1
|
Guo C, Zhang G, Wu C, Lei Y, Wang Y, Yang J. Emerging trends in small molecule inhibitors targeting aldosterone synthase: A new paradigm in cardiovascular disease treatment. Eur J Med Chem 2024; 274:116521. [PMID: 38820853 DOI: 10.1016/j.ejmech.2024.116521] [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: 03/28/2024] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Aldosterone synthase (CYP11B2) is the rate-limiting enzyme in aldosterone production. In recent years, CYP11B2 has become an appealing target for treating conditions associated with excess aldosterone, such as hypertension, heart failure, and cardiometabolic diseases. Several small-molecule inhibitors of CYP11B2 have demonstrated efficacy in both preclinical studies and clinical trials. Among them, the tetrahydroisoquinoline derivative Baxdrostat has entered clinical trial phases and demonstrated efficacy in treating patients with hypertension. However, the high homology (>93 %) between CYP11B2 and steroid-11β-hydroxylase (CYP11B1), which catalyzes cortisol production, implies that insufficient drug specificity can lead to severe side effects. Developing selective inhibitors for CYP11B2 remains a considerable challenge that requires ongoing attention. This review summarizes recent research progress on small-molecule inhibitors targeting CYP11B2, focusing on structure-activity relationships (SAR) and structural optimization. It discusses strategies for enhancing the specificity and inhibitory activity of inhibitors, while also exploring potential applications and future prospects for CYP11B2 inhibitors, providing a theoretical foundation for developing the new generation of CYP11B2-targeted medications.
Collapse
Affiliation(s)
- Cuiyu Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guangbing Zhang
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Lei
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, General Practice Research Institute, West China Hospital, Sichuan University, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China; Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Xu C. Extra-adrenal aldosterone: a mini review focusing on the physiology and pathophysiology of intrarenal aldosterone. Endocrine 2024; 83:285-301. [PMID: 37847370 DOI: 10.1007/s12020-023-03566-6] [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: 07/21/2023] [Accepted: 10/08/2023] [Indexed: 10/18/2023]
Abstract
PURPOSE Accumulating evidence has demonstrated the existence of extra-adrenal aldosterone in various tissues, including the brain, heart, vascular, adipocyte, and kidney, mainly based on the detection of the CYP11B2 (aldosterone synthase, cytochrome P450, family 11, subfamily B, polypeptide 2) expression using semi-quantitative methods including reverse transcription-polymerase chain reaction and antibody-based western blotting, as well as local tissue aldosterone levels by antibody-based immunosorbent assays. This mini-review highlights the current evidence and challenges in extra-adrenal aldosterone, focusing on intrarenal aldosterone. METHODS A narrative review. RESULTS Locally synthesized aldosterone may play a vital role in various physio-pathological processes, especially cardiovascular events. The site of local aldosterone synthesis in the kidney may include the mesangial cells, podocytes, proximal tubules, and collecting ducts. The synthesis of renal aldosterone may be regulated by (pro)renin receptor/(pro)renin, angiotensin II/Angiotensin II type 1 receptor, wnt/β-catenin, cyclooxygenase-2/prostaglandin E2, and klotho. Enhanced renal aldosterone release promotes Na+ reabsorption and K+ excretion in the distal nephron and may contribute to the progress of diabetic nephropathy and salt-related hypertension. CONCLUSIONS Inhibition of intrarenal aldosterone signaling by aldosterone synthase inhibitors or mineralocorticoid receptor antagonists may be a hopeful pharmacological technique for the therapy of diabetic nephropathy and saltrelated hypertension. Yet, current reports are often conflicting or ambiguous, leading many to question whether extra-adrenal aldosterone exists, or whether it is of any physiological and pathophysiological significance.
Collapse
Affiliation(s)
- Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, 330002, Jiangxi, China.
| |
Collapse
|
3
|
Zeng J, Zhang Y, Huang C. Macrophages polarization in renal inflammation and fibrosis animal models (Review). Mol Med Rep 2024; 29:29. [PMID: 38131228 PMCID: PMC10784723 DOI: 10.3892/mmr.2023.13152] [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: 09/04/2023] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
Chronic kidney disease (CKD) is a significant public health concern. Renal fibrosis is the final common pathway in the progression of kidney diseases, irrespective of the initial injury. Substantial evidence underscores the pivotal role of renal inflammation in the genesis of renal fibrosis. The presence of macrophages within normal renal tissue is significantly increased within diseased renal tissue, indicative of their crucial regulatory function in inflammation and fibrosis. Macrophages manifest a high degree of heterogeneity, exhibiting distinct phenotypic and functional traits in response to diverse stimuli within the local microenvironment in various types of kidney diseases. Broadly, macrophages are categorized into two principal groups: Classically activated, designated as M1 macrophages and alternatively activated, designated as M2 macrophages. A number of experimental models are widely used to study the underlying mechanisms driving renal inflammation and fibrosis progression. The present review delineated the phenotypic and functional attributes of macrophages present in diverse induced models, analyzing their disposition in relation to M1 and M2 polarization states.
Collapse
Affiliation(s)
- Ji Zeng
- Department of Pharmacy, Ma'anshan City Hospital of Traditional Chinese Medicine, Ma'anshan, Anhui 243000, P.R. China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
4
|
Buncha V, Cherezova A, Alexander S, Baranovskaya I, Coleman KA, Cherian-Shaw M, Brands MW, Sullivan JC, O'Connor PM, Mamenko M. Aldosterone Antagonism Is More Effective at Reducing Blood Pressure and Excessive Renal ENaC Activity in AngII-Infused Female Rats Than in Males. Hypertension 2023; 80:2196-2208. [PMID: 37593894 PMCID: PMC10528186 DOI: 10.1161/hypertensionaha.123.21287] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AngII (angiotensin II)-dependent hypertension causes comparable elevations of blood pressure (BP), aldosterone levels, and renal ENaC (epithelial Na+ channel) activity in male and female rodents. Mineralocorticoid receptor (MR) antagonism has a limited antihypertensive effect associated with insufficient suppression of renal ENaC in male rodents with AngII-hypertension. While MR blockade effectively reduces BP in female mice with salt-sensitive and leptin-induced hypertension, MR antagonism has not been studied in female rodents with AngII-hypertension. We hypothesize that overstimulation of renal MR signaling drives redundant ENaC-mediated Na+ reabsorption and BP increase in female rats with AngII-hypertension. METHODS We employ a combination of physiological, pharmacological, biochemical, and biophysical approaches to compare the effect of MR inhibitors on BP and ENaC activity in AngII-infused male and female Sprague Dawley rats. RESULTS MR blockade markedly attenuates AngII-hypertension in female rats but has only a marginal effect in males. Spironolactone increases urinary sodium excretion and urinary sodium-to-potassium ratio in AngII-infused female, but not male, rats. The expression of renal MR and HSD11β2 (11β-hydroxysteroid dehydrogenase type 2) that determines the availability of MR to aldosterone is significantly higher in AngII-infused female rats than in males. ENaC activity is ≈2× lower in spironolactone-treated AngII-infused female rats than in males. Reduced ENaC activity in AngII-infused female rats on spironolactone correlates with increased interaction with ubiquitin ligase Nedd4-2 (neural precursor cell expressed developmentally down-regulated protein 4-2), targeting ENaC for degradation. CONCLUSIONS MR-ENaC axis is the primary determinant of excessive renal sodium reabsorption and an attractive antihypertensive target in female rats with AngII-hypertension, but not in males.
Collapse
Affiliation(s)
- Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Alena Cherezova
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Sati Alexander
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Irina Baranovskaya
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Kathleen A Coleman
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University
| | | | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University
| |
Collapse
|
5
|
Maeoka Y, Su XT, Wang WH, Duan XP, Sharma A, Li N, Staub O, McCormick JA, Ellison DH. Mineralocorticoid Receptor Antagonists Cause Natriuresis in the Absence of Aldosterone. Hypertension 2022; 79:1423-1434. [PMID: 35506380 PMCID: PMC9177775 DOI: 10.1161/hypertensionaha.122.19159] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND MR (mineralocorticoid receptor) antagonists are recommended for patients with resistant hypertension even when circulating aldosterone levels are not high. Although aldosterone activates MR to increase epithelial sodium channel (ENaC) activity, glucocorticoids also activate MR but are metabolized by 11βHSD2 (11β-hydroxysteroid dehydrogenase type 2). 11βHSD2 is expressed at increasing levels from distal convoluted tubule (DCT) through collecting duct. Here, we hypothesized that MR maintains ENaC activity in the DCT2 and early connecting tubule in the absence of aldosterone. METHODS We studied AS (aldosterone synthase)-deficient (AS-/-) mice, which were backcrossed onto the same C57BL6/J strain as kidney-specific MR knockout (KS-MR-/-) mice. KS-MR-/- mice were used to compare MR expression and ENaC localization and cleavage with AS-/- mice. RESULTS MR was highly expressed along DCT2 through the cortical collecting duct (CCD), whereas no 11βHSD2 expression was observed along DCT2. MR signal and apical ENaC localization were clearly reduced along both DCT2 and CCD in KS-MR-/- mice but were fully preserved along DCT2 and were partially reduced along CCD in AS-/- mice. Apical ENaC localization and ENaC currents were fully preserved along DCT2 in AS-/- mice and were not increased along CCD after low salt. AS-/- mice exhibited transient Na+ wasting under low-salt diet, but administration of the MR antagonist eplerenone to AS-/- mice led to hyperkalemia and decreased body weight with higher Na+ excretion, mimicking the phenotype of MR-/- mice. CONCLUSIONS Our results provide evidence that MR is activated in the absence of aldosterone along DCT2 and partially CCD, suggesting glucocorticoid binding to MR preserves sodium homeostasis along DCT2 in AS-/- mice.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Xin-Peng Duan
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - Na Li
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland,LeDucq Transatlantic Network of Excellence
| | - James A. McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR,LeDucq Transatlantic Network of Excellence,Veterans Affairs Portland Healthcare System, Portland, Oregon
| |
Collapse
|
6
|
Ackermann D, Vogt B, Bochud M, Burnier M, Martin PY, Paccaud F, Ehret G, Guessous I, Ponte B, Pruijm M, Pechère-Bertschi A, Jamin H, Klossner R, Dick B, Mohaupt MG, Gennari-Moser C. Increased glucocorticoid metabolism in diabetic kidney disease. PLoS One 2022; 17:e0269920. [PMID: 35749380 PMCID: PMC9231809 DOI: 10.1371/journal.pone.0269920] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/31/2022] [Indexed: 11/18/2022] Open
Abstract
Aims
Glomerular damage indicated by proteinuria is a main symptom in diabetic nephropathy. Mineralocorticoid receptor (MR) antagonists (MRAs) are beneficial irrespective of aldosterone availability. Thus, we hypothesized an alternatively activated MR to promote glomerular damage in proteinuric diabetic nephropathy. Specifically, we aimed first to demonstrate the presence of steroid hormones serving as alternative MR targets in type II diabetic patients with proteinuric kidney disease, second whether MR selectivity was modified, third to characterize MR and glucocorticoid receptor (GR) expression and activity in glomerular cell types exposed to eu- and hyperglycemic conditions, fourth to characterize the pro-fibrotic potential of primary human renal mesangial cells (HRMC) upon stimulation with aldosterone and cortisol, and fifth to specify the involvement of the MR and/or GR in pro-fibrotic signaling.
