1
|
Bulger DA, Griendling KK. Novel Mechanism by Which Extracellular Cyclic GMP Induces Natriuresis. Circ Res 2023; 132:1141-1143. [PMID: 37104563 PMCID: PMC10151062 DOI: 10.1161/circresaha.123.322778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- David A Bulger
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA
| |
Collapse
|
2
|
Kemp BA, Howell NL, Gildea JJ, Hinkle JD, Shabanowitz J, Hunt DF, Conaway MR, Keller SR, Carey RM. Evidence That Binding of Cyclic GMP to the Extracellular Domain of NKA (Sodium-Potassium ATPase) Mediates Natriuresis. Circ Res 2023; 132:1127-1140. [PMID: 36919600 PMCID: PMC10171454 DOI: 10.1161/circresaha.122.321693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Extracellular renal interstitial guanosine cyclic 3',5'-monophosphate (cGMP) inhibits renal proximal tubule (RPT) sodium (Na+) reabsorption via Src (Src family kinase) activation. Through which target extracellular cGMP acts to induce natriuresis is unknown. We hypothesized that cGMP binds to the extracellular α1-subunit of NKA (sodium-potassium ATPase) on RPT basolateral membranes to inhibit Na+ transport similar to ouabain-a cardiotonic steroid. METHODS Urine Na+ excretion was measured in uninephrectomized 12-week-old female Sprague-Dawley rats that received renal interstitial infusions of vehicle (5% dextrose in water), cGMP (18, 36, and 72 μg/kg per minute; 30 minutes each), or cGMP+rostafuroxin (12 ng/kg per minute) or were subjected to pressure-natriuresis±rostafuroxin infusion. Rostafuroxin is a digitoxigenin derivative that displaces ouabain from NKA. RESULTS Renal interstitial cGMP and raised renal perfusion pressure induced natriuresis and increased phosphorylated SrcTyr416 and Erk 1/2 (extracellular signal-regulated protein kinase 1/2)Thr202/Tyr204; these responses were abolished with rostafuroxin coinfusion. To assess cGMP binding to NKA, we performed competitive binding studies with isolated rat RPTs using bodipy-ouabain (2 μM)+cGMP (10 µM) or rostafuroxin (10 µM) and 8-biotin-11-cGMP (2 μM)+ouabain (10 μM) or rostafuroxin (10 µM). cGMP or rostafuroxin reduced bodipy-ouabain fluorescence intensity, and ouabain or rostafuroxin reduced 8-biotin-11-cGMP staining. We cross-linked isolated rat RPTs with 4-N3-PET-8-biotin-11-cGMP (2 μM); 8-N3-6-biotin-10-cAMP served as negative control. Precipitation with streptavidin beads followed by immunoblot analysis showed that RPTs after cross-linking with 4-N3-PET-8-biotin-11-cGMP exhibited a significantly stronger signal for NKA than non-cross-linked samples and cross-linked or non-cross-linked 8-N3-6-biotin-10-cAMP RPTs. Ouabain (10 μM) reduced NKA in cross-linked 4-N3-PET-8-biotin-11-cGMP RPTs confirming fluorescence staining. 4-N3-PET-8-biotin-11-cGMP cross-linked samples were separated by SDS gel electrophoresis and slices corresponding to NKA molecular weight excised and processed for mass spectrometry. NKA was the second most abundant protein with 50 unique NKA peptides covering 47% of amino acids in NKA. Molecular modeling demonstrated a potential cGMP docking site in the ouabain-binding pocket of NKA. CONCLUSIONS cGMP can bind to NKA and thereby mediate natriuresis.
