1
|
Lu YY, Li SJ, Zhang Z, He S, Guo YT, Hong MN, Shao S, Wang RQ, Zhang J, Wang JG, Gao PJ, Li XD. C-atrial natriuretic peptide (ANP) 4-23 attenuates renal fibrosis in deoxycorticosterone-acetate-salt hypertensive mice. Exp Cell Res 2023; 431:113738. [PMID: 37572787 DOI: 10.1016/j.yexcr.2023.113738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/08/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
Epithelial-mesenchymal transition (EMT) plays a critical role in hypertension-induced renal fibrosis, a final pathway that leads to end-stage renal failure. C-Atrial natriuretic peptide (ANP)4-23, a specific agonist of natriuretic peptide receptor-C (NPR-C), has been reported to have protective effects against hypertension. However, the role of C-ANP4-23 in hypertension-associated renal fibrosis has not yet been elucidated. In this study, mice were randomly divided into SHAM group, DOCA-salt group and DOCA-salt + C-ANP4-23 group. Renal morphology changes, renal function and fibrosis were detected. Human proximal tubular epithelial cells (HK2) stimulated by aldosterone were used for cell function and mechanism study. The DOCA-salt treated mice exhibited hypertension, kidney fibrosis and renal dysfunction, which were attenuated by C-ANP4-23. Moreover, C-ANP4-23 inhibited DOCA-salt treatment-induced renal EMT as evidenced by decrease of the mesenchymal marker alpha-smooth muscle actin (ACTA2) and vimentin and increase of epithelial cell marker E-cadherin. In HK2 cells, aldosterone induced EMT response, which was also suppressed by C-ANP4-23. The key transcription factors (twist, snail, slug and ZEB1) involved in EMT were increased in the kidney of DOCA-salt-treated mice, which were also suppressed by C-ANP4-23. Mechanistically, C-ANP4-23 inhibited the aldosterone-induced translocation of MR from cytosol to nucleus without change of MR expression. Furthermore, C-ANP4-23 rescued the enhanced expression of NADPH oxidase (NOX) 4 and oxidative stress after aldosterone stimulation. Aldosterone-induced Akt and Erk1/2 activation was also suppressed by C-ANP4-23. Our data suggest that C-ANP4-23 attenuates renal fibrosis, likely through inhibition of MR activation, enhanced oxidative stress and Akt and Erk1/2 signaling pathway.
Collapse
Affiliation(s)
- Yuan-Yuan Lu
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China; Shanghai Geriatric Medical Center, Shanghai, China; Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi-Jin Li
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China; State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zhong Zhang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Shun He
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yue-Tong Guo
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Mo-Na Hong
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Shuai Shao
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Rui-Qi Wang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jia Zhang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Xiao-Dong Li
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
2
|
Johnston JG, Welch AK, Cain BD, Sayeski PP, Gumz ML, Wingo CS. Aldosterone: Renal Action and Physiological Effects. Compr Physiol 2023; 13:4409-4491. [PMID: 36994769 PMCID: PMC11472823 DOI: 10.1002/cphy.c190043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Aldosterone exerts profound effects on renal and cardiovascular physiology. In the kidney, aldosterone acts to preserve electrolyte and acid-base balance in response to changes in dietary sodium (Na+ ) or potassium (K+ ) intake. These physiological actions, principally through activation of mineralocorticoid receptors (MRs), have important effects particularly in patients with renal and cardiovascular disease as demonstrated by multiple clinical trials. Multiple factors, be they genetic, humoral, dietary, or otherwise, can play a role in influencing the rate of aldosterone synthesis and secretion from the adrenal cortex. Normally, aldosterone secretion and action respond to dietary Na+ intake. In the kidney, the distal nephron and collecting duct are the main targets of aldosterone and MR action, which stimulates Na+ absorption in part via the epithelial Na+ channel (ENaC), the principal channel responsible for the fine-tuning of Na+ balance. Our understanding of the regulatory factors that allow aldosterone, via multiple signaling pathways, to function properly clearly implicates this hormone as central to many pathophysiological effects that become dysfunctional in disease states. Numerous pathologies that affect blood pressure (BP), electrolyte balance, and overall cardiovascular health are due to abnormal secretion of aldosterone, mutations in MR, ENaC, or effectors and modulators of their action. Study of the mechanisms of these pathologies has allowed researchers and clinicians to create novel dietary and pharmacological targets to improve human health. This article covers the regulation of aldosterone synthesis and secretion, receptors, effector molecules, and signaling pathways that modulate its action in the kidney. We also consider the role of aldosterone in disease and the benefit of mineralocorticoid antagonists. © 2023 American Physiological Society. Compr Physiol 13:4409-4491, 2023.