Materials and methods
Urinary steroid hormone profiles of patients with diabetic kidney disease were analyzed by gas chromatography–mass spectrometry and compared to an age and gender matched healthy control group taken out of a population study. In both cohorts, the activity of the MR pre-receptor enzyme 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2), which inactivates cortisol to prevent it from binding to the MR, was assessed to define a change in MR selectivity. Expression of HSD11B2, MR and GR was quantified in HRMC and primary human renal glomerular endothelial cells (HRGEC). Activity of MR and GR was explored in HRMC by measuring the MR/GR down-stream signal SGK1 and the pro-fibrotic genes TGFB1, FN1 and COL1A1 in normal and high glucose conditions with the MR/GR agonists aldosterone/cortisol and the MR/GR antagonists spironolactone/RU486.
Results
Patients with diabetic kidney disease excreted more tetrahydroaldosterone than the control group reaching significance in men. The excretion of MR-agonistic steroid hormones was only increased for 18-hydroxytetrahydrocorticosterone in diabetic women. The excretion of most glucocorticoids was higher in the diabetic cohort. Higher apparent systemic HSD11B2 activity suggested less activation of the MR by cortisol in diabetic patients. Both cell types, HRMC and HRGEC, lacked expression of HSD11B2. Hyperglycemic conditions did not change MR and GR expression and activity. Stimulation with both aldosterone and cortisol promoted upregulation of pro-fibrotic genes in HRMC. This effect of MR and/or GR activation was more pronounced in high glucose conditions and partially inhibited by MRAs and GR antagonists.
Conclusions
In patients with diabetic kidney disease alternative MR activation is conceivable as cortisol and cortisone metabolites are increased. Systemic availability of active metabolites is counteracted via an increased HSD11B2 activity. As this cortisol deactivation is absent in HRMC and HRGEC, cortisol binding to the MR is enabled. Both, cortisol and aldosterone stimulation led to an increased expression of pro-fibrotic genes in HRMC. This mechanism was related to the MR as well as the GR and more marked in high glucose conditions linking the benefit of MRAs in diabetic kidney disease to these findings.
Collapse
Affiliation(s)
- Daniel Ackermann
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Murielle Bochud
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Michel Burnier
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Pierre-Yves Martin
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Fred Paccaud
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Georg Ehret
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Idris Guessous
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Belen Ponte
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | - Menno Pruijm
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
| | | | - Heidi Jamin
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Department for BioMedical Research, University of Bern, Berne, Switzerland
| | - Rahel Klossner
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland
| | - Bernhard Dick
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Department for BioMedical Research, University of Bern, Berne, Switzerland
| | - Markus G. Mohaupt
- Swiss Kidney Project on Genes in Hypertension (SKIPOGH) Team, Lausanne, Switzerland
- Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland
- School of Medicine, University of Nottingham, Division of Child Health, Obstetrics & Gynaecology, Nottingham, United Kingdom
| | - Carine Gennari-Moser
- Department of Nephrology and Hypertension, University of Bern, Berne, Switzerland
- Department for BioMedical Research, University of Bern, Berne, Switzerland
- * E-mail:
| |
Collapse
|
7
|
Luther JM, Fogo AB. The role of mineralocorticoid receptor activation in kidney inflammation and fibrosis. Kidney Int Suppl (2011) 2022; 12:63-68. [DOI: 10.1016/j.kisu.2021.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
|
8
|
Sutton EF, Gemmel M, Brands J, Gallaher MJ, Powers RW. Paternal deficiency of complement component C1q leads to a preeclampsia-like pregnancy in wild-type female mice and vascular adaptations postpartum. Am J Physiol Regul Integr Comp Physiol 2020; 318:R1047-R1057. [PMID: 32374620 DOI: 10.1152/ajpregu.00353.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia is a spontaneously occurring, pregnancy-specific syndrome that is clinically diagnosed by new onset hypertension and proteinuria. Epidemiological evidence describes an association between a history of preeclampsia and increased risk for cardiovascular disease in later life; however, the mechanism(s) driving this relationship are unclear. Our study aims to leverage a novel preeclampsia-like mouse model, the C1q-/- model, to help elucidate the acute and persistent vascular changes during and following a preeclampsia-like pregnancy. Female C57BL/6J mice were mated to C1q-/- male mice to model a preeclampsia-like pregnancy ("PE-like"), and the maternal cardiovascular phenotype (blood pressure, renal function, systemic glycocalyx, and ex vivo vascular function) was assessed in late pregnancy and postpartum at 6 and 10 mo of age. Uncomplicated, normotensive pregnancies (female C57BL/6J bred to male C57BL/6J mice) served as age-matched controls. In pregnancy, PE-like dams exhibited increased systolic and diastolic pressure during mid- and late gestation, renal dysfunction, fetal growth restriction, and reduced placental efficiency. Ex vivo wire myography studies of mesenteric arteries revealed severe pregnancy-specific endothelial-dependent and -independent vascular dysfunction. At 3 and 7 mo postpartum (6 and 10 mo old, respectively), hypertension resolved in PE-like dams, whereas mild vascular dysfunction persisted at 3 mo postpartum. In conclusion, the female C57BL/6J-by-male C57BL/6J C1q-/- model recapitulates many aspects of the human preeclampsia syndrome in a low-risk, wild-type female mouse. The pregnancy-specific phenotype results in systemic maternal endothelial-dependent and -independent vascular dysfunction that persists postpartum.
Collapse
Affiliation(s)
- Elizabeth F Sutton
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary Gemmel
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Judith Brands
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Robert W Powers
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Spencer S, Wheeler‐Jones C, Elliott J. Aldosterone and the mineralocorticoid receptor in renal injury: A potential therapeutic target in feline chronic kidney disease. J Vet Pharmacol Ther 2020; 43:243-267. [PMID: 32128854 PMCID: PMC8614124 DOI: 10.1111/jvp.12848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/20/2020] [Accepted: 02/09/2020] [Indexed: 12/24/2022]
Abstract
There is a growing body of experimental and clinical evidence supporting mineralocorticoid receptor (MR) activation as a powerful mediator of renal damage in laboratory animals and humans. Multiple pathophysiological mechanisms are proposed, with the strongest evidence supporting aldosterone-induced vasculopathy, exacerbation of oxidative stress and inflammation, and increased growth factor signalling promoting fibroblast proliferation and deranged extracellular matrix homeostasis. Further involvement of the MR is supported by extensive animal model experiments where MR antagonists (such as spironolactone and eplerenone) abrogate renal injury, including ischaemia-induced damage. Additionally, clinical trials have shown MR antagonists to be beneficial in human chronic kidney disease (CKD) in terms of reducing proteinuria and cardiovascular events, though current studies have not evaluated primary end points which allow conclusions to made about whether MR antagonists reduce mortality or slow CKD progression. Although differences between human and feline CKD exist, feline CKD shares many characteristics with human disease including tubulointerstitial fibrosis. This review evaluates the evidence for the role of the MR in renal injury and summarizes the literature concerning aldosterone in feline CKD. MR antagonists may represent a promising therapeutic strategy in feline CKD.
Collapse
Affiliation(s)
- Sarah Spencer
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| | | | - Jonathan Elliott
- Comparative Biomedical SciencesThe Royal Veterinary CollegeLondonUK
| |
Collapse
|
10
|
|
11
|
Osman W, Al Dohani H, Al Hinai AS, Hannawi S, M Shaheen FA, Al Salmi I. Aldosterone renin ratio and chronic kidney disease. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2020; 31:70-78. [PMID: 32129199 DOI: 10.4103/1319-2442.279963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
As a component of the metabolic syndrome, hypertension (HTN) is increasing throughout the world with variable percentages, but mostly among developing world. Aldosterone plays a role in the relationship between aldosterone and nephropathy. We aimed to evaluate the relationship between aldosterone renin ratio (ARR) and chronic kidney disease (CKD). Variables drawn from the computerized hospital information database were all patients who had an ARR above 35 (if aldosterone reading was above 300 pmol/L). A total of 1584 patients, of whom 777 were male and 807 were female, with a mean [standard deviation (SD)] of 43.3 (16.5) years were studied. The mean ARR was 210.1 (SD: 246.4) in males and 214.3 and 210.1 in females, P = 0.51. The mean estimated glomerular filtration rate (eGFR) was 50.2 (SD 12.6); in males, it was 49.99 (0.90) and in females, it was 50.48 (0.92), P = 0.70. The regression model revealed a negative relationship between ARR and GFR with a coefficient of -2.08, 95% confidence interval: -4.6, 0.21, P = 0.07. CKD population with HTN tends to have a very high level of ARR, and those with advanced CKD have higher ARR. However, high ARR could have low eGFR and kidney dysfunction on follow-up. In view of high prevalence of noncommunicable disease and high early CKD population, there is an important need to consider comprehensive management strategies that involve the blockage of high renin-angiotensin-aldosterone and the use of mineralocorticosteroid receptor blockers.
Collapse
Affiliation(s)
- Wessam Osman
- Department of Internal Medicine, The Royal Hospital, Muscat, Oman
| | - Hayam Al Dohani
- Department of Internal Medicine, The Royal Hospital, Muscat, Oman
| | | | - Suad Hannawi
- Department of Medicine, Ministry of Health and Prevention, Dubai, United Arab Emirates
| | | | - Issa Al Salmi
- Department of Renal Medicine, The Royal Hospital, Muscat, Oman
| |
Collapse
|
12
|
Lu Q, Davel AP, McGraw AP, Rao SP, Newfell BG, Jaffe IZ. PKCδ Mediates Mineralocorticoid Receptor Activation by Angiotensin II to Modulate Smooth Muscle Cell Function. Endocrinology 2019; 160:2101-2114. [PMID: 31373631 PMCID: PMC6735772 DOI: 10.1210/en.2019-00258] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/28/2019] [Indexed: 01/30/2023]
Abstract
Angiotensin II (AngII) and the mineralocorticoid receptor (MR) ligand aldosterone both contribute to cardiovascular disorders, including hypertension and adverse vascular remodeling. We previously demonstrated that AngII activates MR-mediated gene transcription in human vascular smooth muscle cells (SMCs), yet the mechanism and the impact on SMC function are unknown. Using an MR-responsive element-driven transcriptional reporter assay, we confirm that AngII induces MR transcriptional activity in vascular SMCs and endothelial cells, but not in Cos1 or human embryonic kidney-293 cells. AngII activation of MR was blocked by the MR antagonist spironolactone or eplerenone and the protein kinase C-δ (PKCδ) inhibitor rottlerin, implicating both in the mechanism. Similarly, small interfering RNA knockdown of PKCδ in SMCs prevented AngII-mediated MR activation, whereas knocking down of MR blocked both aldosterone- and AngII-induced MR function. Coimmunoprecipitation studies reveal that endogenous MR and PKCδ form a complex in SMCs that is enhanced by AngII treatment in association with increased serine phosphorylation of the MR N terminus. AngII increased mRNA expression of the SMC-MR target gene, FKBP51, via an MR-responsive element in intron 5 of the FKBP51 gene. The impact of AngII on FKBP51 reporter activity and gene expression in SMCs was inhibited by spironolactone and rottlerin. Finally, the AngII-induced increase in SMC number was also blocked by the MR antagonist spironolactone and the PKCδ inhibitor rottlerin. These data demonstrate that AngII activates MR transcriptional regulatory activity, target gene regulation, and SMC proliferation in a PKCδ-dependent manner. This new mechanism may contribute to synergy between MR and AngII in driving SMC dysfunction and to the cardiovascular benefits of MR and AngII receptor blockade in humans.