Collapse
Affiliation(s)
- Brandon A Kemp
- Department of Medicine, Division of Endocrinology and Metabolism (B.A.K., N.L.H., S.R.K., R.M.C.), University of Virginia, Charlottesville
| | - Nancy L Howell
- Department of Medicine, Division of Endocrinology and Metabolism (B.A.K., N.L.H., S.R.K., R.M.C.), University of Virginia, Charlottesville
| | - John J Gildea
- Department of Pathology (J.J.G.), University of Virginia, Charlottesville
| | - Josh D Hinkle
- Department of Chemistry (J.D.H., J.S., D.F.H.), University of Virginia, Charlottesville
| | - Jeffrey Shabanowitz
- Department of Chemistry (J.D.H., J.S., D.F.H.), University of Virginia, Charlottesville
| | - Donald F Hunt
- Department of Chemistry (J.D.H., J.S., D.F.H.), University of Virginia, Charlottesville
| | - Mark R Conaway
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences (M.R.C.), University of Virginia, Charlottesville
| | - Susanna R Keller
- Department of Medicine, Division of Endocrinology and Metabolism (B.A.K., N.L.H., S.R.K., R.M.C.), University of Virginia, Charlottesville
| | - Robert M Carey
- Department of Medicine, Division of Endocrinology and Metabolism (B.A.K., N.L.H., S.R.K., R.M.C.), University of Virginia, Charlottesville
| |
Collapse
|
3
|
Taoro-González L, Cabrera-Pastor A, Sancho-Alonso M, Felipo V. Intracellular and extracelluar cyclic GMP in the brain and the hippocampus. VITAMINS AND HORMONES 2022; 118:247-288. [PMID: 35180929 DOI: 10.1016/bs.vh.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cyclic Guanosine-Monophosphate (cGMP) is implicated as second messenger in a plethora of pathways and its effects are executed mainly by cGMP-dependent protein kinases (PKG). It is involved in both peripheral (cardiovascular regulation, intestinal secretion, phototransduction, etc.) and brain (hippocampal synaptic plasticity, neuroinflammation, cognitive function, etc.) processes. Stimulation of hippocampal cGMP signaling have been proved to be beneficial in animal models of aging, Alzheimer's disease or hepatic encephalopathy, restoring different cognitive functions such as passive avoidance, object recognition or spatial memory. However, even when some inhibitors of cGMP-degrading enzymes (PDEs) are already used against peripheral pathologies, their utility as neurological treatments is still under clinical investigation. Additionally, it has been demonstrated a list of cGMP roles as not second but first messenger. The role of extracellular cGMP has been specially studied in hippocampal function and cognitive impairment in animal models and it has emerged as an important modulator of neuroinflammation-mediated cognitive alterations and hippocampal synaptic plasticity malfunction. Specifically, it has been demonstrated that extracellular cGMP decreases hippocampal IL-1β levels restoring membrane expression of glutamate receptors in the hippocampus and cognitive function in hyperammonemic rats. The mechanisms implicated are still unclear and might involve complex interactions between hippocampal neurons, astrocytes and microglia. Membrane targets for extracellular cGMP are still poorly understood and must be addressed in future studies.
Collapse
Affiliation(s)
- Lucas Taoro-González
- Department of Clinical Psychology, Psychobiology and Methodology, Area of Psycobiology, University of La Laguna, Tenerife, Spain
| | - Andrea Cabrera-Pastor
- Fundación Investigación Hospital Clínico, Instituto de Investigación Sanitaria (INCLIVA), Valencia, Spain; Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María Sancho-Alonso
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
4
|
Sousa LGF, Cortez LUADS, Evangelista JSAM, Xavier-Júnior FAF, Heimark DB, Fonteles MC, Santos CF, Nascimento NRF. Renal protective effect of pinitol in experimental diabetes. Eur J Pharmacol 2020; 880:173130. [PMID: 32360975 DOI: 10.1016/j.ejphar.2020.173130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 01/17/2023]
Abstract
Pinitol is a natural antidiabetic agent shown to prevent or ameliorate metabolic and overall vascular and neural function. In the present study we have evaluated the potential benefits of pinitol on renal function of streptozotocin (STZ)-induced diabetic rats. Both euglycemic or 8-week or 16-week diabetic rats were treated with either saline (1 ml/kg/12h; p.o) or pinitol (20 mg/kg/12h; p.o). The renal function was evaluated by using metabolic cages, renal hemodynamic and tubular parameters measurements. Histological examination and evaluation of the protein expression of renal markers such as nephrin, TGFβ and pERK were also performed. Pinitol decreased by 50% the increased urinary albumin/creatinine ratio in both 8-week and 16 week diabetic rats. In addition, the glomerular volume of 16-week rats increased by 55% and this increase was blunted by pinitol. Remarkably, pressure-natriuresis was completely blunted in both 8 and 16-week diabetic rats but this impairment was prevented by pinitol in both treatment regimens. Pinitol ameliorated renal lesions and also prevented the decrease in nephrin expression and the increase of pERK and TGFβ expression in both diabetic groups. Natriuresis due to high renal perfusion pressure increased 7-fold in control animals but was blocked in 16-week diabetic rats and remarkably pinitol partially restored pressure natriuresis (3-fold increase in sodium excretion during pressure natriuresis). Pinitol prevents and ameliorates albuminuria, glomerular expansion, impairment of pressure-natriuresis, renal structural alterations and changes of renal markers and has the potential to be tested for the prevention of diabetic kidney disease.