Collapse
Affiliation(s)
- Jermaine G Johnston
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Amanda K Welch
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Peter P Sayeski
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| |
Collapse
|
3
|
Temiz-Resitoglu M, Guden DS, Senol SP, Vezir O, Sucu N, Kibar D, Yılmaz SN, Tunctan B, Malik KU, Sahan-Firat S. Pharmacological Inhibition of Mammalian Target of Rapamycin Attenuates Deoxycorticosterone Acetate Salt-Induced Hypertension and Related Pathophysiology: Regulation of Oxidative Stress, Inflammation, and Cardiovascular Hypertrophy in Male Rats. J Cardiovasc Pharmacol 2022; 79:355-367. [PMID: 34840266 DOI: 10.1097/fjc.0000000000001187] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/04/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT The present study aimed to explore the contribution of mammalian target of rapamycin (mTOR) in deoxycorticosterone acetate (DOCA) salt-induced hypertension and related pathophysiological changes in cardiovascular and renal tissues. DOCA salt loading resulted in an increase in systolic blood pressure, diastolic blood pressure, and mean blood pressure along with the activity of ribosomal protein S6, the effector protein of mTOR. Treatment with rapamycin, the selective inhibitor of mTOR, initiated at the fourth week of DOCA- salt administration normalized the systolic blood pressure and attenuated ribosomal protein S6 activity in the heart, aorta, and kidney. Cardiac and vascular hypertrophy, oxidative stress, and infiltration of macrophages (CD68+), the marker of inflammation, were also reduced in rapamycin-treated, DOCA-salt, hypertensive rats. In addition, renal hypertrophy and dysfunction were also reduced with rapamycin-treated hypertensive rats. Moreover, these pathophysiological changes in DOCA-salt hypertensive rats were associated with increased NADPH oxidase (NOX) activity, gp91phox (formerly NOX2) expression, ERK1/2, and p38 MAPK activities in the heart, aorta, and kidney were minimized by rapamycin. These data indicate that mTOR plays an important role in regulating blood pressure and the development of cardiovascular and renal pathophysiological changes, most likely due to increased NOX expression/activity, ERK1/2, and p38 MAPK activity with macrophages infiltration in the heart, kidney, and aorta. Pharmacological inhibition of mTOR and related signaling pathways could serve as a novel target for the treatment of hypertension.