Collapse
Affiliation(s)
- Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Ana P Davel
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Adam P McGraw
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Sitara P Rao
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Brenna G Newfell
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Correspondence: Iris Z. Jaffe, MD, PhD, Tufts Medical Center, Molecular Cardiology Research Institute, 800 Washington Street, Box 80, Boston, Massachusetts 02111. E-mail:
| |
Collapse
|
13
|
Thuesen AD, Finsen SH, Rasmussen LL, Andersen DC, Jensen BL, Hansen PBL. Deficiency of T-type Ca 2+ channels Ca v3.1 and Ca v3.2 has no effect on angiotensin II-induced hypertension but differential effect on plasma aldosterone in mice. Am J Physiol Renal Physiol 2019; 317:F254-F263. [PMID: 31042060 DOI: 10.1152/ajprenal.00121.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
T-type Ca2+ channel Cav3.1 promotes microvessel contraction ex vivo. It was hypothesized that in vivo, functional deletion of Cav3.1, but not Cav3.2, protects mice against angiotensin II (ANG II)-induced hypertension. Mean arterial blood pressure (MAP) and heart rate were measured continuously with chronically indwelling catheters during infusion of ANG II (30 ng·kg-1·min-1, 7 days) in wild-type (WT), Cav3.1-/-, and Cav3.2-/- mice. Plasma aldosterone and renin concentrations were measured by radioimmunoassays. In a separate series, WT mice were infused with ANG II (100 ng·kg-1·min-1) with and without the mineralocorticoid receptor blocker canrenoate. Cav3.1-/- and Cav3.2-/- mice exhibited no baseline difference in MAP compared with WT mice, but day-night variation was blunted in both Cav3.1 and Cav3.2-/- mice. ANG II increased significantly MAP in WT, Cav3.1-/-, and Cav3.2-/- mice with no differences between genotypes. Heart rate was significantly lower in Cav3.1-/- and Cav3.2-/- mice compared with control mice. After ANG II infusion, plasma aldosterone concentration was significantly lower in Cav3.1-/- compared with Cav3.2-/- mice. In response to ANG II, fibrosis was observed in heart sections from both WT and Cav3.1-/- mice and while cardiac atrial natriuretic peptide mRNA was similar, the brain natriuretic peptide mRNA increase was mitigated in Cav3.1-/- mice ANG II at 100 ng/kg yielded elevated pressure and an increased heart weight-to-body weight ratio in WT mice. Cardiac hypertrophy, but not hypertension, was prevented by the mineralocorticoid receptor blocker canrenoate. In conclusion, T-type channels Cav3.1and Cav3.2 do not contribute to baseline blood pressure levels and ANG II-induced hypertension. Cav3.1, but not Cav3.2, contributes to aldosterone secretion. Aldosterone promotes cardiac hypertrophy during hypertension.
Collapse
Affiliation(s)
- Anne D Thuesen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Stine H Finsen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Louise L Rasmussen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Ditte C Andersen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark.,Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Clinical Institute, University of Southern Denmark , Odense , Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Pernille B L Hansen
- Department of Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark.,Cardiovascular and Metabolic Disease, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg , Sweden
| |
Collapse
|
14
|
Yang CT, Kor CT, Hsieh YP. Long-Term Effects of Spironolactone on Kidney Function and Hyperkalemia-Associated Hospitalization in Patients with Chronic Kidney Disease. J Clin Med 2018; 7:jcm7110459. [PMID: 30469400 PMCID: PMC6262621 DOI: 10.3390/jcm7110459] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/19/2018] [Indexed: 01/13/2023] Open
Abstract
Background: Spironolactone, a non-selective mineralocorticoid receptor antagonist, can protect against cardiac fibrosis and left ventricular dysfunction, and improve endothelial dysfunction and proteinuria. However, the safety and effects of spironolactone on patient-centered cardiovascular and renal endpoints remain unclear. Methods: We identified predialysis stage 3–4 chronic kidney disease (CKD) patients between 2000 and 2013 from the Longitudinal Health Insurance Database 2005 (LHID 2005). The outcomes of interest were end-stage renal disease (ESRD), major adverse cardiovascular events (MACE), hospitalization for heart failure (HHF), hyperkalemia-associated hospitalization (HKAH), all-cause mortality and cardiovascular mortality. The Fine and Gray sub-distribution hazards approach was adopted to adjust for the competing risk of death. Results: After the propensity score matching, 693 patients with stage 3–4 CKD were spironolactone users and 1386 were nonusers. During the follow-up period, spironolactone users had a lower incidence rate for ESRD than spironolactone non-users (39.2 vs. 53.69 per 1000 person-years) and a higher incidence rate for HKAH (54.79 vs. 18.57 per 1000 person-years). The adjusted hazard ratios for ESRD of spironolactone users versus non-users were 0.66 (95% CI, 0.51–0.84; p value < 0.001) and 3.17 (95% CI, 2.41–4.17; p value < 0.001) for HKAH. A dose-response relationship was found between spironolactone use and risk of ESRD and HKAH. There were no statistical differences in MACE, HHF, all-cause mortality and cardiovascular mortality between spironolactone users and non-users. Conclusion: Spironolactone represented a promising treatment option to retard CKD progression to ESRD amongst stage 3–4 CKD patients, but strategic treatments to prevent hyperkalemia should be enforced.
Collapse
Affiliation(s)
- Chen-Ta Yang
- Department of Internal Medicine, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Chew-Teng Kor
- Department of Internal Medicine, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Yao-Peng Hsieh
- Department of Internal Medicine, Changhua Christian Hospital, Changhua 50006, Taiwan.
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
| |
Collapse
|
15
|
Ham O, Jin W, Lei L, Huang HH, Tsuji K, Huang M, Roh J, Rosenzweig A, Lu HAJ. Pathological cardiac remodeling occurs early in CKD mice from unilateral urinary obstruction, and is attenuated by Enalapril. Sci Rep 2018; 8:16087. [PMID: 30382174 PMCID: PMC6208335 DOI: 10.1038/s41598-018-34216-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 10/04/2018] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease constitutes the leading cause of mortality in patients with chronic kidney disease (CKD) and end-stage renal disease. Despite increasing recognition of a close interplay between kidney dysfunction and cardiovascular disease, termed cardiorenal syndrome (CRS), the underlying mechanisms of CRS remain poorly understood. Here we report the development of pathological cardiac hypertrophy and fibrosis in early stage non-uremic CKD. Moderate kidney failure was induced three weeks after unilateral urinary obstruction (UUO) in mice. We observed pathological cardiac hypertrophy and increased fibrosis in UUO-induced CKD (UUO/CKD) animals. Further analysis indicated that this cardiac fibrosis was associated with increased expression of transforming growth factor β (TGF-β) along with significant upregulation of Smad 2/3 signaling in the heart. Moreover early treatment of UUO/CKD animals with an angiotensin-converting-enzyme inhibitor (ACE I), Enalapril, significantly attenuated cardiac fibrosis. Enalapril antagonized activation of the TGF-β signaling pathway in the UUO/CKD heart. In summary our study demonstrates the presence of pathological cardiac hypertrophy and fibrosis in mice early in UUO-induced CKD, in association with early activation of the TGF-β/Smad signaling pathway. We also demonstrate the beneficial effect of ACE I in alleviating this early fibrogenic process in the heart in UUO/CKD animals.
Collapse
Affiliation(s)
- Onju Ham
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - William Jin
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Lei Lei
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hui Hui Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ming Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jason Roh
- Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
16
|
Sogawa Y, Nagasu H, Itano S, Kidokoro K, Taniguchi S, Takahashi M, Kadoya H, Satoh M, Sasaki T, Kashihara N. The eNOS-NO pathway attenuates kidney dysfunction via suppression of inflammasome activation in aldosterone-induced renal injury model mice. PLoS One 2018; 13:e0203823. [PMID: 30281670 PMCID: PMC6169882 DOI: 10.1371/journal.pone.0203823] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022] Open
Abstract
Hypertension causes vascular complications, such as stroke, cardiovascular disease, and chronic kidney disease (CKD). The relationship between endothelial dysfunction and progression of kidney disease is well known. However, the relationship between the eNOS-NO pathway and chronic inflammation, which is a common pathway for the progression of kidney disease, remains unexplored. We performed in vivo experiments to determine the role of the eNOS-NO pathway by using eNOS-deficient mice in a hypertensive kidney disease model. All mice were unilateral nephrectomized (Nx). One week after Nx, the mice were randomly divided into two groups: the aldosterone infusion groups and the vehicle groups. All mice also received a 1% NaCl solution instead of drinking water. The aldosterone infusion groups were treated with hydralazine to correct blood pressure differences. After four weeks of drug administration, all mice were euthanized, and blood and kidney tissue samples were collected. In the results, NLRP3 inflammasome activation was elevated in the kidneys of the eNOS-deficient mice, and tubulointerstitial fibrosis was accelerated. Suppression of inflammasome activation by knocking out ASC prevented tubulointerstitial injury in the eNOS knockout mice, indicating that the eNOS-NO pathway is involved in the development of kidney dysfunction through acceleration of NLRP3 inflammasome in macrophages. We revealed that endothelial function, particularly the eNOS-NO pathway, attenuates the progression of renal tubulointerstitial injury via suppression of inflammasome activation. Clinically, patients who develop vascular endothelial dysfunction have lifestyle diseases, such as hypertension or diabetes, and are known to be at risk for CKD. Our study suggests that the eNOS-NO pathway could be a therapeutic target for the treatment of chronic kidney disease associated with endothelial dysfunction.
Collapse
MESH Headings
- Aldosterone/pharmacology
- Animals
- Antihypertensive Agents/administration & dosage
- Disease Models, Animal
- Endothelium/pathology
- Endothelium/physiopathology
- Fibrosis
- Humans
- Hydralazine/administration & dosage
- Hypertension/complications
- Hypertension/metabolism
- Hypertension, Renal/drug therapy
- Hypertension, Renal/metabolism
- Hypertension, Renal/pathology
- Inflammasomes/drug effects
- Inflammasomes/metabolism
- Kidney/pathology
- Macrophages/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Targeted Therapy
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Primary Cell Culture
- Renal Insufficiency, Chronic/chemically induced
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Vasodilator Agents/administration & dosage
Collapse
Affiliation(s)
- Yuji Sogawa
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
- * E-mail:
| | - Seiji Itano
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kengo Kidokoro
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Shun’ichiro Taniguchi
- Department of Molecular Oncology, Shinshu University Graduate School of Medicine, Matsumoto, Nagano, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroyuki Kadoya
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Minoru Satoh
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tamaki Sasaki
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension Kawasaki Medical School, Kurashiki, Okayama, Japan
| |
Collapse
|
17
|
Liao WH, Suendermann C, Steuer AE, Pacheco Lopez G, Odermatt A, Faresse N, Henneberg M, Langhans W. Aldosterone deficiency in mice burdens respiration and accentuates diet-induced hyperinsulinemia and obesity. JCI Insight 2018; 3:99015. [PMID: 30046010 DOI: 10.1172/jci.insight.99015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
Aldosterone synthase inhibitors (ASIs) should alleviate obesity-related cardiovascular and renal problems resulting partly from aldosterone excess, but their clinical use may have limitations. To improve knowledge for the use of ASIs, we investigated physiology in aldosterone synthase-knockout (ASKO) mice. On regular chow diet (CD), ASKO mice ate more and weighed less than WT mice, largely because they hyperventilated to eliminate acid as CO2. Replacing CD with high-fat diet (HFD) lessened the respiratory burden in ASKO mice, as did 12- to 15-hour fasting. The latter eliminated the genotype differences in respiratory workload and energy expenditure (EE). Thus, aldosterone deficiency burdened the organism more when the animals ate carbohydrate-rich chow than when they ate a HFD. Chronic HFD exposure further promoted hyperinsulinemia in ASKO mice that contributed to visceral fat accumulation accompanied by reduced lipolysis, thermogenic reprogramming, and the absence of weight-gain-related EE increases. Intracerebroventricular aldosterone supplementation in ASKO mice attenuated the HFD-induced hyperinsulinemia, but did not affect EE, suggesting that the presence of aldosterone increased the body's energetic efficiency, thus counteracting the EE-increasing effect of low insulin. ASIs may therefore cause acid-overload-induced respiratory burden and promote obesity. Their use in patients with preexisting renal and cardiopulmonary diseases might be contraindicated.
