and progression of hypertension and kidney disease. In angiotensin (Ang) II-dependent hypertension, collecting duct renin synthesis and secretion are stimulated despite suppression of juxtaglomerular (JG) renin. This effect is mediated by the AngII type I receptor (AT1 R), independent of blood pressure. Although the regulation of JG renin has been extensively studied, the mechanisms by which renin is regulated in the collecting duct remain unclear. The augmentation of renin synthesis and activity in the collecting duct may provide a pathway for additional generation of intrarenal and intratubular AngII formation due to the presence of angiotensinogen substrate and angiotensin-converting enzyme in the nephron. The recently described (pro)renin receptor ((P)RR) binds renin or prorenin, enhancing renin activity and fully activating the biologically inactive prorenin peptide. Stimulation of (P)RR also activates intracellular pathways related to fibrosis. Renin and the (P)RR are augmented in renal tissues of AngII-dependent hypertensive rats. However, the functional contribution of the (P)RR to enhanced renin activity in the collecting duct and its contribution to the development of hypertension and kidney disease have not been well elucidated. This review focuses on recent evidence demonstrating the mechanism of renin regulation in the collecting ducts and its interaction with the (P)RR. The data suggest that renin-(P)RR interactions may induce stimulation of intracellular pathways associated with the development of hypertension and kidney disease.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | |
Collapse
|
25
|
Marques FZ, Romaine SP, Denniff M, Eales J, Dormer J, Garrelds IM, Wojnar L, Musialik K, Duda-Raszewska B, Kiszka B, Duda M, Morris BJ, Samani NJ, Danser AJ, Bogdanski P, Zukowska-Szczechowska E, Charchar FJ, Tomaszewski M. Signatures of miR-181a on the Renal Transcriptome and Blood Pressure. Mol Med 2015; 21:739-748. [PMID: 26322847 DOI: 10.2119/molmed.2015.00096] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/17/2015] [Indexed: 01/11/2023] Open
Abstract
MicroRNA-181a binds to the 3' untranslated region of messenger RNA (mRNA) for renin, a rate-limiting enzyme of the renin-angiotensin system. Our objective was to determine whether this molecular interaction translates into a clinically meaningful effect on blood pressure and whether circulating miR-181a is a measurable proxy of blood pressure. In 200 human kidneys from the TRANScriptome of renaL humAn TissuE (TRANSLATE) study, renal miR-181a was the sole negative predictor of renin mRNA and a strong correlate of circulating miR-181a. Elevated miR-181a levels correlated positively with systolic and diastolic blood pressure in TRANSLATE, and this association was independent of circulating renin. The association between serum miR-181a and systolic blood pressure was replicated in 199 subjects from the Genetic Regulation of Arterial Pressure of Humans In the Community (GRAPHIC) study. Renal immunohistochemistry and in situ hybridization showed that colocalization of miR-181a and renin was most prominent in collecting ducts where renin is not released into the systemic circulation. Analysis of 69 human kidneys characterized by RNA sequencing revealed that miR-181a was associated with downregulation of four mitochondrial pathways and upregulation of 41 signaling cascades of adaptive immunity and inflammation. We conclude that renal miR-181a has pleiotropic effects on pathways relevant to blood pressure regulation and that circulating levels of miR-181a are both a measurable proxy of renal miR-181a expression and a novel biochemical correlate of blood pressure.
Collapse
Affiliation(s)
- Francine Z Marques
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Victoria, Australia
| | - Simon Pr Romaine
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - James Eales
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - John Dormer
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Ingrid M Garrelds
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Lukasz Wojnar
- Department of Urology and Oncological Urology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Musialik
- Department of Education and Obesity Treatment and Metabolic Disorders, Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara Duda-Raszewska
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Bartlomiej Kiszka
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Magdalena Duda
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Brian J Morris
- School of Medical Sciences, University of Sydney, New South Wales, Australia
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom.,Leicester National Institute for Health Research Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, United Kingdom
| | - Ah Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Pawel Bogdanski
- Department of Education and Obesity Treatment and Metabolic Disorders, Poznan University of Medical Sciences, Poznan, Poland
| | - Ewa Zukowska-Szczechowska
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Fadi J Charchar
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Victoria, Australia
| | - Maciej Tomaszewski
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom.,Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
26
|
Gonzalez AA, Prieto MC. Roles of collecting duct renin and (pro)renin receptor in hypertension: mini review. Ther Adv Cardiovasc Dis 2015; 9:191-200. [PMID: 25780059 PMCID: PMC4560657 DOI: 10.1177/1753944715574817] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In angiotensin (Ang)-II-dependent hypertension, collecting duct renin synthesis and secretion are stimulated despite suppression of juxtaglomerular (JG) renin. This effect is mediated by Ang II type 1 (AT1) receptor independent of blood pressure. Although the regulation of JG renin is known, the mechanisms by which renin is regulated in the collecting duct are not completely understood. The presence of renin activity in the collecting duct may provide a pathway for intratubular Ang II formation since angiotensinogen substrate and angiotensin converting enzyme are present in the distal nephron. The recently named new member of the renin-angiotensin system (RAS), the (pro)renin receptor [(P)RR], is able to bind renin and the inactive prorenin, thus enhancing renin activity and fully activating prorenin. We have demonstrated that renin and (P)RR are augmented in renal tissues from rats infused with Ang II and during sodium depletion, suggesting a physiological role in intrarenal RAS activation. Importantly, (P)RR activation also causes activation of intracellular pathways associated with increased cyclooxygenase 2 expression and induction of profibrotic genes. In addition, renin and (P)RR are upregulated by Ang II in collecting duct cells. Although the mechanisms involved in their regulation are still under study, they seem to be dependent on the intrarenal RAS activation. The complexities of the mechanisms of stimulation also depend on cyclooxygenase 2 and sodium depletion. Our data suggest that renin and (P)RR can interact to increase intratubular Ang II formation and the activation of profibrotic genes in renal collecting duct cells. Both pathways may have a critical role in the development of hypertension and renal disease.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Chile