Collapse
Affiliation(s)
| | - Demet S Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Sefika P Senol
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Ozden Vezir
- Department of Cardiovascular Surgery, Mersin State Hospital, Mersin, Turkey
| | - Nehir Sucu
- Departments of Cardiovascular Surgery; and
| | - Deniz Kibar
- Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey ; and
| | - Sakir N Yılmaz
- Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey ; and
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
4
|
Shao S, Li XD, Lu YY, Li SJ, Chen XH, Zhou HD, He S, Guo YT, Lu X, Gao PJ, Wang JG. Renal Natriuretic Peptide Receptor-C Deficiency Attenuates NaCl Cotransporter Activity in Angiotensin II-Induced Hypertension. Hypertension 2021; 77:868-881. [PMID: 33486984 DOI: 10.1161/hypertensionaha.120.15636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have identified that NPR-C (natriuretic peptide receptor-C) variants are associated with elevation of blood pressure. However, the mechanism underlying the relationship between NPR-C and blood pressure regulation remains elusive. Here, we investigate whether NPR-C regulates Ang II (angiotensin II)-induced hypertension through sodium transporters activity. Wild-type mice responded to continuous Ang II infusion with an increased renal NPR-C expression. Global NPR-C deficiency attenuated Ang II-induced increased blood pressure both in male and female mice associated with more diuretic and natriuretic responses to a saline challenge. Interestingly, Ang II increased both total and phosphorylation of NCC (NaCl cotransporter) abundance involving in activation of WNK4 (with-no-lysine kinase 4)/SPAK (Ste20-related proline/alanine-rich kinase) which was blunted by NPR-C deletion. NCC inhibitor, hydrochlorothiazide, failed to induce natriuresis in NPR-C knockout mice. Moreover, low-salt and high-salt diets-induced changes of total and phosphorylation of NCC expression were normalized by NPR-C deletion. Importantly, tubule-specific deletion of NPR-C also attenuated Ang II-induced elevated blood pressure, total and phosphorylation of NCC expression. Mechanistically, in distal convoluted tubule cells, Ang II dose and time-dependently upregulated WNK4/SPAK/NCC kinase pathway and NPR-C/Gi/PLC/PKC signaling pathway mediated NCC activation. These results demonstrate that NPR-C signaling regulates NCC function contributing to sodium retention-mediated elevated blood pressure, which suggests that NPR-C is a promising candidate for the treatment of sodium retention-related hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Blood Pressure/genetics
- Blood Pressure/physiology
- Cells, Cultured
- Female
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/physiopathology
- Kidney/metabolism
- Kidney Tubules, Distal/cytology
- Kidney Tubules, Distal/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Atrial Natriuretic Factor/deficiency
- Receptors, Atrial Natriuretic Factor/genetics
- Renin-Angiotensin System/genetics
- Renin-Angiotensin System/physiology
- Signal Transduction/genetics
- Sodium/blood
- Sodium/urine
- Solute Carrier Family 12, Member 3/genetics
- Solute Carrier Family 12, Member 3/metabolism
- Mice
Collapse
Affiliation(s)
- Shuai Shao
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao-Dong Li
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Yuan-Yuan Lu
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Shi-Jin Li
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao-Hui Chen
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Han-Dan Zhou
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Shun He
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Yue-Tong Guo
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao Lu
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Ping-Jin Gao
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Ji-Guang Wang
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
5
|
Alawi LF, Emberesh SE, Owuor BA, Chodavarapu H, Fadnavis R, El‐Amouri SS, Elased KM. Effect of hyperglycemia and rosiglitazone on renal and urinary neprilysin in db/db diabetic mice. Physiol Rep 2020; 8:e14364. [PMID: 32026607 PMCID: PMC7002536 DOI: 10.14814/phy2.14364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/11/2022] Open
Abstract
Alteration in renin-angiotensin system (RAS) has been implicated in the pathophysiology of diabetic kidney disease (DKD). The deleterious actions of angiotensin II (Ang II) could be antagonized by the formation of Ang-(1-7), generated by the actions of angiotensin-converting enzyme 2 (ACE2) and neprilysin (NEP). NEP degrades several peptides, including natriuretic peptides, bradykinin, amyloid beta, and Ang I. Although combination of Ang II receptor and NEP inhibitor treatment benefits patients with heart failure, the role of NEP in renal pathophysiology is a matter of active research. NEP pathway is a potent enzyme in Ang I to Ang-(1-7) conversion in the kidney of ACE2-deficient mice, suggesting a renoprotective role of NEP. The aim of the study is to test the hypothesis that chronic hyperglycemia downregulates renal NEP protein expression and activity in db/db diabetic mice and treatment with rosiglitazone normalizes hyperglycemia, renal NEP expression, and attenuates albuminuria. Mice received rosiglitazone (20 mg kg-1 day-1 ) for 10 weeks. Western blot analysis, immunohistochemistry, and enzyme activity revealed a significant decrease in renal and urinary NEP expression and activity in 16-wk db/db mice compared with lean control (p < .0001). Rosiglitazone also attenuated albuminuria and increased renal and urinary NEP expressions (p < .0001). In conclusion, data support the hypothesis that diabetes decreases intrarenal NEP, which could have a pivotal role in the pathogenesis of DKD. Urinary NEP may be used as an index of intrarenal NEP status. The renoprotective effects of rosiglitazone could be mediated by upregulation of renal NEP expression and activity in db/db diabetic mice.