Collapse
Affiliation(s)
- Wan-Hui Liao
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.,Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Center of Competence in Research "Kidney.CH", Switzerland
| | | | - Andrea Eva Steuer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Gustavo Pacheco Lopez
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.,Department of Health Sciences, Division of Biological and Health Sciences, Metropolitan Autonomous University (UAM), Lerma, Mexico
| | - Alex Odermatt
- National Center of Competence in Research "Kidney.CH", Switzerland.,Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Nourdine Faresse
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Center of Competence in Research "Kidney.CH", Switzerland
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland.,Biological Anthropology and Comparative Anatomy Unit, University of Adelaide, Australia
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
18
|
Schreier B, Hünerberg M, Mildenberger S, Rabe S, Bethmann D, Wickenhauser C, Gekle M. Deletion of the EGF receptor in vascular smooth muscle cells prevents chronic angiotensin II-induced arterial wall stiffening and media thickening. Acta Physiol (Oxf) 2018; 222. [PMID: 29152859 DOI: 10.1111/apha.12996] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
AIM In vivo vascular smooth muscle cell (VSMC) EGF receptor (EGFR) contributes to acute angiotensin II (AII) effects on vascular tone and blood pressure. The ubiquitously expressed EGFR has been implicated in vascular remodelling preceding end-organ damage by pharmacological inhibition, and AII signalling in cultured vascular cells is partly EGFR-dependent. However, the role of VSMC-EGFR in vivo during AII-induced pathophysiological processes is not known. METHODS This study assesses the in vivo relevance of VSMC-EGFR during chronic AII challenge without further stressors, using a mouse model with inducible, VSMC-specific EGFR knock out (VSMC-EGFR-KO). In these mice functional and structural vascular, renal and cardiac effects or biomarkers were investigated in vivo and ex vivo. RESULTS Vascular smooth muscle cell-EGFR-KO prevented AII-induced media hypertrophy of mesenteric arteries, renal arterioles and the aorta, VSMC ERK1/2-phosphorylation as well as the impairment of vascular compliance. Furthermore, induction of vascular fibrosis, creatinineamia, renal interstitial fibrosis as well as the increase in fractional water excretion was prevented. AII-induced increase in systolic blood pressure was mitigated. By contrast, endothelial dysfunction, induction of vascular inflammatory marker mRNA and albuminuria were not inhibited. Cardiac and cardiomyocyte hypertrophy were also not prevented by VSMC-EGFR-KO. CONCLUSION Vascular smooth muscle cell-EGFRs are relevant for pathological AII action in vivo. Our data show in vivo and ex vivo the necessity of VSMC-EGFR for AII-induced structural and functional vascular remodelling, not including endothelial dysfunction. Hereby, VSMC-EGFR gains importance for complete AII-induced renal end-organ damage succeeding vascular remodelling.
Collapse
Affiliation(s)
- B. Schreier
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Hünerberg
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Mildenberger
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Rabe
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - D. Bethmann
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - C. Wickenhauser
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Gekle
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| |
Collapse
|
19
|
Nagarajan S, Vohra T, Loffing J, Faresse N. Protein Phosphatase 1α enhances renal aldosterone signaling via mineralocorticoid receptor stabilization. Mol Cell Endocrinol 2017; 450:74-82. [PMID: 28454724 DOI: 10.1016/j.mce.2017.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/18/2017] [Accepted: 04/22/2017] [Indexed: 01/21/2023]
Abstract
Stimulation of the mineralocorticoid receptor (MR) by aldosterone controls several physiological parameters including blood pressure, inflammation or metabolism. We previously showed that MR turnover constitutes a crucial regulatory step in the responses of renal epithelial cells to aldosterone. Here, we identified Protein Phosphatase 1 alpha (PP1α), as a novel cytoplasmic binding partner of MR that promotes the receptor activity. The RT-PCR expression mapping of PP1α reveals a high expression in the kidney, particularly in the distal part of the nephron. At the molecular level, we demonstrate that PP1α inhibits the ubiquitin ligase Mdm2 by dephosphorylation, preventing its interaction with MR. This results in the accumulation of the receptor due to reduction of its proteasomal degradation and consequently a greater aldosterone-induced Na+ uptake by renal cells. Thus, our findings describe an original mechanism involving a phosphatase in the regulation of aldosterone signaling and provide new and important insights into the molecular mechanism underlying the MR turnover.
Collapse
Affiliation(s)
- Shunmugam Nagarajan
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland; National Center of Competence in Research "Kidney.CH", Switzerland
| | - Twinkle Vohra
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland; National Center of Competence in Research "Kidney.CH", Switzerland
| | - Nourdine Faresse
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland; National Center of Competence in Research "Kidney.CH", Switzerland.
| |
Collapse
|
20
|
Tesch GH, Young MJ. Mineralocorticoid Receptor Signaling as a Therapeutic Target for Renal and Cardiac Fibrosis. Front Pharmacol 2017; 8:313. [PMID: 28611666 PMCID: PMC5447060 DOI: 10.3389/fphar.2017.00313] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022] Open
Abstract
Activation of the mineralocorticoid receptor (MR) plays important roles in both physiological and pathological events. Blockade of MR signaling with MR antagonists (MRAs) has been used clinically to treat kidney and cardiac disease associated with hypertension and other chronic diseases, resulting in suppression of fibrosis in these organs. However, the current use of steroidal MRAs has been limited by off target effects on other hormone receptors or adverse effects on kidney tubular function. In this review, we summarize recent insights into the profibrotic roles of MR signaling in kidney and cardiovascular disease. We review experimental in vitro data identifying the pathological mechanisms associated with MR signaling in cell types found in the kidney (mesangial cells, podocytes, tubular cells, macrophages, interstitial fibroblasts) and heart (cardiomyocytes, endothelial cells, vascular smooth muscle cells, macrophages). In addition, we demonstrate the in vivo importance of MR signaling in specific kidney and cardiac cell types by reporting the outcomes of cell type selective MR gene deletion in animal models of kidney and cardiac disease and comparing these findings to those obtained with MRAs treatment. This review also includes a discussion of the potential benefits of novel non-steroidal MRAs for targeting kidney and cardiac fibrosis compared to existing steroidal MRAs, as well as the possibility of novel combination therapies and cell selective delivery of MRAs.
Collapse
Affiliation(s)
- Greg H Tesch
- Department of Nephrology, Monash Health, ClaytonVIC, Australia.,Monash University Department of Medicine, Monash Health, ClaytonVIC, Australia.,Centre for Inflammatory Diseases, Monash Health, ClaytonVIC, Australia
| | - Morag J Young
- Hudson Institute of Medical Research, ClaytonVIC, Australia
| |
Collapse
|
21
|
Olatunji LA, Usman TO, Seok YM, Kim IK. Activation of cardiac renin-angiotensin system and plasminogen activator inhibitor-1 gene expressions in oral contraceptive-induced cardiometabolic disorder. Arch Physiol Biochem 2017; 123:1-8. [PMID: 26934364 DOI: 10.3109/13813455.2016.1160935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Clinical studies have shown that combined oral contraceptive (COC) use is associated with cardiometabolic disturbances. Elevated renin-angiotensin system (RAS) and plasminogen activator inhibitor-1 (PAI-1) have also been implicated in the development of cardiometabolic events. OBJECTIVE To determine the effect of COC treatment on cardiac RAS and PAI-1 gene expressions, and whether the effect is circulating aldosterone or corticosterone dependent. METHODS Female rats were treated (p.o.) with olive oil (vehicle) or COC (1.0 µg ethinylestradiol and 10.0 µg norgestrel) daily for six weeks. RESULTS COC treatment led to increases in blood pressure, HOMA-IR, Ace1 mRNA, Atr1 mRNA, Pai1 mRNA, cardiac PAI-1, plasma PAI-1, C-reactive protein, uric acid, insulin and corticosterone. COC treatment also led to dyslipidemia, decreased glucose tolerance and plasma 17β-estradiol. CONCLUSION These results demonstrates that hypertension and insulin resistance induced by COC is associated with increased cardiac RAS and PAI-1 gene expression, which is likely to be through corticosterone-dependent but not aldosterone-dependent mechanism.
Collapse
Affiliation(s)
- Lawrence A Olatunji
- a Department of Physiology , Cardiovascular and Molecular Physiology Unit, College of Health Sciences University of Ilorin , P.M.B. 1515 , Ilorin , Nigeria
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
| | - Taofeek O Usman
- a Department of Physiology , Cardiovascular and Molecular Physiology Unit, College of Health Sciences University of Ilorin , P.M.B. 1515 , Ilorin , Nigeria
| | - Young-Mi Seok
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
- c Korea Promotion Institute for Traditional Medicine Industry , Gyeongsan , Gyeongbuk , Republic of Korea , and
| | - In-Kyeom Kim
- b Cardiovascular Research Institute, Kyungpook National University School of Medicine , Daegu , Republic of Korea
- d Department of Pharmacology , Kyungpook National University School of Medicine , Daegu , Republic of Korea
| |
Collapse
|
22
|
Denney JM, Bird C, Gendron-Fitzpatrick A, Sampene E, Bird IM, Shah DM. Renin-angiotensin system transgenic mouse model recapitulates pathophysiology similar to human preeclampsia with renal injury that may be mediated through VEGF. Am J Physiol Renal Physiol 2016; 312:F445-F455. [PMID: 27927648 DOI: 10.1152/ajprenal.00108.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 11/07/2016] [Accepted: 11/26/2016] [Indexed: 11/22/2022] Open
Abstract
Using a transgenic cross, we evaluated features of preeclampsia, renal injury and the sFlt1/VEGF changes. Transgenic hAGT and hREN, or wild-type (WT) C57Bl/6 mice were cross-bred: female hAGT × male hREN for preeclampsia (PRE) model and female WT × male WT for pregnant controls (WTP). Samples were collected for plasma VEGF, sFlt1, and urine albumin. Blood pressures (BP) were monitored by telemetry. Vascular reactivity was investigated by wire myography. Kidneys and placenta were immunostained for sFlt1 and VEGF. Eleven PRE and 9 WTP mice were compared. PRE more frequently demonstrated albuminuria, glomerular endotheliosis (80% vs. 11%; P = 0.02), and placental necrosis (60% vs. 0%; P < 0.01). PRE group demonstrated declining BPs with advancing gestation. Plasma sFlt1 increased across pregnancy in PRE; VEGF did not vary. IHC demonstrated the presence of sFlt1 in glomeruli, lymphatics, and collecting tubules of PRE kidneys, suggesting excretion. VEGF immunostaining was increased specifically in the glomeruli of PRE kidneys. Placenta in PRE showed marked immunostaining for sFlt1. We conclude that this transgenic model of preeclampsia recapitulates human preeclamptic state with high fidelity, and that, vascular adaptation to pregnancy is suggested by declining BPs and reduced vascular response to PE and increased response to acetylcholine. Placental damage with resultant increased release of sFlt1, proteinuria, deficient spiral artery remodeling, and glomerular endotheliosis were observed in this model of PRE. Increased VEGF binding to glomerular endothelial cells in this model of PRE is similar to human PRE and leads us to hypothesize that renal injury in preeclampsia may be mediated through local VEGF.