| | - Minolfa C Prieto
- Department of Physiology, Rm 4061, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| |
Collapse
|
27
|
Tomaszewski M, Eales J, Denniff M, Myers S, Chew GS, Nelson CP, Christofidou P, Desai A, Büsst C, Wojnar L, Musialik K, Jozwiak J, Debiec R, Dominiczak AF, Navis G, van Gilst WH, van der Harst P, Samani NJ, Harrap S, Bogdanski P, Zukowska-Szczechowska E, Charchar FJ. Renal Mechanisms of Association between Fibroblast Growth Factor 1 and Blood Pressure. J Am Soc Nephrol 2015; 26:3151-60. [PMID: 25918036 DOI: 10.1681/asn.2014121211] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/05/2015] [Indexed: 11/03/2022] Open
Abstract
The fibroblast growth factor 1 (FGF1) gene is expressed primarily in the kidney and may contribute to hypertension. However, the biologic mechanisms underlying the association between FGF1 and BP regulation remain unknown. We report that the major allele of FGF1 single nucleotide polymorphism rs152524 was associated in a dose-dependent manner with systolic BP (P = 9.65 × 10(-5)) and diastolic BP (P = 7.61 × 10(-3)) in a meta-analysis of 14,364 individuals and with renal expression of FGF1 mRNA in 126 human kidneys (P=9.0 × 10(-3)). Next-generation RNA sequencing revealed that upregulated renal expression of FGF1 or of each of the three FGF1 mRNA isoforms individually was associated with higher BP. FGF1-stratified coexpression analysis in two separate collections of human kidneys identified 126 FGF1 partner mRNAs, of which 71 and 63 showed at least nominal association with systolic and diastolic BP, respectively. Of those mRNAs, seven mRNAs in five genes (MME, PTPRO, REN, SLC12A3, and WNK1) had strong prior annotation to BP or hypertension. MME, which encodes an enzyme that degrades circulating natriuretic peptides, showed the strongest differential coexpression with FGF1 between hypertensive and normotensive kidneys. Furthermore, higher level of renal FGF1 expression was associated with lower circulating levels of atrial and brain natriuretic peptides. These findings indicate that FGF1 expression in the kidney is at least under partial genetic control and that renal expression of several FGF1 partner genes involved in the natriuretic peptide catabolism pathway, renin-angiotensin cascade, and sodium handling network may explain the association between FGF1 and BP.
Collapse
Affiliation(s)
- Maciej Tomaszewski
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; NIHR Biomedical Research Centre in Cardiovascular Disease, Leicester, United Kingdom;
| | - James Eales
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Stephen Myers
- Faculty of Science and Technology, Federation University Australia, Ballarat, Australia
| | - Guat Siew Chew
- Faculty of Science and Technology, Federation University Australia, Ballarat, Australia
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; NIHR Biomedical Research Centre in Cardiovascular Disease, Leicester, United Kingdom
| | - Paraskevi Christofidou
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Aishwarya Desai
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Cara Büsst
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | | | - Katarzyna Musialik
- Education and Obesity Treatment and Metabolic Disorders, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Jozwiak
- Department of Public Health, Czestochowa University of Technology, Czestochowa, Poland
| | - Radoslaw Debiec
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Anna F Dominiczak
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Wiek H van Gilst
- Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands; and
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; NIHR Biomedical Research Centre in Cardiovascular Disease, Leicester, United Kingdom
| | - Stephen Harrap
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | - Pawel Bogdanski
- Education and Obesity Treatment and Metabolic Disorders, Poznan University of Medical Sciences, Poznan, Poland
| | - Ewa Zukowska-Szczechowska
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | - Fadi J Charchar
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; Faculty of Science and Technology, Federation University Australia, Ballarat, Australia
| |
Collapse
|
28
|
Roy A, Al-bataineh MM, Pastor-Soler NM. Collecting duct intercalated cell function and regulation. Clin J Am Soc Nephrol 2015; 10:305-24. [PMID: 25632105 DOI: 10.2215/cjn.08880914] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Intercalated cells are kidney tubule epithelial cells with important roles in the regulation of acid-base homeostasis. However, in recent years the understanding of the function of the intercalated cell has become greatly enhanced and has shaped a new model for how the distal segments of the kidney tubule integrate salt and water reabsorption, potassium homeostasis, and acid-base status. These cells appear in the late distal convoluted tubule or in the connecting segment, depending on the species. They are most abundant in the collecting duct, where they can be detected all the way from the cortex to the initial part of the inner medulla. Intercalated cells are interspersed among the more numerous segment-specific principal cells. There are three types of intercalated cells, each having distinct structures and expressing different ensembles of transport proteins that translate into very different functions in the processing of the urine. This review includes recent findings on how intercalated cells regulate their intracellular milieu and contribute to acid-base regulation and sodium, chloride, and potassium homeostasis, thus highlighting their potential role as targets for the treatment of hypertension. Their novel regulation by paracrine signals in the collecting duct is also discussed. Finally, this article addresses their role as part of the innate immune system of the kidney tubule.
Collapse
Affiliation(s)
- Ankita Roy
- Renal-Electrolyte Division, Department of Medicine; and
| | | | - Núria M Pastor-Soler
- Renal-Electrolyte Division, Department of Medicine; and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania A.R. and M.M.A. contributed equally to this work.