Collapse
Affiliation(s)
- Laale F. Alawi
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| | - Sana E. Emberesh
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| | - Brenda A. Owuor
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| | - Harshita Chodavarapu
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| | - Rucha Fadnavis
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| | - Salim S. El‐Amouri
- Boonshoft School of MedicineDepartment of NeuroscienceCell Biology and PhysiologyWright State UniversityDaytonOHUSA
| | - Khalid M. Elased
- Department of Pharmacology and ToxicologyBoonshoft School of MedicineWright State UniversityDaytonOHUSA
| |
Collapse
|
6
|
Blaustein MP, Hamlyn JM. Ouabain, endogenous ouabain and ouabain-like factors: The Na + pump/ouabain receptor, its linkage to NCX, and its myriad functions. Cell Calcium 2020; 86:102159. [PMID: 31986323 DOI: 10.1016/j.ceca.2020.102159] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
In this brief review we discuss some aspects of the Na+ pump and its roles in mediating the effects of ouabain and endogenous ouabain (EO): i) in regulating the cytosolic Ca2+ concentration ([Ca2+]CYT) via Na/Ca exchange (NCX), and ii) in activating a number of protein kinase (PK) signaling cascades that control a myriad of cell functions. Importantly, [Ca2+]CYT and the other signaling pathways intersect at numerous points because of the influence of Ca2+ and calmodulin in modulating some steps in those other pathways. While both mechanisms operate in virtually all cells and tissues, this article focuses primarily on their functions in the cardiovascular system, the central nervous system (CNS) and the kidneys.
Collapse
Affiliation(s)
- Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
Zhang C, He X, Murphy SR, Zhang H, Wang S, Ge Y, Gao W, Williams JM, Geurts AM, Roman RJ, Fan F. Knockout of Dual-Specificity Protein Phosphatase 5 Protects Against Hypertension-Induced Renal Injury. J Pharmacol Exp Ther 2019; 370:206-217. [PMID: 31118214 PMCID: PMC6636243 DOI: 10.1124/jpet.119.258954] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/16/2019] [Indexed: 12/28/2022] Open
Abstract
Dual-specificity protein phosphatase 5 (DUSP5) is a member of the tyrosine-threonine phosphatase family with the ability to dephosphorylate and inactivate extracellular signal-related kinase (ERK). The present study investigates whether knockout (KO) of Dusp5 improves renal hemodynamics and protects against hypertension-induced renal injury. The renal expression of DUSP5 was reduced, and the levels of phosphorylated (p) ERK1/2 and p-protein kinase C (PKC) α were elevated in the KO rats. KO of Dusp5 enhanced the myogenic tone of the renal afferent arteriole and interlobular artery in vitro with or without induction of deoxycorticosterone acetate-salt hypertension. Inhibition of ERK1/2 and PKC diminished the myogenic response to a greater extent in Dusp5 KO rats. Autoregulation of renal blood flow was significantly impaired in hypertensive wild-type (WT) rats but remained intact in Dusp5 KO animals. Proteinuria was markedly decreased in hypertensive KO versus WT rats. The degree of glomerular injury was reduced, and the expression of nephrin in the glomerulus was higher in hypertensive Dusp5 KO rats. Renal fibrosis and medullary protein cast formation were attenuated in hypertensive Dusp5 KO rats in association with decreased expression of monocyte chemoattractant protein 1, transforming growth factor-β1, matrix metalloproteinase (MMP) 2, and MMP9. These results indicate that KO of Dusp5 protects against hypertension-induced renal injury, at least in part, by maintaining the myogenic tone