Collapse
Affiliation(s)
- J Morgan Denney
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, University of Wisconsin School of Medicine and Public Health-Madison, Madison, Wisconsin
| | - Cynthia Bird
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, University of Wisconsin School of Medicine and Public Health-Madison, Madison, Wisconsin
| | - Annette Gendron-Fitzpatrick
- Comparative Pathology Laboratory, Research Animal Resource Center, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Emmanuel Sampene
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, University of Wisconsin School of Medicine and Public Health-Madison, Madison, Wisconsin
| | - Ian M Bird
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Wisconsin School of Medicine and Public Health-Madison, Madison, Wisconsin
| | - Dinesh M Shah
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, University of Wisconsin School of Medicine and Public Health-Madison, Madison, Wisconsin;
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW This review will highlight recent developments in mineralocorticoid receptor research which impact aldosterone-associated vascular and cardiometabolic dysfunction. RECENT FINDINGS The mineralocorticoid receptor is also expressed in vascular smooth muscle and vascular endothelium, and contributes to vascular function and remodeling. Adipocyte-derived leptin stimulates aldosterone secretion, which may explain the observed link between obesity and hyperaldosteronism. Adipocyte mineralocorticoid receptor overexpression produces systemic changes consistent with metabolic syndrome. Ongoing studies with novel nonsteroidal mineralocorticoid receptor antagonists may provide a novel treatment for diabetic nephropathy and heart failure in patients with chronic kidney disease, with reduced risk of hyperkalemia. SUMMARY Ongoing research continues to demonstrate novel roles of the vascular and adipocyte mineralocorticoid receptor function, which may explain the beneficial metabolic and vascular benefits of mineralocorticoid receptor antagonists.
Collapse
|
24
|
Abstract
The impaired capacity of the kidney to excrete sodium plays an essential role in the development of hypertension. Adrenal corticosteroids control renal handling of sodium by regulating tubular sodium reabsorption in the distal nephron where both mineralocorticoid receptors (MR) and glucocorticoid receptors are expressed. In addition, cell type- and segment-specific expression of 11β-HSD2 and sodium transporters such as Na-Cl cotransporter (NCC), epithelial sodium channel (ENaC), and pendrin/Na(+)-driven Cl(-)/HCO3 (-) exchanger (NDCBE) builds a distinctive model of sodium transport in the aldosterone-sensitive distal nephron. Aberrant MR activation in the distal nephron triggers salt-sensitive hypertension and hypokalemia through inappropriate sodium reabsorption and potassium secretion. However, MR activity is not necessarily modulated by the ligand alone. Recently, several lines of evidence revealed alternative mechanisms that regulate the activity of MR in a ligand-independent manner or through ligand binding modulation. This review summarizes the disorders related to MR activation in individual tubular cells and highlights the renal mechanism of salt-sensitive hypertension and new approaches for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Nobuhiro Ayuzawa
- Department of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | | |
Collapse
|
25
|
Pollow DP, Romero-Aleshire MJ, Sanchez JN, Konhilas JP, Brooks HL. ANG II-induced hypertension in the VCD mouse model of menopause is prevented by estrogen replacement during perimenopause. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1546-52. [PMID: 26491098 DOI: 10.1152/ajpregu.00170.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
Premenopausal females are resistant to the development of hypertension, and this protection is lost after the onset of menopause, resulting in a sharp increase in disease onset and severity. However, it is unknown how a fluctuating ovarian hormone environment during the transition from perimenopause to menopause impacts the onset of hypertension, and whether interventions during perimenopause prevent disease onset after menopause. A gradual transition to menopause was induced by repeated daily injections of 4-vinylcyclohexene diepoxide (VCD). ANG II (800 ng·kg(-1)·min(-1)) was infused into perimenopausal and menopausal female mice for 14 days. A separate cohort of mice received 17β-estradiol replacement during perimenopause. ANG II infusion produced significantly higher mean arterial pressure (MAP) in menopausal vs. cycling females, and 17β-estradiol replacement prevented this increase. In contrast, MAP was not significantly different when ANG II was infused into perimenopausal and cycling females, suggesting that female resistance to ANG II-induced hypertension is intact during perimenopause. ANG II infusion caused a significant glomerular hypertrophy, and hypertrophy was not impacted by hormonal status. Expression levels of aquaporin-2 (AQP2), a collecting duct protein, have been suggested to reflect blood pressure. AQP2 protein expression was significantly downregulated in the renal cortex of the ANG II-infused menopause group, where blood pressure was increased. AQP2 expression levels were restored to control levels with 17β-estradiol replacement. This study indicates that the changing hormonal environment in the VCD model of menopause impacts the severity of ANG II-induced hypertension. These data highlight the utility of the ovary-intact VCD model of menopause as a clinically relevant model to investigate the physiological mechanisms of hypertension that occur in women during the transition into menopause.
Collapse
Affiliation(s)
- Dennis P Pollow
- Department of Physiology, University of Arizona, Tucson, Arizona; and
| | | | - Jessica N Sanchez
- Department of Physiology, University of Arizona, Tucson, Arizona; and Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - John P Konhilas
- Department of Physiology, University of Arizona, Tucson, Arizona; and Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona; and Sarver Heart Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
26
|
|
27
|
Xiao J, Chen W, Lu Y, Zhang X, Fu C, Yan Z, Zhang Z, Ye Z. Crosstalk between peroxisome proliferator-activated receptor-γ and mineralcorticoid receptor in TNF-α activated renal tubular cell. Inflamm Res 2015; 64:603-14. [PMID: 26072064 DOI: 10.1007/s00011-015-0838-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/31/2015] [Accepted: 06/02/2015] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION In our previous study, we observed the crosstalk between peroxisome proliferator-activated receptor-γ (PPAR-γ) and angiotensin II in activated renal tubular cells. The present study is aimed to further explore the crosstalk between PPAR-γ and mineralocorticoid receptor (MR) in tumor necrosis factor (TNF)-α activated renal tubular cells. METHODS Human proximal renal tubular epithelial cells HK-2 were cultured with the pre-treatment of PPAR-γ agonist, pioglitazone (5 μM), MR antagonist, eplerenone (5 μM), or their combined treatment, followed by activation with TNF-α (20 ng/ml). In the parallel experiment, PPAR-γ inhibitor GW9662 (25 µM) was used to study the independence of PPAR-γ. Gene expression and protein synthesis of intercellular adhesion molecule-1 (ICAM-1), interleukin-6 (IL-6), MR and PPAR-γ were measured by RT-PCR, ELISA and Western blot, respectively; nuclear factor κB (NF-κB) nuclear translocation activity in the nucleus was examined by EMSA assay. RESULTS TNF-α effectively activated HK-2 cells by up-regulating gene expression and protein synthesis of ICAM-1, IL-6 and MR and down-regulating PPAR-γ in a dose-dependent manner. TNF-α also significantly induced NF-κB nuclear translocation in HK-2 cells. Dual treatment of pioglitazone and eplerenone demonstrated synergistic effect on reducing ICAM-1 and IL-6 expression and alleviating NF-κB activation when compared with their monotherapies in TNF-α activated renal tubular cells. PPAR-γ antagonist, GW9662, significantly attenuated protective effect on ICAM-1, IL-6 and PPAR-γ expression by pioglitazone, eplerenone and their combined treatment. CONCLUSIONS Our data suggest that pioglitazone, in a PPAR-γ-dependent manner, trans-represses MR signaling by suppressing NF-κB activation. MR antagonist also restored PPAR-γ expression. Dual treatment of pioglitazone and eplerenone present better efficacy in attenuating excessive inflammatory response in activated renal tubular cells under stimulation of TNF-α than single treatment.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kadoya H, Satoh M, Sasaki T, Taniguchi S, Takahashi M, Kashihara N. Excess aldosterone is a critical danger signal for inflammasome activation in the development of renal fibrosis in mice. FASEB J 2015; 29:3899-910. [DOI: 10.1096/fj.15-271734] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/26/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Hiroyuki Kadoya
- Department of Nephrology and HypertensionKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Minoru Satoh
- Department of Nephrology and HypertensionKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Tamaki Sasaki
- Department of Nephrology and HypertensionKawasaki Medical SchoolKurashikiOkayamaJapan
| | - Shun'ichiro Taniguchi
- Department of Molecular OncologyShinshu University Graduate School of MedicineMatsumotoNaganoJapan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular MedicineJichi Medical UniversityShimotsukeTochigiJapan
| | - Naoki Kashihara
- Department of Nephrology and HypertensionKawasaki Medical SchoolKurashikiOkayamaJapan
| |
Collapse
|
29
|
Takayanagi T, Kawai T, Forrester SJ, Obama T, Tsuji T, Fukuda Y, Elliott KJ, Tilley DG, Davisson RL, Park JY, Eguchi S. Role of epidermal growth factor receptor and endoplasmic reticulum stress in vascular remodeling induced by angiotensin II. Hypertension 2015; 65:1349-55. [PMID: 25916723 DOI: 10.1161/hypertensionaha.115.05344] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/01/2015] [Indexed: 12/18/2022]
Abstract
The mechanisms by which angiotensin II (AngII) elevates blood pressure and enhances end-organ damage seem to be distinct. However, the signal transduction cascade by which AngII specifically mediates vascular remodeling such as medial hypertrophy and perivascular fibrosis remains incomplete. We have previously shown that AngII-induced epidermal growth factor receptor (EGFR) transactivation is mediated by disintegrin and metalloproteinase domain 17 (ADAM17), and that this signaling is required for vascular smooth muscle cell hypertrophy but not for contractile signaling in response to AngII. Recent studies have implicated endoplasmic reticulum (ER) stress in hypertension. Interestingly, EGFR is capable of inducing ER stress. The aim of this study was to test the hypothesis that activation of EGFR and ER stress are critical components required for vascular remodeling but not hypertension induced by AngII. Mice were infused with AngII for 2 weeks with or without treatment of EGFR inhibitor, erlotinib, or ER chaperone, 4-phenylbutyrate. AngII infusion induced vascular medial hypertrophy in the heart, kidney and aorta, and perivascular fibrosis in heart and kidney, cardiac hypertrophy, and hypertension. Treatment with erlotinib as well as 4-phenylbutyrate attenuated vascular remodeling and cardiac hypertrophy but not hypertension. In addition, AngII infusion enhanced ADAM17 expression, EGFR activation, and ER/oxidative stress in the vasculature, which were diminished in both erlotinib-treated and 4-phenylbutyrate-treated mice. ADAM17 induction and EGFR activation by AngII in vascular cells were also prevented by inhibition of EGFR or ER stress. In conclusion, AngII induces vascular remodeling by EGFR activation and ER stress via a signaling mechanism involving ADAM17 induction independent of hypertension.