Collapse
Affiliation(s)
- Luis Gustavo Farias Sousa
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Lôrrainy Umbelina Alves de Souza Cortez
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil; Curso de Nutrição, Universidade de Fortaleza (UNIFOR), Brazil
| | - Janaína Serra Azul Monteiro Evangelista
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil; Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Ceará, Brazil
| | | | - Douglas Biggam Heimark
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Manassés Claudino Fonteles
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Claudia Ferreira Santos
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Nilberto Robson Falcão Nascimento
- Instituto Superior de Ciências Biomédicas, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual do Ceará, Fortaleza, Brazil.
| |
Collapse
|
5
|
Kemp BA, Howell NL, Gildea JJ, Keller SR, Carey RM. Identification of a Primary Renal AT 2 Receptor Defect in Spontaneously Hypertensive Rats. Circ Res 2020; 126:644-659. [PMID: 31997705 DOI: 10.1161/circresaha.119.316193] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Previous studies identified a defect in Ang III (angiotensin III [des-aspartyl1-angiotensin II])-elicited AT2R (Ang type-2 receptor)-mediated natriuresis in renal proximal tubule cells of spontaneously hypertensive rats (SHR). OBJECTIVE This study aimed to delineate in prehypertensive SHR kidneys the receptor or postreceptor defect causing impaired AT2R signaling and renal sodium (Na+) retention by utilizing the selective AT2R agonist compound-21 (C-21). METHODS AND RESULTS Female 4-week-old Wistar Kyoto and SHR rats were studied after 24-hour systemic AT1R (Ang II type-1 receptor) blockade. Left kidneys received 30-minute renal interstitial infusions of vehicle followed by C-21 (20, 40, and 60 ng/[kg·min], each dose 30 minutes). Right kidneys received vehicle infusions. In Wistar Kyoto, C-21 dose-dependently increased urine Na+ excretion from 0.023±0.01 to 0.064±0.02, 0.087±0.01, and 0.089±0.01 µmol/min (P=0.008, P<0.0001, and P<0.0001, respectively) and renal interstitial fluid levels of AT2R downstream signaling molecule cGMP (cyclic guanosine 3',5' monophosphate) from 0.91±0.3 to 3.1±1.0, 5.9±1.2 and 5.3±0.5 fmol/mL (P=nonsignificant, P<0.0001, and P<0.0001, respectively). In contrast, C-21 did not increase urine Na+ excretion or renal interstitial cGMP in SHR. Mean arterial pressure was slightly higher in SHR but within the normotensive range and unaffected by C-21. In Wistar Kyoto, but not SHR, C-21 induced AT2R translocation to apical plasma membranes of renal proximal tubule cells, internalization/inactivation of NHE-3 (sodium-hydrogen exchanger-3) and Na+/K+ATPase (sodium-potassium-atpase) and phosphorylation of AT2R-cGMP downstream signaling molecules Src (Src family kinase), ERK (extracellular signal-related kinase), and VASP (vasodilator-stimulated phosphoprotein). To test whether cGMP could bypass the natriuretic defect in SHR, we infused 8-bromo-cGMP. This restored natriuresis, Na+ transporter internalization/inactivation, and Src and VASP phosphorylation, but not apical plasma membrane AT2R recruitment. In contrast, 8-bromo-cAMP administration had no effect on natriuresis or AT2R recruitment in SHR. CONCLUSIONS The results demonstrate a primary renal proximal tubule cell AT2R natriuretic defect in SHR that may contribute to the development of hypertension. Since the defect is abrogated by exogenous intrarenal cGMP, the renal cGMP pathway may represent a viable target for the treatment of hypertension. Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Brandon A Kemp
- From the Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville (B.A.K., N.L.H., S.R.K., R.M.C.)
| | - Nancy L Howell
- From the Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville (B.A.K., N.L.H., S.R.K., R.M.C.)
| | - John J Gildea
- Department of Pathology, University of Virginia Health System, Charlottesville (J.J.G.)
| | - Susanna R Keller
- From the Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville (B.A.K., N.L.H., S.R.K., R.M.C.)
| | - Robert M Carey
- From the Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville (B.A.K., N.L.H., S.R.K., R.M.C.)