| |
Collapse
|
29
|
Morris BJ. Renin, genes, microRNAs, and renal mechanisms involved in hypertension. Hypertension 2015; 65:956-62. [PMID: 25601934 DOI: 10.1161/hypertensionaha.114.04366] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Brian J Morris
- From the Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
30
|
Angiotensin II increases the expression of (pro)renin receptor during low-salt conditions. Am J Med Sci 2015; 348:416-22. [PMID: 25250989 DOI: 10.1097/maj.0000000000000335] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Evidence indicates that chronic angiotensin II (AngII) infusion increases (pro)renin receptor ((P)RR) expression in renal inner medullary collecting duct (IMCD) cells. Recently, it has been shown that renal (P)RR expression is augmented during a low-salt (LS) diet. However, the role of AngII in mediating the stimulation of (P)RR during LS conditions is unknown. We hypothesized that AngII mediates the increased expression of (P)RR during low-salt conditions in IMCDs. METHODS (P)RR expression and AngII levels were evaluated in Sprague-Dawley rats fed a LS diet (0.03% NaCl) and normal salt (NS; 0.4% NaCl) for 7 days. We examined the effects of sodium reduction (130 mM NaCl) and AngII on (P)RR expression in IMCDs isolated in hypertonic conditions (640 mOsmol/L with 280 mM NaCl). RESULTS Plasma renin activity in LS rats was significantly higher than rats fed with NS (28.1 ± 2.2 versus 6.7 ± 1.1 ng AngI·mL⁻¹·hr⁻¹; P < 0.05), as well as renin content in renal cortex and medulla. The (P)RR mRNA and protein levels were higher in medullary tissues from LS rats but did not change in the cortex. Intrarenal AngII was augmented in LS compared with NS rats (cortex: 710 ± 113 versus 277 ± 86 fmol/g, P < 0.05; medulla: 2093 ± 125 versus 1426 ± 126 fmol/g, P < 0.05). In cultured IMCDs, (P)RR expression was increased in response to LS or AngII treatment and potentiated by both treatments (both at 640 mOsmol/L). CONCLUSIONS These data indicate that (P)RR is augmented in medullary collecting ducts in response to LS and that this effect is further enhanced by the increased intrarenal AngII content.
Collapse
|
31
|
Signaling pathways involved in renal oxidative injury: role of the vasoactive peptides and the renal dopaminergic system. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:731350. [PMID: 25436148 PMCID: PMC4243602 DOI: 10.1155/2014/731350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/16/2014] [Indexed: 12/24/2022]
Abstract
The physiological hydroelectrolytic balance and the redox steady state in the kidney are accomplished by an intricate interaction between signals from extrarenal and intrarenal sources and between antinatriuretic and natriuretic factors. Angiotensin II, atrial natriuretic peptide and intrarenal dopamine play a pivotal role in this interactive network. The balance between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide, by one side, and the prooxidant effect of the renin angiotensin system, by the other side, contributes to ensuring the normal function of the kidney. Different pathological scenarios, as nephrotic syndrome and hypertension, where renal sodium excretion is altered, are associated with an impaired interaction between two natriuretic systems as the renal dopaminergic system and atrial natriuretic peptide that may be involved in the pathogenesis of renal diseases. The aim of this review is to update and comment the most recent evidences about the intracellular pathways involved in the relationship between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide and the prooxidant effect of the renin angiotensin system in the pathogenesis of renal inflammation.
Collapse
|
32
|
Sato W, Sato Y. Midkine in nephrogenesis, hypertension and kidney diseases. Br J Pharmacol 2014; 171:879-87. [PMID: 24106831 DOI: 10.1111/bph.12418] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/31/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Midkine (MK; K; gene abbreviation, Mdk: mus musculus, MDK: homo sapiens) is a multifunctional heparin-binding growth factor that regulates cell growth, survival and migration as well as anti-apoptotic activity in nephrogenesis and development. Proximal tubular epithelial cells are the main sites of MK expression in the kidneys. The pathophysiological roles of MK are diverse, ranging from the development of acute kidney injury (AKI) to the progression of chronic kidney disease, often accompanied by hypertension, renal ischaemia and diabetic nephropathy. The obvious hypertension that develops in Mdk(+/+) mouse models of renal ablation compared with Mdk(-/-) mice eventually leads to progressive renal failure, such as glomerular sclerosis and tubulointerstitial damage associated with elevated plasma angiotensin (Ang) II levels. MK is also induced in the lung endothelium by oxidative stress and subsequently up-regulated by ACE, which hydrolyzes Ang II to induce further oxidative stress, thus accelerating MK generation; this leads to a vicious cycle of positive feedback in the MK-Ang II pathway. Kidney-lung interactions involving positive feedback between the renin-angiotensin system and MK might partly account for the pathogenesis of hypertension and kidney damage. MK is also involved in the pathogenesis of AKI and diabetic nephropathy through the recruitment of inflammatory cells. In contrast, MK plays a protective role against crescentic glomerulonephritis, by down-regulating plasminogen activator inhibitor-1. These diverse actions of MK might open up new avenues for targeted approaches to treating hypertension and various renal diseases. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- Waichi Sato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | | |
Collapse
|
33
|
Gonzalez AA, Green T, Luffman C, Bourgeois CRT, Gabriel Navar L, Prieto MC. Renal medullary cyclooxygenase-2 and (pro)renin receptor expression during angiotensin II-dependent hypertension. Am J Physiol Renal Physiol 2014; 307:F962-70. [PMID: 25143455 DOI: 10.1152/ajprenal.00267.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The (pro)renin receptor [(P)RR] upregulates cyclooxygenase-2 (COX-2) in inner medullary collecting duct (IMCD) cells through ERK1/2. Intrarenal COX-2 and (P)RR are upregulated during chronic ANG II infusion. However, the duration of COX-2 and (P)RR upregulation has not been determined. We hypothesized that during the early phase of ANG II-dependent hypertension, membrane-bound (P)RR and COX-2 are augmented in the renal medulla, serving to buffer the hypertensinogenic and vasoconstricting effects of ANG II. In Sprague-Dawley rats infused with ANG II (0.4 μg·min(-1)·kg(-1)), systolic blood pressure (BP) increased by day 7 (162 ± 5 vs. 114 ± 10 mmHg) and continued to increase by day 14 (198 ± 15 vs. 115 ± 13 mmHg). Membrane-bound (P)RR was augmented at day 3 coincident with phospho-ERK1/2 levels, COX-2 expression, and PGE2 in the renal medulla. In contrast, membrane-bound (P)RR was reduced and COX-2 protein levels were not different from controls by day 14. In cultured IMCD cells, ANG II increased secretion of the soluble (P)RR. In anesthetized rats, COX-2 inhibition decreased the glomerular filtration rate (GFR) and renal blood flow (RBF) during the early phase of ANG II infusion without altering BP. However, at 14 days of ANG II infusions, COX-2 inhibition decreased mean arterial BP (MABP), RBF, and GFR. Thus, during the early phase of ANG II-dependent hypertension, the increased (P)RR and COX-2 expression in the renal medulla may contribute to attenuate the vasoconstrictor effects of ANG II on renal hemodynamics. In contrast, at 14 days the reductions in RBF and GFR caused by COX-2 inhibition paralleled the reduced MABP, suggesting that vasoconstrictor COX-2 metabolites contribute to ANG II hypertension.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile; and