Collapse
Affiliation(s)
- Takehiko Takayanagi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Tatsuo Kawai
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Steven J Forrester
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Takashi Obama
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Toshiyuki Tsuji
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Yamato Fukuda
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Katherine J Elliott
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Douglas G Tilley
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Robin L Davisson
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Joon-Young Park
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Satoru Eguchi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.).
| |
Collapse
|
30
|
Li Y, Wang N, Chen C, He D, Yang J, Zeng C. Inhibitory effect of D3dopamine receptor on migration of vascular smooth muscle cells induced by synergistic effect of angiotensin II and aldosterone. Clin Exp Hypertens 2014; 37:288-93. [DOI: 10.3109/10641963.2014.960971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
31
|
Luther JM. Effects of aldosterone on insulin sensitivity and secretion. Steroids 2014; 91:54-60. [PMID: 25194457 PMCID: PMC4252580 DOI: 10.1016/j.steroids.2014.08.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/25/2014] [Accepted: 08/17/2014] [Indexed: 12/19/2022]
Abstract
Dr. Conn originally reported an increased risk of diabetes in patients with hyperaldosteronism in the 1950s, although the mechanism remains unclear. Aldosterone-induced hypokalemia was initially described to impair glucose tolerance by impairing insulin secretion. Correction of hypokalemia by potassium supplementation only partially restored insulin secretion and glucose tolerance, however. Aldosterone also impairs glucose-stimulated insulin secretion in isolated pancreatic islets via reactive oxygen species in a mineralocorticoid receptor-independent manner. Aldosterone-induced mineralocorticoid receptor activation also impairs insulin sensitivity in adipocytes and skeletal muscle. Aldosterone may produce insulin resistance secondarily by altering potassium, increasing inflammatory cytokines, and reducing beneficial adipokines such as adiponectin. Renin-angiotensin system antagonists reduce circulating aldosterone concentrations and also the risk of type 2 diabetes in clinical trials. These data suggest that primary and secondary hyperaldosteronism may contribute to worsening glucose tolerance by impairing insulin sensitivity or insulin secretion in humans. Future studies should define the effects of MR antagonists and aldosterone on insulin secretion and sensitivity in humans.
Collapse
Affiliation(s)
- James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
32
|
Meinel S, Gekle M, Grossmann C. Mineralocorticoid receptor signaling: crosstalk with membrane receptors and other modulators. Steroids 2014; 91:3-10. [PMID: 24928729 DOI: 10.1016/j.steroids.2014.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/16/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022]
Abstract
The mineralocorticoid receptor (MR) belongs to the steroid receptor superfamily. Classically, it acts as a ligand-bound transcription factor in epithelial tissues, where it regulates water and electrolyte homeostasis and controls blood pressure. Additionally, the MR has been shown to elicit pathophysiological effects including inflammation, fibrosis and remodeling processes in the cardiovascular system and the kidneys and MR antagonists have proven beneficial for patients with certain cardiovascular and renal disease. The underlying molecular mechanisms that mediate MR effects have not been fully elucidated but very likely rely on interactions with other signaling pathways in addition to genomic actions at hormone response elements. In this review we will focus on interactions of MR signaling with different membrane receptors, namely receptor tyrosine kinases and the angiotensin II receptor because of their potential relevance for disease. In addition, GPR30 is discussed as a new aldosterone receptor. To gain insights into the problem why the MR only seems to mediate pathophysiological effects in the presence of additional permissive factors we will also briefly discuss factors that lead to modulation of MR activity as well. Overall, MR signaling is part of an intricate network that still needs to be investigated further.
Collapse
Affiliation(s)
- S Meinel
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - M Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - C Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
33
|
Ma J, Zhang H, Guo H, Xu Y. Effects of low-dose spironolactone combined with irbesartan on cardiac hypertrophy induced by pressure overload in rats. Am J Transl Res 2014; 6:809-819. [PMID: 25628791 PMCID: PMC4297348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/15/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND The mineralocorticoid receptor (MR) blockade in the heart is an attractive therapeutic option for the treatment of heart failure. However, the use of MR antagonist is limited by an increased incidence of hyperkalemia owing to MR blockade in the kidney. This study was designed to evaluate and compare the effectiveness of a low, non-pressure-lowering dose of spironolactone (Sp) with that of a conventional blood pressure-lowering dose combined with irbesartan on pathological cardiac remodeling as well as serum potassium level in pressure-overload rats. METHODS The pressure-overloaded myocardial remodelling was produced by partial abdominal aortic constriction (PAAC) in rats. Four weeks after PAAC, animals were respectively treated with vehicle, irbesartan (15 mg/kg) alone, low-dose Sp (1 mg/kg) or conventional-dose of Sp (20 mg/kg) in combination with irbesartan for consecutive four weeks. RESULTS The result demonstrated that compared to irbesartan monotherapy, the combination of irbesartan and spironolactone both in low- and conventional-dose exhibited additional cardioprotection against PAAC-induced cardiac remodelling. Low-dose spironolactone was as effective in inhibiting cardiac hypertrophy, fibrosis and in improving diastolic function as high dose. Low-dose spironolactone did not lead to a rise in potassium serum levels, but high dose did. CONCLUSIONS This study suggests that combined low dose of spironolactone and irbesartan may be an effective and safety therapeutic strategy for cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jingtao Ma
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and ToxicologyHebei Province, China
- Department of Pharmacology, Hebei Medical UniversityShijiazhuang 050017, China
- The Fourth Hospital of Hebei Medical UniversityShijiazhuang 050051, China
| | - Hongxue Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and ToxicologyHebei Province, China
- Department of Pharmacology, Hebei Medical UniversityShijiazhuang 050017, China
| | - Huicai Guo
- Department of Toxicology, Hebei Medical UniversityShijiazhuang 050017, China
| | - Yanfang Xu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and ToxicologyHebei Province, China
- Department of Pharmacology, Hebei Medical UniversityShijiazhuang 050017, China
| |
Collapse
|
34
|
Barrett Mueller K, Lu Q, Mohammad NN, Luu V, McCurley A, Williams GH, Adler GK, Karas RH, Jaffe IZ. Estrogen receptor inhibits mineralocorticoid receptor transcriptional regulatory function. Endocrinology 2014; 155:4461-72. [PMID: 25051445 PMCID: PMC4197987 DOI: 10.1210/en.2014-1270] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The steroid hormone aldosterone (aldo) contributes to cardiovascular disease in animal models and in humans. Aldo activates the mineralocorticoid receptor (MR), a hormone-activated transcription factor, and indeed, pharmacological MR inhibition improves cardiovascular outcomes. Because the incidence of cardiovascular disease is lower in premenopausal women, we hypothesized that estrogen (E2) signaling through the estrogen receptor (ER) may protect the vasculature by inhibiting the detrimental effects of aldo signaling through the MR. We demonstrate that E2-activated ER inhibits MR-mediated gene transcription from the mouse mammary tumor virus reporter in human embryonic kidney-293 cells. In contrast, aldo-activated MR does not affect ER-mediated gene transcription. The ERα N terminus (amino acids 1-253) containing part of the DNA-binding domain is sufficient to inhibit MR genomic function, although point mutations reveal that DNA binding, ligand-independent activation, and rapid nongenomic ERα signaling are not required for this effect. Furthermore, ERα and MR are part of a complex in cell lysates, with amino acids 1-233 of the ERα N terminus being sufficient to complex with the MR. Overall, the ability of ERα to inhibit MR-mediated gene transcription correlates with the ability of ERα segments to both localize to the nucleus and complex with the MR. In cultured vascular endothelial cells expressing ERα, E2 inhibits aldo induction of the vascular MR target gene intercellular adhesion molecule-1 (ICAM-1). ICAM-1 induction by endothelial MR is known to promote vascular inflammation that could contribute to the mechanism of aldo-induced atherosclerosis. E2 also inhibits aldo induction of ICAM-1 protein and prevents aldo-enhanced leukocyte adhesion to endothelial cells. These studies support a new model in which E2-activated ER in endothelial cells forms a complex with MR in the nucleus to modulate MR regulation of the proinflammatory gene ICAM-1. Estrogen inhibition of MR regulation of genes that contribute to cardiovascular disease may be a new mechanism by which premenopausal women are protected from cardiovascular disease.
Collapse
Affiliation(s)
- Katelee Barrett Mueller
- Molecular Cardiology Research Institute (K.B.M., Q.L., N.N.M., V.L., A.M., R.H.K., I.Z.J.), Tufts Medical Center, and Sackler School of Biomedical Graduate Studies (K.B.M., R.H.K., I.Z.J.), Tufts University School of Medicine, Boston, Massachusetts 02111; and Division of Endocrinology, Diabetes, and Hypertension (G.H.W., G.K.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Faresse N. Post-translational modifications of the mineralocorticoid receptor: How to dress the receptor according to the circumstances? J Steroid Biochem Mol Biol 2014; 143:334-42. [PMID: 24820770 DOI: 10.1016/j.jsbmb.2014.04.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/07/2014] [Accepted: 04/28/2014] [Indexed: 12/22/2022]
Abstract
Aldosterone or glucocorticoid stimulation of the mineralocorticoid receptor (MR) is involved in numerous physiological responses, including ions and water homeostasis, blood pressure control and metabolism. The understanding of MR signaling regulation in the patho/physiological context took a new direction the last few years with a focus on the post-translational modifications of MR. Depending on its environment, cellular expression, activity or its binding partners, the MR is submitted to several post-translational modifications such as phosphorylation, ubiquitylation, sumoylation and acetylation that regulate its localization, activity and/or stability. A complex interplay between all these modifications allows a fine tuning of MR signaling depending on the physiological context. This review reports recent knowledge about post-translational modifications of MR and describes the enzymes and the molecular mechanisms involved.
Collapse
Affiliation(s)
- Nourdine Faresse
- University of Zurich, Institute of Anatomy, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
36
|
Felizardo RJF, Silva MBD, Aguiar CF, Câmara NOS. Obesity in kidney disease: A heavyweight opponent. World J Nephrol 2014; 3:50-63. [PMID: 25332896 PMCID: PMC4202492 DOI: 10.5527/wjn.v3.i3.50] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/18/2014] [Accepted: 06/11/2014] [Indexed: 02/05/2023] Open
Abstract
Obesity is an important worldwide challenge that must be faced in most developed and developing countries because of unhealthy nutritional habits. The consequences of obesity and being overweight are observed in different organs, but the kidney is one of the most affected. Excess adipose tissue causes hemodynamic alterations in the kidney that can result in renal disease. However, obesity is also commonly associated with other comorbidities such as chronic inflammation, hypertension and diabetes. This association of several aggravating factors is still a matter of concern in clinical and basic research because the pathophysiologic mechanisms surrounding chronic kidney disease development in obese patients remain unclear. This review will discuss the consequences of obesity in the context of renal injury.