| |
Collapse
|
6
|
Yokota R, Ronchi FA, Fernandes FB, Jara ZP, Rosa RM, Leite APDO, Fiorino P, Farah V, do Nascimento NRF, Fonteles MC, Casarini DE. Intra-Renal Angiotensin Levels Are Increased in High-Fructose Fed Rats in the Extracorporeal Renal Perfusion Model. Front Physiol 2018; 9:1433. [PMID: 30364140 PMCID: PMC6191567 DOI: 10.3389/fphys.2018.01433] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/20/2018] [Indexed: 11/13/2022] Open
Abstract
Overconsumption of fructose leads to metabolic syndrome as a result of hypertension, insulin resistance, and hyperlipidemia. In this study, the renal function of animals submitted to high fructose intake was analyzed from weaning to adulthood using in vivo and ex vivo methods, being compared with a normal control group. We investigated in ex vivo model of the role of the renin Angiotensin system (RAS) in the kidney. The use of perfused kidney from animals submitted to 8-week fructose treatment showed that high fructose intake caused metabolic and cardiovascular alterations that were consistent with other studies. Moreover, the isolated perfused kidneys obtained from rats under high fructose diet showed a 33% increase in renal perfusion pressure throughout the experimental period due to increased renal vascular resistance and a progressive fall in the glomerular filtration rate, which reached a maximum of 64% decrease. Analysis of RAS peptides in the high fructose group showed a threefold increase in the renal concentrations of angiotensin I (Ang I) and a twofold increase in angiotensin II (Ang II) levels, whereas no change in angiotensin 1-7 (Ang 1-7) was observed when compared with the control animals. We did not detect changes in angiotensin converting enzyme (ACE) activity in renal tissues, but there is a tendency to decrease. These observations suggest that there are alternative ways of producing Ang II in this model. Chymase the enzyme responsible for Ang II formation direct from Ang I was increased in renal tissues in the fructose group, confirming the alternative pathway for the formation of this peptide. Neprilysin (NEP) the Ang 1-7 forming showed a significant decrease in activity in the fructose vs. control group, and a tendency of reduction in ACE2 activity. Thus, these results suggest that the Ang 1-7 vasodilator peptide formation is impaired in this model contributing with the increase of blood pressure. In summary, rats fed high fructose affect renal RAS, which may contribute to several deleterious effects of fructose on the kidneys and consequently an increase in blood pressure.
Collapse
Affiliation(s)
- Rodrigo Yokota
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Zaira Palomino Jara
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Rodolfo Mattar Rosa
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | - Patricia Fiorino
- Laboratory of Renal, Cardiovascular, and Metabolic Physiopharmacology, Center for Health and Biological Sciences, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Vera Farah
- Laboratory of Renal, Cardiovascular, and Metabolic Physiopharmacology, Center for Health and Biological Sciences, Mackenzie Presbyterian University, São Paulo, Brazil
| | | | - Manassés C Fonteles
- Laboratory of Renal, Cardiovascular, and Metabolic Physiopharmacology, Center for Health and Biological Sciences, Mackenzie Presbyterian University, São Paulo, Brazil.,Superior Institute of Biomedical Sciences, Ceará State University, Fortaleza, Brazil
| | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Witte J, Mühlbauer M, Braun D, Steinbach A, Golchert J, Rettig R, Grisk O. Renal Soluble Guanylate Cyclase Is Downregulated in Sunitinib-Induced Hypertension. J Am Heart Assoc 2018; 7:e009557. [PMID: 30371202 PMCID: PMC6222942 DOI: 10.1161/jaha.118.009557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background The tyrosine kinase inhibitor sunitinib causes hypertension associated with reduced nitric oxide (NO) availability, elevated renal vascular resistance, and decreased fractional sodium excretion. We tested whether (1) nitrate supplementation mitigates sunitinib‐induced hypertension and NO contributes less to renal vascular resistance as well as fractional sodium excretion regulation in sunitinib‐treated rats than in controls; and (2) renal soluble guanylate cyclase (sGC) is downregulated and sGC activation lowers arterial pressure in rats with sunitinib‐induced hypertension. Methods and Results Arterial pressure responses to nitrate supplementation and the effects of systemic and intrarenal NO synthase (NOS) inhibition on renal hemodynamics and fractional sodium excretion were assessed in sunitinib‐treated rats and controls. Renal NOS and sGC mRNA as well as protein abundances were determined by quantitative polymerase chain reaction and Western blot. The effect of the sGC activator cinaciguat on arterial pressure was investigated in sunitinib‐treated rats. Nitrate supplementation did not mitigate sunitinib‐induced hypertension. Endothelium‐dependent reductions in renal vascular resistance were similar in control and sunitinib‐treated animals without and with systemic NOS inhibition. Selective intrarenal NOS inhibition lowered renal medullary blood flow in control but not in sunitinib‐treated rats without significant effects on fractional sodium excretion. Renal cortical sGC mRNA and sGC α1‐subunit protein abundance were less in sunitinib‐treated rats than in controls, and cinaciguat effectively lowered arterial pressure by 15‐20 mm Hg in sunitinib‐treated rats. Conclusions Renal cortical sGC is downregulated in the presence of intact endothelium‐dependent renal vascular resistance regulation in developing sunitinib‐induced hypertension. This suggests that sGC downregulation occurs outside the renal vasculature, increases renal sodium retention, and contributes to nitrate resistance of sunitinib‐induced hypertension.