| | - Torrance Green
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Christina Luffman
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Camille R T Bourgeois
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Minolfa C Prieto
- Department of Physiology and Hypertension and Renal Center of Excellence, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
34
|
Ohsawa M, Tamura K, Wakui H, Maeda A, Dejima T, Kanaoka T, Azushima K, Uneda K, Tsurumi-Ikeya Y, Kobayashi R, Matsuda M, Uchida S, Toya Y, Kobori H, Nishiyama A, Yamashita A, Ishikawa Y, Umemura S. Deletion of the angiotensin II type 1 receptor-associated protein enhances renal sodium reabsorption and exacerbates angiotensin II-mediated hypertension. Kidney Int 2014; 86:570-81. [PMID: 24694992 PMCID: PMC4149871 DOI: 10.1038/ki.2014.95] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 12/13/2022]
Abstract
Angiotensin II type 1 receptor (AT1R)–associated protein (ATRAP) promotes AT1R internalization along with suppression of pathological activation of tissue AT1R signaling. However, the functional significance of ATRAP in renal sodium handling and blood pressure regulation under pathological stimuli is not fully resolved. Here we show the blood pressure of mice with a gene-targeted disruption of ATRAP was comparable to that of wild-type mice at baseline. However, in ATRAP-knockout mice, angiotensin II–induced hypertension was exacerbated and the extent of positive sodium balance was increased by angiotensin II. Renal expression of the sodium-proton antiporter 3, a major sodium transporter in the proximal tubules, urinary pH, renal angiotensinogen production, and angiotensin II content was unaffected. Stimulation of the renal expression and activity of the epithelial sodium channel (ENaC), a major sodium transporter in the distal tubules, was significantly enhanced by chronic angiotensin II infusion. The circulating and urinary aldosterone levels were comparable. The blood pressure response and renal ENaC expression by aldosterone were not affected. Thus, ATRAP deficiency exacerbated angiotensin II–mediated hypertension by pathological activation of renal tubular AT1R by angiotensin II. This directly stimulates ENaC in the distal tubules and enhances sodium retention in an aldosterone-independent manner.
Collapse
Affiliation(s)
- Masato Ohsawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akinobu Maeda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toru Dejima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazushi Uneda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuko Tsurumi-Ikeya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyuki Matsuda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiyuki Toya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Kobori
- Department of Pharmacology, Kagawa University School of Medicine, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University School of Medicine, Kagawa, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Umemura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
35
|
Bernstein KE, Giani JF, Shen XZ, Gonzalez-Villalobos RA. Renal angiotensin-converting enzyme and blood pressure control. Curr Opin Nephrol Hypertens 2014; 23:106-12. [PMID: 24378774 PMCID: PMC4028050 DOI: 10.1097/01.mnh.0000441047.13912.56] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review presents novel findings regarding the renal angiotensin-converting enzyme (ACE) and its role in blood pressure (BP) control. RECENT FINDINGS The textbook flow diagram of the renin-angiotensin system (RAS) shows the pulmonary endothelium as the main source of the ACE that converts angiotensin I to angiotensin II. However, ACE is made in large quantities by the kidneys, which raises the important question of what precisely is the function of renal ACE? Recent studies in gene-targeted mice indicates that renal ACE plays a dominant role in regulating the response of the kidney to experimental hypertension. In particular, renal ACE and locally generated angiotensin II affect the activity of several key sodium transporters and the induction of sodium and water retention resulting in the elevation of BP. SUMMARY New experimental data link the renal ACE/angiotensin II pathway and the local regulation of sodium transport as key elements in the development of hypertension.
Collapse
Affiliation(s)
- Kenneth E Bernstein
- Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
36
|
Campbell DJ. Do intravenous and subcutaneous angiotensin II increase blood pressure by different mechanisms? Clin Exp Pharmacol Physiol 2014; 40:560-70. [PMID: 23551142 DOI: 10.1111/1440-1681.12085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 03/22/2013] [Accepted: 03/25/2013] [Indexed: 01/21/2023]
Abstract
Angiotensin (Ang) II plays a key role in blood pressure regulation. Mechanisms of the pressor effect of chronic intravenous AngII administration include vasoconstriction, stimulation of the sympathetic nervous system and aldosterone production, as well as direct effects on renal excretion of sodium and water. Chronic AngII administration by subcutaneous minipump at doses higher than required to increase blood pressure by the intravenous route has identified additional pressor mechanisms, including the immune system, cytokines and matrix metalloproteinases. However, pressor doses of subcutaneous AngII may exceed the angiotensinogen synthesis rate and produce inflammation, fibrosis and necrosis of skin overlying the minipump. Evidence that chronic subcutaneous and intravenous AngII increase blood pressure by different mechanisms includes the prevention of the pressor effects of subcutaneous, but not intravenous, AngII by angiotensin-converting enzyme inhibition. Furthermore, low doses of subcutaneous AngII reduce blood pressure of female, but not male, rodents and higher doses are less pressor in females than in males, whereas intravenous AngII is equally pressor in males and females. Pressor doses of chronic subcutaneous AngII produce greater weight loss, anorexia and reduced kidney weight and cause greater vascular, cardiac and renal pathology than equally pressor doses of chronic intravenous AngII. The different effects of chronic intravenous and subcutaneous AngII suggest that these two models of hypertension give different information and may differ in their relevance to blood pressure regulation in physiological and pathological states such as hypertension in humans.