of the renal vasculature and extending the range of renal blood flow autoregulation to higher pressures, which diminish glomerular injury, protein cast formation, macrophage infiltration, and epithelial-mesenchymal transformation in the kidney. SIGNIFICANCE STATEMENT: Dual-specificity protein phosphatase 5 (DUSP5) is a tyrosine-threonine phosphatase that inactivates extracellular signal-related kinase (ERK). We previously reported that knockout (KO) of Dusp5 enhanced the myogenic response and autoregulation in the cerebral circulation. The present study investigates whether KO of DUSP5 improves renal hemodynamics and protects against hypertension-induced renal injury. Downregulation of DUSP5 enhanced the myogenic tone of renal arteriole and artery and autoregulation of renal blood flow in association with reduced proteinuria, glomerular injury, and interstitial fibrosis after the induction of hypertension. Inhibition of ERK1/2 and protein kinase C diminished the myogenic response to a greater extent in Dusp5 KO rats. These results suggest that DUSP5 might be a viable drug target for the treatment of hypertension nephropathy.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Sydney R Murphy
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Ying Ge
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Aron M Geurts
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center. Jackson, Mississippi (C.Z., X.H., S.R.M., H.Z., S.W., Y.G., W.G., J.M.W., R.J.R., F.F.); Department of Urology, Zhongshan Hospital, Fudan University. Shanghai, China (C.Z., W.G.); and Department of Physiology, Medical College of Wisconsin. Milwaukee, Wisconsin (A.M.G.)
| |
Collapse
|
8
|
Gandhi H, Naik P, Agrawal N, Yadav M. Protective effects of MCR-1329, a dual α1 and angII receptor antagonist, in mineralocorticoid-induced hypertension. Pharmacol Rep 2016; 68:952-9. [PMID: 27371897 DOI: 10.1016/j.pharep.2016.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND With the prototypical structures of losartan and prazosin as the axis of our research, MCR-1329 emerged as a potential designed multiple ligand from a series of compounds designed to possess dual antagonistic activity on the α1 and AT1 receptor. After confirming the activity of MCR-1329 in in vitro and acute in vivo models, the present study was undertaken to determine the efficacy of MCR-1329 in a mammalian test system. METHODS A rat model of deoxycorticosterone acetate (DOCA)-salt induced renal hypertension following unilateral nephrectomy was utilized to determine the effect of MCR-1329. For mechanistic evaluations, MCR-1329 was evaluated on rat aortic strips in vitro and on rat aortic smooth muscle cells to determine the role of MCR-1329 on phosphoinositide 3 kinase (PI3K) signaling. RESULTS Results of the study showed that MCR-1329 prevents development of arterial hypertension. It was also observed that MCR-1329 upheld the intimal structures of major arteries like the thoracic aorta. Acetylcholine (Ach)-mediated relaxation remained intact in arteries from MCR-1329 treated animals. It was observed that MCR-1329 partially prevents Thr-308 phosphorylation of Akt following ligand-mediated receptor stimulation in vascular smooth muscle cells. Addition of LY294002 to the reaction medium caused a near-complete inhibition of Akt-phosphorylation. This suggested that MCR-1329 elicits its antihypertensive role by blocking activation of receptor-mediated PI3K-Akt downstream signaling as well as through preservation of arterial integrity. CONCLUSIONS MCR-1329 has the potential to be evaluated further for clinical development as a potential antihypertensive agent with multiple mechanisms of action.