Collapse
|
37
|
Habibi J, Hayden MR, Ferrario CM, Sowers JR, Whaley-Connell AT. Salt Loading Promotes Kidney Injury via Fibrosis in Young Female Ren2 Rats. Cardiorenal Med 2014; 4:43-52. [PMID: 24847333 DOI: 10.1159/000360866] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 02/24/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/AIMS It is increasingly recognized that there is sexual dimorphism in kidney disease progression; however, this disparity is lost in the presence of diabetes where women progress at a similar rate to men. The renin-angiotensin-aldosterone system (RAAS) is known to regulate diabetes-induced kidney injury, and recent literature would suggest that gender differences exist in RAAS-dependent responses in the kidney. In this regard, these gender differences may be overcome by excessive salt intake. Thereby, we hypothesized that salt would promote proteinuria in transgenic female rats under conditions of excess tissue angiotensin (Ang) II and circulating aldosterone. MATERIALS AND METHODS We utilized young female transgenic (mRen2)27 (Ren2) rats and Sprague-Dawley (SD) littermates and fed a high-salt diet (4%) over 3 weeks. RESULTS Compared to SD and Ren2 controls, female Ren2 rats fed a high-salt diet displayed increases in proteinuria, periarterial and interstitial fibrosis as well as ultrastructural evidence of basement membrane thickening, loss of mitochondrial elongation, mitochondrial fragmentation and attenuation of basilar canalicular infoldings. These findings occurred temporally with increases in transforming growth factor-β but not indices of oxidant stress. CONCLUSIONS Our current data suggest that a diet high in salt promotes progressive kidney injury as measured by proteinuria and fibrosis associated with transforming growth factor-β under conditions of excess tissue Ang II and circulating aldosterone.
Collapse
Affiliation(s)
- Javad Habibi
- Diabetes and Cardiovascular Center, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Endocrinology and Metabolism, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Mo., USA
| | - Melvin R Hayden
- Diabetes and Cardiovascular Center, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Endocrinology and Metabolism, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA
| | - Carlos M Ferrario
- Division of Wake Forest University School of Medicine, Winston-Salem, N.C., USA
| | - James R Sowers
- Diabetes and Cardiovascular Center, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Department of Medical Pharmacology and Physiology, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Endocrinology and Metabolism, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Mo., USA
| | - Adam T Whaley-Connell
- Diabetes and Cardiovascular Center, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Endocrinology and Metabolism, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Nephrology and Hypertension, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA ; Division of Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Mo., USA
| |
Collapse
|
38
|
Imanishi M, Tomita S, Ishizawa K, Kihira Y, Ueno M, Izawa-Ishizawa Y, Ikeda Y, Yamano N, Tsuchiya K, Tamaki T. Smooth muscle cell-specific Hif-1α deficiency suppresses angiotensin II-induced vascular remodelling in mice. Cardiovasc Res 2014; 102:460-8. [DOI: 10.1093/cvr/cvu061] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
39
|
Fujita T. Mechanism of salt-sensitive hypertension: focus on adrenal and sympathetic nervous systems. J Am Soc Nephrol 2014; 25:1148-55. [PMID: 24578129 DOI: 10.1681/asn.2013121258] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
A central role for the kidney among the systems contributing to BP regulation and the development of hypertension has been proposed. Both the aldosterone/mineralocorticoid receptor pathway and the renal sympathetic nervous system have important roles in the regulation of renal excretory function and BP control, but the mechanisms underlying these processes have remained unclear. However, recent studies revealed the activation of two pathways in salt-sensitive hypertension. Notably, Rac1, a member of the Rho-family of small GTP binding proteins, was identified as a novel ligand-independent modulator of mineralocorticoid receptor activity. Furthermore, these studies point to crucial roles for the Rac1-mineralocorticoid receptor-NCC/ENaC and the renal β-adrenergic stimulant-glucocorticoid receptor-WNK4-NCC pathways in certain rodent models of salt-sensitive hypertension. The nuclear mineralocorticoid and glucocorticoid receptors may contribute to impaired renal excretory function and the resulting salt-sensitive hypertension by increasing sodium reabsorption at different tubular segments. This review provides an in-depth discussion of the evidence supporting these conclusions and considers the significance with regard to treating salt-sensitive hypertension and salt-induced cardiorenal injury.
Collapse
Affiliation(s)
- Toshiro Fujita
- Department of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan; and CREST, Tokyo, Japan
| |
Collapse
|
40
|
Hunter RW, Ivy JR, Bailey MA. Glucocorticoids and renal Na+ transport: implications for hypertension and salt sensitivity. J Physiol 2014; 592:1731-44. [PMID: 24535442 PMCID: PMC4001748 DOI: 10.1113/jphysiol.2013.267609] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The clinical manifestations of glucocorticoid excess include central obesity, hyperglycaemia, dyslipidaemia, electrolyte abnormalities and hypertension. A century on from Cushing's original case study, these cardinal features are prevalent in industrialized nations. Hypertension is the major modifiable risk factor for cardiovascular and renal disease and reflects underlying abnormalities of Na+ homeostasis. Aldosterone is a master regulator of renal Na+ transport but here we argue that glucocorticoids are also influential, particularly during moderate excess. The hypothalamic–pituitary–adrenal axis can affect renal Na+ homeostasis on multiple levels, systemically by increasing mineralocorticoid synthesis and locally by actions on both the mineralocorticoid and glucocorticoid receptors, both of which are expressed in the kidney. The kidney also expresses both of the 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes. The intrarenal generation of active glucocorticoid by 11βHSD1 stimulates Na+ reabsorption; failure to downregulate the enzyme during adaption to high dietary salt causes salt-sensitive hypertension. The deactivation of glucocorticoid by 11βHSD2 underpins the regulatory dominance for Na+ transport of mineralocorticoids and defines the ‘aldosterone-sensitive distal nephron’. In summary, glucocorticoids can stimulate renal transport processes conventionally attributed to the renin–angiotensin–aldosterone system. Importantly, Na+ and volume homeostasis do not exert negative feedback on the hypothalamic–pituitary–adrenal axis. These actions are therefore clinically relevant and may contribute to the pathogenesis of hypertension in conditions associated with elevated glucocorticoid levels, such as the metabolic syndrome and chronic stress.
Collapse
Affiliation(s)
- Robert W Hunter
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | | | |
Collapse
|
41
|
Hu Q, Yin L, Hartmann RW. Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal Diseases. J Med Chem 2014; 57:5011-22. [DOI: 10.1021/jm401430e] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Qingzhong Hu
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| | - Lina Yin
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal
Chemistry, Saarland University and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2.3, D-66123 Saarbrücken, Germany
| |
Collapse
|
42
|
McGraw AP, McCurley A, Preston IR, Jaffe IZ. Mineralocorticoid receptors in vascular disease: connecting molecular pathways to clinical implications. Curr Atheroscler Rep 2014; 15:340. [PMID: 23719923 DOI: 10.1007/s11883-013-0340-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The mineralocorticoid receptor (MR), a steroid-hormone-activated transcription factor, plays a substantial role in cardiovascular diseases. MR antagonists (MRAs) have long been appreciated as effective treatments for heart failure and hypertension; however, recent research suggests that additional patient populations may also benefit from MRA therapy. Experimental evidence demonstrates that in addition to its classic role in the regulating sodium handling in the kidney, functional MR is expressed in the blood vessels and contributes to hypertension, vascular inflammation and remodeling, and atherogenesis. MR activation drives pathological phenotypes in smooth muscle cells, endothelial cells, and inflammatory cells, whereas MRAs inhibit these effects. Collectively, these studies demonstrate a new role for extrarenal MR in cardiovascular disease. This review summarizes these new lines of evidence and how they contribute to the mechanisms of atherosclerosis, pulmonary and systemic hypertension, and vein graft failure, and describes new patient populations that may benefit from MRA therapy.
Collapse
Affiliation(s)
- Adam P McGraw
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, USA.
| | | | | | | |
Collapse
|
43
|
Uchiyama M, Maejima S, Wong MKS, Preyavichyapugdee N, Wanichanon C, Hyodo S, Takei Y, Matuda K. Changes in plasma angiotensin II, aldosterone, arginine vasotocin, corticosterone, and electrolyte concentrations during acclimation to dry condition and seawater in the crab-eating frog. Gen Comp Endocrinol 2014; 195:40-6. [PMID: 24184110 DOI: 10.1016/j.ygcen.2013.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
The crab-eating frog Fejervarya cancrivora inhabits mangrove swamps and marshes in Southeast Asia. In the present study, circulating angiotensin II (Ang II), aldosterone (Aldo), arginine vasotocin (AVT), and corticosterone (Cort) concentrations as well as various blood parameters were studied under osmotically stressful conditions. Following acclimation to hyperosmotic seawater and dry condition for 5days, body weight was significantly decreased. Under both conditions, plasma Na(+), Cl(-), and urea concentrations, hematocrit values (Ht; blood volume indicator), and osmolality were significantly increased. Dehydration associated with hypovolemic and hyperosmotic states of body fluids was induced during acclimation to hyperosmotic seawater and dry condition in the crab-eating frogs. Ang II, Aldo, AVT, and Cort were maintained within relatively narrow concentration ranges in the control frogs; however, in frogs under dry and hyperosmotic seawater conditions, large variations were observed among individuals in each group. Mean plasma Ang II and Aldo concentrations significantly increased in hyperosmotic seawater-acclimated and desiccated frogs. Although mean plasma AVT concentrations in dehydrated frogs of both the groups were approximately 2.0-3.5 times higher than those in the control frogs, the differences were not significant because of the variation. There was a significant correlation between plasma osmolality and AVT as well as Ang II but not Aldo. A significant correlation was also observed between Ht and AVT as well as Ang II. Plasma Ang II was significantly correlated with plasma Aldo. These results indicate that the crab-eating frogs may exhibit similar physiological responses to both seawater-acclimated and dry conditions. It appears that under dehydrated conditions, osmoregulatory mechanisms participate in stabilization of the situation. The renin-angiotensin system may have pivotal roles in body fluid regulation under volemic and osmotic stress in the Fejervarya species with unique osmoregulation.
Collapse
Affiliation(s)
- Minoru Uchiyama
- Department of Life and Environmental Science, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan.
| | - Sho Maejima
- Department of Life and Environmental Science, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Marty K S Wong
- Department of Marine Biosciences, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-15 Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| | - Narin Preyavichyapugdee
- Faculty of Animal Sciences and Agricultural Technology, Silpakorn University, Petchaburi IT Campus, Petchaburi 76120, Thailand
| | - Chaitip Wanichanon
- Department of Anatomy, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Susumu Hyodo
- Department of Marine Biosciences, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-15 Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| | - Yoshio Takei
- Department of Marine Biosciences, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-15 Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| | - Kouhei Matuda
- Department of Life and Environmental Science, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| |
Collapse
|
44
|
Nugrahaningsih DAA, Emoto N, Vignon-Zellweger N, Purnomo E, Yagi K, Nakayama K, Doi M, Okamura H, Hirata KI. Chronic hyperaldosteronism in cryptochrome-null mice induces high-salt- and blood pressure-independent kidney damage in mice. Hypertens Res 2013; 37:202-9. [PMID: 24108235 DOI: 10.1038/hr.2013.143] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 11/09/2022]
Abstract
Although aldosterone has an essential role in controlling electrolyte and body fluid homeostasis, aldosterone also exerts certain pathological effects on the kidney. Several previous studies have attempted to examine these deleterious effects. However, the majority of these studies were performed using various injury models, including high-salt treatment and/or mineralocorticoid administration, by which the kidney changes observed were not only due to aldosterone but also due to prior injury caused by salt and hypertension. In the present study, we investigated aldosterone's pathological effect on the kidney using a mouse model with a high level of endogenous aldosterone. We used cryptochrome-null (Cry 1, 2 DKO) mice characterized by high aldosterone levels and low plasma renin activity and observed that even under normal salt exposure conditions, these mice showed increased albumin excretion and kidney tubular injury, decreased nephrin expression and increased reactive oxygen species production in the absence of hypertension. Exposure to high salt levels exacerbated the kidney damage observed in these mice. Moreover, we noted that decreasing blood pressure without blocking aldosterone action did not provide beneficial effects to the kidney in high-salt-treated Cry 1, 2 DKO mice. Thus, our findings support the hypothesis that aldosterone has deleterious effects on the kidney independent of high-salt exposure and high blood pressure.