Collapse
Affiliation(s)
- Duncan J Campbell
- St Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Vic., Australia.
| |
Collapse
|
37
|
Abramova TO, Redina OE, Smolenskaya SE, Markel AL. Elevated expression of the Ephx2 mRNA in the kidney of hypertensive ISIAH rats. Mol Biol 2013. [DOI: 10.1134/s0026893313060022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Prieto MC, Gonzalez AA, Navar LG. Evolving concepts on regulation and function of renin in distal nephron. Pflugers Arch 2012; 465:121-32. [PMID: 22990760 DOI: 10.1007/s00424-012-1151-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 08/24/2012] [Accepted: 08/30/2012] [Indexed: 01/13/2023]
Abstract
Sustained stimulation of the intrarenal/intratubular renin-angiotensin system in a setting of elevated arterial pressure elicits renal vasoconstriction, increased sodium reabsorption, proliferation, fibrosis, and eventual renal injury. Activation of luminal AT(1) receptors in proximal and distal nephron segments by local Ang II formation stimulates various transport systems. Augmented angiotensinogen (AGT) production by proximal tubule cells increases AGT secretion contributing to increased proximal Ang II levels and leading to spillover of AGT into the distal nephron segments, as reflected by increased urinary AGT excretion. The increased distal delivery of AGT provides substrate for renin, which is expressed in principal cells of the collecting tubule and collecting ducts, and is also stimulated by AT(1) receptor activation. Renin and prorenin are secreted into the tubular lumen and act on the AGT delivered from the proximal tubule to form more Ang I. The catalytic actions of renin and or prorenin may be enhanced by binding to prorenin receptors on the intercalated cells or soluble prorenin receptor secreted into the tubular fluid. There is also increased luminal angiotensin converting enzyme in collecting ducts facilitating Ang II formation leading to stimulation of sodium reabsorption via sodium channel and sodium/chloride co-transporter. Thus, increased collecting duct renin contributes to Ang II-dependent hypertension by augmenting distal nephron intratubular Ang II formation leading to sustained stimulation of sodium reabsorption and progression of hypertension.
Collapse
Affiliation(s)
- Minolfa C Prieto
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
39
|
Rands VF, Seth DM, Kobori H, Prieto MC. Sexual dimorphism in urinary angiotensinogen excretion during chronic angiotensin II-salt hypertension. ACTA ACUST UNITED AC 2012; 9:207-18. [PMID: 22795463 DOI: 10.1016/j.genm.2012.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 05/11/2012] [Accepted: 06/14/2012] [Indexed: 01/11/2023]
Abstract
BACKGROUND The intrarenal renin-angiotensin system contributes to hypertension by regulating sodium and water reabsorption throughout the nephron. Sex differences in the intrarenal components of the renin-angiotensin system have been involved in the greater incidence of high blood pressure and progression to kidney damage in males than females. OBJECTIVE This study investigated whether there is a sex difference in the intrarenal gene expression and urinary excretion of angiotensinogen (AGT) during angiotensin II (Ang II)-dependent hypertension and high-salt (HS) diet. METHODS Male and female Sprague-Dawley rats were divided into 5 groups for each sex: Normal-salt control, HS diet (8% NaCl), Ang II-infused (80 ng/min), Ang II-infused plus HS diet, and Ang II-infused plus HS diet and treatment with the Ang II receptor blocker, candesartan (25 mg/L in the drinking water). Rats were evaluated for systolic blood pressure (SBP), kidney AGT mRNA expression, urinary AGT excretion, and proteinuria at different time points during a 14-day protocol. RESULTS Both male and female rats exhibited similar increases in urinary AGT, with increases in SBP during chronic Ang II infusion. HS diet greatly exacerbated the urinary AGT excretion in Ang II-infused rats; males had a 9-fold increase over Ang II alone and females had a 2.5-fold increase. Male rats displayed salt-sensitive SBP increases during Ang II infusion and HS diet, and female rats did not. In the kidney cortex, males displayed greater AGT gene expression than females during all treatments. During Ang II infusion, both sexes exhibited increases in AGT gene message compared with same-sex controls. In addition, HS diet combined with Ang II infusion exacerbated the proteinuria in both sexes. Concomitant Ang II receptor blocker treatment during Ang II infusion and HS diet decreased SBP and urinary AGT similarly in both sexes; however, the decrease in proteinuria was greater in the females. CONCLUSION During Ang II-dependent hypertension and HS diet, higher intrarenal renin-angiotensin system activation in males, as reflected by higher AGT gene expression and urinary excretion, indicates a mechanism for greater progression of high blood pressure and might explain the sex disparity in development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Vicky F Rands
- Department of Physiology, School of Medicine, Tulane University, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
40
|
Huang Y, Yamamoto T, Misaki T, Suzuki H, Togawa A, Ohashi N, Fukasawa H, Fujigaki Y, Ichihara A, Nishiyama A, Senbonmatsu T, Ikegaya N, Hishida A. Enhanced intrarenal receptor-mediated prorenin activation in chronic progressive anti-thymocyte serum nephritis rats on high salt intake. Am J Physiol Renal Physiol 2012; 303:F130-8. [PMID: 22496409 DOI: 10.1152/ajprenal.00275.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Despite suppression of the circulating renin-angiotensin system (RAS), high salt intake (HSI) aggravates kidney injury in chronic kidney disease. To elucidate the effect of HSI on intrarenal RAS, we investigated the levels of intrarenal prorenin, renin, (pro)renin receptor (PRR), receptor-mediated prorenin activation, and ANG II in chronic anti-thymocyte serum (ATS) nephritic rats on HSI. Kidney fibrosis grew more severe in the nephritic rats on HSI than normal salt intake. Despite suppression of plasma renin and ANG II, marked increases in tubular prorenin and renin proteins without concomitant rises in renin mRNA, non-proteolytically activated prorenin, and ANG II were noted in the nephritic rats on HSI. Redistribution of PRR from the cytoplasm to the apical membrane, along with elevated non-proteolytically activated prorenin and ANG II, was observed in the collecting ducts and connecting tubules in the nephritic rats on HSI. Olmesartan decreased cortical prorenin, non-proteolytically activated prorenin and ANG II, and apical membranous PRR in the collecting ducts and connecting tubules, and attenuated the renal lesions. Cell surface trafficking of PRR was enhanced by ANG II and was suppressed by olmesartan in Madin-Darby canine kidney cells. These data suggest the involvement of the ANG II-dependent increase in apical membrane PRR in the augmentation of intrarenal binding of prorenin and renin, followed by nonproteolytic activation of prorenin, enhancement of renin catalytic activity, ANG II generation, and progression of kidney fibrosis in the nephritic rat kidneys on HSI. The origin of the increased tubular prorenin and renin remains to be clarified. Further studies measuring the urinary prorenin and renin are needed.