Collapse
Affiliation(s)
- Hardik Gandhi
- Faculty of Pharmacy, Kalabhavan Campus, The M. S. University of Baroda, Vadodara, Gujarat, India
| | - Prashant Naik
- Faculty of Pharmacy, Kalabhavan Campus, The M. S. University of Baroda, Vadodara, Gujarat, India
| | - Nitesh Agrawal
- Faculty of Pharmacy, Kalabhavan Campus, The M. S. University of Baroda, Vadodara, Gujarat, India
| | - Mangeram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The M. S. University of Baroda, Vadodara, Gujarat, India.
| |
Collapse
|
9
|
Huang X, Dorhout Mees E, Vos P, Hamza S, Braam B. Everything we always wanted to know about furosemide but were afraid to ask. Am J Physiol Renal Physiol 2016; 310:F958-71. [PMID: 26911852 DOI: 10.1152/ajprenal.00476.2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/23/2016] [Indexed: 12/22/2022] Open
Abstract
Furosemide is a widely used, potent natriuretic drug, which inhibits the Na(+)-K(+)-2Cl(-) cotransporter (NKCC)-2 in the ascending limb of the loop of Henle applied to reduce extracellular fluid volume expansion in heart and kidney disease. Undesirable consequences of furosemide, such as worsening of kidney function and unpredictable effects on sodium balance, led to this critical evaluation of how inhibition of NKCC affects renal and cardiovascular physiology. This evaluation reveals important knowledge gaps, involving furosemide as a drug, the function of NKCC2 (and NKCC1), and renal and systemic indirect effects of NKCC inhibition. Regarding renal effects, renal blood flow and glomerular filtration rate could become compromised by activation of tubuloglomerular feedback or by renin release, particularly if renal function is already compromised. Modulation of the intrarenal renin angiotensin system, however, is ill-defined. Regarding systemic effects, vasodilation followed by nonspecific NKCC inhibition and changes in venous compliance are not well understood. Repetitive administration of furosemide induces short-term (braking phenomenon, acute diuretic resistance) and long-term (chronic diuretic resistance) adaptations, of which the mechanisms are not well known. Modulation of NKCC2 expression and activity in kidney and heart failure is ill-defined. Lastly, furosemide's effects on cutaneous sodium stores and on uric acid levels could be beneficial or detrimental. Concluding, a considerable knowledge gap is identified regarding a potent drug with a relatively specific renal target, NKCC2, and renal and systemic actions. Resolving these questions would increase the understanding of NKCCs and their actions and improve rational use of furosemide in pathophysiology of fluid volume expansion.
Collapse
Affiliation(s)
- Xiaohua Huang
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Evert Dorhout Mees
- Department of Medicine/Nephrology, Utrecht University, Vorden, The Netherlands
| | - Pieter Vos
- Dianet Dialysis Centers, Utrecht, The Netherlands; and
| | - Shereen Hamza
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Branko Braam
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
Zhang Y, Sun Y, Ding G, Huang S, Zhang A, Jia Z. Inhibition of Mitochondrial Complex-1 Prevents the Downregulation of NKCC2 and ENaCα in Obstructive Kidney Disease. Sci Rep 2015. [PMID: 26207612 PMCID: PMC4513566 DOI: 10.1038/srep12480] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ureteral obstruction with subsequent hydronephrosis is a common clinical complication. Downregulation of renal sodium transporters in obstructed kidneys could contribute to impaired urinary concentrating capability and salt waste following the release of a ureteral obstruction. The current study was undertaken to investigate the role of mitochondrial complex-1 inhibition in modulating sodium transporters in obstructive kidney disease. Following unilateral ureteral obstruction (UUO) for 7 days, a global reduction of sodium transporters, including NHE3, α-Na-K-ATPase, NCC, NKCC2, p-NKCC2, ENaCα, and ENaCγ, was observed, as determined via qRT-PCR and/or Western blotting. Interestingly, inhibition of mitochondrial complex-1 by rotenone markedly reversed the downregulation of NKCC2, p-NKCC2, and ENaCα. In contrast, other sodium transporters were not affected by rotenone. To study the potential mechanisms involved in mediating the effects of rotenone on sodium transporters, we examined a number of known sodium modulators, including PGE2, ET1, Ang II, natriuretic peptides (ANP, BNP, and CNP), and nitric oxide synthases (iNOS, nNOS, and eNOS). Importantly, among these modulators, only BNP and iNOS were significantly reduced by rotenone treatment. Collectively, these findings demonstrated a substantial role of mitochondrial dysfunction in mediating the downregulation of NKCC2 and ENaCα in obstructive kidney disease, possibly via iNOS-derived nitric oxide and BNP.