Collapse
Affiliation(s)
- Dwi Aris Agung Nugrahaningsih
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriaki Emoto
- 1] Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan [2] Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | | | - Eko Purnomo
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keiko Yagi
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Kazuhiko Nakayama
- 1] Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan [2] Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Masao Doi
- Department of System Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hitoshi Okamura
- Department of System Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
45
|
Lambers Heerspink HJ, de Zeeuw D. Novel drugs and intervention strategies for the treatment of chronic kidney disease. Br J Clin Pharmacol 2013; 76:536-50. [PMID: 23802504 PMCID: PMC3791977 DOI: 10.1111/bcp.12195] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/20/2013] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem. The disease is most often progressive of nature with a high impact on patients and society. It is increasingly recognized that CKD can be detected in the early stages and should be managed as early as possible. Treatment of the cause, but in particular control of the main risk markers, such as high blood pressure, glucose and albuminuria, has been instrumental in delaying the progression to end-stage renal disease (ESRD). However, despite the state of the art therapy, the absolute risk of renal and cardiovascular morbidity and mortality in CKD patients remains devastatingly high. Novel drugs are therefore highly desirable to halt effectively the progressive renal (and cardiovascular) function loss. Recently, several novel strategies have been tested targeting traditional risk factors such as blood pressure (combination therapy of angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) and novel mineralocorticoid receptor antagonists) as well as dyslipidaemia (statins) with surprising results. In addition, drug targets specifically related to the kidney, such as vitamin D, uric acid, erythropoietin and phosphate, have been the subject of clinical trials, in some instances with unexpected results. Finally, novel targets including endothelin receptors and inflammatory pathways are increasingly explored as potential avenues to improve renal and cardiovascular protection, albeit that the drugs tested have not been unequivocally successful. In this article we review novel drugs or intervention strategies for the management of CKD, we try to provide explanations for the failure of some promising drugs and hypothesize on the potential success of new strategies.
Collapse
Affiliation(s)
- Hiddo Jan Lambers Heerspink
- Department of Clinical Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | |
Collapse
|
46
|
Chapman KE, Coutinho AE, Zhang Z, Kipari T, Savill JS, Seckl JR. Changing glucocorticoid action: 11β-hydroxysteroid dehydrogenase type 1 in acute and chronic inflammation. J Steroid Biochem Mol Biol 2013; 137:82-92. [PMID: 23435016 PMCID: PMC3925798 DOI: 10.1016/j.jsbmb.2013.02.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/22/2013] [Accepted: 02/04/2013] [Indexed: 12/18/2022]
Abstract
Since the discovery of cortisone in the 1940s and its early success in treatment of rheumatoid arthritis, glucocorticoids have remained the mainstay of anti-inflammatory therapies. However, cortisone itself is intrinsically inert. To be effective, it requires conversion to cortisol, the active glucocorticoid, by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Despite the identification of 11β-HSD in liver in 1953 (which we now know to be 11β-HSD1), its physiological role has been little explored until recently. Over the past decade, however, it has become apparent that 11β-HSD1 plays an important role in shaping endogenous glucocorticoid action. Acute inflammation is more severe with 11β-HSD1-deficiency or inhibition, yet in some inflammatory settings such as obesity or diabetes, 11β-HSD1-deficiency/inhibition is beneficial, reducing inflammation. Current evidence suggests both beneficial and detrimental effects may result from 11β-HSD1 inhibition in chronic inflammatory disease. Here we review recent evidence pertaining to the role of 11β-HSD1 in inflammation. This article is part of a Special Issue entitled 'CSR 2013'.
Collapse
Affiliation(s)
- Karen E Chapman
- University/BHF Centre for Cardiovascular Sciences, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Escoubet B, Couffignal C, Laisy JP, Mangin L, Chillon S, Laouénan C, Serfaty JM, Jeunemaitre X, Mentré F, Zennaro MC. Cardiovascular Effects of Aldosterone. ACTA ACUST UNITED AC 2013; 6:381-90. [DOI: 10.1161/circgenetics.113.000115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background—
High plasma aldosterone has deleterious cardiovascular effects that are independent of blood pressure, but the role of the mineralocorticoid receptor remains unclear. Renal pseudohypoaldosteronism type 1 is a rare autosomal-dominant disease caused by
NR3C2
loss-of-function mutations, which is characterized by renal salt loss and compensatory high renin and aldo secretion. We aimed to assess the cardiovascular outcomes in adults carrying NR3C2 mutations.
Methods and Results—
In this case-control study, 39
NR3C2
mutation carriers were compared with sex- and age-paired noncarriers. Patients underwent cardiac and vascular ultrasound, cardiac MRI with gadolinium injection, measurement of pulse wave velocity, extracellular water, 24-hour ambulatory blood pressure, and autonomous nervous system activity. Mutation carriers showed increased aldo and renin plasma levels (4.5- and 1.6-fold, respectively;
P
<0.0001), together with increased salt appetite (1.8-fold;
P
=0.002), with normal extracellular water and blood pressure, and no autonomous nervous system activation. Cardiac and vascular parameters were not significantly different between mutation carriers and noncarriers (no left ventricular remodeling or fibrosis, normal left ventricular systolic function, and aorta stiffness). Tissue Doppler showed better diastolic left ventricular function in mutation carriers (e′,
P
=0.001; E/e′,
P
=0.003). Mutation carriers had significantly more frequent history of slow body weight recovery at birth, symptomatic hypotension, and miscarriage in women.
Conclusions—
Despite life-long increase in plasma aldosterone and renin levels, no adverse cardiovascular outcome occurred in pseudohypoaldosteronism type 1, but rather an improved diastolic left ventricular function. This suggests that the cardiovascular consequences of aldosterone excess require full mineralocorticoid receptor signaling.
Clinical Trial Registration—
http://www.clinicaltrials.gov
; unique identifier: NCT00646828.
Collapse
Affiliation(s)
- Brigitte Escoubet
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Camille Couffignal
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Jean-Pierre Laisy
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Laurence Mangin
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Sylvie Chillon
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Cédric Laouénan
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Jean-Michel Serfaty
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Xavier Jeunemaitre
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - France Mentré
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| | - Maria-Christina Zennaro
- From the Assistance Publique-Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France (B.E., C.C., J.-P.L., L.M., S.C., C.L., J.-M.S., F.M.); Université Paris Diderot, Sorbonne Paris Cité (B.E., C.C., J.-P.L., L.M., C.L., J.-M.S., F.M.); Inserm, UMR 872, Centre de Recherche des Cordeliers CRC (B.E.); Inserm UMR-S 738 (C.C., C.L., F.M.); Laboratoire Matière et Systèmes Complexes, UMR-7057 CNRS (L.M.); Inserm, UMR 970 Paris Cardiovascular Research Centre (X.J., M.-C.Z.); Assistance Publique
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The aldosterone/mineralocorticoid receptor system plays an important role in the long-term blood pressure control through Na homeostasis. Its overactivation has been implicated in salt-sensitive hypertension. Excessive salt intake augments the function of mineralocorticoid receptor, despite lowering circulating aldosterone levels, but the mechanism had long been elusive. Recently, Rac1, a member of Rho family small GTP-binding proteins, has emerged as a novel ligand-independent modulator of mineralocorticoid receptor activity. In this review, the roles of Rac1 in the pathogenesis of salt-sensitive hypertension and kidney injury have been summarized. RECENT FINDINGS Genetic engineering studies have highlighted the new aspects of Rac1 and its regulators in salt-sensitive hypertension and cardiac and renal disease. New evidence shows the essential roles of Rac1 in salt-evoked paradoxical mineralocorticoid receptor activation observed in salt-sensitive models and in renal tubular Na reabsorption through reduced nicotinamide-adenine dinucleotide phosphate oxidase-mediated oxidative stress or direct regulation of Na transporters. SUMMARY The emerging concept of 'ligand-independent aberrant mineralocorticoid receptor activation by Rac1' in the pathogenesis of salt-sensitive hypertension and kidney injury has been reviewed. Rac inhibition, in addition to mineralocorticoid receptor blockade and salt restriction, would be a new promising strategy for the treatment of salt-sensitive hypertension.
Collapse
|
49
|
Abstract
The steroid hormone aldosterone regulates sodium and potassium homeostasis. Aldosterone and activation of the mineralocorticoid receptor also causes inflammation and fibrosis of the heart, fibrosis and remodelling of blood vessels and tubulointerstitial fibrosis and glomerular injury in the kidney. Aldosterone and mineralocorticoid-receptor activation initiate an inflammatory response by increasing the generation of reactive oxygen species by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. High salt intake potentiates these effects, in part by activating the Rho family member Rac1, a regulatory subunit of reduced NADPH oxidase that activates the mineralocorticoid receptor. Studies in mice in which the mineralocorticoid receptor has been deleted from specific cell types suggest a key role for macrophages in promoting inflammation and fibrosis. Aldosterone can exert mineralocorticoid-receptor-independent effects via the angiotensin II receptor and via G-protein-coupled receptor 30. Mineralocorticoid-receptor antagonists are associated with decreased mortality in patients with heart disease and show promise in patients with kidney injury, but can elevate serum potassium concentration. Studies in rodents genetically deficient in aldosterone synthase or treated with a pharmacological aldosterone-synthase inhibitor are providing insight into the relative contribution of aldosterone compared with the contribution of mineralocorticoid-receptor activation in inflammation, fibrosis, and injury. Aldosterone-synthase inhibitors are under development in humans.
Collapse
|
50
|
Smooth muscle cell mineralocorticoid receptors: role in vascular function and contribution to cardiovascular disease. Pflugers Arch 2013; 465:1661-70. [PMID: 23636772 DOI: 10.1007/s00424-013-1282-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 04/11/2013] [Indexed: 02/07/2023]
Abstract
The mineralocorticoid receptor (MR), a member of the steroid receptor family, regulates blood pressure by mediating the effects of the hormone aldosterone on renal sodium handling. In recent years, it has become clear that MR is expressed in vascular smooth muscle cells (SMCs), and interest has grown in understanding the direct role of SMC MR in regulating vascular function. This interest stems from multiple clinical studies where MR inhibitor treatment reduced the incidence of cardiovascular events and mortality. This review summarizes the most recent advances in our understanding of SMC MR in regulating normal vascular function and in promoting vascular disease. Many new studies suggest a role for SMC MR activation in stimulating vascular contraction and contributing to vessel inflammation, fibrosis, and remodeling. These detrimental vascular effects of MR activation appear to be independent of changes in blood pressure and are synergistic with the presence of endothelial dysfunction or damage. Thus, in humans with underlying cardiovascular disease or cardiovascular risk factors, SMC MR activation may promote hypertension, atherosclerosis, and vascular aging. Further exploration of the molecular mechanisms for the effects of SMC MR activation has the potential to identify novel therapeutic targets to prevent or treat common cardiovascular disorders.
Collapse
|