Collapse
Affiliation(s)
- Yanjie Huang
- First Department of Medicine, Hamamatsu University School of Medicine, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Vargas F, Rodríguez-Gómez I, Vargas-Tendero P, Jimenez E, Montiel M. The renin-angiotensin system in thyroid disorders and its role in cardiovascular and renal manifestations. J Endocrinol 2012; 213:25-36. [PMID: 22043064 DOI: 10.1530/joe-11-0349] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Thyroid disorders are among the most common endocrine diseases and affect virtually all physiological systems, with an especially marked impact on cardiovascular and renal systems. This review summarizes the effects of thyroid hormones on the renin-angiotensin system (RAS) and the participation of the RAS in the cardiovascular and renal manifestations of thyroid disorders. Thyroid hormones are important regulators of cardiac and renal mass, vascular function, renal sodium handling, and consequently blood pressure (BP). The RAS acts globally to control cardiovascular and renal functions, while RAS components act systemically and locally in individual organs. Various authors have implicated the systemic and local RAS in the mediation of functional and structural changes in cardiovascular and renal tissues due to abnormal thyroid hormone levels. This review analyzes the influence of thyroid hormones on RAS components and discusses the role of the RAS in BP, cardiac mass, vascular function, and renal abnormalities in thyroid disorders.
Collapse
Affiliation(s)
- Félix Vargas
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.
| | | | | | | | | |
Collapse
|
42
|
Reinhold SW, Krüger B, Barner C, Zoicas F, Kammerl MC, Hoffmann U, Bergler T, Banas B, Krämer BK. Nephron-specific expression of components of the renin-angiotensin-aldosterone system in the mouse kidney. J Renin Angiotensin Aldosterone Syst 2012; 13:46-55. [PMID: 22247339 DOI: 10.1177/1470320311432184] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The renin-angiotensin-aldosterone system (RAAS) plays an integral role in the regulation of blood pressure, electrolyte and fluid homeostasis in mammals. The capability of the different nephron segments to form components of the RAAS is only partially known. This study therefore aimed to characterize the nephron-specific expression of RAAS components within the mouse kidney. MATERIALS AND METHODS Defined nephron segments of adult C57B/16 mice were microdissected after collagenase digestion. The gene expression of renin, angiotensinogen (AGT), angiotensin-converting enzyme (ACE), angiotensin II receptors 1a (AT1a), 1b (AT1b), and 2 (AT2) was assessed by reverse transcriptase polymerase chain reaction (RT-PCR). RESULTS Renin mRNA was present in glomeruli, in proximal tubules, in distal convoluted tubules (DCT) and cortical collecting ducts (CCD). AGT mRNA was found in proximal tubules, descending thin limb of Henle's loop (dTL) and in the medullary part of the thick ascending limb (mTAL). ACE mRNA was not detectable in microdissected mouse nephron segments. AT1a, AT1b and AT2 mRNA was detected in glomeruli and proximal convoluted tubules. CONCLUSIONS Our data demonstrate a nephron-specific distribution of RAAS components. All components of the local RAAS - except ACE - are present in proximal convoluted tubules, emphasizing their involvement in sodium and water handling.
Collapse
Affiliation(s)
- Stephan W Reinhold
- Klinik und Poliklinik für Innere Medizin II, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kosugi T, Sato W. Midkine and the kidney: health and diseases. Nephrol Dial Transplant 2011; 27:16-21. [DOI: 10.1093/ndt/gfr652] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
44
|
Abstract
The G protein-coupled succinate and α-ketoglutarate receptors are closely related to the family of P2Y purinoreceptors. Although the α-ketoglutarate receptor is almost exclusively expressed in the kidney, its function is unknown. In contrast, the succinate receptor, SUCRN1, is expressed in a variety of tissues, including blood cells, adipose tissue, liver, retina, and the kidney. Recent evidence suggests SUCRN1 and its succinate ligand are novel detectors of local stress, including ischemia, hypoxia, toxicity, and hyperglycemia. Local levels of succinate in the kidney also activate the renin-angiotensin system and together with SUCRN1 may play a key role in the development of hypertension and the complications of diabetes mellitus, metabolic disease, and liver damage. This makes the succinate receptor a promising drug target to counteract an expanding number of interrelated disorders.