Collapse
Affiliation(s)
- Jeannine Witte
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| | - Melanie Mühlbauer
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| | - Diana Braun
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| | - Antje Steinbach
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| | - Janine Golchert
- 2 Interfaculty Institute for Genetics and Functional Genomics University of Greifswald Greifswald Germany
| | - Rainer Rettig
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| | - Olaf Grisk
- 1 Institute of Physiology University of Greifswald Greifswald Germany
| |
Collapse
|
8
|
Carey RM. AT2 Receptors: Potential Therapeutic Targets for Hypertension. Am J Hypertens 2017; 30:339-347. [PMID: 27664954 DOI: 10.1093/ajh/hpw121] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022] Open
Abstract
The renin-angiotensin system (RAS) is arguably the most important and best studied hormonal system in the control of blood pressure (BP) and the pathogenesis of hypertension. The RAS features its main effector angiotensin II (Ang II) acting via its 2 major receptors, angiotensin type-1(AT1R) and type-2 (AT2R). In general, AT2Rs oppose the detrimental actions of Ang II via AT1Rs. AT2R activation induces vasodilation and natriuresis, but its effects to lower BP in hypertension have not been as clear as anticipated. Recent studies, however, have demonstrated that acute and chronic AT2R stimulation can induce natriuresis and lower BP in the Ang II infusion model of experimental hypertension. AT2R activation induces receptor recruitment from intracellular sites to the apical plasma membranes of renal proximal tubule cells via a bradykinin, nitric oxide, and cyclic guanosine 3',5' monophosphate signaling pathway that results in internalization and inactivation of sodium (Na+) transporters Na+-H+ exchanger-3 and Na+/K+ATPase. These responses do not require the presence of concurrent AT1R blockade and are effective both in the prevention and reversal of hypertension. This review will address the role of AT2Rs in the control of BP and Na+ excretion and the case for these receptors as potential therapeutic targets for hypertension in humans.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
9
|
Abstract
Cyclic GMP (cGMP) is a ubiquitous intracellular second messenger that mediates a wide spectrum of physiologic processes in multiple cell types within the cardiovascular and nervous systems. Synthesis of cGMP occurs either by NO-sensitive guanylyl cyclases in response to nitric oxide or by membrane-bound guanylyl cyclases in response to natriuretic peptides and has been shown to regulate blood pressure homeostasis by influencing vascular tone, sympathetic nervous system, and sodium and water handling in the kidney. Several cGMPs degrading phosphodiesterases (PDEs), including PDE1 and PDE5, play an important role in the regulation of cGMP signaling. Recent findings revealed that increased activity of cGMP-hydrolyzing PDEs contribute to the development of hypertension. In this review, we will summarize recent research findings regarding the cGMP/PDE signaling in the vasculature, the central nervous system, and the kidney which are associated with the development and maintenance of hypertension.
Collapse
Affiliation(s)
- Evanthia Mergia
- Department of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
10
|
Ouabain Contributes to Kidney Damage in a Rat Model of Renal Ischemia-Reperfusion Injury. Int J Mol Sci 2016; 17:ijms17101728. [PMID: 27754425 PMCID: PMC5085759 DOI: 10.3390/ijms17101728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/06/2016] [Accepted: 10/10/2016] [Indexed: 01/19/2023] Open
Abstract
Warm renal ischemia performed during partial nephrectomy has been found to be associated with kidney disease. Since endogenous ouabain (EO) is a neuro-endocrine hormone involved in renal damage, we evaluated the role of EO in renal ischemia-reperfusion injury (IRI). We measured plasma and renal EO variations and markers of glomerular and tubular damage (nephrin, KIM-1, Kidney-Injury-Molecule-1, α1 Na-K ATPase) and the protective effect of the ouabain inhibitor, rostafuroxin. We studied five groups of rats: (1) normal; (2) infused for eight weeks with ouabain (30 µg/kg/day, OHR) or (3) saline; (4) ouabain; or (5) saline-infused rats orally treated with 100 µg/kg/day rostafuroxin for four weeks. In group 1, 2-3 h after IRI, EO increased in ischemic kidneys while decreased in plasma. Nephrin progressively decreased and KIM-1 mRNA increased starting from 24 h. Ouabain infusion (group 2) increased blood pressure (from 111.7 to 153.4 mmHg) and ouabain levels in plasma and kidneys. In OHR ischemic kidneys at 120 h from IRI, nephrin, and KIM-1 changes were greater than those detected in the controls infused with saline (group 3). All these changes were blunted by rostafuroxin treatment (groups 4 and 5). These findings support the role of EO in IRI and suggest that rostafuroxin pre-treatment of patients before partial nephrectomy with warm ischemia may reduce IRI, particularly in those with high EO.