Collapse
Affiliation(s)
- Yue Zhang
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| | - Ying Sun
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| | - Guixia Ding
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| | - Songming Huang
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| | - Aihua Zhang
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| | - Zhanjun Jia
- 1] Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China [2] Institute of Pediatrics, Nanjing Medical University, Nanjing, China [3] Nanjing Key Laboratory of Pediatrics, Nanjing Children Hospital, Affiliated with Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
11
|
Aleksinskaya MA, van Faassen EEH, Nelissen J, Janssen BJA, De Mey JGR, Hanemaaijer R, Rabelink T, van Zonneveld AJ. Identification of free nitric oxide radicals in rat bone marrow: implications for progenitor cell mobilization in hypertension. PLoS One 2013; 8:e57761. [PMID: 23554866 PMCID: PMC3595254 DOI: 10.1371/journal.pone.0057761] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/24/2013] [Indexed: 01/01/2023] Open
Abstract
Nitric oxide (NO) has been implicated in matrix metallopeptidase 9 (MMP9)-dependent mobilization of hematopoietic stem and progenitor cells from bone marrow (BM). However, direct measurement of NO in the BM remained elusive due to its low in situ concentration and short lifetime. Using NO spin trapping and electron paramagnetic resonance (EPR) spectroscopy we give the first experimental confirmation of free NO radicals in rodent BM. NO production was quantified and attributed to enzymatic activity of NO synthases (NOS). Although endothelial NOS (eNOS) accounts for most (66%) of basal NO, we identified a significant contribution (23%) from inducible NOS (iNOS). Basal NO levels closely correlate with MMP9 bioavailability in BM of both hypertensive and control rats. Our observations support the hypothesis that inadequate mobilization of BM-derived stem and progenitor cells in hypertension results from impaired NOS/NO/MMP9 signalling in BM, a condition that may be corrected with pharmacological intervention.
Collapse
Affiliation(s)
- Marina A Aleksinskaya
- Department of Nephrology and the Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Ma SK, Choi JS, Joo SY, Kim HY, Kim CS, Bae EH, Lee JU, Kim SW. Activation of the Renal PI3K/Akt/mTOR Signaling Pathway in a DOCA-Salt Model of Hypertension. Chonnam Med J 2012; 48:150-4. [PMID: 23323219 PMCID: PMC3539094 DOI: 10.4068/cmj.2012.48.3.150] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 10/28/2012] [Accepted: 10/29/2012] [Indexed: 11/24/2022] Open
Abstract
The present study investigated the changes that occurred in the mammalian target of rapamycin (mTOR) signaling pathway in the kidney as a result of deoxycorticosterone acetate (DOCA)-salt hypertension. Rats were implanted with DOCA strips (200 mg/kg) 1 week after unilateral nephrectomy and were then supplied with 0.9% saline to drink. Four weeks after DOCA implantation, systolic blood pressure (SBP) was measured by use of the tail-cuff method. The expression levels of phosphorylated phosphatidylinositol-3-kinase (PI3K), Akt, and mTOR, as well as the protein expression levels of ED-1 and cyclooxygenase-2 (COX-2), transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (SMA), caspase-3, Bax, and Bcl-2, were then examined in the kidney by semiquantitative immunoblotting. DOCA-salt hypertensive rats were found to have significantly increased SBP as well as an increased kidney weight-to-body weight ratio. Moreover, the phosphorylation of PI3K, Akt, and mTOR was increased in the kidney of DOCA-salt hypertensive rats compared with the control, as was the protein expression of ED-1, COX-2, TGF-β1, and α-SMA. The expression levels of caspase-3 and Bax were increased significantly, whereas Bcl-2 expression was decreased. In conclusion, the phosphorylation of PI3K/Akt/mTOR was increased in the kidney of DOCA-salt hypertensive rats.