Collapse
Affiliation(s)
- Peter M T Deen
- Department of Physiology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, The Netherlands
| | | |
Collapse
|
45
|
Lara LS, McCormack M, Semprum-Prieto LC, Shenouda S, Majid DSA, Kobori H, Navar LG, Prieto MC. AT1 receptor-mediated augmentation of angiotensinogen, oxidative stress, and inflammation in ANG II-salt hypertension. Am J Physiol Renal Physiol 2011; 302:F85-94. [PMID: 21900456 DOI: 10.1152/ajprenal.00351.2011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Augmentation of intrarenal angiotensinogen (AGT) synthesis, secretion, and excretion is associated with the development of hypertension, renal oxidative stress, and tissue injury during ANG II-dependent hypertension. High salt (HS) exacerbates hypertension and kidney injury, but the mechanisms remain unclear. In this study, we determined the consequences of HS intake alone compared with chronic ANG II infusion and combined HS plus ANG II on the stimulation of urinary AGT (uAGT), renal oxidative stress, and renal injury markers. Sprague-Dawley rats were subjected to 1) a normal-salt diet [NS, n = 5]; 2) HS diet [8% NaCl, n = 5]; 3) ANG II infusion in NS rats [ANG II 80 ng/min, n = 5]; 4) ANG II infusion in HS rats [ANG II+HS, n = 5]; and 5) ANG II infusion in HS rats treated with ANG II type 1 receptor blocker (ARB) [ANG II+HS+ARB, n = 5] for 14 days. Rats fed a HS diet alone did not show changes in systolic blood pressure (SBP), proteinuria, cell proliferation, or uAGT excretion although they did exhibit mesangial expansion, collagen deposition, and had increased NADPH oxidase activity accompanied by increased peroxynitrite formation in the kidneys. Compared with ANG II rats, the combination of ANG II infusion and a HS diet led to exacerbation in SBP (175 ± 10 vs. 221 ± 8 mmHg; P < 0.05), proteinuria (46 ± 7 vs. 127 ± 7 mg/day; P < 0.05), and uAGT (1,109 ± 70 vs.. 7,200 ± 614 ng/day; P < 0.05) associated with greater collagen deposition, mesangial expansion, interstitial cell proliferation, and macrophage infiltration. In both ANG II groups, the O(2)(-) levels were increased due to increased NADPH oxidase activity without concomitant increases in peroxynitrite formation. The responses in ANG II rats were prevented or ameliorated by ARB treatment. The results indicate that HS independently stimulates ROS formation, which may synergize with the effect of ANG II to limit peroxynitrite formation, leading to exacerbation of uAGT and greater injury during ANG II salt hypertension.
Collapse
Affiliation(s)
- Lucienne S Lara
- Tulane Univ., School of Medicine, Dept. of Physiology, Rm. 4061, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The distal nephron plays a critical role in the renal control of homeostasis. Until very recently most studies focused on the control of Na(+), K(+), and water balance by principal cells of the collecting duct and the regulation of solute and water by hormones from the renin-angiotensin-aldosterone system and by antidiuretic hormone. However, recent studies have revealed the unexpected importance of renal intercalated cells, a subtype of cells present in the connecting tubule and collecting ducts. Such cells were thought initially to be involved exclusively in acid-base regulation. However, it is clear now that intercalated cells absorb NaCl and K(+) and hence may participate in the regulation of blood pressure and potassium balance. The second paradigm-challenging concept we highlight is the emerging importance of local paracrine factors that play a critical role in the renal control of water and electrolyte balance.
Collapse
Affiliation(s)
- Dominique Eladari
- Centre de Recherche des Cordeliers, Université Paris Descartes, INSERM UMRS 872, Equipe 3, F-75006, Paris, France; ,
- Université Pierre et Marie Curie, CNRS ERL7226, F-75006, Paris, France
- Département de Physiologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, F-75015, Paris, France
| | - Régine Chambrey
- Centre de Recherche des Cordeliers, Université Paris Descartes, INSERM UMRS 872, Equipe 3, F-75006, Paris, France; ,
- Université Pierre et Marie Curie, CNRS ERL7226, F-75006, Paris, France
| | - Janos Peti-Peterdi
- Department of Physiology and Biophysics, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California 90033;
| |
Collapse
|
47
|
Liu L, Gonzalez AA, McCormack M, Seth DM, Kobori H, Navar LG, Prieto MC. Increased renin excretion is associated with augmented urinary angiotensin II levels in chronic angiotensin II-infused hypertensive rats. Am J Physiol Renal Physiol 2011; 301:F1195-201. [PMID: 21865264 DOI: 10.1152/ajprenal.00339.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Renin expression in principal cells of collecting ducts (CD) is upregulated in angiotensin II (ANG II)-dependent hypertensive rats; however, it remains unclear whether increased CD-derived renin undergoes tubular secretion. Accordingly, urinary levels of renin (uRen), angiotensinogen (uAGT), and ANG II (uANG II) were measured in chronic ANG II-infused Sprague-Dawley rats (80 ng/min for 14 days, n = 10) and sham-operated rats (n = 10). Systolic blood pressure increased in the ANG II rats by day 5 and continued to increase throughout the study (day 13; ANG II: 175 ± 10 vs. sham: 116 ± 2 mmHg; P < 0.05). ANG II infusion increased renal cortical and medullary ANG II levels (cortical ANG II: 606 ± 72 vs. 247 ± 43 fmol/g; P < 0.05; medullary ANG II: 2,066 ± 116 vs. 646 ± 36 fmol/g; P < 0.05). Although plasma renin activity (PRA) was suppressed in the ANG II-infused rats (0.3 ± 0.2 vs. 5.5 ± 1.8 ng ANG I·ml(-1)·h(-1); P < 0.05), renin content in renal medulla was increased (12,605 ± 1,343 vs. 7,956 ± 765 ng ANG I·h(-1)·mg(-1); P < 0.05). Excretion of uAGT and uANG II increased in the ANG II rats [uAGT: 1,107 ± 106 vs. 60 ± 26 ng/day; P < 0.0001; uANG II: 3,813 ± 431 vs. 2,080 ± 361 fmol/day; P < 0.05]. By day 13, despite suppression of PRA, urinary prorenin content increased in ANG II rats [15.7 ± 3 vs. 2.6 ± 1 × 10(-3) enzyme units excreted (EUE)/day, P < 0.01] as was the excretion rate of renin (8.6 ± 2 × 10(-6) EUE/day) compared with sham (2.8 ± 1 × 10(-6) EUE/day; P < 0.05). Urinary renin and prorenin protein levels examined by Western blot were augmented ∼10-fold in the ANG II-infused rats. Concomitant AT(1) receptor blockade with candesartan prevented the increase. Thus, in ANG II-dependent hypertensive rats with marked PRA suppression, increased urinary levels of renin and prorenin reflect their augmented secretion by CD cells into the luminal fluid. The greater availability of renin and AGT in the urine reflects the capability for intratubular ANG II formation which stimulates sodium reabsorption in distal nephron segments.