Collapse
|
11
|
Kemp BA, Howell NL, Keller SR, Gildea JJ, Padia SH, Carey RM. AT2 Receptor Activation Prevents Sodium Retention and Reduces Blood Pressure in Angiotensin II-Dependent Hypertension. Circ Res 2016; 119:532-43. [PMID: 27323774 DOI: 10.1161/circresaha.116.308384] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022]
Abstract
RATIONALE Compound 21 (C-21) is a highly selective nonpeptide angiotensin AT2 receptor (AT2R) agonist. OBJECTIVE To test the hypothesis that chronic AT2R activation with C-21 induces natriuresis via an action at the renal proximal tubule (RPT) and lowers blood pressure (BP) in experimental angiotensin II (Ang II)-dependent hypertension. METHODS AND RESULTS In rats, Ang II infusion increased both sodium (Na(+)) retention and BP on day 1, and BP remained elevated throughout the 7-day infusion period. Either intrarenal or systemic administration of C-21 prevented Ang II-mediated Na(+) retention on day 1, induced continuously negative cumulative Na(+) balance compared with Ang II alone, and reduced BP chronically. The effects of C-21 are likely to be mediated by action on the RPT as acute systemic C-21-induced natriuresis was additive to that induced by chlorothiazide and amiloride. At 24 hours of Ang II infusion, AT2R activation with C-21, both intrarenally and systemically, translocated AT2Rs from intracellular sites to the apical plasma membranes of RPT cells without altering the total cellular pool of AT2Rs and internalized/inactivated major RPT Na(+) transporters Na(+)-H(+)-exchanger-3 and Na(+)/K(+)ATPase. C-21 lowered BP to a similar degree whether administered before or subsequent to the establishment of Ang II-dependent hypertension. CONCLUSIONS Chronic AT2R activation initiates and sustains receptor translocation to RPT apical plasma membranes, internalizes/inactivates Na(+)-H(+)-exchanger-3 and Na(+)/K(+)ATPase, prevents Na(+) retention resulting in negative cumulative Na(+) balance, and lowers BP in experimental Ang II-induced hypertension. Acting uniquely at the RPT, C-21 is a promising candidate for the treatment of hypertension and Na(+)-retaining states in humans.
Collapse
Affiliation(s)
- Brandon A Kemp
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Nancy L Howell
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Susanna R Keller
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - John J Gildea
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Shetal H Padia
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Robert M Carey
- From the Division of Endocrinology and Metabolism, Department of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Department of Pathology (J.J.G.), University of Virginia Health System, Charlottesville.
| |
Collapse
|
12
|
Kemp BA, Howell NL, Gildea JJ, Keller SR, Padia SH, Carey RM. AT₂ receptor activation induces natriuresis and lowers blood pressure. Circ Res 2014; 115:388-99. [PMID: 24903104 DOI: 10.1161/circresaha.115.304110] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE Compound 21 (C-21) is a highly selective nonpeptide AT2 receptor (AT2R) agonist. OBJECTIVE To test the hypothesis that renal proximal tubule AT2Rs induce natriuresis and lower blood pressure in Sprague-Dawley rats and mice. METHODS AND RESULTS In rats, AT2R activation with intravenous C-21 increased urinary sodium excretion by 10-fold (P<0.0001); this natriuresis was abolished by direct renal interstitial infusion of specific AT2R antagonist PD-123319. C-21 increased fractional excretion of Na(+) (P<0.05) and lithium (P<0.01) without altering renal hemodynamic function. AT2R activation increased renal proximal tubule cell apical membrane AT2R protein (P<0.001) without changing total AT2R expression and internalized/inactivated Na(+)-H(+) exchanger-3 and Na(+)/K(+)ATPase. C-21-induced natriuresis was accompanied by an increase in renal interstitial cGMP (P<0.01); C-21-induced increases in urinary sodium excretion and renal interstitial cGMP were abolished by renal interstitial nitric oxide synthase inhibitor l-N(6)-nitroarginine methyl ester or bradykinin B2 receptor antagonist icatibant. Renal AT2R activation with C-21 prevented Na(+) retention and lowered blood pressure in the angiotensin II infusion model of experimental hypertension. CONCLUSIONS AT2R activation initiates its translocation to the renal proximal tubule cell apical membrane and the internalization of Na(+)-H(+) exchanger-3 and Na(+)/K(+)ATPase, inducing natriuresis in a bradykinin-nitric oxide-cGMP-dependent manner. Intrarenal AT2R activation prevents Na(+) retention and lowers blood pressure in angiotensin II-dependent hypertension. AT2R activation holds promise as a renal proximal tubule natriuretic/diuretic target for the treatment of fluid-retaining states and hypertension.
Collapse
Affiliation(s)
- Brandon A Kemp
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Nancy L Howell
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - John J Gildea
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Susanna R Keller
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Shetal H Padia
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville
| | - Robert M Carey
- From the Departments of Medicine (B.A.K., N.L.H., S.R.K., S.H.P., R.M.C.) and Pathology (J.J.G.), University of Virginia Health System, Charlottesville.