Collapse
Affiliation(s)
- Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Jia Z, Aoyagi T, Kohan DE, Yang T. mPGES-1 deletion impairs aldosterone escape and enhances sodium appetite. Am J Physiol Renal Physiol 2010; 299:F155-66. [PMID: 20335314 DOI: 10.1152/ajprenal.90702.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldosterone (Aldo) is a major sodium-retaining hormone that reduces renal sodium excretion and also stimulates sodium appetite. In the face of excess Aldo, the sodium-retaining action of this steroid is overridden by an adaptive regulatory mechanism, a phenomenon termed Aldo escape. The underlying mechanism of this phenomenon is not well defined but appeared to involve a number of natriuretic factors such prostaglandins (PGs). Here, we investigated the role of microsomal prostaglandin E synthase-1 (mPGES-1) in the response to excess Aldo. A 14-day Aldo infusion at 0.35 mg x kg(-1) x day(-1) via an osmotic minipump in conjunction with normal salt intake did not produce obvious disturbances in fluid metabolism in WT mice as suggested by normal sodium and water balance, plasma sodium concentration, hematocrit, and body weight, despite the evidence of a transient sodium accumulation on days 1 or 2. In a sharp contrast, the 14-day Aldo treatment in mPGES-1 knockoute (KO) mice led to increased sodium and water balance, persistent reduction of hematocrit, hypernatremia, and body weight gain, all evidence of fluid retention. The escaped wild-type (WT) mice displayed a remarkable increase in urinary PGE(2) excretion in parallel with coinduction of mPGES-1 in the proximal tubules, accompanied by a remarkable, widespread downregulation of renal sodium and water transporters. The increase in urinary PGE(2) excretion together with the downregulation of renal sodium and water transporters were all significantly blocked in the KO mice. Interestingly, compared with WT controls, the KO mice exhibited consistent increases in sodium and water intake during Aldo infusion. Together, these results suggest an important role of mPGES-1 in antagonizing the sodium-retaining action of Aldo at the levels of both the central nervous system and the kidney.
Collapse
Affiliation(s)
- Zhanjun Jia
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah 84132, USA
| | | | | | | |
Collapse
|
14
|
Jia Z, Aoyagi T, Yang T. mPGES-1 protects against DOCA-salt hypertension via inhibition of oxidative stress or stimulation of NO/cGMP. Hypertension 2010; 55:539-46. [PMID: 20065149 DOI: 10.1161/hypertensionaha.109.144840] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is a recently characterized cytokine-inducible enzyme critically involved in pain and inflammatory response. However, its role in blood pressure regulation is still debatable. The present study was undertaken to examine the effect of mPGES-1 deletion on DOCA-salt hypertension. After 2 weeks of DOCA plus 1% NaCl as drinking fluid, hypertension and sodium retention were more severe in mPGES-1 knockout (KO) mice than in wild-type (WT) controls. The indices of oxidative stress including urinary 8-isprostane and renal thiobarbituric acid-reactive substances were only modestly increased or unchanged in the WT mice but more significantly increased in the KO mice after DOCA-salt. Conversely, in response to DOCA-salt, the indices of antioxidant systems including renal expression of superoxide dismutase-3 and urinary nitrate/nitrite excretion were all significantly elevated in the WT mice but remarkably suppressed in the KO mice. Tempol treatment (50 mg/kg per day) in DOCA-salt KO mice produced a marked attenuation of hypertension, sodium retention, and kidney injury. Immunoblotting demonstrated increased renal expression of mPGES-1 in DOCA-salt WT mice. DOCA-salt induced a nearly 5-fold increase in urinary PGE(2) excretion in the WT mice, and this increase was completely abolished in the KO mice. Together, these results suggest that mPGES-1-derived PGE(2) confers protection against DOCA-salt hypertension likely via inhibition of oxidative stress or stimulation of superoxide dismutase-3 and urinary nitrate/nitrite system.
Collapse
Affiliation(s)
- Zhanjun Jia
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah 84132, USA
| | | | | |
Collapse
|