Collapse
Affiliation(s)
- Liu Liu
- Dept. of Physiology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Facemire CS, Nguyen M, Jania L, Beierwaltes WH, Kim HS, Koller BH, Coffman TM. A major role for the EP4 receptor in regulation of renin. Am J Physiol Renal Physiol 2011; 301:F1035-41. [PMID: 21835766 DOI: 10.1152/ajprenal.00054.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prostaglandins have been implicated as paracrine regulators of renin secretion, but the specific pathways and receptor(s) carrying out these functions have not been fully elucidated. To examine the contributions of prostanoid synthetic pathways and receptors to regulation of renin in the intact animal, we used a panel of mice with targeted disruption of several key genes: cyclooxygenase-2 (COX-2), microsomal PGE synthases 1 and 2 (mPGES1, mPGES2), EP2 and EP4 receptors for PGE(2), and the IP receptor for PGI(2). To activate the macula densa signal for renin stimulation, mice were treated with furosemide over 5 days and renin mRNA levels were determined by real-time RT-PCR. At baseline, there were no differences in renin mRNA levels between wild-type and the various strains of mutant mice. Furosemide caused marked stimulation of renin mRNA expression across all groups of wild-type control mice. This response was completely abrogated in the absence of COX-2, but was unaffected in mice lacking mPGES1 or mPGES2. The absence of G(s)/cAMP-linked EP2 receptors had no effect on stimulation of renin by furosemide and there was only a modest, insignificant reduction in renin responses in mice lacking the IP receptor. By contrast, renin stimulation in EP4(-/-) mice was significantly reduced by ∼70% compared with wild-type controls. These data suggest that stimulation of renin by the macula densa mechanism is mediated by PGE(2) through a pathway requiring COX-2 and the EP4 receptor, but not EP2 or IP receptors. Surprisingly, mPGES1 or mPGES2 are not required, suggesting other alternative mechanisms for generating PGE(2) in response to macula densa stimulation.
Collapse
Affiliation(s)
- Carie S Facemire
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Singh RR, Moritz KM, Wintour EM, Jefferies AJ, Iqbal J, Bertram JF, Denton KM. Fetal uninephrectomy in male sheep alters the systemic and renal responses to angiotensin II infusion and AT1R blockade. Am J Physiol Renal Physiol 2011; 301:F319-26. [DOI: 10.1152/ajprenal.00139.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fetal uninephrectomy (uni-x) at 100 days of gestation results in compensatory nephrogenesis in the remaining kidney, resulting in a 30% reduction in total nephron number in male sheep. Recently, we showed that uni-x males at 6 mo of age have elevated arterial pressure, reduced renal blood flow (RBF), glomerular filtration rate (GFR), and low plasma renin levels (Singh R, Denton K, Bertram J, Jefferies A, Head G, Lombardo P, Schneider-Kolsky M, Moritz K. J Hypertens 27: 386–396, 2009; Singh R, Denton K, Jefferies A, Bertram J, Moritz K. Clin Sci (Lond) 118: 669–680, 2010). We hypothesized this was due to upregulation of the intrarenal renin-angiotensin system (RAS). In this study, renal responses to ANG II infusion and ANG II type 1 receptor (AT1R) blockade were examined in the same 6-mo-old male sheep. Uni-x animals had reduced levels of renal tissue and plasma renin and ANG II. Renal gene expression of renin, and gene and protein levels of AT1R and AT2R, were significantly lower in uni-x animals. In response to graded ANG II infusion, sham animals had the expected decrease in conscious RBF and GFR. Interestingly, the response was biphasic in uni-x sheep, with GFR initially decreasing, but then increasing at higher ANG II doses (34 ± 7%; Pgroup × treatment < 0.001), due to a paradoxical decrease in renal vascular resistance ( Pgroup × treatment < 0.001). In response to AT1R blockade, while GFR and RBF responded similarly between groups, there was a marked increase in sodium excretion in uni-x compared with sham sheep (209 ± 35 vs. 25 ± 12%; P < 0.001). In conclusion, in 6-mo-old male sheep born with a single kidney, these studies demonstrate that this is a low-renin form of hypertension, in which responses to ANG II are perturbed and the intrarenal RAS is downregulated.
Collapse
Affiliation(s)
| | - Karen M. Moritz
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland, Australia
| | - E. Marelyn Wintour
- Departments of 1Anatomy and Developmental Biology and
- Physiology, Monash University, Victoria, and
| | | | - Javed Iqbal
- Physiology, Monash University, Victoria, and
| | | | | |
Collapse
|
50
|
Abstract
In the adult organism, systemically circulating renin almost exclusively originates from the juxtaglomerular cells in the afferent arterioles of the kidneys. These cells share similarities with pericytes and myofibro-blasts. They store renin in a vesicular network and granules and release it in a regulated fashion. The release mode of renin is not understood; in particular, the involvement of SNARE proteins is unknown. Renin release is acutely increased via the cAMP signaling pathway, which is triggered mainly by catecholamines and other G(s)-coupled agonists, and is inhibited by calcium-related pathways that are commonly activated by vasoconstrictors. Renin release from juxtaglomerular cells is directly modulated in an inverse fashion by the blood pressure inside the afferent arterioles and by the chloride content in the tubule fluid at the macula densa segment of the distal tubule. Renin release is stimulated by nitric oxide and by prostanoids released by neighboring endothelial and macula densa cells. Steady-state renin concentrations in the plasma are determined essentially by the number of renin-producing cells in the afferent arterioles, which changes in parallel with challenges to the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Armin Kurtz
- Physiologisches Institut der Universität, Regensburg, Germany.
| |
Collapse
|