| |
Collapse
|
13
|
Citterio L, Ferrandi M, Delli Carpini S, Simonini M, Kuznetsova T, Molinari I, Dell' Antonio G, Lanzani C, Merlino L, Brioni E, Staessen JA, Bianchi G, Manunta P. cGMP-dependent protein kinase 1 polymorphisms underlie renal sodium handling impairment. Hypertension 2013; 62:1027-33. [PMID: 24060892 DOI: 10.1161/hypertensionaha.113.01628] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Defective pressure-natriuresis related to abnormalities in the natriuretic response has been associated with hypertension development. A major signaling pathway mediating pressure natriuresis involves the cGMP-dependent protein kinase 1 (PRKG1) that, once activated by Src kinase, inhibits renal Na(+) reabsorption via a direct action on basolateral Na-K ATPase and luminal Na-H exchanger type 3, as shown in renal tubuli of animals. Because a clear implication of PRKG1 in humans is still lacking, here we addressed whether PRKG1 polymorphisms affect pressure-natriuresis in patients. Naive hypertensive patients (n = 574), genotyped for PRKG1 rs1904694, rs7897633, and rs7905063 single nucleotide polymorphisms (SNPs), underwent an acute Na(+) loading, and the slope of the pressure-natriuresis relationship between blood pressure and Na(+) excretion was calculated. The underlying molecular mechanism was investigated by immunoblotting protein quantifications in human kidneys. The results demonstrate that the PRKG1 risk haplotype GAT (rs1904694, rs7897633, rs7905063, respectively) associates with a rightward shift of the pressure-natriuresis curve (0.017 ± 0.004 μEq/mm Hg per minute) compared with the ACC (0.0013 ± 0.003 μEq/mm Hg per minute; P = 0.001). In human kidneys, a positive correlation of protein expression levels between PRKG1 and Src (r = 0.83; P<0.001) or α1 Na-K ATPase (r = 0.557; P<0.01) and between α1 Na-K ATPase and Na-H exchanger type 3 (r = 0.584; P<0.01) or Src (r = 0.691; P<0.001) was observed in patients carrying PRKG1 risk GAT (n = 23) but not ACC (n = 14) variants. A functional signaling complex among PRKG1, α1 Na-K ATPase, and Src was shown by immunoprecipitation from human renal caveolae. These findings indicate that PRKG1 risk alleles associate with salt-sensitivity related to a loss of the inhibitory control of renal Na(+) reabsorption, suggestive of a blunt pressure-natriuresis response.
Collapse
Affiliation(s)
- Lorena Citterio
- San Raffaele Scientific Institute, Nephrology and Dialysis, Università Vita Salute San Raffaele, Via Olgettina 60, 20132 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Nakagawa T, Touhara K. Extracellular modulation of the silkmoth sex pheromone receptor activity by cyclic nucleotides. PLoS One 2013; 8:e63774. [PMID: 23755109 PMCID: PMC3670925 DOI: 10.1371/journal.pone.0063774] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 04/06/2013] [Indexed: 11/25/2022] Open
Abstract
Odorants and pheromones are essential to insects as chemical cues for finding food or an appropriate mating partner. These volatile compounds bind to olfactory receptors (Ors) expressed by olfactory sensory neurons. Each insect Or functions as a ligand-gated ion channel and is a heteromeric complex that comprises one type of canonical Or and a highly conserved Orco subunit. Because there are many Or types, insect Ors can recognize with high specificity a myriad of chemical cues. Cyclic nucleotides can modulate the activity of insect Or-Orco complexes; however, the mechanism of action of these nucleotides is under debate. Here, we show that cyclic nucleotides, including cAMP and cGMP, interact with the silkmoth sex pheromone receptor complex, BmOr-1-BmOrco, from the outside of the cell and that these nucleotides act as antagonists at low concentrations and weak agonists at high concentrations. These cyclic nucleotides do not compete with the sex pheromone, bombykol, for binding to the BmOr-1 subunit. ATP and GTP also weakly inhibited BmOr-1-BmOrco activity, but D-ribose had no effect; these findings indicated that the purine moiety was crucial for the inhibition. Only the bombykol receptors have been so far shown to be subject to modulation by nucleotide-related compounds, indicating that this responsiveness to these compounds is not common for all insect Or-Orco complexes.
Collapse
Affiliation(s)
- Tatsuro Nakagawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazushige Touhara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- JST ERATO Touhara Chemosensory Signal Project, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
15
|
Gabor A, Leenen FHH. Central neuromodulatory pathways regulating sympathetic activity in hypertension. J Appl Physiol (1985) 2012; 113:1294-303. [PMID: 22773773 DOI: 10.1152/japplphysiol.00553.2012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The classical neurotransmitters, glutamate and GABA, mediate fast (milliseconds) synaptic transmission and modulate its effectiveness through slow (seconds to minutes) signaling processes. Angiotensinergic pathways, from the lamina terminalis to the paraventricular nucleus (PVN)/supraoptic nucleus and rostral ventrolateral medulla (RVLM), are activated by stimuli such as circulating angiotensin type II (Ang II), cerebrospinal fluid (CSF) sodium ion concentration ([Na(+)]), and possibly plasma aldosterone, leading to sympathoexcitation, largely by decreasing GABA and increasing glutamate release. The aldosterone-endogenous ouabain (EO) pathway is a much slower neuromodulatory pathway. Aldosterone enhances EO release, and the latter increases chronic activity in angiotensinergic pathways by, e.g., increasing expression for Ang I receptor (AT(1)R) and NADPH oxidase subunits in the PVN. Blockade of this pathway does not affect the initial sympathoexcitatory and pressor responses but to a large extent, prevents chronic responses to CSF [Na(+)] or Ang II. Recruitment of these two neuromodulatory pathways allows the central nervous system (CNS) to shift gears to rapidly cause and sustain sympathetic hyperactivity in an efficient manner. Decreased GABA release, increased glutamate release, and enhanced AT(1)R activation in, e.g., the PVN and RVLM contribute to the elevated blood pressure in a number of hypertension models. In Dahl S rats and spontaneous hypertensive rats, high salt activates the CNS aldosterone-EO pathway, and the salt-induced hypertension can be prevented/reversed by specific CNS blockade of any of the steps in the cascade from aldosterone synthase to AT(1)R. Further studies are needed to advance our understanding of how and where in the brain these rapid, slow, and very slow CNS pathways are activated and interact in models of hypertension and other disease states associated with chronic sympathetic hyperactivity.
Collapse
Affiliation(s)
- Alexander Gabor
- Hypertension Unit, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | |
Collapse
|
16
|
Gildea JJ, Wang X, Shah N, Tran H, Spinosa M, Van Sciver R, Sasaki M, Yatabe J, Carey RM, Jose PA, Felder RA. Dopamine and angiotensin type 2 receptors cooperatively inhibit sodium transport in human renal proximal tubule cells. Hypertension 2012; 60:396-403. [PMID: 22710646 DOI: 10.1161/hypertensionaha.112.194175] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Little is known regarding how the kidney shifts from a sodium and water reclaiming state (antinatriuresis) to a state where sodium and water are eliminated (natriuresis). In human renal proximal tubule cells, sodium reabsorption is decreased by the dopamine D(1)-like receptors (D(1)R/D(5)R) and the angiotensin type 2 receptor (AT(2)R), whereas the angiotensin type 1 receptor increases sodium reabsorption. Aberrant control of these opposing systems is thought to lead to sodium retention and, subsequently, hypertension. We show that D(1)R/D(5)R stimulation increased plasma membrane AT(2)R 4-fold via a D(1)R-mediated, cAMP-coupled, and protein phosphatase 2A-dependent specific signaling pathway. D(1)R/D(5)R stimulation also reduced the ability of angiotensin II to stimulate phospho-extracellular signal-regulated kinase, an effect that was partially reversed by an AT(2)R antagonist. Fenoldopam did not increase AT(2)R recruitment in renal proximal tubule cells with D(1)Rs uncoupled from adenylyl cyclase, suggesting a role of cAMP in mediating these events. D(1)Rs and AT(2)Rs heterodimerized and cooperatively increased cAMP and cGMP production, protein phosphatase 2A activation, sodium-potassium-ATPase internalization, and sodium transport inhibition. These studies shed new light on the regulation of renal sodium transport by the dopaminergic and angiotensin systems and potential new therapeutic targets for selectively treating hypertension.
Collapse
Affiliation(s)
- John J Gildea
- University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Althaus M. Gasotransmitters: novel regulators of epithelial na(+) transport? Front Physiol 2012; 3:83. [PMID: 22509167 PMCID: PMC3321473 DOI: 10.3389/fphys.2012.00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 03/20/2012] [Indexed: 11/13/2022] Open
Abstract
The vectorial transport of Na(+) across epithelia is crucial for the maintenance of Na(+) and water homeostasis in organs such as the kidneys, lung, or intestine. Dysregulated Na(+) transport processes are associated with various human diseases such as hypertension, the salt-wasting syndrome pseudohypoaldosteronism type 1, pulmonary edema, cystic fibrosis, or intestinal disorders, which indicate that a precise regulation of epithelial Na(+) transport is essential. Novel regulatory signaling molecules are gasotransmitters. There are currently three known gasotransmitters: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H(2)S). These molecules are endogenously produced in mammalian cells by specific enzymes and have been shown to regulate various physiological processes. There is a growing body of evidence which indicates that gasotransmitters may also regulate Na(+) transport across epithelia. This review will summarize the available data concerning NO, CO, and H(2)S dependent regulation of epithelial Na(+) transport processes and will discuss whether or not these mediators can be considered as true physiological regulators of epithelial Na(+) transport biology.
Collapse
Affiliation(s)
- Mike Althaus
- Institute of Animal Physiology, Justus Liebig University of Giessen Giessen, Germany
| |
Collapse
|